TO: Randy Smith, Vice Provost for Academic Programs FROM: Anika Anthony, Associate Dean of Academic Affairs, Graduate School DATE: January 21,2021 RE: Proposal for a new Master of Science in ad Microbial Pathogenesis, College of

The College of Medicine is proposing a new Master of Science in Immunology and Microbial Pathogenesis.

The proposal was received by the Graduate School on November 6, 2020. The combined GS/CAA subcommittee first reviewed the proposal on November 17, 2020 and requested revisions. Revisions were received on December 2, 2020. GS/CAA conducted a second review of the proposal and recommended it for approval by the Graduate Council on December 9. The proposal was approved by the Graduate Council on January 11, 2021.

Mark E. Drew Assistant Professor Director of Graduate Education Department of Microbial Infection and Immunity OSU College of Medicine

December 2, 2020

GS/CAA curricular subcommittee Ohio State University

Re: Master of Science (MS) in Immunology and Microbial Pathogenesis

Dear Members of the Subcommittee,

Thank you for your kind and positive review of our proposal to offer a new Master of Science in Immunology and Microbial Pathogenesis program at OSU. Your comments and suggestions were extremely helpful in refining the proposal. Specifically, in response to your comments, we have made the following changes:

1. “Clarity on which courses are new vs. those that are already existing.”

Page 34 of the proposal now includes brief descriptions of the three courses under development to support this program in our expectation of its approval. The complete syllabi are being developed by four key program faculty (Drew, Deora, Root, Novias) with planned entry into the new course portal on or before January 1, 2021.

2. “Provide an advising sheet that students will use to track their progress through the program.”

This was an especially helpful suggestion and is included as an attachment along with this letter and our revised proposal.

3. “Please provide additional details on how the program will measure direct measures of student learning and program quality.”

As recommended, Appendix F “Program Goals and Plan for Program Assessment” has been expanded to include details on this point. Ultimately, the assessment plan is designed to maximize the success of both current and future students in the program as well as to ensure that the program is ultimately meeting student expectations as measured by their post-graduation, career success outcomes. A new column has been added to Table 2 to help clarify the cases in which these assessments will directly impact this overall goal.

4. “Please provide Appendix I, letters of concurrence and support from the Department of Microbiology, the Department of Veterinary Biosciences, and the Biomedical Sciences Graduate Program.”

These letters are now included into Appendix I of the revised proposal.

Again, thank you for you review of this proposal and we look forward to your support during the application and review process at both the University and State levels.

Sincerely,

Mark E. Drew, Ph.D. Department of Microbial Infection and Immunity The Ohio State University College of Medicine

Full name of Proposed Degree Program: MS in Immunology and Microbial Pathogenesis Mode of Instruction: Mixed mode of delivery. On campus (Columbus) and hybrid asynchronous Offered by: The Ohio State University College of Medicine, Department of Microbial Infection and Immunity

MS in Immunology and Microbial Pathogenesis Full Proposal

Table of Contents

BASIC CHARACTERISTICS OF THE EDUCATIONAL PROGRAM ...... 3 Brief description of the disciplinary purpose and significance of proposed degree ...... 3 Definition of the focus of the program ...... 3 Rationale for degree name ...... 3 Duration of the program ...... 3 Admission timing ...... 4 Primary target audience for the program and admission requirements ...... 4 Special efforts to enroll and retain underrepresented groups ...... 4 INSTITUTIONAL PLANNING FOR THE PROGRAM ...... 5 Physical facilities, equipment and staff needed to support the program ...... 5 Projected additional costs associated with the program and evidence of institutional commitment and capacity to meet these costs...... 5 Availability and adequacy of the faculty and facilities for the new degree program...... 5 Evidence that a market exists for a new program ...... 5 STATEWIDE ALTERNATIVES ...... 6 GROWTH OF THE PROGRAM ...... 7 Current and future demand ...... 7 Program assessment ...... 7 CURRICULUM AND INSTRUCTIONAL DESIGN ...... 7 Curricular content ...... 7 Mixed mode of delivery ...... 8 Table 1 – Master of Science in Immunology and Microbial Pathogenesis required courses ...... 8 Description of a required integrated, or culminating learning, experience ...... 9 Appendices ...... 10 Appendix A: Proposed Curriculum for the Master of Science in Immunology and Microbial Pathogenesis Degree Program ...... 10 Appendix B: Selected Relevant Elective Course offerings at Ohio State University ...... 11 Appendix C: Physical Research Needs of the Program ...... 12 Appendix D: Five-year Projected Master’s Program Budget ...... 17 Appendix E: Statewide Alternatives ...... 18 Appendix F: Program Goals and Plan for Program Assessment ...... 20 Table 2 – Program Assessment ...... 20 Appendix G: Course Descriptions/Syllabi ...... 21 2 MS in Immunology and Microbial Pathogenesis Full Proposal

Core courses currently approved and offered: ...... 21 Descriptions of courses under design and to be approved: ...... 34 Appendix H: Program Faculty ...... 36 Appendix I: Letters of Concurrence and Support ...... 37

BASIC CHARACTERISTICS OF THE EDUCATIONAL PROGRAM

Brief description of the disciplinary purpose and significance of proposed degree

We are seeking approval for a graduate program to award students a Master of Science degree in Immunology and Microbial Pathogenesis. The program’s primary goal is to supply highly-skilled individuals to the biomedical workforce in Ohio. Graduates will support ongoing academic and private sector research needs within the state and serve as a talented labor pool to attract and meet future growth in these areas. The program will offer focused, formal education with focused, hands-on research training for people seeking careers in academic, pharmaceutic, biotech, agriculture, government, and fields while benefiting from Ohio State University’s environment of diversity, education, ethics, honesty, integrity, personal and professional growth, professionalism, and responsibility.

The program will be offered at the Columbus campus and will fill a geographical need for students who wish to pursue relevant scientific careers. Graduates of the program will gain a highly marketable skillset in the practicalities of working within a top-tier research environment along with a foundational knowledge of health- related human immunology and microbial pathogenesis. Graduates will be prepared to contribute to the State of Ohio’s efforts especially in this time of emerging pathogens and need for rapid development of diagnostics and therapeutic interventions. Similarly, the current revolution of cancer immunotherapy research and discovery predicts a need for talented labor in these fields as well.

Definition of the focus of the program

The focus of the program will be to educate and train students in the areas of Immunology and Microbial Pathogenesis with a particular focus on becoming proficient at working within a laboratory-based setting. The required curriculum will be a combination of didactic, journal club, seminar, and research-based coursework culminating in the successful defense of a master’s thesis. The curriculum will be consistent with the program’s mission to provide the practical knowledge necessary to be employed in a research-related environment and contribute to the health needs of the people of Ohio. In particular, students will have the option to concentrate their curriculum in one of the two focus areas by selection of advanced core courses and electives in immunology or microbial pathogenesis.

Rationale for degree name

The Master of Science degree in Immunology and Microbial Pathogenesis has been chosen for this program as it reflects that the student has, as a culminating experience, completed their thesis research and is prepared to carry out original research in Immunology and Microbial pathogenesis and their associated fields.

Duration of the program

Total Credit Hours: A minimum of 40 semester credit hours will be required to earn the Master of Science in Immunology and Microbial Pathogenesis. This minimum is higher than some statewide alternatives for an MS in related fields (similar however to U. of Akron and U. of Toledo, Appendix E) it is reflective of the hands-on, research component necessary for completion of a culminating thesis document. These credit hours consist of 3 MS in Immunology and Microbial Pathogenesis Full Proposal

30 hours core courses (including 15 from laboratory research practicum) and 10 hours of elective courses (see Table 1 and Appendix A).

Length of Time for Completion: The curriculum is designed to be completed in five terms initiating in Autumn of Year 1 (AuY1) and culminating with Spring of Year 2 (SpY2). Students can elect to begin early by enrolling in their research laboratory rotation course during the summer term prior to AuY1.

Admission timing

The program is expected to be implemented beginning Autumn 2021. It is anticipated that the program will admit one cohort of six students per year until a planned rolling average enrollment of 12 students is reached.

Primary target audience for the program and admission requirements

The program consists of five terms of 8-credit hours each, delivered on campus with a required research-based component. For these reasons we project the primary target audience to be students with proximity and availability to the Columbus campus during daytime hours, Monday-Friday. Although the program is formally full- time at 8-credit hours per term, it is conceivable that a student may have time to maintain part-time employment while pursing the degree.

Students accepted to the program would be expected to hold a Bachelor’s degree in the biological sciences and seeking to advance their knowledge and skills to increase their chances for employment and/or increase their earning potential in relevant research positions.

Recruitment and admissions are to be handled through the Office of Graduate Education and adhere to an application process with the following qualifications:

• A personal statement of why the applicant is applying to the program • An official transcript with proof of completed Bachelor's Degree (or higher) in any of the biological sciences or related areas, or a combination of related major along with successful completion of relevant prerequisite for the curriculum (see Table 1 for core-course requirements) • Three letters of recommendation. • All international applicants whose native language is not English will be required to take the Test of English as a Foreign Language (TOEFL) and have an official score report sent directly to the Associate Dean for Graduate Studies from Educational Testing Service. The recommended minimum TOEFL scores are 560 (written) or 220 (electronic) or 89 (internet based).

Evaluation of applicants for admission to the program will adhere to the principles of individualized holistic review. Therefore, GPA and test scores will be considered as but single metrics in the admissions process, with no score to be considered a sole criterion for admission into the program.

Special efforts to enroll and retain underrepresented groups

We plan to work with our collaborating institutions to facilitate recruitment and retention of minority students. Special efforts will be made to recruit and retain underrepresented groups in the Immunology and Microbial Pathogenesis Master of Science degree program. The Institution’s Office for Diversity and Cultural Affairs is committed to enhancing the recruitment, admission and retention of students from underrepresented groups. This office currently offers services including professional and personal guidance, summer research 4 MS in Immunology and Microbial Pathogenesis Full Proposal

opportunities, career development and pipeline programs, visiting student programs, interaction with other minority students, and networking and mentorship opportunities throughout the Institution’s affiliations.

We also plan to reduce costs through fee-waivers and tuition scholarships/offsets for URM students. The program will seek funding for these efforts through re-investment of program capital, College and University assets, and private-sector industry partners.

INSTITUTIONAL PLANNING FOR THE PROGRAM

Physical facilities, equipment and staff needed to support the program

The Immunology and Microbial Pathogenesis graduate program will be housed in The Ohio State University College of Medicine and implemented through the Department of Microbial Infection and Immunity. The department currently has 27 principal investigator-led laboratories which will serve the physical research needs of the program. Facilities include over 23,000 square feet of well-equipped laboratory space within the OSU Biomedical Research Tower (Appendix C).

One PhD-level, OSU appointed faculty member will serve as the Program Director and will oversee and administer the program. One part-time (50% FTE) Administrative Assistant will be needed to help with program- related responsibilities such as general communications, course and room scheduling, and recruiting.

Projected additional costs associated with the program and evidence of institutional commitment and capacity to meet these costs.

We have developed a five-year budget projection with the assistance the OSU College of Medicine for the Master of Science degree (Appendix D). The budget includes the standard state subsidy for graduate programs. The program is predicted to be self-sufficient by year 2 and revenue generating in year 3.

Availability and adequacy of the faculty and facilities for the new degree program.

OSU is one of the largest research universities in the nation with more health sciences colleges and extensive laboratory and clinical infrastructure located in close proximity to one another than any other U.S. university. The Master of Science Graduate Program in Immunology and Microbial Pathogenesis will have an excellent infrastructure to support training and research. The OSU College of Medicine’s Department of Microbial Infection and Immunity is currently located within the Biomedical Research Tower on the Medical Center Campus. Our teaching and research operations are at the intersection of all fields relevant to immunology and infectious diseases, including but not limited to, bacterial, viral, and parasitic infectious diseases, emerging pathogens, cancer immunotherapies, transplant immunology, autoimmune diseases, basic immunology, and neurodegenerative diseases. The Department’s faculty researchers work in close collaboration with, for example: clinical faculty, faculty from the Department of Biomedical Engineering, the College of , and industrial partners in all relevant fields (see appendices B and D for detailed descriptions of the facilities and faculty). Additionally, numerous University Institutes, such as the Infectious Disease Institute, the Pelotonia Institute for Immuno-, the Dorothy M. Davis Heart & Lung Research Institute, and OSU Centers such as the Center for Biostatistics, Center for Retrovirus Research, the Battelle Center for Science, Engineering and Public Policy, and the OSU Comprehensive Cancer Center all have research efforts that will complement and support the program.

Students will also have access to the OSU Medical Library, providing physical and electronic resources that include many of the books, periodicals, journals, and other learning resources needed to support the teaching and scholarly activities of this proposed program.

Evidence that a market exists for a new program 5 MS in Immunology and Microbial Pathogenesis Full Proposal

Scientists trained in the fields of Immunology and Microbial Pathogenesis are actively engaged in research and developing technologies to benefit the health and wellness of Ohio’s residents. The need to ensure a safe food supply, the development of advanced immunology drugs for cancer and chronic inflammatory diseases, for combating the threat of infectious diseases caused by emerging pathogens, and antimicrobial resistance of existing pathogens are just a few examples of ongoing efforts within the state.

Accordingly, research and expertise in these fields are of increasing demand. According to the 2019 report by the state’s bioscience organization, BioOhio: “Ohio’s bioscience industry continues its strong history of innovation and discovery, with at least 3,300 organizations manufacturing products, providing essential services, or researching the next breakthrough at nearly 4,100 facilities around the state. This is an increase of 215 (6%) locations in the last five years and 889 (28%) since 2001.” The report categorizes this industry into six areas, all of which employ workers trained in the fields of microbiology and/or immunology: agricultural biotechnology, medical and testing laboratories, medical devices & equipment, medical product distribution, pharmaceuticals & therapeutics, and research & development. Additionally, the report projects nearly 5,000 new jobs in these areas and workers in these fields earn well above the average living wage for Ohio. For example, according to the most recent data released by the U.S. Bureau of Labor Statistics (May 2018), microbiologists in central Ohio earn an average annual salary of $68,030.

Students earning the MS degree will also be competitive for higher compensation and supervisory roles in the workforce. According to the American Society of Microbiology, the addition of a Master of Science to a relevant Bachelor’s degree will afford the graduate the opportunity to enter at a higher pay-scale and be considered for non-entry level specialties such as Clinical or Research Laboratory Managers, Biosafety Officers, and Instructor/laboratory coordinators (ASM publications, Careers in Microbiology and Microbial Sciences, 2019).

To maximize career prospects, it is expected that each student will interface regularly with the OSU Office of Student Life – Career Counseling and Support Services (https://ccss.osu.edu). This office provides counseling and support services, both on and off campus, to assist graduate and professional students in their career decision making processes. Additionally, the Program Director will meet with each student individually at the beginning of each term to discuss progress and career related goals.

STATEWIDE ALTERNATIVES

The program is highlighted by a laboratory-focused approach to graduate training that will serve a growing pool of students without detracting from existing programs across the state or local-partnership graduate programs. Students will benefit from education in laboratory management skills, host-pathogen interactions, infection and immunology-based bioinformatics, a world-class seminar series, and the variety of research carried out in the laboratories of our outstanding faculty. Additionally, MI&I provides significant interaction with PhD graduate students and post-doctoral scientists in an engaging training environment.

A comparison of related MS programs offered through University System of Ohio Institutions is outlined in Appendix E. The MS in Microbiology offered by the Department of Microbiology, College of Arts and Sciences at Ohio State University is offered on the Columbus campus and will serve as an alternative choice for prospective OSU students. The two programs differ however as the proposed program is designed specifically as a research-based, Thesis-awarded MS with a biomedical focus. Microbiology also offers a Thesis-MS as a terminal degree but also allows a non-thesis option for students (see attached letter of support and concurrence from the OSU Department of Microbiology, Appendix I). Relevant to this degree’s area of focus, within the University System of Ohio’s 14 four-year research universities, nine schools (including OSU) offer MS programs in microbiology and/or immunology related areas, only three of which are offered through university units conferring medical degrees (University of Cincinnati and University of Toledo). Geographically, the MS in Immunology and Microbiology at Wright State University is nearest at approximately 60 miles from the greater 6 MS in Immunology and Microbial Pathogenesis Full Proposal

Columbus metropolitan area. For this program, the proximity and relationship to a major academic medical center will afford trainees significant opportunity in clinical research. One ongoing example is the Department of Microbial Infection and Immunity’s recent collaboration with the OSU Wexner Medical Center’s Department of in leading the design and implementation of serological and micro diagnostic assays during the COVID-19 pandemic.

This program is unique, however, as it addresses the need for a work-force not only trained in the specialized knowledge of these areas of biomedical research, but also in the skill-set necessary to oversee and manage a productive research laboratory. Through required course content, graduates will gain knowledge of lab management practices such as staffing, purchasing, and safety oversight. This will set these graduates apart as they prepare to enter the work-force.

GROWTH OF THE PROGRAM

Current and future demand

Given the proximity to some of the state’s largest bioscience employers (e.g. OSU, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Abbott, Battelle, Cardinal Health), we expect our program to be in demand. Based on estimates of laboratory space, classroom availability, and the 27 full-time faculty conducting research within the department, we believe the program is well-positioned to achieve a running average enrollment of 12 students. The program plans to admit 6 new students each academic year and, based on a budget estimate, to be financially self-sufficient by year 3. To meet these projections, we do not anticipate the need for additional faculty, staff, or space. However, If the program interest exceeds expectations, further review of enrollment limits and institutional needs will take place.

Program assessment

To maximize the success of each enrolled student, graduate, and future student, the program will maintain an active self-assessment process (Appendix F) This will include: annual recording of application and admission data; student academic performance indices; student evaluations of instruction (course satisfaction), semester- based student performance evaluations (reviewed by the program director and a committee of program faculty); annual evaluations of the program by member faculty; annual student evaluations of the program; exit surveys; time-to-degree tracking; and career recording of alumni. These assessment data will be reviewed annually by the program committee and used to continually refine the program. These data will also serve as support of applications seeking program funding.

CURRICULUM AND INSTRUCTIONAL DESIGN

Curricular content

The coursework for the proposed Master of Science in Immunology and Microbial Pathogenesis is designed to provide foundational learning through a defined set of required core courses (Table 1) delivered within a proposed 5-term series initiating each Autumn term, with an optional early Summer start to laboratory rotations (Appendix A). The curriculum will further concentrate sub-disciplines through recommended elective courses offered at Ohio State University (Appendix B) and thesis research mentorship by program faculty (Appendix H). Therefore, the curriculum is designed to provide a solid educational, technical, and experiential foundation for graduate students entering their choice of academic, industrial, regulatory, or related work forces. To accommodate the individual scheduling and health-related needs of each student, with the exception of the necessary lab-based learning, all coursework is compatible with Meeting OSU guidelines for optional, remotely accessed asynchronous learning should that be necessary.

7 MS in Immunology and Microbial Pathogenesis Full Proposal

Mixed mode of delivery

Given the hands-on, research experience mission of this program, a full, distance-learning option is not planned to be offered at this time and the preferred mode of delivery for the program is designed as an on campus, in- person learning format. In the event however that in-person learning is limited due to public health concerns, all lecture-based courses, with the exception of laboratory-based learning (MEMCIM 7998) will be offered via an online, synchronous mode of delivery. Currently all existing lecture-based core courses are being taught in a synchronous, online format (BSGP 7240, MEDMCIM 7500, 7010, 7400, and 8010) following the OSU Office of Distance Education and eLeanrning’s Best Practices For Online Teaching Checklist (https://odee.osu.edu/instructors/distance-education/best-practices-online-teaching). In-person, laboratory research courses, which are an essential component of the intent of this program will be necessary and coordinated with guidance from the OSU College of Medicine and the Graduate School.

Table 1 – Master of Science in Immunology and Microbial Pathogenesis required courses

Additional core and elective courses are available within appropriate programs at OSU (Appendix B).

Course # TITLE CREDITS MEDMCIM (# TBD) MI&I Laboratory Scientific and Management Skills 2 MEDMCIM (#TBD)a, c MI&I Laboratory Rotations 2a MEDMCIM 7998a, c MI&I Graduate Research 13a MEDMCIM 7500c, d Recent Discoveries in Immunology and Microbial Pathogenesis 4 BSGP 7240/MICRBIO 7724 d Molecular Pathogenesis 3 MEDMCIM 7010 d Molecular and Cellular Immunology 3 MEDMCIM 7400b, d Advanced Topics in Microbial Pathogenesis 2b MEDMCIM 8010b, d Advanced Topics in Immunology 2b MEDMCIM (#TBD)c, d Thesis Writing 1

Total core credit hours 30

Elective courses relevant to degree ≥10

Total elective credit hours 10

Total credit hours required for degree 40

a. Indicates research-based laboratory practicum course. b. Students are required to select either 7400 or 8010 based on individual thesis focus under advisement of their research mentor/PI. c. Course is graded pass/no pass d. Meets OSU guidelines for remotely accessed asynchronous learning

8 MS in Immunology and Microbial Pathogenesis Full Proposal

Description of a required integrated, or culminating learning, experience

All students will be required to complete the OSU CARE Training in Responsible Conduct of Research Program (https://cehv.osu.edu/caretrainingprogram) or equivalent. This is a workshop-format program involving 8 discussion-based training sessions led and moderated by faculty ethicists from the OSU Center for Ethics and Human Values with expertise in research ethics and integrity. Participants will watch a video of a related CARE panel discussion and read a curated set of readings prior to engaging in substantive face-to-face (or remotely- arranged equivalent) discussions of case studies that highlight the distinctive ethical challenges facing researchers. Topics covered in this training include: Conflicts of interest, protection of human subjects, mentorship relationships, collaborative research, authorship and publication, data sharing and privacy, the researcher as a responsible member of society, and environmental and societal impacts of research.

A thesis project culminating with a written thesis document and successful defense will be required to educate students in research, professional writing, and continued self-education to promote their personal and professional growth. As part of the core curriculum, students will enroll in a laboratory rotation course (MI&I Laboratory Rotations, MEDMCIM XXXX) during AuY1 to aid in choosing a research mentor who will direct their project and guide them in developing their thesis. A thesis-writing core course will be required during SpY2 (MEDMCIM XXXX).

Should a student not be able to successfully complete their curriculum requirements during the five-term academic period, they will be allowed to petition the Immunology and Microbial Pathogenesis Program’s Curriculum Committee to request additional time in which to complete their proposed project. If the petition is approved, the student will be required to enroll in additional laboratory research and thesis writing courses as necessary. Their degree will not be conferred until they have completed their research project and successfully defended their thesis document.

9 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendices

Appendix A: Proposed Curriculum for the Master of Science in Immunology and Microbial Pathogenesis Degree Program

10 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix B: Selected Relevant Elective Course offerings at Ohio State University

Department of Microbiology, OSU College of Arts and Sciences

MICRBIOL 5147 (3 credits): Eukaryotic Pathogens MICRBIOL 5161 (3 credits): Bioinformatics and Molecular Microbiology MICRBIOL 5270 (3 credits): Antibiotics and Natural Products MICRBIOL 6010 (2 credits): Principles of Microbiology MICRBIOL 6020 (3 credits): Microbial Physiology and Biochemistry MICRBIOL 6080 (3 credits): Advanced Microbial Genetics MICRBIOL 6155 (3 credits): Microbial Ecology and Evolution MICRBIOL 7536 (2 credits): Advanced Food Microbiology

Department of Bioethics, OSU College of Medicine

BIOETHC 6010 (3 credits): Biomedical Research Ethics

Department of Biomedical Informatics, OSU College of Medicine

BMI 5710 (3 credits): Introduction to Biomedical Informatics BMI 5760 (3 credits): Public Health Informatics BMI 5770 (3 credits): Health Analytics: Data to Discovery to Dissemination

11 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix C: Physical Research Needs of the Program

OSU is one of the largest research universities in the nation with more health sciences colleges and extensive laboratory and clinical infrastructure located in close proximity to one another than any other U.S. university. The Master of Science Graduate Program in Immunology and Microbial Pathogenesis

The Department of Microbial Infection and Immunity is located within the Biomedical Research Tower of the Wexner Medical Center at The Ohio State University (OSUMC). The MI&I laboratories occupy approximately 23,000 sq. feet on the seventh floor of the Biomedical Research Tower, a 403,000-square-foot state-of-the-art building, across the street from the Medical Center. Major themes of the department are respiratory infectious diseases, intracellular parasitism, granulomatous inflammation, immunology, epigenetic control of innate and adaptive immunity.

MI&I Departmental Space and Resources MI&I space includes several common user areas, with 2 cold rooms and multiple procedure rooms outfitted for molecular and tissue culture work, as well as rooms dedicated to microscopy (including live imaging microscopy) and radioactive studies (including one certified radioactive procedure room with 4 large working stations).

MI&I Core Equipment Core MI&I equipment in the BRT includes: Class IIA 6 feet Biosafety Cabinets, double water-jacketed CO2 incubators, multiple incubators and shakers for bacteria culture, a Sheldon Bactron anaerobic chamber, a Beckman Optima L-100 XP Ultracentrifuge with numerous rotors, a Beckman Optima™ TLX Ultracentrifuge, an Avanti J-25I High Performance centrifuge, a Beckman J2-21 centrifuge, low-speed Beckman Coulter X- 14R/X15R refrigerated centrifuges, refrigerated microcentrifuges, several non-refrigerated microcentrifuges, a Molecular Devices SPECTRAmax M2e and a Molecular Devices SPECTRAmax M5 Multi-Mode spectrophotometer/luminometer/fluorometer microplate reader, a BioRad Bioplex Luminex-based multiplex system, a BioRad Tetrad 2 thermocycler, three eppendorf Mastercycler gradient thermocyclers, two BioRad MyCycler thermocyclers, BioRad iCycler thermocycler, two BioRad CFX96 and one Applied Biosystems real- time PCR systems, a BioRad Molecular Imager ChemiDoc XRS Imaging system, a Fotodyne Imaging system, a BD FACS Canto II Flow Cytometer, a Purelab and a Millipore Ultra water purification systems, a Beckman Biomek 2000 robotic system, two NanoDrop Spectrophotometers, a Savant speed-vac and gel dryer system, a size-exclusion chromatography system consisting of a HPLC connected to different sizing columns to perform lipoglycan purifications, silica gel column chromatography systems for lipid purifications, thin layer chromatography systems to allow for visualization and identification of lipids and carbohydrates, inverted microscopes with cameras, Olympus fluorescence microscope with DIC optics and software for camera, an IVIS Lumina Camera system, an Olympus FV10i confocal camera capable of life cell imaging, liquid nitrogen storage system, ATR/Heto Freeze-dryer lyophilizer, a BioRad Experion Automated Electrophoresis Station, liquid chromatography systems, a UV-crosslinking oven, UV transilluminator, a blue light transilluminator, two electroporators, two pH meters, water baths, shaking incubators, refrigerators, -20oC and -80oC freezers, balances, phosphoroimager, two autoclaves, and two automated dishwashers.

Additional Core Research Resources

The Department of Microbial Infection and Immunity has access to many state-of-the-art shared core research facilities (see https://medicine.osu.edu/research/resources/core-facilities for a full listing). Some examples relevant to the proposed degree program are:

OSU BSL-3 Research Core

Research projects involving risk group 3 (RG3) pathogens, such as ongoing SARS-CoV-2 takes place in the BSL3 facilities/resources available at The Ohio State University (OSU) and the OSU College of Medicine (OSU 12 MS in Immunology and Microbial Pathogenesis Full Proposal

COM). The BSL3 Facility focuses on RG3 respiratory pathogens including emerging pathogens (e.g. SARS- CoV-2), and pathogens that can cause worldwide chronic and antibiotic-resistant infections (e.g. Mycobacterium tuberculosis). BSL3 facilities available for research include: a 3350 sq. ft lab space located in OSU Biomedical Research Tower (BRT), which consists of 6 separate laboratories for safely handling and processing infected tissues and cultures. These laboratories contain biosafety cabinets, centrifuges, microcentrifuges, light and fluorescent microscopes, ELISA and microplate readers, CO2 and humidified incubators. Computer stations facilitate the safe removal of notes and data from the facility.

Access to BSL3 facilities is granted only when personnel receive thorough biosafety training and appropriate on- site training. Training materials are reviewed by the BSL3 Program leadership, Institution Biosafety Officer, and EH&S leadership. Biosafety training emphasizes the facility design and systems in place and the current rules for best practices and regulations that users must follow. Refresher training is provided on an annual basis and time sensitive issues are discussed in the monthly BSL3 user group (BUG). Further emphasis on administrative controls (e.g. BSL3 protocols), alongside on-site training focused on demonstrating proper application of personal protective equipment (PPE), facility features, and proper usage of equipment is given before authorization. Junior scientists must be accompanied and supervised by senior scientists who are experienced in BSL3-related techniques to ensure proper handling of infectious materials. The BSL3 program at OSU ensures that all BSL3 users are fully trained and supported by operational staff.

OSU University Laboratory for Animal Resources (ULAR) Animals to be used for this study are covered by an institutional protocol. The basement level of the Biomedical Research Tower (BRT) houses the animal facility. This facility provides resources for the performance of experiments involving animal models of human diseases. ULAR is responsible for the animal care program that is AAALAC-accredited since 1962 (Accreditation # 028). Over 100,000 sq. ft of animal housing space in 15 facilities can accommodate rodents, rabbits, swine, ruminants, and dogs as well as other species. Rodent facilities have over 70 dedicated rooms, which include barrier housing, sterile housing, phenotyping, and GEM production facilities. ULAR consists of 3 veterinary ACLAM diplomats, 4 clinical veterinarians, and over 70 fulltime animal care staff.

OSU Flow Cytometry Shared Resource (FSCR) - This core facility assists in the analysis and sorting of cell populations according to the expression of selective cellular markers. Software available for use offline includes: WinMDI, Modfit, Cellquest Pro and FACSDiva. Imaging output software used is Microsoft office for both PC and Macintosh systems. Instrumentation includes the BD FACS Aria, and FACS Vantage and i-Cyt Reflection. Bectin-Dickinson FACS Caliber, equipped with 4 MPT’s allowing for 4 color-analysis, using a 488 nm air-cooled Argon and 633 nm helium-neon laser as excitation wavelengths. The Becton-Dickinson FACS Vantage SE, capable of 6-color analysis, utilizing a Krypton 302C Inova laser for multi-line excitation 350-600 nm. This instrument has a turbo-sort option and a CLONECYT single-cell or multi-cell deposition system for microtiter plates or microscopy slides.

Campus Microscopy and Imaging Facility (CMIF, www.cmif.osu.edu) on the 2nd floor of the BRT offers a full range of microscopes, and support instrumentation allows cell and tissue preparation with immunocytochemistry, in situ hybridization, freeze-fracture, cryo-ultramicrotomy, scanning and transmission electron microscopy (FEI Nova 400 Nano SEM, FEI Tecnai G2 Bio Twin TEM). This facility also has a Zeiss LSM510 Scanning Confocal Microscope, an Olympus FV1000 Multiphoton, and a Visitech Infinity 3 Live-Cell Confocal Microscope. All microscopes are staff-operated or self-operated after training.

Laser Microdissection Pressure Catapulting Molecular Analysis Facility- This core facility contains a robotized PALM MicroLaser system with PALM MicroBeam IV instrument from Carl Zeiss MicroImaging GmbH and PALM RoboStage/RoboMover for high throughput sample collection. The facility enables molecular analyses of laser captured tissue material. Services include standardization of novel techniques related to tissue processing, staining, fixation and capture, with the goal of preserving nucleic acid and protein integrity of the laser-captured tissue. Capture and analysis of tissue down to the resolution of a single cell population (cutting 13 MS in Immunology and Microbial Pathogenesis Full Proposal

precision 0.6 micron) from in vivo tissue sections is routinely performed. In addition, the facility has developed a way to rapidly identify and capture human blood vessels from clinical samples in a manner that makes high- density screening of the transcriptome possible.

The Genomics Shared Resource - This resource occupies about 2,400 square feet on the 2nd floor of the Biomedical Research Tower (BRT). The Genomics Shared Resource provides both Nucleic Acid services and Microarray services. It offers instrumentation and expertise for DNA and RNA analysis using sequencing, genotyping, real-time PCR, Affymetrix GeneChips, nCounter Analysis, next-generation sequencing, DNA synthesis support and genome-wide analysis using the Illumina NGS platform and Affymetrix and customizable gene chips. Affymetrix GeneChip System including two GeneChip Hyb-Station Oven 320/640, Two Affy. Fluidics Station 450 and One Affy. GeneChip Scanner 3000. The system for in-house custom microarray including GeneMachine OminiGrid 100 Arrayer; Tecan TeMo Liquid Handling Workstation and four Tecan HS4800 Hybridization Stations; two Axon 4000B and 4200A Microarray Scanners, two MJ Tetrad thermocycler and PE 9700 PCR Machines. Applied Biosystems 3730 DNA Analyzers, Illumina Genome Analyzers IIx, 4 Applied Biosystems 7900HT sequence detection systems, NanoString Technologies’ nCounter System, Sequenom Compact MassArray, Transgenomic Wave DHPLC Systems, Beckman Biomek FX liquid handler, Typhoon 9410 imager and Personal Densitometer from GE Healthcare, Agilent Bioanalyzer.

The OSU Campus Chemical Instrument Center (CCIC, www.ccic.ohio-state.edu), located on the 2nd floor of the BRT, provides state-of-the-art research facilities in three areas: Nuclear Magnetic Resonance (NMR), Mass Spectrometry (MS) and Proteomics Facility. The Mass Spectrometry and Proteomics facility is directed by Dr. Liwen Zhang and is equipped to offer a broad range of services with seven state-of-the-art mass spectrometers: a Thermo LTQ-Orbitrap, a Thermo LTQ, a Bruker Esquiere LC/MS, a Micromass LC-TOF, a Bruker Reflex III MALDI-TOF, a Thermo Trace GCMS, and a Micromass Q-TOF II. The lab is also equipped with an Ettan Spot Handling Workstation and a Dalt12 system for complete proteomic analysis including gel electrophoresis separation and subsequent protein identification, post-translational modification analysis and MudPIT. These instruments provide for accurate mass determination, sequence determination of biomolecules, oligonucleotides analysis, molecular weight analysis by mass assignment (ESI, EI, MALDI), quantification using GC-MS, and peak detection and identification by LC/MS.

Comparative Pathology & Mouse Phenotyping Shared Resource (CPMPSR) Facilities- The CPMPSR provides expert, readily available and affordable experimental pathology support to investigators utilizing animal models to study human disease. Comparative pathologists affiliated with the CPMPSR are familiar with normal anatomy and physiology, as well as background age- and strain-related lesions of various animal models. Recognition of lesions and their interpretation in the context of individual investigations provides a critical component to research incorporating animal models. Services include comprehensive macroscopic and microscopic examinations of various species of laboratory animals with an emphasis on the phenotypic characterization of newly produced lines of genetically engineered mice. Additional services include , , radiography, routine frozen and paraffin slide preparation as well as tissue microarray preparation and special histochemical and immunohistochemical staining. The main laboratory for the CPMPSR is located on the 4th floor (467/471) of the Veterinary Medicine Academic Building (VMAB). The core has: a Euthanex SMARTBOX unit; 7’ TBJ, Inc. 36-84-S down-draft, height-adjustable necropsy table; a 4’ Pacific Southwest Prep Station Lab down-draft tissue trimming station; necropsy equipment; an Olympus SZ-6145TR stereozoom microscope with attached Altra 20 digital camera; Hewlett Packard Faxitron Series Cabinet Xray System; and, photographic equipment (Nikon D90 digital SLR with Nikon 60 2.8 micro lens, photo stand and lighting). The necropsy room is also equipped with a MOPEC LD500 ventilated tissue storage cabinet. The laboratory is equipped with automated benchtop hematology (FORCYTE Autosampler 10 with OSI Data Management System) and chemistry (VetAce) analyzers, as well as an Aerospray hematology slide stainer-centrifuge with Cytopro rotor. The laboratory also includes a Fisher double door refrigerator, 2 Thermo Forma freezers (-70oC), and, 2 refrigerated centrifuges (Beckman Allegra X-22, Eppendorf). In addition, the lab has a doubled-headed Olympus BX41 light microscope with attached Altra 20 digital camera for performing blood differential counts and evaluation of urine/fluid/cytology samples. The histology laboratory (302 Goss Laboratory) 14 MS in Immunology and Microbial Pathogenesis Full Proposal

occupies approximately 1160 sq. ft. and includes Tissue Tek VIP and Fischer Histomatic 266 MP tissue processors; Shandon HistoCentre 2 and Tissue Tek embedding stations; 6 microtomes (Olympus 4055 micro, Leitz 1512, HM315); a Dako Universal Training Center autostainer with Seymour slide labeler; a Leica IPS modular histology slide printer; Microm HM500 OM and Leica CM1950 cryostats; and, an Olympus BH2 immunofluorescence light microscope. Other support equipment includes pH meter, balances, centrifuges, FG- 311 refractometer and vortex mixers. Room 933 in the Biomedical Research Tower is used for image analysis and discussing pathologic findings with investigative staff. The room includes a 6-headed Olympus BX51 light microscope with attached Altra 20 digital camera and MicroSuite software linked to a 42” Panasonic plasma television. Reference laboratories, including AniLytics, Incorporated in Gaithersburg, MD and Rules Based Medicine in Austin, TX provide specialized testing such as hormone and cytokine assays.

OSU Human Tissue Resource Network (HTRN), Pathology Core Facility. The core has: Microtomes (4), Cryostat (1), Tissue Processor (1), Water Baths (5), Automated Slide Stainer (1), Automated Immunohistochemistry Instrument (DAKO) (2), Automated Slide Labeler (TBS) (1), Tissue Matrix Array (Beecher Instruments) (1), Vacuum Processor (1), Refrigerators (2), Freezers (3), Real(time PCR (Roche) (1), ABI(3130XL DNA Sequencer (1), Microcentrifuge (2), Balances (2), Bioview Accord Semi(automated Scanning System (1), Biosafety Hood (1), Incubators (3), Drying Oven (1 ), Thermomixers (2).

The Center for Biostatistics, Department of Biomedical Informatics, is located at 1800 Kenny Rd, Columbus, OH, 43210. The Center for Biostatistics is equipped with a diverse palette of statistical software including SAS 9.4 (SAS Institute Inc., Cary, NC), STATA 13 (StataCorp, College Station, TX), Minitab (Minitab, Inc., State College, PA), R (open resource) and PASS 12 (NCSS, Kaysville, UT),) and specialized freeware Bio-conductor. The support of the office management software includes site-licensed Microsoft Office Professional. Through the Medical Center computer network, statisticians are provided with e-mail support, access to the Internet, and immediate back- up of all files.

The BRT is connected by an enclosed walking bridge with the Dorothy M. Davis Heart and Lung Research Institute (DHLRI), which contains several additional core research laboratories (Bioinformatics, Microarray- Genetics, EPR-NMR, Proteomics, and Integrative Cardiovascular Physiology). Each of these Cores is directed by a faculty scientist who is a leading expert in the specific technology of the Core. Each also has an experienced full-time manager who supports the application needs of the scientific users. DHLRI Animal Core- This core offers support for mouse colony management as well as performing specialized procedures and providing technical assistance for experiments. Our collaborator, Dr. Amer oversees the breeding of transgenic animals. Services also provided, but not limited to, include administration of reagents via various routes; tissue, blood, and bone marrow isolation; procedure training; genotyping; and special feeding. DHLRI Microscopy Core- This microscopy core laboratory with several fluorescent microscopes, time-lapse video microscopy and multi- channel visualization of fluorescence cellular antigens and other cell markers. (Nikon Eclipse 800 with DIC optics microscope and a Zeiss LSM510 multiphoton confocal inverted microscope).

Also, within the BRT is The Ohio State University Comprehensive Cancer Center (OSUCCC, www.osuccc.osu.edu). Located on the 8th and 9th floors, and part of the 10th floor, the OSUCCC houses core facilities for DNA sequencing radiochemistry, biostatistics and informatics, real-time PCR, Transgenic Animal Shared Resources, microarray, spectroscopy, electrophoresis, centrifugation, liquid scintillation counters, gamma counter chromatography (including HPLC), and microscopy.

Next to the BRT is the Biological Sciences building, which contains the Department of Microbiology (undergraduate Microbiology) and the Plant-Microbe Genomics Facility (PMGF, pmgf.osu.edu). The Plant- Microbe Genomics Facility provides services for DNA sequencing, DNA microarray work, and proteomics protocols.

The Research Institute at Nationwide Children’s Hospital (10 minutes from OSU) has a Vaccine and Immunity Research Group with core facilities containing DNA sequencing, Microarray, and Transgenic capabilities. 15 MS in Immunology and Microbial Pathogenesis Full Proposal

16 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix D: Five-year Projected Master’s Program Budget

Master of Science (MS) in Immunology and Microbial Pathogenesis

FY2022 FY2023 FY2024 FY2025 FY2026 FY2027 FY2028 Budget estimate is based upon:

Tuition Revenue Completion of the required minimum of 48 credit hours for the MS Full Time Track Number of Students 6 12 12 12 12 12 12 6 students in year 1 with a total enrollment of 12 thereafter Average Annual Credit Hours per student 24 20 20 20 20 20 20 Total Annual Credit Hours 144 239 239 239 239 239 239 Full time students complete in 5 semesters Average Annual Hours (prior 2 year window) 72 192 239 239 239 239 (minimum 8 credits/semester) Tuition per student, Au + Sp (in-state, 5% annual increase) 12,425 13,046 13,698 14,383 15,102 15,858 16,650 Tuition per student, Summer (in-state, 5% annual increase) 6,134 6,441 6,763 7,101 7,456 7,829 8,220 Fees for supplies based on the projected weekly research credit hours relative to a Tuition per FT student (assume year 2+ 0.83 FTE, 5% annual increase) 18,559 16,174 16,983 17,832 18,723 19,660 20,643 current departmental estimate of $2,500/year for a full-time PhD trainee Full Time Totals 111,353 194,088 203,792 213,982 224,681 235,915 247,711 75% effort by Program Director University Tax on Tuition (35% per Dan Clinchot) 27,838 48,522 50,948 53,495 56,170 58,979 61,928 50% effort by Administrative Assistant

Revenue from Tuition 83,515 145,566 152,844 160,486 168,511 176,936 185,783

Subsidy Revenue

Effective Rate (estimate, 2% annual increase) 270 275 281 287 292 298 304 Subsidy Generated 0 19,829 53,800 68,491 69,861 71,258 72,683 Tax on Subsidy (35%) 0 6,940 18,830 23,972 24,451 24,940 25,439

Revenue from Subsidy 0 12,889 34,970 44,519 45,410 46,318 47,244

Student Fee Revenue

Annual Lab Supply Fees (40 hour week) 2,500 2,500 2,500 2,500 2,500 2,500 2,500 Research hours/week 12 12 12 12 12 12 12 Revenue from Fees 4,500 7,470 7,470 7,470 7,470 7,470 7,470

Expenses

Personnel

Program Director (assuming 75% FTE +24.6% fringe, 2.5% annual increase) 93,450 95,786 98,181 100,635 103,151 105,730 108,373 Administrative Assistant (assuming 50% FTE + 32.3% fringe, 2.5% annual increase)33,075 33,902 34,749 35,618 36,509 37,421 38,357 Graduate/Postdoc TA Stipends (5% annual increase) 3,000 3,150 3,308 3,473 3,647 3,829 4,020

Misc. (5% increase) 2,000 2,100 2,205 2,315 2,431 2,553 2,680

Marketing (5% annual increase) 3,000 3,150 3,308 3,473 3,647 3,829 4,020 Travel (5% annual increase) 3,000 3,150 3,308 3,473 3,647 3,829 4,020

Total Expenses 137,525 141,238 145,058 148,987 153,030 157,191 161,471

FY2022 FY2023 FY2024 FY2025 FY2026 FY2027 FY2028

Total Revenue (tuition, subsidy, fees) 88,015 165,925 195,284 212,476 221,390 230,724 240,497 Total Expenses (personnel, misc.) 137,525 141,238 145,058 148,987 153,030 157,191 161,471 Total Income -49,510 24,687 50,226 63,488 68,360 73,534 79,026

17 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix E: Statewide Alternatives

Institution Degree Designation Required Research Thesis? Credit Hours requirement?

Kent State Univ. MS in Biology w/ microbiology ≥32 Yes Yes emphasis Miami of Ohio MS in Microbiology ≥30 Optional Optional OSU (Dept. of MS in Microbiology, thesis >44 Yes No Microbiology) MS in Microbiology, non-thesis >30 Ohio University MS in Biological Sciences ≥30 Yes Yes Univ. of Akron MS in Biology ≥40 (≥12 in Yes Optional research) Univ. of Cincinnati MS in Molecular Genetics, ≥30 Yes Yes Biochemistry & Microbiology

Univ. of MS in Immunology ≥30 Yes Yes Cincinnati/Cincinnati Children’s

Univ. of Toledo MS in Medical Immunology and ≥40 Yes Yes Microbial Pathogenesis Wright State Univ. MS in Immunology and ≥30 No No Microbiology Youngstown State MS in Biological Sciences with ≥36 Optional Optional specialization in Immunology or Microbial Pathogenesis

18 MS in Immunology and Microbial Pathogenesis Full Proposal

19 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix F: Program Goals and Plan for Program Assessment

The goal of the MS Program in Immunology and Microbial Pathogenesis is to provide focused, formal, hands-on research training for Ohio residents seeking careers in academic, pharmaceutic, biotech, agriculture, government, and public health fields. At the completion of the degree, students will be able to demonstrate a depth of knowledge and practical skill-set in the following key areas:

• Laboratory management practices including the day-to-day logistics of laboratory operations (e.g. scheduling, ordering, personnel management, safety guidelines)

• Foundational and advanced knowledge of the fields of immunology and microbial pathogenesis

• Capacity to properly design, conduct, interpret, and present independent research experiments and projects

• Ability to convey, in written form, scientific hypotheses, research proposals, and findings as demonstrated through the successful completion of a written thesis

The program will have an active self-assessment process as outlined in Table 2 with data to be maintained in a secure database administered by the program, accessible by only the Program Director and Administrative Assistant. Relevant public data will also be posted on the program’s website. These assessment data will be reviewed annually by the program committee to continually refine the program and identify weaknesses in meeting the program’s overall goal of providing a student the highest possible chance for a rewarding career following graduation. These data will also serve as support for applications seeking program funding sources to aid in enrollment in the form of student scholarships. The program will track direct measures of student learning (e.g. course and cumulative GPA, graduation rates, time-to completion) to serve as indicators of ongoing program performance and program quality. For example, research-related measures of student authorship on scientific publications as well as oral and poster-based research presentations will be collected and evaluated as an indicator of both student and program faculty performance and enthusiasm. Similarly, advisors and program leadership will monitor student academic performance regularly through advisory one-on-one meetings each semester with both the student and research mentor. Advising sheets will be completed to summarize and record these meetings and signed by the student, their research advisor (if applicable), and the program director.

Table 2 – Program Assessment

Assessment Primary Metrics Reporting/Review Administered by Reviewed by Alignment between Frequency Program Goals and Assessments Program Tracking of of Annually, Sp term Program Program Faculty Assessment of program application and applications, applicant Director/Administrative Committee and posted strengths and enrollment data GPAs, applicant Assistant on Program website weaknesses in diversity, offer and recruitment (e.g. low acceptance rates applicant diversity, trends in average GPA) to help meet program enrollment goals Program Time-to-degree Annually, Sp term Program Program Faculty Assessment to evaluate academic tracking, average director/Administrative Committee program performance performance GPA, publication and Assistant (e.g. presentation data employment/placement rates) to meet quality of program Student GPA, research advisor Beginning of each Administrative Program Director Assessment of student academic and thesis committee academic term (Au, Assistant progress to meet GPA performance evaluations Sp, Su) and expected graduation date. Student Student evaluations of Beginning of each Administrative Program Faculty Assessment used by satisfaction instruction (SEIs), one- academic term (Au, Assistant and Program Committee the program to provide Sp, Su) and upon Director feedback to the 20 MS in Immunology and Microbial Pathogenesis Full Proposal

on-one advisory graduation (exit program and course meetings, exit surveys surveys) directors to ensure student expectations are met Faculty Program faculty Annually, Sp term Administrative Program Faculty Assessment to monitor satisfaction reviews Assistant Committee and maintain faculty enthusiasm and support of the program to continually improve the student course and lab research experience Program Alumni career Annually, Au term Administrative Program Faculty Assessment to evaluate Performance recording Assistant Committee, Posted on program graduate Program website employment success to help meet the overall goal of the program.

Appendix G: Course Descriptions/Syllabi

Core courses currently approved and offered:

MEDMCIM 7010 - Cellular and Molecular Immunology

Eligibility/Prerequisites:

Graduate students and advanced undergraduates are eligible. Auditing is welcome.

Course Goals and Outcomes: (try to list at least three)

1. Students will learn about innate and adaptive immunity and role of the immune system in host defense.

2. Students will be able to identify immune cells and their functions.

3. Students will be able to recall cytokines and chemokines and their function and the signaling pathways that are induced

4. Students will understand how the immune system determines self from non-self and the consequences when this fails.

5. Students will be able to read and understand immunology papers and seminars

Textbooks/Materials: Students should have online computer access, which is freely available throughout campus (e.g. Thompson Library).

Textbook is Cellular and Molecular Immunology by Abbas, Lichtman, and Pilla

Lectures will be posted on the MEDMCIM 7010 Carmen site.

Assignments:

Assignments will be posted on the MEDMCIM 7010 Carmen site. It is your responsibility to check Carmen and have the assignments completed before class.

21 MS in Immunology and Microbial Pathogenesis Full Proposal

Course Structure and Grading: (example)

The course is lecture-based focusing on cellular and immunology. The course covers all areas of innate and adaptive immunity. Lectures will be provided by numerous faculty with expertise in the area of lecture. (see Course Schedule, below).

Three exams, graded A-E will be given, contributing equal weight to the overall grade

To do well, come to class on time, participate, and complete the assignments. Above all, ask questions when you do not understand or need more information. The course is designed for you to succeed.

Grading Scale 90-100 = A; 87-89 = A-; 84-87 = B+; 86-80 = B; 79-75 = B-; 74-70 = C+; 69-65 = C; 64-60 = C ; 59-55 = D; 0-59 = E

Course Policies:

Students are expected to attend lectures and arrive before start lectures as tardiness is disruptive. Students are expected to take exams when scheduled or make arrangements with the instructor prior to the exam to take the exam at an earlier time. Failure to take an exam will result in a zero score for that exam.

Academic Misconduct

Academic integrity is essential to maintaining an environment that fosters excellence in teaching, research, and other educational and scholarly activities. Thus, The Ohio State University and the Committee on Academic Misconduct (COAM) expect that all students have read and understand the University's Code of Student Conduct, and that all students will complete all academic and scholarly assignments with fairness and honesty. Students must recognize that failure to follow the rules and guidelines established in the University's Code of Student Conduct and this syllabus may constitute Academic Misconduct. The Ohio State University's Code of Student Conduct (Section 3335-23-04) defines academic misconduct as: Any activity that tends to compromise the academic integrity of the University, or subvert the educational process. Examples of academic misconduct include (but are not limited to) plagiarism, collusion (unauthorized collaboration), copying the work of another student, and possession of unauthorized materials during an examination. Ignorance of the University's Code of Student Conduct is never considered an excuse for academic misconduct, so I recommend that you review the Code of Student Conduct and, specifically, the sections dealing with academic misconduct. If I suspect that a student has committed academic misconduct in this course, I am obligated by University Rules to report my suspicions to the Committee on Academic Misconduct. If COAM determines that you have violated the University's Code of Student Conduct (i.e., committed academic misconduct), the sanctions for the misconduct could include a failing grade in this course and suspension or dismissal from the University. If you have any questions about the above policy or what constitutes academic misconduct in this course, please contact me.

Disability Services:

The University strives to make all learning experiences as accessible as possible. If you anticipate or experience academic barriers based on your disability (including mental health, chronic or temporary medical conditions), please let me know immediately so that we can privately discuss options. To establish reasonable accommodations, I may request that you register with Student Life Disability Services. After registration, make 22 MS in Immunology and Microbial Pathogenesis Full Proposal

arrangements with me as soon as possible to discuss your accommodations so that they may be implemented in a timely fashion. SLDS contact information: [email protected]; 614-292-3307; slds.osu.edu; 098 Baker Hall, 113 W. 12th Avenue.

Grievances and Solving Problems:

According to University Policies, if you have a problem with this class, you should seek to resolve the grievance concerning a grade or academic practice by speaking first with the instructor or professor. Then, if necessary, take your case to the department chairperson, college dean or associate dean, and to the provost, in that order. Specific procedures are outlined in Faculty Rule 3335-7-23. Grievances against graduate, research, and teaching assistants should be submitted first to the supervising instructor, then to the chairperson of the assistant's department.

Diversity:

The Ohio State University affirms the importance and value of diversity in the student body. Our programs and curricula reflect our multicultural society and global economy and seek to provide opportunities for students to learn more about persons who are different from them. We are committed to maintaining a community that recognizes and values the inherent worth and dignity of every person; fosters sensitivity, understanding, and mutual respect among each member of our community; and encourages each individual to strive to reach his or her own potential. Discrimination against any individual based upon protected status, which is defined as age, color, disability, gender identity or expression, national origin, race, religion, sex, sexual orientation, or veteran status, is prohibited.

Counseling and Consultation Services/Mental Health Statement:

As a student you may experience a range of issues that can cause barriers to learning, such as strained relationships, increased anxiety, alcohol/drug problems, feeling down, difficulty concentrating and/or lack of motivation. These mental health concerns or stressful events may lead to diminished academic performance or reduce a student's ability to participate in daily activities. The Ohio State University offers services to assist you with addressing these and other concerns you may be experiencing. If you or someone you know are suffering from any of the aforementioned conditions, you can learn more about the broad range of confidential mental health services available on campus via the Office of Student Life Counseling and Consultation Services (CCS) by visiting ccs.osu.edu or calling (614) 292- 5766. CCS is located on the 4th Floor of the Younkin Success Center and 10th Floor of Lincoln Tower. You can reach an on-call counselor when CCS is closed at (614) 292-5766 and 24 hour emergency help is also available through the 24/7 National Prevention Hotline at 1-(800)-273-TALK or at suicidepreventionlifeline.org

Copyright Statement:

The materials used in connection with this course may be subject to copyright protection and are only for the use of students officially enrolled in the course for the educational purposes associated with the course. Copyright law must be considered before copying, retaining, or disseminating materials outside of the course.

23 MS in Immunology and Microbial Pathogenesis Full Proposal

BSGP 7240/MICRBIO 7724 - Microbial Pathogenesis

Eligibility/Prerequisites:

Graduate students and advanced undergraduates are eligible. Auditing is welcome.

Course Goals and Outcomes:

1. Students will learn about methods to study host-microbe interactions.

2. Students will be introduced to a wide variety of pathogens and the mechanisms by which they cause disease.

3. Students will learn how pathogens deal with nutritional, innate, and adaptive immunity.

Textbooks/Materials:

Students should have online computer access, which is freely available throughout campus (e.g. Thompson Library). Materials will be posted on the MEDMCIM 7724 Carmen site. It is your responsibility to check Carmen and read the materials.

Assignments:

There are no graded assignments.

Course Structure and Grading:

The course is lecture-based focusing on host-pathogen interactions and specific examples of major bacterial, viral, fungal, and parasitic pathogens. Lectures will be provided by numerous faculty with expertise in these pathogens.

Five exams, graded 0-100 will be given, contributing equal weight to the overall grade.

To do well, come to class on time, participate, and read the materials. Above all, ask questions when you do not understand or need more information. The course is designed for you to succeed.

Grading:

5 exams, 20% each.

Grading Scale 93-100 = A; 90-92 = A-; 87-89 = B+; 83-86 = B; 80-82 = B-; 77-79 = C+; 73-76 = C; 70-72 = C-; 67-69 = D+; 63-66 = D; 60-62 = D-; 0-59 = F

Course Policies:

Being respectful to the instructors of this course is appreciated. This includes showing up on time, silencing electronics, paying attention to the instructor and participating in discussions. Exams are during class time. Exams can be taken at other times with advance notice and agreement by the instructor.

Academic Misconduct

Academic integrity is essential to maintaining an environment that fosters excellence in teaching, research, and other educational and scholarly activities. Thus, The Ohio State University and the Committee on Academic 24 MS in Immunology and Microbial Pathogenesis Full Proposal

Misconduct (COAM) expect that all students have read and understand the University's Code of Student Conduct, and that all students will complete all academic and scholarly assignments with fairness and honesty. Students must recognize that failure to follow the rules and guidelines established in the University's Code of Student Conduct and this syllabus may constitute Academic Misconduct. The Ohio State University's Code of Student Conduct (Section 3335-23-04) defines academic misconduct as: Any activity that tends to compromise the academic integrity of the University, or subvert the educational process. Examples of academic misconduct include (but are not limited to) plagiarism, collusion (unauthorized collaboration), copying the work of another student, and possession of unauthorized materials during an examination. Ignorance of the University's Code of Student Conduct is never considered an excuse for academic misconduct, so I recommend that you review the Code of Student Conduct and, specifically, the sections dealing with academic misconduct. If I suspect that a student has committed academic misconduct in this course, I am obligated by University Rules to report my suspicions to the Committee on Academic Misconduct. If COAM determines that you have violated the University's Code of Student Conduct (i.e., committed academic misconduct), the sanctions for the misconduct could include a failing grade in this course and suspension or dismissal from the University. If you have any questions about the above policy or what constitutes academic misconduct in this course, please contact me.

Disability Services:

The University strives to make all learning experiences as accessible as possible. If you anticipate or experience academic barriers based on your disability (including mental health, chronic or temporary medical conditions), please let me know immediately so that we can privately discuss options. To establish reasonable accommodations, I may request that you register with Student Life Disability Services. After registration, make arrangements with me as soon as possible to discuss your accommodations so that they may be implemented in a timely fashion. SLDS contact information: [email protected]; 614-292-3307; slds.osu.edu; 098 Baker Hall, 113 W. 12th Avenue.

Grievances and Solving Problems:

According to University Policies, if you have a problem with this class, you should seek to resolve the grievance concerning a grade or academic practice by speaking first with the instructor or professor. Then, if necessary, take your case to the department chairperson, college dean or associate dean, and to the provost, in that order. Specific procedures are outlined in Faculty Rule 3335-7-23. Grievances against graduate, research, and teaching assistants should be submitted first to the supervising instructor, then to the chairperson of the assistant's department.

Diversity:

The Ohio State University affirms the importance and value of diversity in the student body. Our programs and curricula reflect our multicultural society and global economy and seek to provide opportunities for students to learn more about persons who are different from them. We are committed to maintaining a community that recognizes and values the inherent worth and dignity of every person; fosters sensitivity, understanding, and mutual respect among each member of our community; and encourages each individual to strive to reach his or her own potential. Discrimination against any individual based upon protected status, which is defined as age, color, disability, gender identity or expression, national origin, race, religion, sex, sexual orientation, or veteran status, is prohibited.

Counseling and Consultation Services/Mental Health Statement:

As a student you may experience a range of issues that can cause barriers to learning, such as strained relationships, increased anxiety, alcohol/drug problems, feeling down, difficulty concentrating and/or lack of 25 MS in Immunology and Microbial Pathogenesis Full Proposal

motivation. These mental health concerns or stressful events may lead to diminished academic performance or reduce a student's ability to participate in daily activities. The Ohio State University offers services to assist you with addressing these and other concerns you may be experiencing. If you or someone you know are suffering from any of the aforementioned conditions, you can learn more about the broad range of confidential mental health services available on campus via the Office of Student Life Counseling and Consultation Services (CCS) by visiting ccs.osu.edu or calling (614) 292- 5766. CCS is located on the 4th Floor of the Younkin Success Center and 10th Floor of Lincoln Tower. You can reach an on-call counselor when CCS is closed at (614) 292-5766 and 24 hour emergency help is also available through the 24/7 National Prevention Hotline at 1-(800)-273-TALK or at suicidepreventionlifeline.org

Copyright Statement:

The materials used in connection with this course may be subject to copyright protection and are only for the use of students officially enrolled in the course for the educational purposes associated with the course. Copyright law must be considered before copying, retaining, or disseminating materials outside of the course.

26 MS in Immunology and Microbial Pathogenesis Full Proposal

MEDMCIM 7500 - Departmental Seminar - Recent Discoveries in Immunology and Microbial Pathogenesis

Course Description:

The Department of Microbial Infection and Immunity Seminar Series will host nationally and internationally recognized speakers on a biweekly basis throughout the semester. This course will educate students in how to prepare for and comprehend these seminars through classroom journal club-format discussions and submission of written-format summary statements. Students are expected to attend all seminars and journal club sessions.

Eligibility/Prerequisites:

Graduate students and advanced undergraduates are eligible. Auditing is welcome.

Course Goals and Outcomes:

Learning Goal Measurable Outcomes

Students will become skilled in the 1. Knowledge of research article types (e.g., primary research, methodology to select, access, and read reviews, abstracts, proceedings) research articles 2. Proficiency in literature search and access strategies 3. Knowledge of the components of a research article (e.g., sections, figures, legends)

Students will develop the ability to 1. Completion of a seminar evaluation form addressing appraise a research seminar presentation aspects such as style, layout, and clarity 2. As a component of a written summary statement, assesses a research presentation in its overall effectiveness in conveying the content

Students will learn to comprehend and 1. Verbally describe and interpret data contained in research convey the impact of recent scientific articles discoveries in the fields of Immunology 2. Participate in a group discussion on the strengths and and Microbial Pathogenesis weaknesses of a research article 3. As a component of a written summary statement, assesses the discovery in the context of prior research gaps addressed, new technologies developed or used, and its potential for better health outcomes.

Textbooks/Materials:

No textbooks are required. Journal club papers will be posted on the course’s OSU Carmen website. Students should have online computer access for reading and literature searches, which is freely available throughout campus (e.g., Wexner Medical Library, Thompson Library).

Assignments:

27 MS in Immunology and Microbial Pathogenesis Full Proposal

Students will be assigned journal club papers approximately one week in advance of class discussion and, following each seminar, will be required to submit a completed seminar evaluation form along with a one-half page, single-spaced, 1-inch margin, summary of each seminar by 5:00 PM Monday of the following week. It is the student’s responsibility to have sufficiently prepared for the journal club session and to make certain that the summary assignments are completed and turned in on time. The written summaries must be original work though students are welcome to discuss the papers and seminars with classmates, colleagues, advisors, etc.

Course Structure and Grading:

The course is designed for the student to succeed and will be graded Satisfactory or Unsatisfactory (S/U). Students are expected to arrive to class/seminar on time, complete assignments on time, and to prepare for and contribute to the journal club discussions.

A grade of Satisfactory, based on meeting the above expectations, will be issued at the discretion of the course director. A grade of Unsatisfactory will be issued for any of the following:

1. Students that have accrued more than one unexcused absence 2. Students that have not completed a written summary for each seminar attended

Course Policies:

Attendance and participation are mandatory as per the grading structure (above). Students who are absent for an excused reason will be allowed to make up assigned components of the class missed due to the excused absence. Students should contact the course director as soon as possible to determine how the missed work should be completed. Students should let the course director know as soon as possible that they will be absent from a class session/seminar.

Disability Services:

The University strives to make all learning experiences as accessible as possible. If you anticipate or experience academic barriers based on your disability (including mental health, chronic or temporary medical conditions), please let me the course director know immediately so that we can privately discuss options. To establish reasonable accommodations, we may request that you register with Student Life Disability Services. After registration, make arrangements with the course director as soon as possible to discuss your accommodations so that they may be implemented in a timely fashion. SLDS contact information: [email protected]; 614-292-3307; slds.osu.edu; 098 Baker Hall, 113 W. 12th Avenue.

Grievances and Solving Problems:

According to University Policies, if you have a problem with this class, you should seek to resolve the grievance concerning a grade or academic practice by speaking first with the instructor or professor. Then, if necessary, take your case to the department chairperson, college dean or associate dean, and to the provost, in that order. Specific procedures are outlined in Faculty Rule 3335-7-23.

Diversity:

The Ohio State University affirms the importance and value of diversity in the student body. Our programs and curricula reflect our multicultural society and global economy and seek to provide opportunities for students to learn more about persons who are different from them. We are committed to maintaining a community that recognizes and values the inherent worth and dignity of every person; fosters sensitivity, understanding, and mutual respect among each member of our community; and encourages each individual to strive to reach his or 28 MS in Immunology and Microbial Pathogenesis Full Proposal

her own potential. Discrimination against any individual based upon protected status, which is defined as age, color, disability, gender identity or expression, national origin, race, religion, sex, sexual orientation, or veteran status, is prohibited.

Counseling and Consultation Services/Mental Health Statement:

As a student you may experience a range of issues that can cause barriers to learning, such as strained relationships, increased anxiety, alcohol/drug problems, feeling down, difficulty concentrating and/or lack of motivation. These mental health concerns or stressful events may lead to diminished academic performance or reduce a student's ability to participate in daily activities. The Ohio State University offers services to assist you with addressing these and other concerns you may be experiencing. If you or someone you know are suffering from any of the aforementioned conditions, you can learn more about the broad range of confidential mental health services available on campus via the Office of Student Life Counseling and Consultation Services (CCS) by visiting ccs.osu.edu or calling (614) 292- 5766. CCS is located on the 4th Floor of the Younkin Success Center and 10th Floor of Lincoln Tower. You can reach an on-call counselor when CCS is closed at (614) 292-5766 and 24 hour emergency help is also available through the 24/7 National Prevention Hotline at 1-(800)-273-TALK or at suicidepreventionlifeline.org

Academic Misconduct

Academic integrity is essential to maintaining an environment that fosters excellence in teaching, research, and other educational and scholarly activities. Thus, The Ohio State University and the Committee on Academic Misconduct (COAM) expect that all students have read and understand the University's Code of Student Conduct, and that all students will complete all academic and scholarly assignments with fairness and honesty. Students must recognize that failure to follow the rules and guidelines established in the University's Code of Student Conduct and this syllabus may constitute Academic Misconduct. The Ohio State University's Code of Student Conduct (Section 3335-23-04) defines academic misconduct as: Any activity that tends to compromise the academic integrity of the University, or subvert the educational process. Examples of academic misconduct include (but are not limited to) plagiarism, collusion (unauthorized collaboration), copying the work of another student, and possession of unauthorized materials during an examination. Ignorance of the University's Code of Student Conduct is never considered an excuse for academic misconduct, so it is recommended that you review the Code of Student Conduct and, specifically, the sections dealing with academic misconduct. If it is suspected that a student has committed academic misconduct in this course, Professors are obligated by University Rules to report suspicions to the Committee on Academic Misconduct. If COAM determines that a student has violated the University's Code of Student Conduct (i.e., committed academic misconduct), the sanctions for the misconduct could include a failing grade in this course and suspension or dismissal from the University. If you have any questions about the above policy or what constitutes academic misconduct in this course, please contact the course director.

Copyright Statement:

The materials used in connection with this course may be subject to copyright protection and are only for the use of students officially enrolled in the course for the educational purposes associated with the course. Copyright law must be considered before copying, retaining, or disseminating materials outside of the course.

29 MS in Immunology and Microbial Pathogenesis Full Proposal

MEDMCIM 7998 - Graduate Research in Microbial Infection and Immunity

Course Description: This course provides an opportunity for individualized study in the fields of microbial infection and immunity. This research-focused course will allow students the opportunity to participate in a research environment under the supervision of one of the department’s faculty members.

Prerequisites: Permission of instructor.

Credit hours: 1-8

Course Learning Outcomes: Student will be able to demonstrate knowledge of identified research experience by appropriate means (e.g. lab meetings, oral/poster presentations, research paper, etc.) to be determined in advance by student and instructor.

Course Materials: Selected readings on research topics will be provided by the instructor.

Grading: This course is graded S/U.

Course Policies:

Students are expected to reliably attend and participate in the faculty member’s lab as agreed upon. Students are expected to successfully complete mandatory laboratory safety training and carry out research and laboratory duties in a responsibly safe manner. Disregard for the department’s safety guidelines and/or engaging in unsafe behavior will result in expulsion from the class.

Academic Misconduct

Academic integrity is essential to maintaining an environment that fosters excellence in teaching, research, and other educational and scholarly activities. Thus, The Ohio State University and the Committee on Academic Misconduct (COAM) expect that all students have read and understand the University's Code of Student Conduct, and that all students will complete all academic and scholarly assignments with fairness and honesty. Students must recognize that failure to follow the rules and guidelines established in the University's Code of Student Conduct and this syllabus may constitute Academic Misconduct. The Ohio State University's Code of Student Conduct (Section 3335-23-04) defines academic misconduct as: Any activity that tends to compromise the academic integrity of the University, or subvert the educational process. Examples of academic misconduct include (but are not limited to) plagiarism, collusion (unauthorized collaboration), copying the work of another student, and possession of unauthorized materials during an examination. Ignorance of the University's Code of Student Conduct is never considered an excuse for academic misconduct, so I recommend that you review the Code of Student Conduct and, specifically, the sections dealing with academic misconduct. If I suspect that a student has committed academic misconduct in this course, I am obligated by University Rules to report my suspicions to the Committee on Academic Misconduct. If COAM determines that you have violated the University's Code of Student Conduct (i.e., committed academic misconduct), the sanctions for the misconduct could include a failing grade in this course and suspension or dismissal from the University. If you have any questions about the above policy or what constitutes academic misconduct in this course, please contact me.

Disability Services:

30 MS in Immunology and Microbial Pathogenesis Full Proposal

The University strives to make all learning experiences as accessible as possible. If you anticipate or experience academic barriers based on your disability (including mental health, chronic or temporary medical conditions), please let me know immediately so that we can privately discuss options. To establish reasonable accommodations, I may request that you register with Student Life Disability Services. After registration, make arrangements with me as soon as possible to discuss your accommodations so that they may be implemented in a timely fashion. SLDS contact information: [email protected]; 614-292-3307; slds.osu.edu; 098 Baker Hall, 113 W. 12th Avenue.

Grievances and Solving Problems:

According to University Policies, if you have a problem with this class, you should seek to resolve the grievance concerning a grade or academic practice by speaking first with the instructor or professor. Then, if necessary, take your case to the department chairperson, college dean or associate dean, and to the provost, in that order. Specific procedures are outlined in Faculty Rule 3335-7-23. Grievances against graduate, research, and teaching assistants should be submitted first to the supervising instructor, then to the chairperson of the assistant's department.

Diversity:

The Ohio State University affirms the importance and value of diversity in the student body. Our programs and curricula reflect our multicultural society and global economy and seek to provide opportunities for students to learn more about persons who are different from them. We are committed to maintaining a community that recognizes and values the inherent worth and dignity of every person; fosters sensitivity, understanding, and mutual respect among each member of our community; and encourages each individual to strive to reach his or her own potential. Discrimination against any individual based upon protected status, which is defined as age, color, disability, gender identity or expression, national origin, race, religion, sex, sexual orientation, or veteran status, is prohibited.

Counseling and Consultation Services/Mental Health Statement:

As a student you may experience a range of issues that can cause barriers to learning, such as strained relationships, increased anxiety, alcohol/drug problems, feeling down, difficulty concentrating and/or lack of motivation. These mental health concerns or stressful events may lead to diminished academic performance or reduce a student's ability to participate in daily activities. The Ohio State University offers services to assist you with addressing these and other concerns you may be experiencing. If you or someone you know are suffering from any of the aforementioned conditions, you can learn more about the broad range of confidential mental health services available on campus via the Office of Student Life Counseling and Consultation Services (CCS) by visiting ccs.osu.edu or calling (614) 292- 5766. CCS is located on the 4th Floor of the Younkin Success Center and 10th Floor of Lincoln Tower. You can reach an on-call counselor when CCS is closed at (614) 292-5766 and 24 hour emergency help is also available through the 24/7 National Prevention Hotline at 1-(800)-273-TALK or at suicidepreventionlifeline.org

Copyright Statement:

The materials used in connection with this course may be subject to copyright protection and are only for the use of students officially enrolled in the course for the educational purposes associated with the course. Copyright law must be considered before copying, retaining, or disseminating materials outside of the course.

31 MS in Immunology and Microbial Pathogenesis Full Proposal

MEDMCIM 8010 - Selected Topics in Advanced Immunology

Eligibility/Prerequisites:

Students will have passed MEDMCIM 7010 or obtained permission to enroll by course director

Course Goals and Outcomes:

1. Students will learn about complex immunological concepts and how they relate to specific diseases.

2. Students will expand their knowledge of general immune mechanisms as learned in MEDMCIM 7010 to understand the plasticity and specificity of immune responses in health and diseases.

3. Students will develop their critical thinking skills via literature search, reading, and preparing an oral presentation on selected topics of immunology and via group discussions.

4-Students will develop scientific writing and analytical skills through preparing a written commentary for each topic (one page).

Textbooks/Materials:

Lecture notes, relevant research manuscripts and reviews will be posted on Carmen. Students should have online computer access for literature search, which is freely available throughout campus (e.g. Thompson Library).

Assignments:

Assignments will be performed in class and will consist of preparing power point-based oral presentations and written commentaries. It is your responsibility to have the assignments completed.

Course Structure and Grading:

For each topic, the course includes a lecture (by faculty) followed by the presentation (by a student) and group discussion of an article. Lectures will be given by faculty with expertise in the selected topics (see Course Schedule, below).

Satisfactory/unsatisfactory based upon class participation and writing of a one-page summary on each topic discussed during the semester.

To do well, come to class on time and participate. Above all, ask questions when you do not understand or need more information. The course is designed for you to succeed.

Course Policies:

Attendance and participation are mandatory. Students should inform the course director and the professor if they can’t attend a class. Repeated absences and failure to complete the written assignments may lead to a failing grade.

Disability Services:

32 MS in Immunology and Microbial Pathogenesis Full Proposal

The University strives to make all learning experiences as accessible as possible. If you anticipate or experience academic barriers based on your disability (including mental health, chronic or temporary medical conditions), please let me the course director know immediately so that we can privately discuss options. To establish reasonable accommodations, we may request that you register with Student Life Disability Services. After registration, make arrangements with the course director as soon as possible to discuss your accommodations so that they may be implemented in a timely fashion. SLDS contact information: [email protected]; 614-292-3307; slds.osu.edu; 098 Baker Hall, 113 W. 12th Avenue.

Grievances and Solving Problems:

According to University Policies, if you have a problem with this class, you should seek to resolve the grievance concerning a grade or academic practice by speaking first with the instructor or professor. Then, if necessary, take your case to the department chairperson, college dean or associate dean, and to the provost, in that order. Specific procedures are outlined in Faculty Rule 3335-7-23. Grievances against graduate, research, and teaching assistants should be submitted first to the supervising instructor, then to the chairperson of the assistant's department.

Diversity:

The Ohio State University affirms the importance and value of diversity in the student body. Our programs and curricula reflect our multicultural society and global economy and seek to provide opportunities for students to learn more about persons who are different from them. We are committed to maintaining a community that recognizes and values the inherent worth and dignity of every person; fosters sensitivity, understanding, and mutual respect among each member of our community; and encourages each individual to strive to reach his or her own potential. Discrimination against any individual based upon protected status, which is defined as age, color, disability, gender identity or expression, national origin, race, religion, sex, sexual orientation, or veteran status, is prohibited.

Counseling and Consultation Services/Mental Health Statement:

As a student you may experience a range of issues that can cause barriers to learning, such as strained relationships, increased anxiety, alcohol/drug problems, feeling down, difficulty concentrating and/or lack of motivation. These mental health concerns or stressful events may lead to diminished academic performance or reduce a student's ability to participate in daily activities. The Ohio State University offers services to assist you with addressing these and other concerns you may be experiencing. If you or someone you know are suffering from any of the aforementioned conditions, you can learn more about the broad range of confidential mental health services available on campus via the Office of Student Life Counseling and Consultation Services (CCS) by visiting ccs.osu.edu or calling (614) 292- 5766. CCS is located on the 4th Floor of the Younkin Success Center and 10th Floor of Lincoln Tower. You can reach an on-call counselor when CCS is closed at (614) 292-5766 and 24 hour emergency help is also available through the 24/7 National Prevention Hotline at 1-(800)-273-TALK or at suicidepreventionlifeline.org

Academic Misconduct

Academic integrity is essential to maintaining an environment that fosters excellence in teaching, research, and other educational and scholarly activities. Thus, The Ohio State University and the Committee on Academic Misconduct (COAM) expect that all students have read and understand the University's Code of Student Conduct, and that all students will complete all academic and scholarly assignments with fairness and honesty. Students must recognize that failure to follow the rules and guidelines established in the University's Code of Student Conduct and this syllabus may constitute Academic Misconduct. The Ohio State University's Code of Student Conduct (Section 3335-23-04) defines academic misconduct as: Any activity that tends to compromise the academic integrity of the University, or subvert the educational process. 33 MS in Immunology and Microbial Pathogenesis Full Proposal

Examples of academic misconduct include (but are not limited to) plagiarism, collusion (unauthorized collaboration), copying the work of another student, and possession of unauthorized materials during an examination. Ignorance of the University's Code of Student Conduct is never considered an excuse for academic misconduct, so it is recommended that you review the Code of Student Conduct and, specifically, the sections dealing with academic misconduct. If it is suspected that a student has committed academic misconduct in this course, Professors are obligated by University Rules to report suspicions to the Committee on Academic Misconduct. If COAM determines that a student has violated the University's Code of Student Conduct (i.e., committed academic misconduct), the sanctions for the misconduct could include a failing grade in this course and suspension or dismissal from the University. If you have any questions about the above policy or what constitutes academic misconduct in this course, please contact the course director.

Copyright Statement:

The materials used in connection with this course may be subject to copyright protection and are only for the use of students officially enrolled in the course for the educational purposes associated with the course. Copyright law must be considered before copying, retaining, or disseminating materials outside of the course.

Descriptions of courses under design and to be approved:

MEDMCIM 7XXX MI&I Laboratory Scientific and Management Skills (2 credits) Graded A-E Course under design by program faculty (Drew, Deora, Novias, Root) Expected submission to Unit and Graduate School – January 2021

Brief Course Summary:

This required, two-credit course will serve as a key introductory course for each incoming MS program class. It will prepare students in the basic fundamentals of modern laboratory research skills necessary for their safe and productive integration into their chosen research laboratory. This will serve to provide foundational knowledge needed to maximize their productive success as they enter into the research environment. It will be taught in a modular format by program faculty using a combination of lecture-based and hands-on modalities. Each module will involve in-class lectures and quizzes introducing each subject area followed by small-group (4-6 student) in- lab learning/reinforcement experiences followed by graded lab exercises. Modules are expected to include: essentials of laboratory safety; biomedical research ethics, bioinformatics, laboratory personnel management, data analysis and presentation, essentials of sterile technique and cell culture, optical microscopy techniques, flow cytometry analysis. Two one-hour sessions will be attended each week with an additional expected six hours of self-paced at-home and in-lab study weekly. Grading will be based on attendance (25%), in-class quizzes (25%), and lab-based exercises (50%).

MEDMCIM 7XXX MI&I Laboratory Rotations (2 credits) Graded P/NP Course under design by program faculty (Drew, Deora, Novias, Root) Expected submission to Unit and Graduate School – January 2021 34 MS in Immunology and Microbial Pathogenesis Full Proposal

Brief Course Summary:

This required, two-credit course will provide the student an individualized research opportunity in at least two laboratories of participating program faculty. This research-focused course will allow students initial exposure to the Program’s laboratory environment through in-lab research experiences. It is assumed that one of these rotation experiences will culminate with a commitment to one of these laboratories for their thesis research. Grading will be pass/no pass.

MEDMCIM 7XXX Thesis Writing (1 credit) Graded P/NP Course under design by program faculty (Drew, Deora, Novias, Root) Expected submission to Unit and Graduate School – January 2021

This required, 1 credit course will guide students through the process of writing their required thesis document. Students will meet for one hour each week in a lecture format with course faculty to develop skills in scientific writing style, formatting, layout, and editing. In addition to support with their thesis document, students will become proficient with a variety of relevant software packages (MS Word, EndNote, GraphPad Prism, and Excel).

35 The Ohio State University MS in Immunology and Microbial Pathogenesis

Advising Sheet: OSU Master of Science in Immunology and Microbial Pathogenesis Total Credits Required: 40

Date: ______Student Name: ______OSU email address: ______Entering Term: ______Research Advisor ______

A: Core Courses (27 credits – complete all courses)

Course Hours Term and Year Grade MEDMCIM 7XXX: MI&I Scientific and Laboratory 2 Management Skills MEDMCIM 7XXX: MI&I Laboratory Rotations 2 MEDMCIM 7010: Molecular and Cellular Immunology 3 MEDMCIM 7020: Molecular Pathogenesis 3 MEDMCIM 7500: Recent Discoveries in Immunology and 4 (total hours) Microbial Pathogenesis MEDMCIM 7998: MI&I Graduate Research 13 (total hours)

B: EMPHASIS AREA CORE COURSES (2 credits – choose one course from the following list)

Course Hours Term and Year Grade BSGP 7400: Advanced Topics in Microbial Pathogenesis 2 MEDMCIM 8010: Advanced Topics in Immunology 2

C: ELECTIVE COURSES (≥10 credits selected from list below or relevant courses by permission of advisor)

Course Hours Term and Year Grade MICRBIOL 5161: Bioinformatics and Molecular 3 Microbiology MICRBIOL 5270: Antibiotics and Natural Products 3 MICRBIOL 6010: Principles of Microbiology 2 MICRBIOL 6020: Microbial Physiology and Biochemistry 3 MICRBIOL 6080: Advanced Microbial Genetics 3 MICRBIOL 6155: Microbial Ecology and Evolution 3 MICRBIOL 7536: Advanced Food Microbiology 2 BIOETHC 6010: Biomedical Research Ethics 3 BMI 5710: Introduction to Biomedical Informatics 3 BMI 5760: Public Health Informatics 3 MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix I: Letters of Concurrence and Support

Below please see the letters of support from:

1. Dr. Igor Jouline, Chair Department of Microbiology OSU College of Arts and Sciences

2. Dr. Prosper Boyaka, Chair and Microbial Biology Program co-director Department of Veterinary Biosciences OSU College of Veterinary Medicine

3. Dr. Jeffrey Parvin, Associate Dean for Graduate Studies Director, Biomedical Sciences Graduate Prog. OSU College of Medicine

37 Department of Microbiology 105 Biological Sciences Building 484 W. 12th Ave. Columbus, OH 43210 614-292-2301

Eugene Oltz, PhD Chair, Department of Microbial Infection & Immunity Samuel Saslaw Professor of Infectious Diseases The Ohio State University Wexner School of Medicine October 16, 2020

Dear Dr. Oltz,

I am writing to provide support for your application to the OSU Graduate School to establish a Master of Science degree program eniled Masers of Science in Immnology and Microbial Pathogenesis. I have read your application and I have also consulted with our Graduate Program Committee regarding its goals and overall context. We all agree that this will be a welcome addition to a suite of professional educational programs in Life Sciences at Ohio State. While there is a small overlap with the Master of Science track in our own Microbiology Graduate Program, your proposed program has a distinct, research-oriented focus on Immunology and Pathogenesis, which makes it unique and attractive to students seeking careers in academic and applied Life Sciences

I am sure your application will be well received, and I look forward to continue our joint efforts in offerring Ohio State students the best educational opportunities in Basic and .

Sincerely,

Igor B. Jouline, Ph.D. Rod Sharp Professor of Microbiology Interim Chair, Department of Microbiology College of Arts and Sciences The Ohio State University

Department of Veterinary Biosciences Office of the Chair 207 Goss Lab 1925 Coffey Road Columbus, OH 43210-1093 614-247.4671 Phone 614-292.6473 FAX vet.osu.edu/biosciences

October 19, 2020

Email: [email protected] Eugene Oltz, PhD Chair, Dept. of Microbial Infection & Immunity Samuel Saslaw Professor of Infectious Diseases College of Medicine The Ohio State University

Re: Master of Science (MS) in Immunology and Microbial Pathogenesis

Dear Gene,

This letter is to provide my support to the proposal for the Science (MS) Master’s degree in Immunology and Microbial Pathogenesis.

The last couple of decades have seen an increasing demand for high-caliber scientists trained in the fields of Immunology and Microbial Pathogenesis and who are capable of supporting the development of new immunology drugs for combating the threat of infectious diseases, as well as cancer and chronic inflammatory diseases. Thus, it is important to develop dedicated non-PhD programs that will provide laboratory-focused graduate training, and make the students highly marketable for positions such as clinical or research laboratory managers, biosafety officers, and instructor/laboratory coordinators.

As a longtime member of the immunology community and co-director of the Host Defense and Microbial Biology Program of the Infectious Diseases Institute (IDI) at The Ohio State University (OSU), I believe that the proposed Master’s degree in Immunology and Microbial Pathogenesis will fill an important gap and will be a good addition to the portfolio of graduate trainings provided by OSU. Therefore, I offer my full support to this proposal.

Sincerely,

Prosper Boyaka, PhD Professor, and Interim Chair Host Defense and Microbial Biology Program co-director, OSU IDI

Jeffrey D. Parvin, MD, PhD Louis Levy Professor for Cancer Associate Dean for Graduate Studies Director, Biomedical Sciences Graduate Program Dept. of Biomedical Informatics The Ohio State University [email protected] https://u.osu.edu/parvinlab/

July 31,2020

Eugene Oltz, PhD Chair, Dept. of Microbial Infection & Immunity Samuel Saslaw Professor of Infectious Diseases The Ohio State University College of Medicine

Dear Gene, As Associate Dean for Graduate Studies for the College of Medicine and as a Director of the Biomedical Graduate Program (PhD) at Ohio State University, I am writing in support of the Microbial Infection & Immunity department proposal to establish a Master’s of Science degree program titled, “Masters of Science in Immunology and Microbial Pathogenesis.” I am excited by the prospect of your department offering a stand-alone, terminal MS degree in this field as it will build the workforce specializing in immunology and microbiology, a specialty made important by the current pandemic and the success of immune-oncology. Regarding your proposed curriculum, we welcome your future students to enroll in BSGP 7240 (Molecular Pathogenesis) and BSGP 7400 (Selected Topics in Microbial Pathogenesis) as well as other appropriate BSGP courses. We look forward to your proposal’s success and its much-needed addition in training graduate students at Ohio State for vital careers in the biomedical sciences. Sincerely,

Jeffrey Parvin Associate Dean for Graduate Studies Director, Biomedical Sciences Graduate Prog.

MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix H: Program Faculty

Program faculty descriptions will be made available as a separate pdf document

36 BMI 5770: Health Analytics: Data to Discovery to 3 Dissemination

Total Credit Hours Completed to Date: ______Cumulative GPA to Date: ______

Total Credit Hours Remaining for Graduation ______Expected Date of Graduation ______

Student Signature: ______Date: ______

Program Advisor Signature ______Date: ______

Exit Requirements:

• Fulfillment of credit hour requirements • Fulfillment of course requirements • Achievement of a cumulative GPA of at least 3.0 in all courses taken for graduate credit • Successful completion of a Master’s Written Thesis Document • Submission of the Application to Graduate to the Graduate School, via the GradForms system, by the deadline communicated by the College of Medicine • Please review the Graduate School Handbook for more detail on these requirements Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Appendix H: Program Faculty

AMAL O. AMER, MD, PhD

706 Biomedical Research Tower, 460 West 12th Avenue, Columbus, Ohio 43210

QUALIFICATIONS Ph.D Microbiology and Immunology University of Western Ontario, London, Ontario, Canada. 2002 M.Sc. Clinical Pathology Cairo University, Egypt, Faculty of Medicine. 1990 MD Medicine Cairo University, Egypt, Faculty of Medicine. 1986 POSITIONS Current position THE OHIO STATE UNIVERSITY, Columbus, Ohio, 2007-present Professor (Tenured) 2016-present Department of Microbial Infection and Immunity Manage a research laboratory, train graduate, undergraduate, and medical students, obtain federal and private funds, teach immunology and microbiology, and participate in committees. Cystic Fibrosis Foundation RDP C3 Immune core co-director 2019-2023

Prior positions Associate Professor, Department of Microbial Infection and Immunity, Ohio State 2012- University. 2016 Assistant Professor, Department of , Ohio State University. 2007- 2012 Post-doctoral Fellow, University of Michigan, Cancer Center, Ann Arbor. 2004- 2007 Post-doctoral Fellow, University of Michigan, Department of Microbiology and 2001- Immunology. 2004 Doctoral Student, University of Western Ontario, Department of Microbiology and Immunology, London, Ca 1997-2001 nad a. Me Rese Associate, He Organiza (W Collaborating Center dic arch World alt tion HO for al h ) Heamoglobinopathies, Saudi Arabia. 1993- 1995 Research Associate, The National Research Center, Department of Biochemistry, Cairo, 1988- Egypt. 1993 Internship , Faculty of Medicine, Cairo University, Egypt 1987- 1988

HONORS AND AWARDS Press release: Research shows why one bacterial infection is so deadly in cystic fibrosis patients. http://phys.org/news/2012-04-bacterial-infection-deadly-cystic-fibrosis.html 2012 Press release: Research Shows Why One Bacterial Infection is so Deadly in Cystic Fibrosis URL: http://newsblaze.com/story/2012042214453500002.wi/topstory.html 2012 Press release: Harmless Bacteria Becomes Fatal in CF/ DailyRx.com (62,761 unique monthly visitors) 30 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

(4/22) DailyRx.com story: http://bit.ly/J6Szu. 2012 American Lung Association Career Investigator Award. 2009 Davis Heart and Lung Research Institute Thematic Grants Program. Ohio State University. 2007 Collip Medal Award. Nominated by The Department of Microbiology and Immunology. University of Western Ontario. 2002 Student Travel Award. Department of Microbiology and Immunology. University of Western Ontario. 2000 Ontario Graduate Scholarship (OGS). University of Western Ontario. 1999-2001 Special University Scholarship (SUS). University of Western Ontario. 1997-1998 TEACHING Graduate Lecturer Concepts in Biomedical Science IBGP7000. 2012-present

31 Course director Course Organizer: IBGP 7400. 2011-present Lecturer Cellular Mechanism and Pathogenesis of Inflammation. MVIMG847. 2010-present Lecturer Molecular pathogenesis. IBGP 7240. 2010-present Lecturer Signature Program Translational Science. Immunology and Inflammation. 2010-present Lecturer Molecular Biology of Bacterial Pathogens. 2010- 2011 Course director IBGP795. 2009-2012 Lecturer Biology of human diseases. 2011-2012 Lecturer Microbiology 524. 2008-2011 Lecturer Select Topics in Microbial Pathogenesis-IBGP 740. 2008-2011 Lecturer Microbial Pathogenesis Course-Department of Microbiology and Immunology, The University of Louisville, Louisville, Kentucky. 2008 Small Group Leader Infectious Disease Sequence, Department of Microbiology andImmunology, University of Michigan . 2002-2004 Teaching Assistant Biology of the Protists, Department of Microbiology and Immunology, University of Western Ontario. 1997-2000 Undergraduate Lecturer Microbial Pathogenesis and Immunobiology. Micro 4110. 2016

Medical Lecturer Host Defense Block, Infectious Disease. E Module. 2013-present Lecturer Infection and host/Pathogen Science. IBGP 8800.02. 2010-present

Graduate advising Kylene Daily, MSTP program 2017-present

Kaitlin Hamilton, MCDB program. 2016-present

Asmaa Ali Badr, MCDB program: Graduate Fellowship from the Egyptian Government. 2016-present

Kyle Caution, IBGP Program: 2009-2015 (graduation) Kyle published 13 co-author papers and one first author paper in Nature Scientific Reports. Kyle presented 32 abstracts, 7 oral presentations and obtained 16 awards and is now on a postdoctoral fellow with a Cystic Fibrosis foundation for 2 years.

Mia Tazi, IBGP Program: 2011-2015 (graduation) Mia published 8 co-author papers and1 first author paper in Autophagy. She obtained fellowships from the OSU T32 bridge program, American Association of immunologists and Cystic fibrosis Foundation. She is now a senior scientist in Vertex working on CF.

Basant Abdulrahman, MCDB Program: 2010 - 2012 (graduation) Basant moved from another lab without publications and within 2 years in Dr. Amer’s lab, she published 3 co-author and 2 first author publications. She obtained 3 awards and was supported by a fellowship from the Egyptian Education Bureau. She is now a postdoctoral fellow in Calgary Canada working on autophagy and viral infections.

Dalia Abdelaziz. Graduate student, Egypt education channel: 2009 - 2011 (graduation) Dalia published 3 co-author and 2 first author publications. She obtained 2 awards and was supported by a fellowship from the Egyptian Education Bureau. She is now a postdoctoral fellow in Canada working on autophagy and prions.

Postdoctoral and Clinical Fellows Kylene Daily. MSTP. The role of autophagy in Alzheimer’s Disease 2018-2022 Kyle Caution. Epigenetic regulation of autophagy in cystic fibrosis. 2015-2018 Cystic Fibrosis Foundation postdoctoral fellowship Kathrin Krause. Characterization of the macrophage innate immune response in cystic fibrosis. (Fellowship from the German Government). 2015-2017 Cystic Fibrosis Foundation postdoctoral fellowship Hany Khalil. The role of autophagy in Legionella pneumophila infection of the elderly population. (Fellowship from the Egyptian Bureau of Education). In 1 year, he published 1 paper and a review and was supported by a training grant from the Egyptian Government. 2014-2015 Arwa Abu Khweek. The role of caspase-7 substrates in pulmonary infection. She published 3 first author and 7 co-author papers and received 1 award. 2009-2012 Benjamin Kopp. The characterization of the inflammatory response to Burkholderia infection in human phagocytes. Ben published 3 papers and obtained a KO8 under the supervision of Dr. Amer. He is now a faculty at Nationwide Children’s Hospital 2009-2011 Sheetal Kotrange. The role Pyrin Inflammasome in Burkholderia infection. She published 4 papers and proceeded to obtain medical training. 2009-2011 Doctoral Students (Dissertation Committee Member) Isabel Menendez, DVM PhD (K01 co-mentor) 2019- LaurenSiavash Johnson , Azarimicrobiology. Microbiology. program (PI: Stephanie Seveau) 2017-present 2018 Songyu(PI: Dong. Seveau) Biochemistry program (PI:Dmitri Kudryashov) - Stephanie Ray. Microbiology program (PI: Chad Rappley) 2017-present Nichole Zalles. MSTP Program (PI: Croce). 2017-present Abbie Zewdu. MCDB Program (PI: Guttridge). 2016-2018 Hannah Bekebrede. CVM Program. OSU Veterinary School. (PI: Yasuko Rikihisa). 2016-present Sankalp Malhotra. MSTP Program (PI: Dan Wozniak). 2016-2018 Nicholas Chesarino. IBGP Program (PI: Jacob Yount). 2014-2017 Christian Harding. (PI: Bob Munson). 2012-2015 Parker Woods. IBGP program (PI: Ian Davis). 2013-2016 Charlie Pyle. (PI: Larry Schlesinger, Daren Knoell). 2012-2016 Bin Ni. MSTP program (PI: Larry Schlesinger). 2011-2014 Tamer Abdelghany. MCDP program (PI: Jay Zweier). 2010-2012 Dawn Walker. IBGP program (PI: Mark Drew). 2009-2013 Tom Cremer. IBGP program (PI: Susheela Tridandapani) 2009-2010

SUCCESS program students Maria Ortiz. The role of bacterial secretion systems in inflammation in cystic fibrosis. Summer 2014 Natalia S. Fernández Dávila. How biofilm-derived Legionella pneumophila evades the innate immune response in macrophages. Summer 2013

TRAINEE AWARDS Kaitlin Hamilton (GRA) T32 Infectious Disease Institute 2020- 2021 Kylene Daily (MSTP) BSGP Travel award for best oral presentation (2nd) 2019 Kaitlin Hamilton (GRA) C3 Cure Cystic Fibrosis Columbus award 2018 Kaitlin Hamilton (GRA) Oral and Poster presentation at NACFC 2018 Asmaa Badr (GRA) Fellowship from the Egyptian Government. 2015- 2019 Kathrin Krause (Postdoctoral Fellow). Cystic Fibrosis Foundation Training grant. Benjamin Kopp (clinical fellow) now a faculty at Nationwide children’s hospital) K08. Burkholderia- mediated defective killing mechanisms in macrophages from CF patients 2015-2019 Kyle Caution (Postdoctoral fellow). Cystic Fibrosis Foundation Training grant. 2016-2018 Kyle Caution (GRA). T32 Training grant (declined) 2016-2017 Kyle Caution (GRA). CMIB best poster presentation. Ohio State University. 2016 Kathrin Krause (Postdoctoral Fellow). Fellowship from the German Government. 2015-2017 Mia Tazi (GRA). ASBMB Travel award. 2015 Mia Tazi (GRA). AAI oral presentation. American Association of Immunologists Annual meeting. 2015 Kyle Caution (GRA). AAI oral poster presentation. AAI Annual meeting. 2015 Kyle Caution (GRA). AAI travel Award. American Association of Immunologists Annual meeting. 2015 Mia Tazi (GRA). St. Jude National Graduate Student Symposium Travel Award Winner. 2014 Mia Tazi (GRA). Edward F. Hayes Graduate research Forum Oral Presentation. 2014 Kyle Caution (GRA). Edward F. Hayes Graduate research Forum Poster Presentation. 2014 Kyle Caution (GRA). St. Jude National Graduate Student Symposium Travel Award Winner. 2014 Kyle Caution (GRA). ASBMB travel award. Saint Diego. California. 2014 Kyle Caution (GRA). ASM oral presentation and travel award. 2014 Mia Tazi (GRA). Hayes Graduate Research Forum best oral presentation (3rd). 2014 Youssra Saqr (Undergard). Cystic Fibrosis summer fellowship. 2014 Mia Tazi (GRA). Midwest Microbial Pathogenesis Conference invited talk. 2014 Seeshidra Hoque (Undergard). Ohio State University STEP award. (undergrad). 2014 Mia Tazi (GRA). Ohio State University Jane Addams Award for Civic engagement. 2014 Kyle Caution (GRA). CMIB Research Day oral presentation. Ohio State University. 2014 Mia Tazi (GRA). Sigma Xi research grant award. Ohio State University. 2014 Kyle Caution (GRA). Honorable mention Sigma Xi awards. Ohio State University. 2014 Mia Tazi (GRA). American Association of Immunologists Research Grant (AAI). 2014 Mia Tazi (GRA). Center for Microbial Interface Biology NIH Training Grant T32. 2013-2014 Mia Tazi (GRA). Midwest Microbial Pathogenesis Travel Award. 2013 Mia Tazi (GRA). Biomedical Sciences Graduate Program Oral Presentation Honorable Mention. 2013 Arwa Khweek (Postdoctoral fellow). ASBMB Travel award. 2013 Kyle Caution (GRA). Nomination for The Ohio State University Presidential Fellowship by Biomedical Science Graduate program. 2012, 2013 Mia Tazi (GRA). 27th Edward F. Hayes Graduate Research Forum Poster Presentation. 2013 Mia Tazi (GRA). Cystic Fibrosis summer fellowship. 2013 Basant Abdulrahman (GRA). ASBMB Travel Award. 2012 Mia Tazi (GRA). Immunology Round Table Travel Award for best Immunology Presentation. 2012 Basant Abdelrahman (GRA). Center for Microbial Interface Biology Research Day Travel Award. 2011 Kyle Caution (GRA). Dorothy Heart and Lung (DHLRI) Research Day. Best poster presentation. 2011 Kyle Caution (GRA). Center for Microbial Interface Biology Retreat Travel Award. 2010 Dalia Abdelaziz (GRA). Center for Microbial Interface Biology Oral Presentation. 2010

SERVICE University service Member of: E&F MI&I committee 2019- present The Promotion and Tenure Department of Microbial Infection and Immunity committee 2017- present The Promotion and Tenure COM committee 2018- present The T32 predoctoral Training Grant 2016- present The T32 postdoctoral Training Grant 2016- present The College of Medicine Admissions Committee. 2014-2018 The subcommittee for evaluation of MD-PhD candidates. 2014-2018 Executive Board of the Ohio State chapter of Sigma Xi. 2014-2016 Executive Board of the Ohio State chapter of Sigma Xi. 2014-2016 The University Senate (alternate). 2014-2017 The Faculty Council (alternate). 2014-2017 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

The University Senate (alternate). 2014-2017 Faculty Council (alternate). 2014-2017 Advisor for the MI&I Graduate Committee MIIGSS. Columbus. Ohio. 2012-2014 The MI&I Award committee. 2008-2009 Co-director of the CMIB Research Retreat. 2012-2014

35 Chair of the Award Committee Department of Microbial Infection and Immunity. 2013-2016 The Organization Committee for the Pulmonary Research Day. Columbus. Ohio. 2010-2011 The Organization Committee for the Dorothy Heart and Lung Research Institute 10th Annual Research Day. 2010-2011 The Educational Committee for the Dorothy Heart and Lung Research Institute. Columbus. Ohio. 2010-2012 Co-director of the CMIB Research Retreat. Columbus. Ohio. 2009-2010

National service Federal Advisory Committees: Service on NIH Study Section: Ad-hoc Member, Study Section: Basic Neuroscience of Aging Review Committee NIA 2020 Chair, Innate Immunity and Inflammation (III) Study Section. 2018 - 2020 Ad-hoc Chair, NIAID Investigator Initiated Program Project Applications (P01) ZAI1 P TM-I 2020 (M2) Co-Chair, Cystic Fibrosis Center ZDK1 GRB-M (J1) P30 program 2019 Ad-hoc Chair, NIAID Investigator Initiated Program Project Applications (P01) ZAI1 P TM-I 2018 (S2) Permanent member, Innate Immunity and Inflammation (III) Study Section. 2016 - 2018 Ad-hoc Member, Study Section ZRG1-BCMB-A 51. 2016 Permanent member, MTI (Mentored Transition to Independence; K99, K22) NHLBI. 2014 - 2016 Ad-hoc Member, Study Section ZRG1 DM. 2015 Ad-hoc Member, Study Section IHD. 2015 Ad-hoc Member, Study Section III. 2015 Ad-hoc Member, Study Section NIAID K99 special emphasis. 2015 Ad-hoc Member, Study Section NHLBI K99 special emphasis. 2014 Ad-hoc Member, Study Section LCMI Lung Cellular, Molecular, and Immunobiology. 2014 Ad-hoc Member, Study Section Topics in Bacterial pathogenesis and virulence IDM-V02. 2014 Ad-hoc Member, Special Emphasis Panel. 2013 Ad-hoc Member and Co-Chair, Special Emphasis Panel. 2013 Ad-hoc Member, Study Section LCMI. Lung Cellular, Molecular, and Immunobiology Study 2012 Section. Ad-hoc Member, Study Section HIBP. Host Interaction with Bacterial Pathogens Study Section. 2011

Associations Adhoc reviewer for the Alzheimer’s Association 2017, 2018, 2019

International service International grant review Reviewer for Cystic Fibrosis Foundation. Italy. 2 0 1 8 Reviewer for University of Camerino with the European Charter of Researchers, Italy. 2 0 1 5 Reviewer for Fonds Wetenschappelijk Onderzoek Research Foundation FWO Flanders, Belgium. 2 0 1 4 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Reviewer for The Medical research Council (MRC). United Kingdom. 2 0 1 4 Reviewer for The Fulbright postdoctoral fellowships. 2 0 1 3 Reviewer for Cystic Fibrosis Foundation. United Kingdom. 2 0 1 3 The Health research Board. Postdoctoral fellowships. Ireland. 2 0 0 9 Reviewer for Fonds Wetenschappelijk Onderzoek Research Foundation FWO Flanders, Belgium. 2 0 1 2 Reviewer for Cystic Fibrosis Foundation “Fondazione per la Ricerca sulla Fibrosi Cistica”, Italy. 2 0 1 2 Member of the Advisory Committee of the European Immunology Summit. 2 0 1 6 Conference organization Session Chair North American Cystic Fibrosis Meeting. Denver. Colorado. 2 0 1 8 Session Chair International Burkholderia cenocepacia working group meeting. Dublin. Ireland. 2 0 1 8 Session Chair Keystone Meeting Benf. Canada 2 0 1 7 Co-chair. Bacterial and parasitic Infection and Immunity. American Association of Immunologists 2 (AAI). 0 1 6 Session Co-chair: The 39th European Cystic Fibrosis Meeting. Basel. Switzerland. 2 0 1 6 Chair for the Organizing Committee for The International Burkholderia cenocepacia Conference. Columbus. Ohio. 2016 Co-chair of the workshop: Pathogenesis of Infection at North American Cystic Fibrosis Conference (NACFC) Georgia World Congress Center, Atlanta, Georgia. 2014

37 Member of the Organizing Committee for The International Burkholderia cenocepacia Conference. Ann Arbor. Michigan. 2012-2013 Member of the Organizing committee for Midwest Microbial Pathogenesis Conference. Columbus. 2012- 2013 Member and Chair of the Award Committee Department of Microbial Infection and Immunity. 2013- 2016

Journal review Chief editor: Frontiers Microbes and Innate Immunity. Host editor: Special issue on The Inflammasome. Frontiers in Cellular and Infection Microbiology Journal. Special issue on Microbial Modulation of Host Apoptosis and Pyroptosis. Frontiers in Microbiology Journal. Associate Editor: Nature Scientific Reports, The Journal of Frontiers in Cellular and Infection Microbiology, The International Journal of Biochemistry and Molecular Biology. Reviewer: Science, Nature, Nature Communications, Journal of Experimental Medicine, PNAS, Plos Pathogens, Autophagy Journal, Plos One, Trends in Microbiology, Cellular Microbiology Journal, Virulence Journal, Journal of Immunology, Infection and Immunity Journal, The Journal of Biological Chemistry, Cell Hosts and Microbes, Cellular Microbiology, The International Journal of Biochemistry and Cell Biology, The Journal of Leukocyte Biology, American Journal of Respiratory Cell and Molecular Biology, clinical and Experimental Immunology, Infection and Immunity.

RESEARCH SUPPORT Current project support -R01HL127651 (MPI: Amer/Cormet-Boyaka) 0.30 calendar 2.5% 07/20/2015-06/30/2020 NIH/NHLBI $254,966 The role of microRNA-calibrated autophagy in innate immunity and inflammation The goal of this project is to determine the epigenetic changes and other mechanisms leading to increased expression of miRNAs in CF patients and CF mice.

-R01AI124121 (MPI: Amer/Cormet-Boyaka) 2.52 calendar 21% 02/01/2016-01/31/2021 NIH/NIAID $337,252 Restoring macrophage function in cystic fibrosis The goal of this project is to understand the basic biological mechanisms linking ion transport to autophagy and also offer new therapeutic targets for CF patients.

NIH supplement: NOT-AG-18-039 (Alzheimer's-focused administrative supplements for NIH grants that are not focused on Alzheimer's disease). 10% 2019-2022 Total Direct Costs $250,000 Total F&A (Indirect) Costs $140,000 Total Funds Requested $390,000 -HALLST18GO (PI: Luanne Hall-Stoodley) 0.48 4% 11/01/2018-10/31/2020 calendar Cystic Fibrosis Foundation $124,968 Xenophagy and clearance of nontuberculous mycobacteria in CF macrophages The goal of this project is to gain insights into the molecular mechanisms underlying unrestricted MaRg growth and subsequent pro-inflammatory signals in CF-MΦs to ultimately restore clearance by these innate immune cells. Role: Co-I

-MCCOY19R0 (PI: Amer / Kopp) 1.20 calendar 10% 07/01/2019-06/30/2023 NCH / Cystic Fibrosis Foundation (Prime) $ 29,946 Cure CF Columbus (C3 RDP); Core C Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

The goal of this project is to study the impact of CF Immune Dysfunction on Infection, and to develop treatments that prevent this pathophysiology.

39 - DFA Startup Reinvestment Funding $125,000 2019-2023 - T32 infectious disease institute: Kaitlin Hamilton

Pending: - R01 R01AI154553-01 TheInflammasome and Host-Legionella Interaction - R21 R21AG067755-01A1Alzheimer’s Disease Biomarker for Diagnosis and Prognosis

- R21 R21AG064899-01A1 Mechanistic basis of inflammation in Alzheimers Disease - R21R21DA047748-01A1 Using reduced representation bisulfite sequencing (RRBS) to unravel susceptibility determinants to Legionella pneumophila in smokers

Previous project support - CFF postdoctoral fellowship Kathrin Krause - CFF Postdoctoral fellowship Kyle Caution - R21 Amer (PI) Human susceptibility to Legionella infection 2015-2018 - R01 HL094586 Amer (PI) NIH: Role of caspases in Legionella pneumophila pulmonary infection. 2009-2014 R01HL094586-04 Supplement: 2013-2014 No cost extension 2014-2015 - R21 Amer (PI) 2009-2011 How can caspases control intracellular pulmonary infection. - Non NIH American Lung Association (ALA) Amer (PI): The role of caspases in Legionella pneumophila pulmonary infection. 2009-2011 Thematic Grants Program Amer (PI): DHLRI 2007 Cystic Fibrosis Grant, Cystic Fibrosis Foundation (CFF) Amer (PI) 2014-2016 Ohio State University Center for Clinical and Translational Science (CCTS) Amer (PI) 2014-2015 Public Health Preparedness for Infectious Diseases (PHPID) Amer (PI) 2013-2015

PUBLICATIONS (63) h-index 33, i10-index 47, citations 11,815 1. Wang C, Timari I, Zhang B, Li DW, Leggett A, Amer AO, Bruschweiler-Li L, Kopec RE, Bruschweiler R. 2020. COLMAR Lipids Web Server and Ultrahigh-Resolution Methods for 2D NMR- and MS-Based Lipidomics. J Proteome Res. PMID: 32073269 2. Kathrin Krause1, Kylene Daily, Shady Estfanous, Kaitlin Hamilton, Asmaa Badr, Arwa Abu Khweek, Rana Hegazi, Midhun N. K. Anne, Brett Klamer, Xiaoli Zhang , Mikhail A. Gavrilin, Vijay Pancholi, and Amal O. Amer. 2019. Caspase-11 counteracts mitochondrial ROS- mediated clearance of Staphylococcus aureus in macrophages. EMBO Reports. PMID:31637841, PMCID:6893291 3. Kyle Caution, Nicholas Young, Frank Robledo-Avila, Kathrin Krause, Arwa Abu-Kweek, Kaitlin Hamilton, Asmaa Badr, Anup Vaidya, Moses Gosu, Duaa Dakhlalla, Sudha Argwal, Xiaoli Zhang, Santiago Partida-Sanchez, Mikhail Gavrilin, Wael Jarjour, and Amal O. Amer. 2019. Caspase-11 mediates migration of neutrophils and necroptosis during acute gouty arthritis. Frontiers in Immunology. PMID:31803174 PMCID: PMC6874099 4. Juan Tang, Sha Tu, Guoxin Lin, Hui Guo, Chengkai Yan , Qingjun Liu, Ling Huang, Na Tang, Yizhi Xiao, Marshall R. Pope, Murugesan V.S. Rajaram, Amal O. Amer, Brian M. Ahmer, John S. Gunn, Daniel J. Wozniak, Lijian Tao, Coppola Vincenzo, Liwen Zhang, Wallace Y. Langdon, Jordi B. Torrelles, Stanley Lipkowitz and Jian Zhang. 2019. Sequential Ubiquitination of NLRP3 by RNF125 and Cbl-b Limits Inflammasome Activation and Endotoxemia. Journal of Experimental Medicine. PMID: 31999304 5. Kyle Caution, Alexander Pan, Kathrin Krause, Asmaa Badr, Kaitlin Hamilton, Anup Vaidya, Hawin Gosu, Kylene Daily, Shady Estfanous, Mikhail A. Gavrilin, Mark E. Drew, Estelle Cormet- Boyaka, Xi Chen, David E. Frankhouser, Ralf Bundschuh, Pearlly Yan, Duaa Dakhlallah and Amal O. Amer. 2019. Methylomic correlates of autophagy activity in cystic fibrosis. Journal of Cystic Fibrosis. PMID:30737168 PMCID: PMC6591064 6. Kathrin Krause, Kyle Caution, Asmaa Badr, Kaitlin Hamilton, Abdulmuti Saleh, Khushbu Patel, Rana Hegazi, Xiaoli Zhang, Mikhail A. Gavrilin and Amal O. Amer. 2018. Caspase-11 promotes autophagosome formation in response to bacterial infection. Autophagy. DOI: 10.1080/ 15548627. 2018.1491494 PMID:30165781 PMCID: PMC6152595 7. Sheng-Wei Chang, Ph.D; Jack Wellmerling; Xiaoli Zhang; Rachael Rayner; Wissam Osman; Sara Mertz; Amal Amer; Mark Peeples; Prosper Boyaka; Estelle Cormet-Boyaka. The psychoactive substance of cannabis 9-tetrahydrocannabinol (THC) negatively regulates CFTR in airway cells. 2018. Biochimica et Biophysica Acta. Accepted. 8. Arwa Abu Khweek and Amal O. Amer. 2018. Factors Mediating Environmental Biofilm Formation by Legionella pneumophila. Front. Cell Infect. Microbiol. 8: 38 PMID: 29535972 9. Frank H Robledo-Avila, Juan de Dios Ruiz-Rosado, Kenneth L Brockman, Benjamin Kopp, Amal O. Amer, Karen McCoy, Lauren O Bakaletz and Santiago Partida-Sanchez. 2018. Dysregulated calcium homeostasis in Cystic fibrosis neutrophils leads to deficient ROS/NETs antimicrobial responses. Frontiers Microbiology. Journal of Immunology. 201:2016-2027. 10. Seveau S, Turner J, Gavrilin MA, Torrelles JB, Hall-Stoodley L, Yount J, Amer AO. Checks and Balances between Autophagy and the Inflammasomes during Infection. J Mol Biol. 2017 430(2): 174- 192 PMID: 29162504 11. Shrestha CL, Assani KD, Rinehardt H, Albastroiu F, Zhang S, Shell R, Amer AO, Schlesinger LS, Kopp BT. Cysteamine-mediated clearance of antibiotic-resistant pathogens in human cystic fibrosis macrophages. PLoS One. 2017 5;12(10): e0186169. PMID: 28982193; PMCID: PMC5642023. 12. Kathrin Krause, Benjamin T. Kopp, Mia F. Tazi, Kyle Caution, Kaitlin Hamilton, Asmaa Badr, Chandra Shrestha, Dmitry Tumin, Don Hayes, Frank Robledo, Luanne Hall-Stoodley, Brett G. Klamer, Xiaoli Zhang, Santiago Partida-Sanchez, Narasimham L. Parinandi, Stephen E. Kirkby, Duaa Dakhlallah, Karen S. McCoy, Estelle Cormet-Boyaka and Amal O. Amer. 2017. The expression of Mirc1/Mir17-92 cluster in sputum samples correlates with pulmonary exacerbations in cystic fibrosis patients. Journal of cystic fibrosis. S1569-1993 PMID: 29241629 13. Kyle Caution and Amal O. Amer. 2016. Caspase-11: mediator of pyroptotic and non-pyroptotic host responses. Trends in Cell and Molecular Biology. Vol 11 In press. 14. Kathrin Krause and Amal O. Amer. 2016. Caspase exploitation by Legionella pneumophila. Frontiers in Microbiology. 7: 515 PMID: 27148204 15. Mia F. Tazi, Duaa A. Dakhlallah, Kyle Caution, Amr Ahmed, Hany Khalil, Ian Davis, Clay Marsh, Larry S. Schlesinger, Estelle Cormet-Boyaka and Amal O. Amer. 2016. Elevated miR-17~92 cluster expression negatively regulates autophagy and cystic fibrosis transmembrane conductance regulator (CFTR) function in CF macrophages. Autophagy Journal. 12: 2026-2037 16. Hany Khalil, Mia Tazi, Kyle Caution, Amr Ahmed, Apurva Kanneganti, Kaivon Assani, Benjamin Kopp and Amal O. Amer. 2016. Aging is associated with hypermethylation of autophagy genes in macrophages. Epigenetics. 3;11: 381-8 PMID: 26909551 17. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, .., Amer AO., et al. 2016. Guidelines for the use and interpretation of assays for monitoring autophagy. 2016. Autophagy. 12: 1. 18. Abu Khweek A, Apurvua Kanneganti and Amer O. Amer. 2016. The Sphingosine-1-Phosphate Lyase (LegS2) Contributes to the Restriction of Legionella pneumophila in Murine Macrophages. Plos One. 11: e0146410. 19. Kyle Caution, Mikhail A. Gavrilin, Mia Tazi, K. Apurva, Daniel Layman, Sheshadri Hoque, Kathrin Krause and Amal O. Amer. 2015. Caspase-11 and caspase-1 differentially modulate actin polymerization via RhoA and Slingshot proteins to promote bacterial clearance. Nature Scientific reports. 5:18479 PMID: 26686473 20. Parker Woods, Mia Tazi, Nicholas Chesarino, Amal Amer, and Ian Davis. 2015. TGF-β-induced IL-6 prevents development of acute lung injury in influenza A virus-infected F508del CFTR- heterozygous mice. American Journal of Physiology - Lung Cellular and Molecular Physiology. 308: L1136. 21. Dalia H. A. Abdelaziz, Hany Khalil, Estelle Cormet-Boyaka and Amal O. Amer. 2015. The cooperation between the autophagy machinery and the inflammasome to implement an appropriate innate immune response: Do they regulate each other? Immunological Reviews. 265: 194-204 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

22. He Y, Amer AO. 2014. Microbial modulation of host apoptosis and pyroptosis. Front Cell Infect Microbiol 4: 83 23. Assani K, Tazi MF, Amer AO, Kopp BT. 2014. IFN-gamma stimulates autophagy-mediated clearance of Burkholderia cenocepacia in human cystic fibrosis macrophages. PLoS One 9: e96681

44 24. Novotny LA, Amer AO, Brockson ME, Goodman SD, Bakaletz LO. 2013. Structural stability of Burkholderia cenocepacia biofilms is reliant on eDNA structure and presence of a bacterial nucleic acid binding protein. PLoS One 8: e67629 25. Khweek AA, Caution K, Akhter A, Abdulrahman BA, Tazi M, Hassan H, Majumdar N, Doran A, Guirado E, Schlesinger LS, Shuman H, Amer AO. 2013. A bacterial protein promotes the recognition of the Legionella pneumophila vacuole by autophagy. Eur J Immunol 43: 1333-44 26. Amer AO. 2013. The many uses of autophagosomes. Autophagy 9: 633-4 27. Khweek A, Fernandez Davila NS, Caution K, Akhter A, Abdulrahman BA, Tazi M, Hassan H, Novotny LA, Bakaletz LO, Amer AO. 2013. Biofilm-derived Legionella pneumophila evades the innate immune response in macrophages. Front Cell Infect Microbiol 3: 18 28. Famke Aeffner; Basant Abdulrahman; Judy M. Hickman-Davis; Paul Janssen; Amal Amer; David M. Bedwell; Eric J. Sorscher; Ian C. Davis. Heterozygosity for the F508del mutation in the cystic fibrosis transmembrane conductance regulator anion channel attenuates influenza severity. Journal of Infectious Diseases 2013; doi: 10.1093/infdis/jit251 29. Abdulrahman BA, Khweek AA, Akhter A, Caution K, Tazi M, Hassan H, Zhang Y, Rowland PD, Malhotra S, Aeffner F, Davis IC, Valvano MA, Amer AO. 2013. Depletion of the Ubiquitin- binding Adaptor Molecule SQSTM1/p62 from Macrophages Harboring cftr DeltaF508 Mutation Improves the Delivery of Burkholderia cenocepacia to the Autophagic Machinery. J Biol Chem. PMCID:3548511 288: 2049-58 30. Rosales-Reyes R, Aubert DF, Tolman JS, Amer AO, Valvano MA. 2012. Burkholderia cenocepacia type VI secretion system mediates escape of type II secreted proteins into the cytoplasm of infected macrophages. PLoS One. PMCID:3405007 7: e41726 31. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, .., Amer AO., et al. 2012. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. PMCID:3404883 8: 445-544 32. Gavrilin MA, Abdelaziz DH, Mostafa M, Abdulrahman BA, Grandhi J, Akhter A, Abu Khweek A, Aubert DF, Valvano MA, Wewers MD, Amer AO. 2012. Activation of the Pyrin Inflammasome by Intracellular Burkholderia cenocepacia. J Immunol. PMCID:3482472 188: 3469-77 33. Benjamin T. Kopp, Basant A. Abdulrahman, Arwa Abu Khweek, Surender B. Kumar, Anwari Akhter, Richard Montione, Mia F. Tazi, Kyle Caution, Karen McCoy, Amer AO. 2012. Exaggerated inflammatory responses mediated by Burkholderia cenocepacia in macrophages derived from cystic fibrosis patients. Biochem Biophys Res Commun.PMCID:3408781 424: 221- 7 34. Akhter A, Caution K, Abu Khweek A, Tazi M, Abdulrahman BA, Abdelaziz DH, Voss OH, Doseff AI, Hassan H, Azad AK, Schlesinger LS, Wewers MD, Gavrilin MA, Amer AO. 2012. Caspase- 11 Promotes the Fusion of Phagosomes Harboring Pathogenic Bacteria with Lysosomes by Modulating Actin Polymerization. Immunity. PMCID:3408798 37: 35-47 35. Kotrange S, Kopp B, Akhter A, Abdelaziz D, Abu Khweek A, Caution K, Abdulrahman B, Wewers MD, McCoy K, Marsh C, Loutet SA, Ortega X, Valvano MA, Amer AO. 2011. Burkholderia cenocepacia O polysaccharide chain contributes to caspase-1-dependent IL- 1beta production in macrophages. J Leukoc Biol. PMCID:3040464 89: 481-8 36. Hoda Hassan, Amer AO. 2011. Cell Intrinsic Roles of Apoptosis-Associated Speck-Like Protein in Regulating Innate and Adaptive Immune Responses. The Scientific World Journal. PMCID:3236380 11: 2418-23 37. Brooks MN, Rajaram MV, Azad AK, Amer AO, Valdivia-Arenas MA, Park JH, Nunez G, Schlesinger LS. 2011. NOD2 controls the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis and M. bovis BCG in human macrophages. Cell Microbiol. PMCID:3259431 13: 402-18 38. Anand PK, Tait SW, Lamkanfi M, Amer AO, Nunez G, Pages G, Pouyssegur J, McGargill MA, Green DR, Kanneganti TD. 2011. TLR2 and RIP2 pathways mediate autophagy of Listeria monocytogenes via extracellular signal-regulated kinase (ERK) activation. J Biol Chem. PMCID:3234870 286: 42981-91 39. Abdulrahman BA, Khweek AA, Akhter A, Caution K, Kotrange S, Abdelaziz DH, Newland C, Rosales- Reyes R, Kopp B, McCoy K, Montione R, Schlesinger LS, Gavrilin MA, Wewers MD, Valvano MA, Amer AO. 2011. Autophagy stimulation by rapamycin suppresses lung Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

inflammation and infection by Burkholderia cenocepacia in a model of cystic fibrosis. Autophagy. PMCID:3359483 7: 1359-70 40. Abdelaziz DH, Gavrilin MA, Akhter A, Caution K, Kotrange S, Khweek AA, Abdulrahman BA, Hassan ZA, El-Sharkawi FZ, Bedi SS, Ladner K, Gonzalez-Mejia ME, Doseff AI, Mostafa M, Kanneganti TD, Guttridge D, Marsh CB, Wewers MD, Amer AO. 2011. Asc-dependent and independent mechanisms

46 contribute to restriction of legionella pneumophila infection in murine macrophages. Front Microbiol. PMCID:3112328 2: 18 41. Abdelaziz DH, Gavrilin MA, Akhter A, Caution K, Kotrange S, Khweek AA, Abdulrahman BA, Grandhi J, Hassan ZA, Marsh C, Wewers MD, Amer AO. 2011. Apoptosis-associated speck- like protein (ASC) controls Legionella pneumophila infection in human monocytes. J Biol Chem. PMCID:3030324 286: 3203-8 42. Lamkanfi M, Sarkar A, Vande Walle L, Vitari AC, Amer AO, Wewers MD, Tracey KJ, Kanneganti TD, Dixit VM. 2010. Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J Immunol. PMCID:3428148 185: 4385-92 43. Khweek AA, Amer A. 2010. Replication of Legionella Pneumophila in Human Cells: Why are We Susceptible? Front Microbiol. PMCID:3109522 1: 133 44. Butchar JP, Mehta P, Justiniano SE, Guenterberg KD, Kondadasula SV, Mo X, Chemudupati M, Kanneganti TD, Amer A, Muthusamy N, Jarjoura D, Marsh CB, Carson WE, 3rd, Byrd JC, Tridandapani S. 2010. Reciprocal regulation of activating and inhibitory Fc{gamma} receptors by TLR7/8 activation: implications for tumor immunotherapy. Clin Cancer Res. PMCID:2848878 16: 2065- 75 45. Amer AO. 2010. Modulation of caspases and their non-apoptotic functions by Legionella pneumophila. Cell Microbiol. PMID:19863553 12: 140-7 46. Abdelaziz DH, Amr K, Amer AO. 2010. Nlrc4/Ipaf/CLAN/CARD12: more than a flagellin sensor. Int J Biochem Cell Biol. PMCID:2862870 42: 789-91 47. Rajaram MV, Butchar JP, Parsa KV, Cremer TJ, Amer A, Schlesinger LS, Tridandapani S. 2009. Akt and SHIP modulate Francisella escape from the phagosome and induction of the Fas- mediated death pathway. PLoS One. PMCID:2775408 4: e7919 48. Cremer TJ, Ravneberg DH, Clay CD, Piper-Hunter MG, Marsh CB, Elton TS, Gunn JS, Amer A, Kanneganti TD, Schlesinger LS, Butchar JP, Tridandapani S. 2009. MiR-155 induction by F. novicida but not the virulent F. tularensis results in SHIP down-regulation and enhanced pro- inflammatory cytokine response. PLoS One. PMCID:2794384 4: e8508 49. Cremer TJ, Amer A, Tridandapani S, Butchar JP. 2009. Francisella tularensis regulates autophagy- related host cell signaling pathways. Autophagy. PMCID:2774735 5: 125-8 50. Akhter A, Gavrilin MA, Frantz L, Washington S, Ditty C, Limoli D, Day C, Sarkar A, Newland C, Butchar J, Marsh CB, Wewers MD, Tridandapani S, Kanneganti TD, Amer AO. 2009. Caspase- 7 activation by the Nlrc4/Ipaf inflammasome restricts Legionella pneumophila infection. PLoS Pathog. PMCID: 2657210 5: e1000361 51. Valdivia-Arenas M, Amer A, Henning L, Wewers M, Schlesinger L. 2007. Lung infections and innate host defense. Drug Discov Today Dis Mech 4: 73-81 52. Lamkanfi M, Amer A, Kanneganti TD, Munoz-Planillo R, Chen G, Vandenabeele P, Fortier A, Gros P, Nunez G. 2007. The Nod-like receptor family member Naip5/Birc1e restricts Legionella pneumophila growth independently of caspase-1 activation. J Immunol 178: 8022-7 53. Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park JH, Franchi L, Whitfield J, Barchet W, Colonna M, Vandenabeele P, Bertin J, Coyle A, Grant EP, Akira S, Nunez G. 2006. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 440: 233-6 54. Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, Taraporewala ZF, Miller D, Patton JT, Inohara N, Nunez G. 2006. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem 281: 36560-8 55. Franchi L, McDonald C, Kanneganti TD, Amer A, Nunez G. 2006. Nucleotide-binding oligomerization domain-like receptors: intracellular pattern recognition molecules for pathogen detection and host defense. J Immunol 177: 3507-13 56. Franchi L, Amer A, Body-Malapel M, Kanneganti TD, Ozoren N, Jagirdar R, Inohara N, Vandenabeele P, Bertin J, Coyle A, Grant EP, Nunez G. 2006. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1beta in salmonella-infected macrophages. Nat Immunol 7: 576-82 57. Amer A, Franchi L, Kanneganti TD, Body-Malapel M, Ozoren N, Brady G, Meshinchi S, Jagirdar R, Gewirtz A, Akira S, Nunez G. 2006. Regulation of Legionella phagosome maturation and infection through flagellin and host Ipaf. J Biol Chem 281: 35217-23 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

58. Amer AO, Swanson MS. 2005. Autophagy is an immediate macrophage response to Legionella pneumophila. Cell Microbiol 7: 765-78

48 59. Amer AO, Byrne BG, Swanson MS. 2005. Macrophages rapidly transfer pathogens from lipid raft vacuoles to autophagosomes. Autophagy 1: 53-8 60. Amer AO, Valvano MA. 2002. Conserved aspartic acids are essential for the enzymic activity of the WecA protein initiating the biosynthesis of O-specific lipopolysaccharide and enterobacterial common antigen in Escherichia coli. Microbiology 148: 571-82 61. Amer AO, Swanson MS. 2002. A phagosome of one's own: a microbial guide to life in the macrophage. Curr Opin Microbiol 5: 56-61 62. Amer AO, Valvano MA. 2001. Conserved amino acid residues found in a predicted cytosolic domain of the lipopolysaccharide biosynthetic protein WecA are implicated in the recognition of UDP-N- acetylglucosamine. Microbiology 147: 3015-25 63. Amer AO, Valvano MA. 2000. The N-terminal region of the Escherichia coli WecA (Rfe) protein, containing three predicted transmembrane helices, is required for function but not for membrane insertion. J Bacteriol 182: 498-503

Complete list of publication can be found at: https://www.ncbi.nlm.nih.gov/pubmed?term=%28amer%20ao%5bAuthor%5d%29

BOOK CHAPTERS Book chapter - Biofilm, a cosy structure for Legionella pneumophila growth and persistence in the environment. Arwa Abu Khweek and Amal O. Amer. DOI: 10.5772/intechopen.89156. InTech. October 2019 - Biofilm derived Legionella pneumophila evades activation of the Nlrc4 inflammasome. Arwa Abu Khweek, Natalia S Fernández Dávila, Kyle Caution, Anwari Akhter, Basant A Abdulrahman, Mia Tazi , Hoda Hassan, Laura A Novotny , Lauren O Bakaletz and Amal O Amer. Top 10 Contributions on Microbiology: 2nd Edition. July, 2019 - Estelle Cormet-Boyaka, Kyle Caution and Amal O. Amer. Chapter title: Autophagy in Cystic Fibrosis pathogenesis and treatment. The book "Autophagy in Current Trends in Cellular Physiology and Pathology" ISBN 978-953-51-2727-7, Print ISBN 978-953-51-2726-0, InTech. November, 2016

Book editor Pulmonary Infection. Edited by Amal Amer. ISBN 978-953-51-0286-1, 136 pages, Publisher: InTech, Chapters published March 14, 2012 DOI: 10.5772/1494 2012 INTERNATIONAL, NATIONAL AND REGIONAL CONFERENCE PRESENTATIONS National and international invited talks - International Burkholderia Cepacia Working Group (IBCWG) 2020. Toronto. Canada 2020 - Cleveland Clinic. "The role of lysosomal dysfunction in cystic fibrosis pathobiology". 9/2020 - 2nd Global Conference on Infectious Diseases, Dubai, UAE. 2020 -The Talks on Infectious Diseases. San Antonio, TX. 2020 "The interplay between inflammasome activation and bacterial infection". - Keystone Symposium. Taiwan. 2019 - The North American Cystic Fibrosis Conference. Chair. Denver. Colorado. 2018 - International Burkholderia Cepacia Working Group (IBCWG) 2018. Dublin. Ireland. 2018 - The 8th International Symposium in Autophagy. Autophagy in health and disease. Japan. Nara. 2017 - Keystone Meeting. The unconventional functions of caspases. Benf. Canada. 2017 - International Congress on Science and Technology for Health Care. Inflammation Research. Antalya. Turkey. 2016 - The 39th European Cystic Fibrosis Meeting. Autophagy and Inflammation. Basel. Switzerland. 2016 - The 2015 Innate Immunity Summit. The role of microRNA and autophagy in innate immune responses to pathogens. London, England. 2015 - Transcriptomics. Epigenetic regulation of autophagy by Burkholderia cenocepacia. Orlando. USA. 2015 - Heleopolis University. The role of the inflammasome in health and disease. Cairo. Egypt. 2015 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

- Helwan University. Research abroad. Helwan. Egypt. 2015

50 - 11th International Conference on Molecular Biology and Biochemistry. The role of autophagy in health and disease. Cairo. Egypt. 2015 - The International Burkholderia Cepacia Working Group meeting 2015. How Burkholderia cenocepacia suppresses autophagy? Vancouver. Canada. 2015 - The 18th Annual Meeting for the International Burkhoderia Cepacia Working Group. MicroRNA, autophagy and Burkholderia cenocepacia. Is there a link? Nimes. France. 2014 - Cleveland clinic. Defective autophagy in cystic fibrosis. Cincinnati. Ohio. 2014 - The 8th International Meeting on Legionella pneumophila. Failure to Fuse: Legionella manipulates actin via caspases. Melbourne. Australia. 2013 - Genomics-2013. International Conference on Functional and Comparative Genomics & Pharmacogenomics” Chicago. USA. 2013 - International Burkholderia Cepacia Working Group 2013. Defective autophagy activity contributes to increased IL-1β production during Burkholderia cenocepacia infection. Ann Arbor, Michigan. 2013 - Case Western Reserve University. How caspases modulate bacterial infections. Pathobiology, Molecular Medicine, Cleveland Clinic Lerner College of Medicine, 2013 - The European Society for Clinical Investigation (ESCI). How macrophages contribute to inflammation in Burkholderia cenocepacia infected patients. Budapest. Hungary. 2012 - Midwest Microbial Pathogenesis Conference. How macrophages detect Burkholderia cepacia. Wisconsin in Milwaukee. 2012 - Zink Autophagy Meeting. How Burkholderia establish infection in cystic fibrosis patients? Blame autophagy. Mexico. 2011 - The Phagocyte Gordon Research Conference. Human pyrin detects Burkholderia cepacia in macrophages. Davidson. North Carolina. 2011 - The 45th Annual Scientific Meeting of the European Society for Clinical Investigations. Legionella pneumophila modulates the expression of the inflammasome members to establish infection in human monocytes. Crete. Greece. 2011 - ATS International Conference Thematic presentation: Host defense mechanisms against bacterial respiratory infections. Burkholderia cenocepacia O polysaccharide chain contributes to caspase-1- dependent IL-1β production in macrophages. Denver. Colorado. 2011 - The 96th Annual Meeting of The American Association of Immunologists. How caspase-7 controls intracellular infection in macrophages. Seattle, Washington. 2009 - University of Louisville. Department of Microbiology and Immunology.- The role of caspases in Legionella pneumophila infections. Louisville. Kentucky. 2008 Regional talks Research in Progress Series Dorothy Heart and Lung Research Institute. A novel use of Rapamycin in cystic fibrosis patients infected with Burkholderia cepacia. Columbus, Ohio. 2011 Nationwide Children's Hospital. Columbus. The role of the inflammasome in Burkholderia cepacia infection. Ohio, United States. 2011 - Division of Pulmonary, , Critical Care and 4th Annual Research Day. DHLRI. Legionella pneumophila: the atypical pulmonary pathogen. 2009 - Division of Pulmonary, Allergy, Critical Care and Sleep Medicine 3rd Annual Research Day. Caspase activation does not always mean cell death. DHLRI, OSU. 2008 - Immunology Seminar Series OSU. How caspases regulate the survival of intracellular pathogens. 2008 - 2nd Annual Center for Microbial Interface Biology Retreat OSU. How caspases dictate the fate of the intracellular pathogen Legionella pneumophila. 2007

Career development 1. Keeping sanity in academia workshop 2020 2. Implicit bias 2018 3. Commercialization REACH Course 2017 4. Negotiation workshop. 2017 5. Strengths-based management and supervision workshop. 2014 6. OSU Suicide prevention. 2014 7. Mission Possible workshop. 2014

Amy Elizabeth Lovett-Racke, Ph.D.

Current Position Professor Department of Microbial Infection and Immunity The Ohio State University Wexner Medical Center 460 West 12th Avenue, 684 Biomedical Research Tower Columbus, Ohio 43210 Phone: (614) 688-5647 FAX: (614) 292-7544 Email: [email protected]

Personal Information Home address: 134 Mendolin Way, Powell, OH 43065 Date of Birth: March 3, 1965 Place of Birth: Kansas City, MO Family: Married to Michael K. Racke, M.D.; six children

Education Texas A&M University, College Station, TX B.S. in Microbiology August 1983 – May 1987 University of Texas Health Science Center, Houston, TX Graduate School of Biomedical Sciences Ph.D. in Biomedical Science – Program in Immunology August 1988 – May 1993

Doctoral - Postdoctoral Training and Relevant Employment Research Technician Baylor College of Medicine – Houston, TX Department of Supervisor: Cesar Fermin, Ph.D. June 1987 – August 1988 Graduate Student University of Texas Health Science Center – Houston, TX Graduate School of Biomedical Sciences Dissertation: Mapping T-cell epitopes of the rubella virus structural proteins Mentor: Jerry S. Wolinsky, M.D. August 1988 – May 1993 Postdoctoral Fellow – Intramural Research Training Award National Institutes of Health – Bethesda, MD National Institutes of Neurological Diseases and Stroke Neuroimmunology Branch Mentor: Henry F. McFarland, M.D. July 1993 – April 1995 Postdoctoral Fellow – Lucille P. Markey Special Emphasis Pathway in Human Pathobiology Washington University School of Medicine – St. Louis, MO Department of – Division of Multiple Sclerosis Research Mentor – Michael K. Racke, M.D. Clinical Mentor – Susan McKinnon, MD July 1995 – August 1997 Post-doctoral Research Associate Washington University School of Medicine – St. Louis, MO Department of Neurology – Division of Multiple Sclerosis September 1997 – May 1999 Amy E. Lovett-Racke, PhD - 2

Instructor University of Texas Southwestern Medical Center – Dallas, TX Department of Neurology July 1999 – October 2002 Assistant Professor University of Texas Southwestern Medical Center – Dallas, TX Department of Neurology October 2002 – May 2006 Assistant Professor The Ohio State University Medical Center – Columbus, OH Department of Molecular Virology, Immunology and July 2006 – September 2010 Associate Professor The Ohio State University Medical Center – Columbus, OH Department of Molecular Virology, Immunology and Medical Genetics October 2010 – August 2011 Associate Professor The Ohio State University Medical Center – Columbus, OH Department of Microbial Infection and Immunity (primary) Department of Neuroscience (secondary) September 2011 – 2016 Professor The Ohio State University Medical Center – Columbus, OH Department of Microbial Infection and Immunity (primary) Department of Neuroscience (secondary) June 2016 – present

Grants and Fellowships Current Grants National Institutes of Health – RO1-AI140741 Principal Investigator - Defining the role of molecules unique to encephalitogenic T cells in MS Co-Investigators: Yuhong Yang, Jacob Yount, and Chad Rappleye Direct Costs: $1,000,000 September 2019 – August 2023 National Institutes of Health – R01-AG059711 Impact of senescence on T-cell function and immunotherapeutic response PI: Christin Burd Co-Investigators: Lovett-Racke, Oakes, Kendra, and Yu Direct Costs: $1,366,195 September 2018 – August 2023 National Institutes of Health - RO1 Principal Investigator – Role of miRNA dysregulation on T cell differentiation and function in MS Co-Investigator: Yuhong Yang, MD Direct Costs: $1,547,284 February 2020 – January 2025 Completed Grants National Institutes of Health Intramural Research Training Award Postdoctoral Fellowship July 1993 – April 1995 Lucille P. Markey Special Emphasis Pathway in Human Pathobiology Postdoctoral Fellowship September 1995 – August 1997 National Institutes of Health R01-AI047133 Co-investigator – Mechanism of action of Copaxone in MS Principal Investigator: Michael K. Racke, MD Direct Costs: $549,835 Amy E. Lovett-Racke, PhD - 3

September 1999 – August 2003 Yellow Rose Foundation Principal Investigator – Characterization of clonally expanded CSF T cells in multiple sclerosis Direct Costs: $200,000 January 2002 – December 2005 Once Upon A Time Foundation Principal Investigator – Identification of genes transcribed by GATA-3 in the central nervous system Direct Costs: $50,000 May 2003-May 2005 National Multiple Sclerosis Society Principal Investigator - Silencing genes in the central nervous system Direct Costs: $40,000 August 2004-July 2005 National Multiple Sclerosis Society Principal Investigator – Role of T-bet in immune-mediated demyelinating disease Harry Weaver Neuroscience Scholar Award Direct Costs: $442,184 August 2004 – September 2009 National Multiple Sclerosis Society Principal Investigator – Characterizing therapeutic targets for multiple sclerosis Direct Costs: $343,910 April 2006 – September 2009 National Multiple Sclerosis Society Co-Investigator - PPARalpha agonists as treatment for autoimmune demyelination Principal Investigator: Michael K. Racke, MD Direct Costs: $401,893 April 2007 – March 2010 National Institutes of Health R01-NS037513 Co-Investigator – T cell activation requirement in autoimmune demyelination Principal Investigator: Michael K. Racke, MD Direct Costs: $250,000 per year December 2002 – November 2011 National Institutes of Health R21-NS067383 Principal Investigator – Mining the multiple sclerosis miRNAome for disease markers Direct Costs: $275,000 September 2009 – November 2011 National Institutes of Health R21-NS072813 Co-Investigator – Encapsulated vitamin D3 vaccine for the treatment of multiple sclerosis Principal Investigator – Kristy Ainslie, PhD; Co-I – Eric Bachelder, PhD Direct Costs: $275,000 September 2011- August 2013 National Institutes of Health R21-AI092417 Co-Investigator - MicroRNA modulation of immune function in multiple sclerosis Principal Investigator – Caroline Whitacre, PhD Direct Costs: $275,000 August 2011 – July 2014 National Institutes of Health R21-NS078390 Principal Investigator - Role of dysregulated miRNA in Tregs in multiple sclerosis Co-I: Michael K. Racke, MD Direct Costs: $275,000 May 2012 – October 2014 Amy E. Lovett-Racke, PhD - 4

National Institutes of Health R21-AI095773 Co-Investigator – Regulatory myeloid cells in inflammatory disease: and targeted generation Principal Investigator – Tracey Papenfuss, DVM, PhD; Co-Is – Kristy Ainslie and Eric Bachelder Direct Costs: $275,000 August 2012 – Dec 2014 Juvenile Diabetes Research Foundation Co-Investigator - Tolerogenic nanoparticle vaccine for the treatment of type I diabetes Principal Investigator – Eric Bachelder, PhD Direct Costs: $405,000 September 2012 – August 2014 Ohio State University College of Medicine Discovery Grant Co-Investigator – Nedd4 in T cell tolerance Principal Investigator – Jian Zhang Direct Costs: $30,000 March 2014 – February 2015 National Institutes of Health R01-NS067441 Principal Investigator – Molecular analysis of genes uniquely expressed by encephalitogenic T cells Direct Costs: $984,375 July 2010 – September 2015 National Institutes of Health – RO1-AI090901 (PI moved to a new institution) Role: Co-Investigator - Nedd4 in T helper cell development and autoimmunity’ PI: Jian Zhang Direct Costs: $1,500,000 June 2016 – June 2017 Strategic Pharma-Academic Research Consortium - 15-04-B Co-Principal Investigator - Microbiome marker discovery in multiple sclerosis using metagenomic sequencing technology Co-Principal Investigators – Yanjiao Zhou and Laura Piccio Direct Costs: $280,308 January 2015 – December 2017 National Multiple Sclerosis Society – RG-5241 Principal Investigator - Defining the role of miRNA in defective Tregs in multiple sclerosis Direct Costs: $532,551 October 2014 – March 2018 National Institutes of Health - R01-NS040433 Co-Investigator – Immune regulation of neuronal injury and repair Principal Investigator – Kathryn Jones, PhD; Co-I – Virginia Sanders, PhD Direct Costs: $1,772,000 January 2013 – December 2018 National Institutes of Health – R21-NS095008 Principal Investigator - Defining the role of vitamin D in multiple sclerosis Direct Costs: $275,000 April 2016 – March 2019 TG Therapeutics – TG1101-RMS201 Co-Principal Investigator - A multi-center phase IIa dose finding study of ublituximab, a third-generation anti-CD20 monoclonal antibody, in paitents with relapsing forms of multiple sclerosis National PI: Edward Fox Site Co-Principal Investigators: Michael Racke and Amy Lovett-Racke Direct Costs: $435,910 (my budget only) May 2016 – April 2019 National Multiple Sclerosis Society – RG-1507-05275 Amy E. Lovett-Racke, PhD - 5

Principal Investigator - Neuroprotective role of vitamin D during childhood Co-I: Jonathan Godbout, PhD Direct Costs: $248,293 April 2016 – September 2019 National Institutes of Health – R21-HD086368 Principal Investigator - Neuroprotective role of vitamin D during childhood Co-Investigator: Jonathan Godbout, PhD Direct Costs: $275,000 August 2016 – July 2019

Memberships, Committees and Leadership 1994-present International Society for Neuroimmunology 2000-present Manuscript Reviewer for Journal of Immunology, Journal of Neuroimmunology, Journal of Leukocyte Biology, Acta Pharmaceutica, Arthritis and Rheumatism, PPAR Research, Proceedings of the National Academy of Science, Frontiers in Multiple Sclerosis and Neuroimmunology, Annals of Neurology, Brain Behavior and Immunity, Immunobiology, European Journal of Immunology, Brain, Nature, PLoS One 2003-2006 Institutional Animal Care and Use Committee, University of Texas Southwestern Medical Center, Dallas, TX 2004-2006 Immunology Graduate Studies Program – University of Texas Southwestern Medical Center, Dallas, TX 2004-2007 National Multiple Sclerosis Society, Pilot Project Reviewer 2004-present American Association of Immunologists 2005 Medical Research Council, Ad hoc Grant Reviewer 2006 National Institutes of Health, Clinical Neuroimmunology and Brain Tumor Study Section, Ad hoc member 2007-present Integrated Biomedical Science Graduate Program – The Ohio State University, Columbus, OH 2007-present Neuroscience Graduate Studies Program – The Ohio State University, Columbus, OH 2007-present Molecular, Cellular and Developmental Biology Graduate Program – The Ohio State University, Columbus, OH 2009 National Institutes of Health Challenge Grant Reviewer 2009-2012 OSU College of Medicine – Director of Immunology education for medical students 2009-2014 National Multiple Sclerosis Society Research Advisory Committee 2009-2011 OSU College of Medicine Faculty Council Representative for the Department of Molecular Virology, Immunology and Medical Genetics 2010-2013 OSU College of Medicine Assembly – Faculty Council Representative 2010-present Journal of Neuroimmunology – Deputy Editor 2010-present Frontiers in Neurology: Multiple Sclerosis and Neuroimmunology – Associate Editor 2010-2013 College of Medicine Lead Serve and Inspire Host Defense Block Curriculum Development Committee 2011-2012 OSU College of Medicine Lead Serve and Inspire Medical Practice and Patient Care Block Curriculum Development Committee – Lead for Week 7: Immunology and Pneumonia; Lead for Week 8: Immunology and HIV/AIDS 2011-2016 Journal of Immunology – Associate Editor 2011-2013 OSU College of Medicine Faculty Council Representative for the Department of Microbial Infection and Immunity 2011-2014 OSU College of Medicine Academic Review Board 2011-2012 OSU Department of Microbial Infection and Immunity Faculty Recruitment Committee 2012-2014 OSU College of Medicine Executive Curriculum Committee 2012-2019 OSU Department of Microbial Infection and Immunity Internal Grant Review Committee Amy E. Lovett-Racke, PhD - 6

2013 American Association of Immunologists Annual Meeting Session Chair, Honolulu, HI 2013 National Institutes of Health – Neuroscience Training Study Section, Ad hoc member 2013 National Institutes of Health – Autoimmunity Center of Excellence Study Section 2013-2014 Chair, OSU College of Medicine Academic Review Board 2014 American Association of Immunologists Annual Meeting Session Chair, Pittsburgh, PA 2014 National Institutes of Health - Immunology Fellowship (F30, F31, F32) and Academic Research Enhancement Award (R15) Study Section, Ad hoc member 2014-present Brain, Behavior and Immunity – Editorial Board member 2015-present National Multiple Sclerosis Society Immunology Pilot Project Grant Review Committee, Chair 2015-2019 NIH Hypersensitivity and Autoimmune and Immune-mediated Diseases (HAI) Study Section member 2017 NIH Brain Disorders and Clinical Neuroscience (BDCN) Special Emphasis Panel Study Section – Co-Chair 2018-present American Association of Immunologists – Abstract Programming Chair for AAI annual meetings 2019-present National Multiple Sclerosis Society – Pathways to Cure ‘Stop MS’ Committee member 2019 NIH Neuroimmunology, Neuroinflammation, and Brain Tumor Special Emphasis Panel Study Section member

Honors and Awards 1993- Intramural Research Training Award, National Institutes of Neurological 1995 Disorders and Stroke, National Institutes of Health, Bethesda, MD 1995- Lucille P. Markey Special Emphasis Pathway in Human Pathobiology 1997 Fellowship, Lucille P. Markey Foundation, Washington University School of Medicine, St. Louis, MO 2004- Harry Weaver Neuroscience Scholar Award, National Multiple 2009 Sclerosis Society 2013 OSU College of Medicine Distinguished Educator Award 2015 National Multiple Sclerosis Society Research Volunteer Hall of Fame Award 2015 OSU COM Faculty Achievement Award

Graduate and Professional Courses Taught (The Ohio State University) 2006 NeuroGSP797 – Seminars in Neuroscience, Lecturer (10%), Credit=1 2007 NeuroGSP750 – Principles of Neuroimmunology, Lecturer (10%), Credit=3 IBGP703 – Host Defense, Lecturer (10%), Credit=3 MVIMG793 – Molecular Virology, Immunology and Medical Genetics Research in Progress Seminars, Coordinator/Moderator (100%), Credit=1 2008 MCDB800/890 – Seminars in Molecular, Cellular and Developmental Biology, Moderator (100%), Credit=2 IBGP703 – Host Defense, Lecturer (10%), Credit=3 MVIMG793 – Molecular Virology, Immunology and Medical Genetics Research in Progress Seminars, Coordinator/Moderator (100%) NeuroSC705 – Neurobiology of Disease, Lecturer (7%), Credit=3 MVIMG701 – Cellular and Molecular Immunology, Lecturer (2%), Credit=5 Medical School Host Defense, Lecturer (10%) 2009 NeuroGSP750 – Principles of Neuroimmunology, Lecturer (10%), Credit=3 IBGP703 – Host Defense, Lecturer (10%), Credit=3 MVIMG793 – Molecular Virology, Immunology and Medical Genetics Research in Progress Seminars, Coordinator/Moderator (100%), Credit=1 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

NeuroSC705 – Neurobiology of Disease, Lecturer (7%), Credit=3 MVIMG701 – Cellular and Molecular Immunology, Lecturer (10%), Credit=5 MVIMG845 – History of Immunology, Lecturer (10%), Credit=3 Medical School Host Defense, Lecturer (10%)

59 Amy E. Lovett-Racke, PhD - 7

2010 IBGP703 – Host Defense, Lecturer (10%), Credit=3 MVIMG793 – Molecular Virology, Immunology and Medical Genetics Research in Progress Seminars, Coordinator/Moderator (100%), Credit=1 NeuroSC705 – Neurobiology of Disease, Lecturer (7%), Credit=3 MVIMG701 – Cellular and Molecular Immunology, Lecturer (10%), Credit=5 MVIMG845 – History of Immunology, Lecturer (10%), Credit=3 Medical School Host Defense, Lecturer (10%) Microbiology 522 – Lecturer (2%), Credit=5 US Medical License Exam Step 1 – Immunology Review Session Lecturer 2011 IBGP703 – Host Defense, Lecturer (10%), Credit=3 MVIMG793 – Molecular Virology, Immunology and Medical Genetics Research in Progress Seminars, Coordinator/Moderator (100%), Credit=1 NeuroSC705 – Neurobiology of Disease, Lecturer (7%), Credit=3 MVIMG701 – Cellular and Molecular Immunology, Lecturer (10%), Credit=5 Medical School Host Defense, Lecturer (10%) Microbiology 522 – Lecturer (2%), Credit=5 Pathology 650 – Unit coordinator and lecturer (20%), Credit=5 US Medical License Exam Step 1 – Immunology Review Session Lecturer 2012 Medical School Host Defense, Lecturer (10%) Medical School Medical Practice and Patient Care, Lead Week 7 (Immunology and Pneumonia) and Week 8 (Immunology and HIV/AIDS) and Lecturer US Medical License Exam Step 1 – Immunology Review Session Lecturer Biomsci 2892H - Mastering the Biomedical Literature II, Lecturer (8%) IBGP 7000 – Biomedical Sciences Survey, Lecturer (20%) 2013 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroGS 7887 – Seminar Topics in Neuroscience, Lecturer (2%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) NeuroSC 7500 – Principles of Neuroimmunology, Lecturer (8%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) Medical School Host Defense, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) IBGP 7972 – Senior Seminar, Coordinator (100%) 2014 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) Medical School Host Defense, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) 2015 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) NeuroSC 7500 – Principles of Neuroimmunology, Lecturer (8%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) Medical School Host Defense, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) BSGP 7080 - Grant Writing 2016 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) Medical School Host Defense, Lecturer (5%) BSGP 7000 - Biomedical Sciences Survey BSGP 7080 - Grant Writing IBGP 8800 – Signature Program Translational Science Curriculum ‘Immunology and Inflammation’ Medical School Host Defense, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) 2017 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) NeuroSC 7500 – Principles of Neuroimmunology, Lecturer (8%) Medical School Host Defense, Lecturer (5%) Amy E. Lovett-Racke, PhD - 8

BSGP 7000 – Biomedical Sciences Survey, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) IBGP 8800 – Signature Program Translational Science Curriculum ‘Immunology and Inflammation’ 2018 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) Medical School Host Defense, Lecturer (5%) BSGP 7000 – Biomedical Sciences Survey, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) IBGP 8800 – Signature Program Translational Science Curriculum ‘Immunology and Inflammation’ 2019 MVIMG 7010 – Cellular and Molecular Immunology, Lecturer (4%) NeuroSC 4100 – Basic and Clinical Foundations of Neurological Disease (3%) NeuroSC 7050 – Neurobiology of Disease, Lecturer (3%) NeuroSC 7500 – Principles of Neuroimmunology, Lecturer (8%) Medical School Host Defense, Lecturer (5%) BSGP 7000 – Biomedical Sciences Survey, Lecturer (5%) Medical School Foundations 2, Lecturer (15%) IBGP 8800 – Signature Program Translational Science Curriculum ‘Immunology and Inflammation’

Trainees High School 2015-2016 Matthew Xin – The Wellington School, Research Thesis Advisor Undergraduate 2007-2008 Rachel Zang – Boston College and The Ohio State University, Research Advisor 2012-2013 Nicholas Jarjour – Biomedical Sciences Program, Honors Thesis Advisor, The Ohio State University, Columbus, OH 2012-2013 Matt Koerbel – Biomedical Science Program, Research Advisor, The Ohio State University, Columbus, OH 2013-2016 Amanda Selhorst – Biomedical Science Program, Honors Thesis Advisor, The Ohio State University, Columbus, OH 2014 Christina Sabin - Bryan College – Summer internship at OSU. 2014-2016 Varun Shah – Neuroscience Program, Research Advisor, The Ohio State University, Columbus, OH 2015-2016 Vikas Pulluru – Neuroscience Program, Research Advisor, The Ohio State University, Columbus, OH 2015 Phyllis Donkor – Biochemistry Program, Research Advisor, The Ohio State University, Newark, OH 2015-2016 James Oberhaus – Research Advisor, The Ohio State University, Columbus, OH 2017-present Matthew Xin – Vanderbilt University Student, Research Advisor, Summer Internship at The Ohio State University, Columbus, OH 2017-2018 Gabriella Wittbrod – Research Advisor, The Ohio State University, Columbus, OH 2017-2018 Michael Mousou – Research Advisor, The Ohio State University, Columbus, OH 2018-present Calsey Graham – Research Advisor, The Ohio State University, Columbus, OH 2018-present Lizzie Sawdai – Research Advisor, The Ohio State University, Columbus, OH 2019-present Matthew Patterson – Research Advisor, The Ohio State University, Columbus, OH 2019-present Josh Deffenbaugh – Research Advisor, The Ohio State University, Columbus, OH Pre-doctoral 2002-2006 Anne Rocchini Gocke, PhD – Program in Immunology, University of Texas Southwestern Medical Center, Dallas,TX 2008-2011 David J. Huss, PhD candidate – Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH Amy E. Lovett-Racke, PhD - 9

2008-2011 Alan J. Smith, MD/PhD candidate – Integrated Biomedical Science Graduate Program, The Ohio State University, Columbus, OH 2011-2015 Mary Severin, PhD candidate – Integrated Biomedical Science Graduate Program, The Ohio State University, Columbus, OH 2011-2015 Priscilla Lee, PhD candidate – Molecular, Cellular and Development Biology Graduate Program, The Ohio State University, Columbus, OH 2013-2014 Patrick Nuro-Gyina – Discovery Prep Program, The Ohio State University, Columbus, OH Post-doctoral 2004-2006 Petra Cravens, PhD – University of Texas Southwestern Medical Center, Dallas, TX 2006-2011 Yuhong Yang, MD – The Ohio State University Medical Center, Columbus, OH 2008-2011 Mireia Guerau de Arellano Vilanova, PhD – The Ohio State University Medical Center, Columbus, OH 2016-2017 Priscilla Lee, PhD – The Ohio State University, Columbus, OH 2016-2019 Sara Gombash Lampe – PhD - The Ohio State University, Columbus, OH

Invited National and International Presentations (selected) 1. September 2004 - Silencing T-bet prevents autoimmune demyelinating disease – 7th Congress of the International Society for Neuroimmunology, Venice, Italy. 2. May 2005 – Therapeutic benefit of silencing T-bet in autoimmune demyelinating disease – National Institutes of Neurological Disorders and Stroke, Bethesda, MD. 3. June 2005 – Therapeutic potential of siRNA for the treatment of multiple sclerosis – Consortium of Multiple Sclerosis Centers Annual Meeting, Orlando, FL 4. October 2006 – T-bet regulates the fate of Th1 and Th17 lymphocytes in autoimmunity – Americas Committee for the Treatment and Research in Multiple Sclerosis Meeting, Chicago, IL. 5. March 2007 – Therapeutic potential of siRNA for multiple sclerosis – 8th Annual Frontiers in Neuroscience Research Day, Charleston, SC. 6. April 2008 – Silencing T-bet suppresses Th1 and Th17 cell differentiation – Experimental Biology 2008, San Diego, CA. 7. October 2008 - Role of T-bet in regulating autoimmune Th1 and Th17 cells – 9th Congress of the International Society for Neuroimmunology, Fort Worth, TX. 8. November 2008 – Defining the role of T-bet in encephalitogenic Th1 and Th17 cells – Tykeson Fellows Conference on Multiple Sclerosis, Chicago, IL. 9. April 2010 – T-bet or not T-bet: That is the question in autoimmunity – University of Texas at San Antonio, TX. 10. May 2010 – Identifying microRNA differentially expressed in T cells of MS patients – American Association of Immunologists Annual Meeting, Baltimore, MD. 11. August 2010 – Therapeutic potential of small RNAs in multiple sclerosis, Kyoto Neuroimmunology Conference, Kyoto, Japan. 12. January 2011 – Small RNAs…big results: potential roles of siRNA and miRNA in multiple sclerosis, University of Texas Southwestern Medical Center, Dallas, TX. 13. October 2011 – Defining the role of miRNA in susceptibility to multiple sclerosis, John Hopkins University, Baltimore, MD. 14. January 2012 – Small RNAs: Susceptibility factors and potential for multiple sclerosis, University of Michigan, Ann Arbor, MI. 15. June 2012 – Role of miRNA in T cells in susceptibility to multiple sclerosis, Americas Committee for the Treatment and Research in Multiple Sclerosis meeting, San Diego, CA. 16. October 2012 – Potential of small interfering RNAs as therapies for multiple sclerosis, University of Toledo, Toledo, OH. 17. March 2014 – Are miRNAs susceptibility factors for multiple sclerosis?, University of Connecticut Health Center, Farmington, CT. 18. April 2015 – Contribution of EAE to understanding and treating multiple sclerosis. National Institutes of Health 40 Years of Neuroimmunology meeting, Warrenton, VA. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

19. August 2015 – Are miRNAs responsible for the Treg defects observed in multiple sclerosis? World Congress on Inflammation, Boston, MA. 20. July 2016 - Intersection between Neuroimmunology and Neuroendocrinology: Application to Multiple Sclerosis, FASEB Summer Conference on Translational Neuroimmunology, Big Sky, MT.

63 Amy E. Lovett-Racke, PhD - 10

21. July 2016 - RNA-based Approaches for the Treatment of Multiple Sclerosis, FASEB Summer Conference on Translational Neuroimmunology, Big Sky, MT. 22. January 2017 – microRNAs: Potential Risks Factors for the Development of Multiple Sclerosis. Immunology Seminar Series, University of Louisville, KY. 23. May 2017 – miRNAs as Potential Risks Factors for Multiple Sclerosis. Whitaker Research Track Invited Speaker. Consortium of Multiple Sclerosis Centers, New Orleans, LA. 24. May 2017 – Rapid and Robust B Cell Depletion in Preliminary Results of a Phase 2 Study of Ublituximab, a Novel Glycoengineered anti-CD20 mAb, in RMS. Consortium of Multiple Sclerosis Centers, New Orleans, LA. 25. May 2018 – Immune Consequences of Current MS Therapies. Consortium of Multiple Sclerosis Centers, Nashville, TN.

Publications Book Chapters 1. Arredondo LR, Lovett-Racke AE and Racke MK. (2002) Experimental autoimmune encephalomyelitis. Multiple Sclerosis: Immunology and Pathology, pg 75-99. 2. Lovett-Racke AE and Racke MK. (2004) The role of growth factors in experimental autoimmune encephalomyelitis. Experimental Models of Multiple Sclerosis (eds. Lavi E. and Constantinescu C.S.), New York, NY: Springer, pg 343-362. 3. Racke MK, Ratts RB, Stuart RW, Deng C and Lovett-Racke AE. (2004) The role of costimulation in experimental autoimmune encephalomyelitis. Experimental Models of Multiple Sclerosis (eds. Lavi E and Constantinescu CS), New York, NY: Springer, pg 471-490. 4. Lovett-Racke AE, Gocke AR and Cravens PD. (2007) The basics of cellular and molecular immunology. Neuroimmunology in Clinical Practice (eds. Kalman B and Brannagan T). Hoboken, NJ: Wiley Blackwell Publishing, pg 1-16. 5. Lovett-Racke AE. (2009) Therapeutic potential of small interfering RNA for brain diseases. Therapeutic Ribonucleic Acids for Brain Tumors (eds. VA Erdmann, G Reifenberger, J Barciszewski) New York, NY: Springer, pg 275-296. 6. Lovett-Racke AE and Racke MK. (2013) T-bet as a key transcription factor in autoimmune inflammatory demyelination. Multiple Sclerosis Immunology – A Foundation for Current and Future Treatments. New York, NY:Springer. 7. Lovett-Racke AE and Racke MK. (2018) Role of IL-12/IL-23 in the Pathogenesis of Multiple Sclerosis. Neuroinflammation – 2nd Edition. Elsevier. Amersterdam, The Netherlands. Journal Articles 8. Fermin CD, Lovett AE, Igarashi M and Dunner K. (1990) Immunochemistry and histochemistry of the inner ear gelatinous membranes and statoconia of the chick (Gallus domesticus). Acta Anatomica 138(1):75-83. 9. Wolinsky JS, McCarthy M, Allen-Canady O, Moore WT, Jin R, Cao SN, Lovett AE and Simmons, D. (1991) Monoclonal antibody-defined map of expressed rubella virus protein domains. Journal of Virology 65(1):3986-3994. 10. Lovett AE, McCarthy M and Wolinsky JS. (1993) Mapping cell-mediated immunodominant domains of the rubella virus structural proteins using recombinant proteins and synthetic peptides. Journal of General Virology 74(3):445-452. 11. McCarthy M, Lovett AE, Kerman RH, Overstreet A and Wolinsky JS. (1993) Immunodominant T- cell epitopes of rubella virus structural proteins and synthetic peptides. Journal of Virology 67(2):673-681. 12. Wolinsky JS, Sukholutsky E, Moore WT, Lovett AE, McCarthy M and Adame B. (1993) An antibody and synthetic peptide defined rubella virus E1 glycoprotein neutralization domain. Journal of Virology 67(2):961-968. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

13. Lovett AE, Hahn CS, Rice CM, Frey TK and Wolinsky JS. (1993) Rubella-specific CTL responses: identification of the capsid as a target of MHC class I-restricted lysis and definition of two epitopes. Journal of Virology 67(10):5849-5858. 14. Lovett-Racke AE, Martin R, McFarland HF, Racke MK and Utz U. (1997) Longitudinal study of myelin basic protein-specific T cell receptors during the course of multiple sclerosis. Journal of Neuroimmunology 78(1-2):162-171. 15. Lovett-Racke AE, Trotter JL, Lauber J, Perrin PJ, June CH and Racke MK. (1998) Decreased dependence of myelin basic protein-reactive T cells on CD28-mediated costimulation in multiple

65 Amy E. Lovett-Racke, PhD - 11 sclerosis patients: a marker of activation/memory T cells. Journal of Clinical Investigation 101(4):725-730 (Rapid Publication). PMC508618 16. Racke MK and Lovett-Racke AE. (1998) Bystander suppression in experimental autoimmune encephalomyelitis: where and how does it occur? Research in Immunology 149(9):820-827. 17. Lovett-Racke AE, Bittner P, Cross AH, Carlino JA and Racke MK. (1998) Regulation of experimental autoimmune encephalomyelitis with insulin-like growth factor (IGF-1) and IGF-1/IGF binding protein-3 complex (IGF-1/IGFBP3). Journal of Clinical Investigation 101(8):1797-1804. PMC508763 18. Perrin PJ, Lovett-Racke AE, Phillips SM and Racke MK. (1999) Differential requirements of naïve and memory T cells for CD28 costimulation in autoimmune pathogenesis. Histology and Histopathology 14(4):1269-1276. 19. Ratts RB, Arredondo LR, Bittner P, Perrin PJ, Lovett-Racke AE and Racke MK. (1999) The role of CTLA-4 in tolerance induction and T cell differentiation in experimental autoimmune encephalomyelitis: intravenous antigen administration. International Immunology 11(12):1889- 1895. 20. Ratts RB, Arredondo LR, Bittner P, Perrin PJ, Lovett-Racke AE and Racke MK. (1999) The role of CTLA-4 in tolerance induction and T cell differentiation in experimental autoimmune encephalomyelitis: intraperitoneal antigen administration. International Immunology 11(12):1881- 1888. 21. Peters TR, Bodine DM, McDonagh KT, Lovett-Racke AE, McFarland HF, McFarlin DE, Nienhuis AW and Racke MK. (2000) Retrovirus mediated gene transfer of the self antigen MBP into the bone marrow of B10.PL mice does not confer resistance to experimental autoimmune encephalomyelitis. Journal of Neuroimmunology 103(1):51- 62. 22. Racke MK, Ratts RB, Arredondo L, Perrin PJ and Lovett-Racke AE. (2000) The role of costimulation in autoimmune demyelination. Journal of Neuroimmunology 107(2):205- 215. 23. Lovett-Racke AE, Smith ME, Arredondo LR, Bittner PS, Ratts RB, Shive CL, Forsthuber TG and Racke MK. (2000) Developmentally regulated expression of Th2 cytokines in the brain. Brain Research 870(1-2):27-35. 24. Arredondo LR, Deng C, Ratts RB, Lovett-Racke AE, Holtzman DM and Racke MK. (2001 Role of nerve growth factor in experimental autoimmune encephalomyelitis. European Journal of Immunology 31(2):625-633. 25. Racke MK, Lovett-Racke AE, Karandikar NJ, Wilson B and Monson NL. (2001) Myelin- reactive T cells and their regulation in multiple sclerosis. NeuroScience News 4:29-38. 26. Frohman EM, Monson NL, Lovett-Racke AE and Racke MK. (2001) Autonomic regulation of neuroimmunological responses: Implications for multiple sclerosis. Journal of Clinical Immunology 21(2):61-73. 27. Lovett-Racke AE and Racke MK. (2002) Retinoic acid promotes the development of Th2-like human myelin basic protein-reactive T cells. Cellular Immunology 215(1):5460- 5466. 28. Karandikar, N.J., Crawford, M.P., Ratts, Robert B., Brenchley, J.M., Ambrozak, D.R., Lovett- Racke AE, Frohman EM, Stastney P, Douek DC, Koup RA and Racke MK. (2002) Glatiramer acetate (Copaxone®) therapy induces CD8+ T cell responses in patients with multiple sclerosis. Journal of Clinical Investigation 109(5):641-649. PMC150895 29. Diab A, Deng C, Smith JD, Hussain RZ, Phanavanh B, Lovett-Racke AE, Drew PD and Racke MK. (2002) Peroxisone proliferator-activated receptor-γ agonist 15-deoxy-Δ12,14- PGJ2 ameliorates experimental autoimmune encephalomyelitis. Journal of Immunology 168(5):2508-2515. 30. Deng C, Minguela A, Hussain RZ, Lovett-Racke AE, Radu C, Ward ES and Racke MK. (2002) Expression of the tyrosine phosphatase SRC homology 2 domain-containing protein tyrosine phosphatase 1 determines T cell activation threshold and severity of Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

experimental autoimmune encephalomyelitis. Journal of Immunology 168(9):4511- 4518. 31. Deng C, Wu B, Yang H, Hussain RZ, Lovett-Racke AE, Christadoss P and Racke MK (2003) Decreased expression of Scr homology 2 domain-containing protein tyrosine phosphatase 1 reduces T cell activation threshold but not the severity of experimental autoimmune myasthenia gravis. Journal of Neuroimmunology 138(1-2):76-82. 32. Diab A, Hussain RZ, Lovett-Racke AE, Chavis JA, Drew PD and Racke MK. (2004) Ligands for the peroxisome proliferators-activated receptor-γ and retinoind X receptor exert additive effects on experimental autoimmune encephalomyelitis. Journal of Neuroimmunology 148(1-2):116-126.

67 Amy E. Lovett-Racke, PhD - 12

33. Lovett-Racke AE, Hussain RZ, Northrop S, Choy J, Rocchini A, Matthes L, Chavis JA, Diab A, Drew PD and Racke MK (2004) Peroxisome proliferators-activated receptor α agonists as therapy for autoimmune disease. Journal of Immunology 172(9):5790-5798. 34. Lovett-Racke AE, Rocchini AE, Choy J, Northrop SC, Hussain RZ, Sikder D and Racke MK. (2004) Silencing T-bet defines a critical role in the differentiation of autoreactive T lymphocytes. Immunity 21(5):719-731. 35. Racke MK, Hu W and Lovett-Racke AE. (2005) PTX Cruiser: Driving Autoimmunity via TLR4. Trends in Immunology 26(6):289-291. 36. Frohman EM, Brannon K, Frohman TC, Giesser B, Langer-Gould A, Levalds C, Lovett- Racke AE, Treadaway K, Waubant E, Winslow H and Wuermser LA. (2005) Multiple sclerosis and the female patient, Part 2: Clinical manifestations and symptomatic treatments strategies. Female Patient 30:45-50. 37. Frohman EM, Brannon K, Frohman TC, Giesser B, Langer-Gould A, Levalds C, Lovett- Racke AE, Treadaway K, Waubant E, Winslow H and Wuermser LA. (2005) Multiple sclerosis and the female patient, Part 1: Characterizing the disease process. Female Patient 30:41-46. 38. Lovett-Racke AE, Cravens PD, Gocke AR, Racke MK and Stuve O. (2005) Therapeutic potential of small interfering RNA for central nervous system diseases. Archives of Neurology 62(12):1810- 1813. 39. Muir MT, Lovett-Racke AE and Racke MK. (2005) Novel therapeutic strategies targeting the pathogenic T-cells in multiple sclerosis. Expert Review of Clinical Immunology 1:345-355. 40. Racke MK, Gocke AR, Muir M, Diab A, Drew PD and Lovett-Racke AE. (2006) Nuclear receptors and autoimmune disease: The potential of PPAR agonists to treat multiple sclerosis. Journal of Nutrition 136(3):700-703. PMC2819754 41. Hansen BS, Hussain RZ, Lovett-Racke AE, Thomas JA and Racke MK. (2006) Multiple toll-like receptor agonists act as potent adjuvants in the induction of autoimmunity. Journal of Neuroimmunology 172(1-2):94-103. 42. Lovett-Racke AE and Racke MK. (2006) Epstein-Barr virus and multiple sclerosis. Archives of Neurology 63(6):810-811. 43. Ratts RB, Lovett-Racke AE, Choy J, Northrop SC, Hussain RZ, Karandikar NJ and Racke MK. (2006) CD28-CD57+ T cells predominate in chronic CD8 responses to glatiramer acetate. Journal of Neuroimmunology 178(1-2):117-129. 44. Ratts RB, Karandikar NJ, Hussain RZ, Choy J, Northrop SC, Lovett-Racke AE and Racke MK. (2006) Phenotypic characterization of autoreactive T cells in multiple sclerosis. Journal of Neuroimmunology 178(1-2):100-110. 45. Stuve O, Marra CM, Cravens PD, Singh MP, Hu W, Lovett-Racke AE, Monson NL, Phillips JT, Tervaert JW, Nash RA, Hartung HP, Kiesseier BC, Racke MK, Frohman EM and Hemmer B. (2007) Potential risk of progressive multifocal leukoencephalopathy with natalizumab therapy: possible interventions. Archives of Neurology 64(2):169-176. 46. Gocke AR, Northrop SC, Ben L, Hussain RZ, Cravens PD, Racke MK and Lovett-Racke AE. (2007) T-bet regulates the fate of TH1 and TH17 lymphocytes in autoimmunity. Journal of Immunology 178(3):1341-1348. 47. Yang Y, Ratts RB, Hussain RZ, Northrop SC, Ben L., Lovett-Racke AE and Racke MK. (2007) CD28:B7 interaction is necessary for the protective effect of T cell vaccination in EAE. European Journal Immunology 37(7):2032-42. 48. Stuart R, Lovett-Racke AE, Frohman EM, Hawker K and Racke MK. (2007) Genetic analysis of the exon 1 position 49 CD152 dimorphism in multiple sclerosis. Journal of Neuroimmunology 191(1-2):45-50. PMC2812429 49. Harp CT, Lovett-Racke AE, Racke MK, Frohman EM and Monson NL. (2008) Impact of myelin- specific antigen presenting B cells on T cell activation in multiple sclerosis. Clinical Immunology 128(3):382-391. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

50. Novack J, Lovett-Racke AE and Racke MK. (2008) Monoclonal therapies and neurological disorders. Archives of Neurology 65(9):1162-1165. 51. Yang Y, Gocke AR, Lovett-Racke AE, Drew PD and Racke MK. (2008) PPAR regulation of the immune response and autoimmune encephalomyelitis. PPAR Research 2008:546753. PMC2465123 52. Gocke AR, Hussain RZ. Ben L, Drew PD, Lovett-Racke AE and Racke MK. (2009) Transcription modulation of the immune response by PPARα agonists amerliorates autoimmune encephomyelitis. Journal of Immunology 182(7):4479-4487. PMC2959196

69 Amy E. Lovett-Racke, PhD - 13

53. Yang Y, Weiner J, Liu Y, Smith AJ, Huss DJ, Winger R, Peng H, Cravens PD, Racke MK and Lovett-Racke AE. (2009) T-bet is essential for encephalitogenicity of both Th1 and Th17 cells. Journal of Experimental Medicine 206(7):1549-1564. PMC2715092 54. Racke MK, Lovett-Racke AE and Karandikar NJ. (2010) The mechanism of action of glatiramer acetate treatment in multiple sclerosis. Neurology 74 (Suppl 1):S25-30. 55. Hu W, Nessler S, Hemmer B, Eagar TN, Kane LP, Leliveld SR, Müller-Schiffmann A, Gocke AR, Lovett-Racke A, Ben L, Hussain R, Breil A, Elliott JL, Puthaparthi K, Cravens PD, Singh MP, Petsch B, Stitz L, Racke MK, Korth C and Stüve O. (2010) Pharmacological PrP-silencing accelerates CNS autoimmune disease via TCR signaling. Brain 133(Pt 2):375-388. PMC2822628 56. Yang Y, Liu Y, Wei P, Peng H, Winger R, Hussain RZ, Ben L., Cravens PD, Gocke AR, Puttaparthi K, Racke MK, McTigue DM and Lovett-Racke AE. (2010) Silencing Nogo- A promotes functional recovery in demyelinating disease. Annals of Neurology 67(4):498-507. PMC2929680 57. Huss D, Winger RC, Peng H, Yang Y, Racke MK and Lovett-Racke AE. (2010) TGF-β enhances effector Th1 cell activation and promotes self-regulation via IL-10. Journal of Immunology 184:5628-5636. PMC3804066 58. Harp CT, Ireland S, Davis LS, Remington G, Cassidy B, Cravens PD, Stuve O, Lovett- Racke AE, Eagar TN, Greenbery BM, Racke MK, Cowell LG, Karandikar NJ, Frohman EM and Monson NL. (2010) Memory B cells from a subset of treatment-naïve relapsing- remitting multiple sclerosis patients elicit CD4(+) T-cell proliferation and IFN-γ production in response to myelin basic protein and myelin oligodendrocyte glycoprotein. European Journal of Immunology 40:2942-2956. PMC3072802 59. Guerau-de-Arellano M, Lovett-Racke AE and Racke MK. (2010) miRNAs in multiple sclerosis: regulating the regulators. Journal of Neuroimmunology 229:3-4. PMC2991561 60. Yang Y, Lovett-Racke AE and Racke MK. (2010) Regulation of immune responses and autoimmune encephalomyelitis by PPARs. PPAR Research 2010:104705. PMC3014678 61. Racke MK and Lovett-Racke AE. (2011) Glatiramer acetate treatment of multiple sclerosis: an immunological perspective. Journal of Immunololgy 186:1887-1890. 62. Lovett-Racke AE, Yang Y and Racke MK. (2011) Th1 versus Th17: Are T cell cytokines relevant in multiple sclerosis? Biochim Biophys Acta 1812:246-251. PMC3004998 63. Smith AJ, Lui Y, Peng H, Beers R, Racke MK and Lovett-Racke AE. (2011) Comparison of a classical Th1 bacteria versus a Th17 bacteria as adjuvant in the induction of experimental autoimmune encephalomyelitis. Journal of Neuroimmunology 237:33-38. PMC3156447 64. Huss DJ, Winger R, Cox GM, Guerau-de-Arellano M, Yang Y, Racke MK, Lovett-Racke AE. (2011) TGF-β signaling via smad4 drives IL-10 production in effector Th1 cells and reduces host cell recruitment in EAE. European Journal of Immunology 41:2987-2996. PMC3478765 65. Ghoreschi K, Weigert C, Deng C, Bruck J, Hussain RZ, Gocke AR, Respa A, Valtcheva N, Feil S, Feil R, Rupec RA, Lovett-Racke AE, Dringen R, Racke MK and Rocken M. (2011) Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. Journal of Experimental Medicine 208:2291-2303. PMC3201195 66. Almad A, Lash AT, Pei P, Lovett-Racke AE and McTigue DM. (2011) The PPAR alpha agonist gemfibrozil is an ineffective treatment for spinal cord injured mice. Experimental Neurology 232:309-317. 67. Guerau-de-Arellano M, Smith KM, Godlewski J, Liu Y, Winger R, Lawler SE, Whitacre CC, Racke MK and Lovett-Racke AE. (2011) miRNA dysregulation in multiple sclerosis favors pro- inflammatory autoimmunity. Brain 134:3578-89. PMC3235556 68. Lovett-Racke AE. (2012) Research Highlights: Highlights from the latest articles in optimizing vaccine development. Immunotherapy4:469-71. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

69. Guerau-de-Arellano M, Alder H, Ozer HG, Lovett-Racke A and Racke MK. (2012) miRNA profiling for biomarker discovery in multiple sclerosis: from microarray to deep sequencing. Journal of Neuroimmunology 248:32-9. PMC3288464 70. Jukkola P, Lovett-Racke AE, Zamvil SS and Gu C. (2012) K+ channel alterations in the progression of experimental autoimmune encephalomyelitis. Neurobiology of Disease 47:280-93. PMC3367054 71. Smith KM, Guerau-de-Arellano M, Bottoni A, Williams JL, Satoskar AR, Croce CM, Racke MK, Lovett-Racke AE and Whitacre CC. (2012) MicroRNA-29b controls inflammatory bias of CD4+ T cells in multiple sclerosis through a negative feedback loop targeting T-bet and IFNγ. Journal of Immunology189:1567-76. PMC3779520

71 Amy E. Lovett-Racke, PhD - 14

72. Peng H, Guerau-de-Arellano M, Mehta VB, Yang Y, Huss DJ, Papenfuss TL, Lovett- Racke AE and Racke MK. (2012) Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor κB (NF-κB) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. Journal of Biological Chemistry 287:28017-26. PMC3431702 73. Fenn AM, Smith KM, Lovett-Racke AE, Guerau-de-Arellano M, Whitacre CC, Godbout JP. (2013) Increased micro-RNA 29b in the aged brain correlates with the reduction of insulin-like growth factor-1 and fractalkine ligand. Neurobiology of Aging. 34(12):2748- 58. 74. Peine KJ, Guerau-de-Arellano M, Lee P, Kanthamneni N, Severin M, Probst GD, Peng H, Yang Y, Vangundy Z, Papenfuss TL, Lovett-Racke AE, Bachelder EM, Ainslie KM. (2014) Treatment of Experimental Autoimmune Encephalomyelitis by Codelivery of Disease Associated Peptide and Dexamethasone in Acetalated Dextran Microparticles. Molecular Pharmacology 11:828-35. PMC3993881 75. Racke MK, Yang Y, Lovett-Racke AE. (2014) Is T-bet a potential therapeutic target in multiple sclerosis? Journal of Interferon and Cytokine Research 34:623-32. 76. Yang Y, Winger R, Lee PW, Nuro-Gyina PK, Minc A, Larson M, Liu Y, Pei W, Racke MK, Lovett- Racke AE. (2015) Impact of suppressing RORγt in ameliorating central nervous system autoimmunity. Clinical and Experimental Immunology 179:108-118. 77. Lee PW, Yang Y, Racke MK, Lovett-Racke AE. (2015) Analysis of TGFβ1 and TGFβ3 as a regulator of encephalitogenic Th17 cells: implications for multiple sclerosis. Brain Behavior and Immunity 46:44-49. 78. Ding X,Yan Y, Li X, Li K, Ciric B, Yang J, Zhang Y, Wu S, Xu H, Wanjun Chen W, Lovett- Racke AE, Zhang GX, Rostami A. (2015) Silencing IFN-γ binding/signaling in astrocytes versus microglia leads to opposite effects on central nervous system autoimmunity. J Immunol 194:4251-64. 79. Jones K, Lovett-Racke AE, Walker CL and Sanders VM. (2015) CD4+ T cells and neuroprotection: relevance to motoneuron injury and disease. J Neuroimmune Pharmacol 2015;10:587-94. 80. Guerau-de-Arellano M, Liu Y, Meisen WH, Pitt D, Racke MK and Lovett-Racke AE. (2015) Analysis of miRNA in normal appearing white matter to identify altered CNS pathways in multiple sclerosis. J Autoimmune Disorders 1:6. 81. Severin ME, Lee PW, Liu Y, Selhorst AJ, Gormley MG, Pei W, Guerau-de-Arellano M, Racke MK and Lovett-Racke AE. (2016) MicroRNAs Targeting TGFβ Signaling Underlie the Regulatory T Cell Defect in Multiple Sclerosis. Brain 139:1747-61. 82. Tazi MF, Dakhlallah DA, Caution K, Gerber MM, Chang SW, Khalil H, Kopp B, Ahmed A, Krause K, Davis I, Marsh C, Lovett-Racke AE, Schlesinger LS, Cormet-Boyaka E, Amer AO. (2016) Elevated Mirc1/Mir17-92 cluster expression negatively regulates autophagy and CFTR (cystic fibrosis transmembrane conductance regulator) function in CF macrophages. Autophagy. 2016;12:2026-37. 83. Lovett-Racke AE. (2016) Contribution of EAE to understanding and treating multiple sclerosis. J Neuroimmunol. 2016 Aug 12. pii: S0165-5728(16)30195-3. doi: 10.1016/j.jneuroim.2016.08.013. 84. Nuro-Gyina P, Rieser EL, Granitto MC, Pei W, Liu Y, Lee PW, Zhang J, Lovett-Racke AE, Racke MK and Yang Y. (2016) Regulation of effector function of CNS autoreactive CD4 T cells through inhibitory receptors and IL-7Rα. J Neuroinflammation. 13:302. 85. Young NA, Valiente GR, Hampton JM, Wu LC, Burd CJ, Willis WL, Bruss M, Steigelman H, Gotsatsenko M, Amici RA, Severin M, Claverie LM, Guerau-de-Arellano M, Lovett- Racke A, Ardoin S and Jarjour WN. Estrogen-regulated STAT1 activation promotes TLR8 expression to facilitate signaling via microRNA-21 in systemic lupus erythematosus. Clin Immunol 2017;176:12- 22. 86. Lee PW, Smith AJ, Yang Y, Selhorst AJ, Liu Y, Racke MK and Lovett-Racke AE. IL- 23R- activated STAT3/STAT4 is essential for Th1/Th17-mediated CNS autoimmunity. JCI Insight 2017;2: pii: 91663 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

87. Lee PW, Severin ME and Lovett-Racke AE. TGF-β regulation of T cells in multiple sclerosis. Eur J Immunol 2017;47:446-453. 88. Cossarizza A, et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies. Eur J Immunol. 2017;47:1584-1797. 89. Jukkola P, Gu Y, Lovett-Racke AE and Gu C. Suppression of inflammatory demyelination and axon degeneration through inhibiting Kv3 channels. Front Mol Neurosci 2017;10:344.

73 Amy E. Lovett-Racke, PhD - 15

90. Lee PW, Xin M, Wei P, Yang Y, and Lovett-Racke AE. IL-3 is a marker of encephalitogenic T cells, but not essential for CNS autoimmunity. Front Immunol. 2018;9:1255, doi: 10.3389/fimmu.2018.01255. 91. Aqel SI, Granitto MC, Nuro-Gyina PK, Pei W, Liu Y, Lovett-Racke AE, Racke MK, and Yang Y. Distinct roles for Blimp-1 in autoreactive CD4 T cells during priming and effector phase of autoimmune encephalomyelitis. J Neuroimmunol. 2018 Dec 15;325:20-28. 92. Aqel SI, Kraus EE, Jena N, Kumari V, Granitto MC, Mao L, Farinas MF, Zhao EY, Perottino G, Pei W, Lovett-Racke AE, Racke MK, Fuchs JR, Li C, and Yang Y. Novel small molecule IL-6 inhibitor suppresses autoreactive Th17 development and promotes Treg development. Clin Exp Immunol. 2019;196(2):215-225. 93. Lovett-Racke AE, Gormley M, Liu Y, Yang Y, Graham C, Wray S, Racke MK, Shubin R, Twyman C, Alvarez E, Bass A, Eubanks JL, and Fox E. B cell depletion with ublituximab reshapes the T cell profile in multiple sclerosis patients. J Neuroimmunol 2019;332:187-197. 94. Cossarizza A, et al., Guidelines for the use of flow cytometry and cell sorting in immunological studies (second edition). Eur J Immunol. 2019 (10):1457-1973. 95. Lee PW, Selhorst A, Gombash Lampe S, Liu Y, Yang Y, and Lovett-Racke AE. Neuron- specific vitamin D signaling attenuates microglia activation and CNS autoimmunity. Front Neurol 2020 Jan 31;11:19. doi: 10.3389/fneur.2020.00019. William P. Lafuse Curriculum Vitae and Bibliography May 19, 2020

Biographical Information Born: November 1, 1950 - Liberty, Indiana

Educ ation Year Degree Institution 1973 BS (Biology, with honors) Purdue University 1978 PhD Johns Hopkins University Major: Biology Minor: Biochemistry

Professional Experience:

Year Position Institution 1978- Postdoctoral Department of Immunology, in laboratory of Dr. Chella 1982 research fellow S. David, Mayo Clinic, Rochester, MN. 1982- Research Associate Department of Immunology, Mayo Clinic, Rochester, 1983 MN. 1983- Associate Consultant Department of Immunology, Mayo Clinic, Rochester, 1986 MN. 1986- Assistant Professor Department of Medical Microbiology and Immunology, 1992 Ohio State University, Columbus, OH. 1992- Associate Professor Department of Molecular Virology, Immunology, and 2002 Medical Genetics, Ohio State University, Columbus, OH. 2002- Professor Departments of Molecular Virology, Immunology, and 2011 Medical Genetics, Ohio State University, Columbus, OH. 2011- Professor Department of Microbial Infection and Immunity, present Ohio State University, Columbus, OH. 1996- Member Institute of Behavioral Medicine Research, 2005 Ohio State University, Columbus, OH. 2002- Member Center for Microbial Interface Biology 2017 Ohio State University, Columbus, OH. 202017- Member OSU infectious Diseases Institute present Ohio State University, Columbus, OH.

Honors 1979-1980 American Cancer Postdoctoral Fellowship 1999 Simson Faculty Research Award for outstanding contributions in research by a member of the OSU College of Medicine Faculty

Memberships in Professional Societies American Association of Immunologists Society for Leukocyte Biology

Service Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Departmental Committees: 1986-1991, 1993-2005 Immunology Advisory Committee 1989-1990 Immunology Faculty Search Committee 1992 Faculty Work Load Policy Committee 1993-1994 Molecular Virologist Faculty Search Committee 1987-1998, 2002-present Graduate Studies Committee (member) 1993-1996 Graduate Studies Committee (Chairman)

76 1992-1997 Promotion and Tenure Committee 2001 PNI Faculty Search Committee 2012-present MII Department Education Committee 2012-2015 MII Department Professional Development and Mentoring Committee College Committees: 1987-1989 Molecular Biology Advisory Committee 1988-1999 Laboratory Animal Facilities and Finance Advisory Committee 1993-1994 Dept. of Medical Microbiology and Immunology Chair Search Committee 1993-1996; 2001- 2003 Graduate Education Committee 1998-1999, 2013-2016 Alternate, Faculty Council 2011-present Biomedical Science Graduate Studies Committee 2019 Analytical Flow Cytometry Core Users Committee

University Committees: 2014-2017 Institutional Biosafety Committee Student Organizations Judge of Bennet, ISCABER, and Medical Scientist student presentations National Service Feb 1989, Jun 1990 NIH Special Study Section for Small Business Innovative Research Grants June 1994 NIH NIAID Special Study Section for Review of grants in response to an RFA on Neonatal Immunity.

Professional Activities Journal Reviewer: J. Immunology, J. Leukocyte Biology, Brain, Behavior and Immunity, Blood, J. Experimental Medicine, Cellular Immunology, Clinical and Experimental Immunology, American Journal of Physiology-Lung, Cellular, and Molecular Physiology, FEMD Microbiology Letters, PLOS One, PLOS Pathogen International Service: Reviewer of Wellcome Trust Postdoctoral Fellowships Reviewed grant applications for: The Welcome Trust The French Muscluar Dystrophy Association Science and Technology Center in Ukraine (US Civilian Research and Development Foundation)

Teaching 1987-2012 MVIMG701 and Micro 701 Molecular and Cellular Immunology, Course Director 2013-present CBG7010 (MEDMCIM 7010) and Micro 7010 Molecular and Cellular Immunology, Course Director

1987, 1989, 1991, 1993, 1995, 1997, 1999, 2001, 2003, 2005, 2007 MMI833 Current Immunology Methods, Course Director 2002-2010 IBG Biology of Host Disease III, Module 10 Host Defense. Lectures on Innate Immunity and M. tuberculosis Graduate Programs: Biomedical Graduate program Graduate Research Students Supervised: • Ming Ling Kuo, Med. Micro (MS, 1988) • Dennis Lanning, Med.Micro (PhD, 1995) • Jaeho Jung, Med.Micro.(PhD, 1996) • Shabbir Hussain, Med. Micro. (PhD, 1999) • Chad Boomershine, MVIMG/MSP (PhD, 2000) • Fatou Sow, Microbiology (PhD, 2007) • Kris Van Zandt, Microbiology (PhD, 2007)

Postdoctoral Fellows Supervised: • Yue Wang, PhD June 2002-February 2004 Graduate Student Committees: Past: • Jyotsna Nath MS 1988, Microbiology • Linda Vespa PhD 1989, Microbiology • Elizabeth Fitzpatrick PhD 1990, Med.Micro-Immuno. • Yoshinori Ishii MS 1991, Med.Micro-Immuno. • Cha-Gyun Shin PhD 1991, Med.Micro-Immuno. • Mary Faris PhD 1991, Microbiology • Laural Baglia PhD 1993, Med.Micro-Immuno. • Paskasari Permana PhD 1993, Med.Micro.-Immuno. • Mary Hilburger PhD 1994, Microbiology • Dancella Fernandez PhD 1994, Microbiology • Jackie Ward PhD 1995, Pathology • Melissa Myers PhD 1995, Med-Micro-Immuno • Alexa Konstantinos MS 1995, Med Micro-Immuno • Yuhong Liu PhD 1995, Med.Micro.Immuno • Andy Oberyszyn MS 1995, Med.Micro.Immuno. • Linda Harvey MS (non-thesis) 1995, Med.Micro.Immuno • Kelly Donelson MS (non-thesis) 1996, Med.Micro.Immuno. • David Brown PhD 1996, Microbiology • Kimberley Wilcox MS 1996, Microbiology • Zehra Demiral MS (non-thesis) 1996, Microbiology • Abbie Meyers PhD 1996, Med.Micro.Immuno. • Maria Ariza PhD 1996, Med.Micro.Immuno. • Marilyn Fernandez PhD 1997, Med.Micro.Immuno. • Ben Whitlock PhD 1997, Med.Micro.Immuno. • Gatum Bijur PhD 1997, Med.Micro.Immuno. • Jackie Benson PhD 1998, Med.Micro. Immuno. • Leigh Ann Kutz PhD 1998, Med. Micro. Immuno. • Shanxiang Zhang MS 1998, Med.Micro.Immuno. • Richard Wardrop PhD 2000, Mol. Virol, Immuno, Med Genetics • Maria Lara-Marquez PhD 1999, Med. Micro. Immuno. • Tianyi Wang Ph.D 2001, Microbiology • Matt Cannon Ph.D 2001, Animal Sciences • Traci Wilgus Ph.D 2001, Mol. Viro., Immuno., Med Gen. • Adam Studebaker PhD 2003, Mol. Viro., Immuno., Med Gen. • Yong-Sung Bahn PhD 2003, Mol. Viro., Immuno., Med Gen. • Erwin Chung PhD 2003, Mol Viro, Immunol, Med Gen. • Minqquin Lin PhD 2003, Vet. Biosciences • Hui-Chen Chen PhD 2003, Mol. Viro., Immuno., Med Gen. • John Hunzeker PhD 2003, Mol. Viro., Immuno., Med Gen. • Tianyi Yang PhD 2003, Pathology • Melanie McClain PhD 2005, Mol Viro, Immuno, Med Gen • Neeti Bhardwaj MS 2005, Microbiology • Nicole Damico PhD 2006, Mol. Viro, Immunol, Med Gen • Negin Gitban PhD 2006, Integrated Biology Graduate Program • Rajeev Nepal PhD 2006, Microbiology • Yan Ge PhD 2006, Vet Biosciences • Mike Farrow PhD 2007, Oral Biology • Nicholas Kin PhD 2007, Integrated Biology Graduate Program • Hua Niu PhD 2008, Vet Biosciences • Qingming PhD 2009, Vet Biosciences Xiong • Steve PhD 2010, Microbiology Oghumu • Hannah PhD 2011, Microbiology Cummings • Samir PhD 2011, MCDB Salama Rebeca Allen PhD 2011 IGBP • Heidi PhD 2012 Denistry Steinkamp • Amy PhD 2013 Oral Biology Hufnagle • Ran Dong MS 2012 Microbiology • Rashmi MS 2014 Microbiology Tuladhar • MIA Tazi PhD 2015 BGP • Alicia PhD 2019 MD/PhD Blaszczak

Research Currently Funded Grants: Title: Stress effects on virus protein induced inflammation and sickness behavior PI: Marshall Williams Co-investigator William Lafuse 20% effort Dates 4/01/2015-03/31/2020 Agency: National Institute of Allergy and Infectious Disease 2RO1AI084898 Total funding: $3,046,338 (year 1: Direct $384,606; Indirect : $199,313, year 2 total $568,411 )

PO1 Grant: Exploring the impact of inflammaging on immune function during tuberculosis Title: Project 2: Biology of alveolar macrophages in aging and tuberculosis PI: William Lafuse , Co-investigator Murugesan Rajaram Period of funding 4/01/18-2/28/19, no cost extension until 2/28/20 Source of funding NIH 7PO1AG051428- 03 subaward PO#61005 Total funding $95,385 (direct $61,539)

PO1 Grant: Exploring the impact of inflammaging on immune function during tuberculosis Title: Disparity supplement: Social stress as a co-morbidity of age-associated tuberculosis PI: William Lafuse, Co- investigator: Murugesan Rajaram Period of funding 4/01/18-2/28/19, no cost extension until 2/28/20 Source of funding NIH 7PO1AG051428- 03 subaward PO#61004 Total Funding $70,525 (direct 45,500)

Previously Funded Grants as Principal Investigator: Title: Role of commensal microbiota in stressor-induced immunomodulation PI: William Lafuse (25%) Period of Funding : 1/01/16-1/31/2017 Source of Funding: NIH National Institute of Allergy and Infectious Disease Subcontract to 1R21AI107238- 01A1 Total Funding $48,646 Title: Inhibition of IFN-gamma signaling by Mycobacteria Agency: NIH/AID R01 AIO45673 Principal Investigator: William Lafuse (25%) Dates 2/15/01-1/31/05 with no cost extension to 1/31/06 Annual Direct Costs: $175,00 Total Direct Costs: $1,032,500 Title:Molecular Biology of Murine Macrophage Activation Agency: NIH/AID R01 AI131529 Principal Investigator: William Lafuse (35%) Dates 7/15/92-6/30/95 Annual Direct Costs: $123,273 Total: Direct Costs $493,092

Title: Characterization of a mouse I Region Recomination Hot Spot Agency: NIH/GM R01 GM41786 Principal Investigator: William Lafuse (25% effort) Dates: 1/1/90-12/31/92 Annual Direct Costs: $84.305 Total Direct Costs: $394,102

Title: Characterization of mouse I Region Recombination Hot Spots Agency: American Cancer Society IM471A Principal Investigator: William Lafuse (25% effort) Dates: 1/1/89-12/31/89 Total Direct Costs: $69,000

Title: Analysis of a mouse I Region Recombination Hot Spot Agency: American Cancer Society, Ohio Divison Principal Investigator: William Lafuse Dates 11/1/86-12/31/86 Direct Costs: $3,000 Title: Polymorphism of Ia genes of Inbred Strains of Mice Agency:Bremer Foundation Principal Investigator: William Lafuse Dates 11/1/86-10/31/87 Direct Costs: $5,000

Funded Grants where I shared equal responsibility for the grant as a Co-Principal Investigator: Title: Regulation of NRAMP1 mediated iron transport Agency NIH/NIDDKD R01 DK57667 Principal Investigator: Bruce Zwilling (25% effort) Co-Principal Investigator: William Lafuse (25% effort) Dates: 05/01/00-2/31/05, no cost extension to 2/31/06 Annual Direct Costs $190,000 Total Direct Costs $990,000

Title: Nramp: Mycobacterial resistance, mRNA stability and iron Agency: NIH/HL R01 HL59795 Principal Investigator: Bruce S. Zwilling (25%) Co-Principal Investigator: William Lafuse (25%) Dates 9/30/97-8/30/02 Annual Direct Costs: $175,000 Total Direct Costs: $875,000 Title: Catecholamine Regulation of Macrophage Function Agency: NIH/NIAID R01 AI042901 Principal Investigator: Bruce S. Zwilling (25%) Co-Principal Investigator: William Lafuse (25%) Dates: 12/01/97-11/30/02 Annual Direct Costs: $194,224 Total Direct Costs: $1,001,126 Funded research grants on which I have been a co-investigator:

Title: Role of commensal microbiota in stressor-induced immunomodulation PI: Michael Bailey Co- investigator: William Lafuse 25% effort Dates: 2/1/2014 -1/31/2017 Agency: National Institute of Allergy and Infectious Disease 1R21AI107238-01A1 Total Funding: $380,869 (Direct $247,500 Indirect $133,368)

Julie Martin Mid-Career award (AFAR) Turner (PI) 6/01/10 – 06/30/15 Divergent macrophage-pathogen interactions in old age The major goal of this project is to determine the receptor mediate uptake mechanism of M.tb in macrophages form old mice. Role: Co-Investigator

Title: Neuromodulation of the antibody response Agency NIH/NAID R01 AI037326 Principal Investigator: Virginia M. Sanders Co-Principal Investigator: William Lafuse (5%) Dates 9/1/94-8/30/08 Title: Neuromodulation of the antibody response Agency NIH/NAID R01 AI037326 Principal Investigator: Virginia M. Sanders Co-Principal Investigator: William Lafuse (5%) Dates 9/1/94-8/30/08

Title: Restraint Stress: Modulation of Mycbacterial Resistance Agency: NIH MH45679-04 Principal Investigator: Bruce Zwilling Co-Investigator William Lafuse (5% effort) Dates: 8/01/92-7/31/97 Annual Direct Costs: $194,962 Total Direct Costs: $1,085,169 Title: Ethanol Suppression of Mycobacterial Resistance Agency: NIH AA09321 Principal Investigator: Bruce Zwilling Co-Investigator: William Lafuse (15% effort) Dates: 9/30/92-8/31/95 Annual Direct Costs: $115,239 Total Direct Costs: $332,591 Title: Reactivation of Tuberculosis by Restraint Stress Agency: NIH MH54966 Principal Investigator: Bruce Zwilling Co-Investigator: William Lafuse (5% effort) Dates: 9/1/95-4/30/00 Annual Direct Costs: $151,805

Title: Stress induced immunosuppression of anti-viral immunity Agency: NIH MH46801 Principal Investigator: John Sheridan Co- Investigator: William Lafuse (5% effort) Dates: 5/1/90-4/31/94 Annual Direct Costs: $133,316 Total Direct Costs: $369,761

Title: Restraint stress, suppression of Ia expression Agency: NIH MH45679 Principal Investigator: Bruce Zwilling Co-Investigator: William Lafuse (10% effort) Dates:8/1/89-7/31/92 Annual Direct Costs: $135,727 Total Direct Costs:$433,033

Title: Ia expression by macrophages of Bcgr and Bcgs mice. Agency: NIH AI22249 Principal Investigator: Bruce Zwilling Co-Investigator: William Lafuse (10% effort) Dates 4/1/87-3/31/90 Title: Effects of HTLV-III/LAV infection on Ia gene expression by human monocytes (Project 5) Agency: NIH AI25924 Aids Clinical Study Group Program Director: Robert Fass Principal Investigator: Bruce Zwilling Co-Investigator: William Lafuse (10% effort) Dates: 9/30/87-9/29/89 Annual Direct Costs: $75,000 Title: Personality, psychological stress, and immunocompetence Agency: NIH MH40787 Principal Investigator: Ronald Glaser Co-Investigator: William Lafuse (5% effort) Dates:9/1/88-8/31/92 Annual Direct Costs: $196,979 Total Direct Costs: $987,096

Non-Funded Grants submitted as Principal Investigator since 2005 Title Period NIH 2R01AI045673-05 02/15/2001-03/31/2010 Macrophage Gene Repression by Mycobacteria 27.2 percentile NIH 2R01AI45673-05A1 02/15/2001-11/30/2010 Macrophage Gene Repression by Mycobacteria 50.3 percentile NIH 1RO1AI069247-01 06/01/2005-11/01/2011 Induction of Hepcidin in macrophages and control of tuberculosis NS NIH 1RO1AG028315-01 07/01/2006-06/30/2009 Iron Homeostasis in Aging: Role of IL-6 and Hepcidin NS Grant application was in response to a RFA NIH 1RO1AI075067-01 07/01/2007-06/30/2012 Regulation and function of hepcidin in M. tuberculosis infected macrophages. NS NIH 1RO1AI075067-01A1 04/01/2008-03/31/2013 Regulation and function of hepcidin in M. tuberculosis infected macrophages 43.3 percentile NIH 1RO1AI75067-01A2 04/01/2009-03/31/2014 Regulation and function of hepcidin in M. tuberculosis infected macrophages. NS NIH 1RO1AI076293-01 12/01/2007-11/30/2012 Iron transport proteins and the Macrophage Phagosome NS NIH 1RO1AI085153-01 12/01/2009-11/30/2014 Macrophage Iron Homeostasis and M. tuberculosis Infection. NS NIH 1R01DK088535-01 04/01/2010-03/31/2015 Psychological Stress and Anemia. NS NIH1RO1DK088535-01A1 12/01/2010-11/30/2015 Psychological Stress: Enhanced Innate Immunity and Iron Deficiency. NS NIH 1R21AI094087-01 04/01/2011-03/31/2013 Dysregulation of Erythropoiesis by Visceral Leishmanisis. NS NIH 1RO1AI098193-01 09/01/2011-08/31/2016 Hepcidin: Anti-mycobacterial activity and its potential as a TB therapeutic agent. NS NIH 1R21 AI107238-01 07/1/2013-6/30/2015 Role of Commensal Microbiota in Stressor-Induced Immunomodulation impact score 36 NIH 1PO1 AG044298-01 4/01/2013-3/31/18 Project 2: Biology of Alveolar macrophages in aging and tuberculosis impact score 33 NIH 1R01 AI107220-01 7/01/2013-6/30/18 Human Macrophage MicroRNAs and Tuberculosis NS

NIH 1PO1 AG044298-01A Project 2: Biology of Alveolar macrophages in aging and tuberculosis impact score 20 NIH 1R01 AI107220-01A Human Macrophage MicroRNAs and Tuberculosis NS

NIH 1 RO1 AG059699-01 NS Bacterial translocation and inflammaging in the aged spleen 7/01/2018-6/30/2023

NIH1 R21 AI44224-01 NS Molecular Mechanisms of Mycobacteriurm tuberculosis-mediated macrophage Gene transcriptional repression 4/01/2019-3/31/2021

NIH1 AI145384-01, NIH 1R21 AI145384-01A1 NS Characterization of phenotypes in persistent Mycobacterium abscessus pulmonary Infections using Collaborative Cross (CC) and CC founder strains to develop mouse Models that recapitulate human infection 4/01/2019-3/31/2021

NIH 1 R21 AG064155-01 NS Bacterial translocation and spleen monocyte production and activation in aged mice 7/01/2019-6/30/2021

Invited Seminars: Ohio State University: October 21, 1986 OSU Immunology Seminar “Recombinant hot spots in the mouse H-2 Complex” March 22, 1988 OSU Immunology Seminar “Recombination sites in the mouse H-2 major histocompatibilty complex” April 25, 1989 OSU Immunology Seminar “Recombination sites and chromosome walking in the mouse H- 2 major histocomaptibility complex” April 24, 1990 OSU Immunology Seminar “The search for genes linked to the BCG resistance gene in the mouse” October 15, 1991 OSU Immunology Seminar “Molecular biology of macrophage IFN-g induced genes” March 9, 1993 OSU Immunology Seminar “Molecular biology of genes induced in macrophages” May 31, 1994 OSU Immunology Seminar “Molecular biology of mouse macrophage activation by interferon-gamma” April 18, 1995 OSU Immunology Seminar “Molecular studies of gene induction and expression in activated mouse macrophages” October 17, 1995 OSU Immunology Seminar “ IFN-g signaling in mouse macrophages: autoregulation of Stat I expression and activation” April 22, 1997 OSU Immunology Seminar “mRNA stability and RNA binding activity in IFN-gamma activated mouse macrophages” April 30, 1997 Dept. of Medical Microbiology and Immunology Seminar, mRNA stability and RNA binding activity in IFN-gamma activated mouse macrophages” March 10, 1998 OSU Immunology Seminar “Inhibition of interferon-gamma induced gene expression and signaling in mouse macrophages by infection with Mycobacterium avium” June 1 1999 OSU Immunology Seminar “The role of oxidants and protein kinase C in mRNA stability of mouse Nramp1 (Natural Resistance Associated Macrophage Protein 1)” March 13, 2001 OSU Immunology Seminar “The role of oxidant generated signaling pathways in Nramp1 mRNA stability and RNA binding proteins” January 30, 2002 OSU MVIMG Seminar “Role of Oxidant signaling pathways in Natural Resistance Associated Macrophage Protein1 (Nramp1) mRNA stability” February 17, 2004 Modulation of macrophage functions by catecholamines OSU Immunology seminar. October 31, 2006 Expression and function of hepcidin and the iron export protein ferroportin 1 in Mycobacteria Infected Macrophages. OSU Immunology Seminar March 19, 2007 Expression and function of hepcidin and the iron export protein ferroportin 1 in Mycobacteria infected macrophages. MVIMG faculty seminar October 12, 2008 Hepcidin and mycobacteria: expression, anti-mycobacterial activity, and phagosome localization of hepcidin in infected macrophages. OSU Immunology Seminar November 30, 2009 Hepcidin and Anemia during psychological stress and infection. MVIMG Seminar January 18, 2011 Activity of microbial peptide hepcidin against Mycobacterium tuberculosis OSU Immunology Seminar. Feb13, 2012 Mechanisms of Anemia during Visceral Leishmaniasis in Syrian Golden Hamsters CMIB Work in Progress seminar July, 2012 William Lafuse: Road to PI talk. MII seminar Aug, 2014 Influence of microbiaota in stress induced macrophage priming and inflammaging. CMIB Work in Progress seminar May 2016 Changes in alveolar macrophage phenotype and response to M. tuberculosis due to aging, World TB Meeting 3/25/16

Other: April 26, 2001 National Institutes of Health, Bethesda, MD; Biology of iron overload and new approaches to therapy Meeting “Regulation of Nramp1 Mediated Iron Transport” June 2004 PNIRS 2003 Meeting; Macrophage resistance to Mycobacterium avium is induced by α2- Adrenergic stimulation. March 2, 2005 Dept of Microbiology, Medical College of Ohio, Toledo, OH; Inhibition of IFN-gamma induced gene expression in macrophages by mycobacteria infection. September 20, 2017 National Institute of Aging Health Disparities Meeting, Bethesda, Md. Social stress as a co- morbidity of age-associated tuberculosis

Publications 1. Lafuse, W., Mereulo, D. and Edidin, M. The genetic control of liver cAMP levels in mice. Immunogenetics 9: 57, 1979. 2. Lafuse, W.P. and Edidin, M. Influence of the mouse major histocompatibility complex, H-2, on liver adenylate cyclase and on glucagon binding to liver cell membranes. Biochemistry 19: 49, 1980. 3. David, C.S., Beisel, K.W., Krco, C.J., and Lafuse, W.P. Genetic organization, tissue expression and functional role of murine Ia antigens. In: Current Trends in Histocompatibility, Eds. R.A. Reisfeld and S. Ferrone, Plenum Press, New York, 1981. 4. Lafuse, W.P., Neely, B.C. and David, C.S. Biochemical studies of mouse Ia molecules with xenoantisera. I. Identification of a second Ia molecule (I-E) in H-2s haplotype. Immunogenetics 10: 311, 1980. 5. Lafuse, W.P., McCormick, J.F., and David, C.S. Serological and biochemical identification of hybrid Ia antigens. J. Exp. Med. 151: 709, 1980. 6. Lafuse, W.P., McCormick, J.F., and David, C.S. Lack of gene complementation prevents expression of I-Ek in recombinant, A.TFR5. J. Immunol. 124: 2511, 1980. 7. Krco, C.J., Challacombe, S.J., Lafuse, W.P., David, C.S. and Tomasi, T.B. Expression of Ia antigens by mouse Peyer's patch cells. Cellular Immunology 57: 420, 1981. 8. Lafuse, W.P., Yokota, S. and David, C.S. Hybrid Ia antigens: genetic, serologic and biochemical analyses. Journal of Supramolecular Structure 14: 97, 1980. 9. Lafuse, W.P., Yokota, S. and David, C.S. Hybrid Ia antigens: genetic, serologic and biochemical analyses. In: Control of Cellular Division and Development, Part B, ed. D. Cunningham, E. Goldwasser, J. Watson and C.F. Fox. Alan R. Liss, New York, p. 127, 1981. 10. Beisel, K.W., Lafuse, W.P., Stimpfling, J.H. and David, C.S. Identification of private H-2 and Ia specificities of H-2r haplotype. Immunogenetics 12: 191, 1981. 11. Lafuse, W.P., McCormick, J.F., Corser, P.S. and David, C.S. Gene complementations to generate Ia antigens (Ia.23) on hybrid molecules. Transplantation 30: 341, 1980. 12. David, C.S., Lafuse, W.P. and Yokota, S. Hybrid antigens. Identification characterization and role in immune response. In: Immunology of Major Histocompatibility System, Eds. M. Zaleski and J.F. Mohn, S. Karger, Basel, pp. 28-36, 1981. 13. Beisel, K.W., Lafuse, W.P., Stimpfling, J.H. and David, C.S. Description of H-2K, H-21 and H-2D gene products of H-2v haplotype. Immunogenetics 11: 407, 1980. 14. Yokota, S., Lafuse, W.P., McCormick, J.F. and David, C.S. Detection of hybrid (combinatorial) Ia antigens using parent anti-Fl sera. J. Immunol. 126: 371, 1981. 15. Lafuse, W.P., McCormick, J.F., Corser, P.S. and David, C.S. Hybrid (combinatorial Ia specificities: Gene complementations to generate Ia.22. Immunogenetics 14: 115, 1981. 16. Lafuse, W.P., Melvold, R. and David, C.S. Serological and biochemical analysis of Ia molecules in the I-A mutant B6.C-H-2bm12. Transplantation 31: 434, 1981. 17. Long, P.M., Lafuse, W.P. and David, C.S. Serological and biochemical identification of minor histocompatibility antigen (H-4). J. Immunol. 127: 825, 1981. 18. Lafuse, W.P., Yokota, S. and David, C.S. Gene complementations generate hybrid Ia specificities involved in immune response. Transplantation Proc. 3: 1017, 1981. 19. Lafuse, W.P. and David, C.S. Hybrid (combinatorial) Ia antigens mediate histocompatibility auto and alloreactivity, and immune recognition. Transplantation Proc. 13: 1846, 1981. 20. Lafuse, W.P., Hendrickson, H., Corser, P.S. and David, C.S. Serological and biochemical studies of a variant Ae beta Ia polypeptide chain in D2.GD (H-2g2). J. Immunol. 127: 811, 1981. 21. Lafuse, W.P. and David, C.S. Murine Ia antigens: Studies using hybrid and mutant mice. In: H-2-Ia and HLA-Dr Antigens. Vol. 1 Mice, S. Ferrone and C.S. David, eds. CRC Press, pp. 105-142. 22. Lafuse, W.P., Corser, P., and David, C.S. Biochemical evidence for multiple I-E Ia molecules. Immunogenetics 15: 365-375, 1982. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

23. Pierres, M., Lafuse, W.P., Dosseto, M., Devaux, C., Birnbaum, C.Z. and David, C.S. Molecular specificity of a monoclonal anti-Ik alloantibody identifying a highly conserved determinant on mouse I-A, I-E and human Dr antigens. Tissue Antigens 20: 208, 1982. 24. Lafuse, W.P., Pierres, M. and David, C.S. Crossreactions of anti-I-Ek monoclonal antibodies with subsets of

86 I-Ab molecules. Transplantation 35: 49, 1983. 25. David, C.S., Lafuse, W.P. and Pierres, M. Biochemical evidence for multiple Ia molecules. In: Macrophages and Natural Killer Cells, S.J. Normann and E. Sorkin, Eds., Plenum Press, pp. 569-577, 1982. 26. David, C.S. and Lafuse, W.P. Ia genes, gene products and determinants. In: Ir Genes, Past, Present and Future, D.W. Pierce, S.E. Cullen, J.A. Kapp, B.D. Schwartz and D.C. Shreffler, eds., Humana Press, Clifton, NJ, pp. 3-12, 1983. 27. Lafuse, W.P., Pierres, M. and David, C.S. Isolation and analysis of subpopulations of I-Ab molecules with monoclonal antibodies. In: Ir Genes, Past, Present and Future, C.W. Pierce, S.E. Cullen, J.A. Kapp, B.D. Schwartz, D.C. Shreffler, Eds., Humana Press, Clifton, NJ, pp. 4549, 1983. 28. Singh, S., Lafuse, W.P., McKean, D.J. and David, C.S. Structural characterization of I-A subsets using monoclonal antibodies. In: Ir Genes, Past, Present and Future, C.W. Pierce, S.E. Cullen, J.A. Kapp, B.D. Schwartz, D.C. Shreffler, Eds., Humana Press, Clifton, NJ, pp. 63-67, 1983. 29. Lafuse, W.P. and David, C.S. Ia antigens: Genes, molecules and function. Transplantation 38: 443, 1984. 30. Wooley, P.H., Luthra, H.S., Lafuse, W.P., Huse, A., Stuart, J.M. and David, C.S. Type II collagen-induced arthritis in mice. III. Suppression of arthritis by using monoclonal and polyclonal anti-Ia antisera. J. Immunol. 134: 2366, 1985. 31. David, C.S, McCormick, J.F., Lafuse, W.P., and Hirose, S. Serological confirmation in Ia mutant B6.C-H- 2bm12 of gene conversions. Transplantation 42: 429, 1986. 32. Rodriquez, M., Lafuse, W.P., Leibowitz, J. and David, C.S. Partial suppression of Theiler's virus-induced demyelination in vivo by administration of monoclonal antibodies to immune response gene products. J. Neurology 36: 964, 1986. 33. Lafuse, W.P., Savarirayan, S., McCormick, J.F. and David, C.S. Identification of I-E alpha genes in H-2 recombinant mouse strains by Fl complementation. Transplantation 43: 297, 1986. 34. Lafuse, W.P., Berg, N., Savarirayan, S. and David, C.S. Mapping of a second recombination hot spot within the I-E region of the mouse H-2 gene complex. J. Exp. Med. 163: 1518, 1986. 35. Lafuse, W.P., Singh, S., Savarirayan, S. and David, C.S. DNA restriction fragment analysis of I-E genes in I- E negative H-2 reombinant mouse strains. Immunogenetics 24: 68, 1986. 36. Lafuse, W.P., Pierres, M. and David, C.S. Molecular heterogeneity of I-Ab molecules: Analysis of I-Ab subsets with monoclonal antibodies. Immunological Investigations 15: 735, 1986. 37. Lafuse, W.P. and David, C.S. Recombination hot spots within the I region of the mouse H-2 complex map to the Eα and Eβ genes. Immunogenetics 24: 352, 1986. 38. Savariayan, S., Lafuse, W.P., and David, C.S. Recombination between H-2D and H-2L Genes: Identification, Characterization and Gene Order Transplant Proc. 18:702, 1985. 39. Lafuse, W.P. and David, C.S. Recombination sites within the I region of the mouse H-2 complex. In H-2 antigens, genes, molecules, function, Ed. C.S. David. Plenum Press, New York, p. 41, 1988. 40. Zwilling, B.S., Massie, M., Vespa, L., Kwaniewski, M., Nath, J. and Lafuse, W.P. Continuous expression of I-A is linked to the Bcg gene. Current Topics in Microbiology and Immunology 137: 316, 1988. 41. Nath, J., Lafuse, W.P., and Zwilling, B.S. Regulation of Class II MHC gene expression by macrophages from Bcgr and Bcgs mice. Cellular Immunology 117: 120, 1988. 42. Lafuse, W.P., Lee, S.T., Castle, L. and David, C.S. Restriction fragment analysis of H-2 recombinant mouse strains with crossovers between Eα and C4. Immunogenetics 30: 387, 1989. 43. Lafuse, W.P., Castle, L., and David, C.S. Restriction fragment length polymorphism analysis of Factor B and C2 genes in H-2 recombinant mouse strains. Immunogenetics 31: 275, 1990. 44. Glaser, R., Kennedy S., Lafuse, W.P., Bonneau, R.H., Speicher, C., Hillhouse, J., and Kiecolt- Glaser, J. Psychological stress-induced modulation of IL-2 receptor gene expression and IL- 2 production in peripheral blood leukocytes. Arch. Gen. Psych. 47: 707, 1990. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

45. Lafuse, W.P., Lee, S.T., and David, C.S. Molecular analysis of I region recombinants. RFLP analysis of two recombinant mouse strains with crossovers in the Eα recombination hot spot. J. Immunogenetics 17:169, 1990. 46. Lafuse, W.P. Molecular biology of mouse MHC class II genes. Critical Reviews in Immunology 11:167, 1991. 47. Lafuse, W.P., Lanning, D., Spies, T. and David, C.S. PFGE and RFLP analysis of the S/D region of the mouse H-2 complex. Immunogenetics 36:110, 1992. 48. Lafuse, W.P., Brown, D., and Zwilling, B. Assignment of the microtubule-associated protein 2 gene to mouse chromosome 1. Mammalian Genome 3: 48, 1992.

88 49. Zwilling, B.S, Lafuse, W.P., Brown, D., and Pearl, D. Characterization of ACTH mediated suppression of MHC class II expression by murine peritoneal macrophages. J. Neuroimmunol 39:133, 1992. 50. Sant Angelo, D. B., Lafuse, W.P., and Passmore, H.C. Evidence that nucleotide sequence identity is a requirement for meiotic crossing over within the mouse Eb recombinational hotspot. Genomics 13:1334, 1992. 51. Glaser, R. Lafuse, W.P., Bonneau, R.H., Atkinson, C., and Kiecolt-Glaser, J. Stress associated modulation of proto-oncogene expression in human peripheral blood leukocytes. Behavioral Neuroscience 107:525, 1993. 52. Sabharwal, P., Glaser, R., Lafuse, W.P., Varma, S., Liu, Q., Arkins, S., Kooijman, R., Kutz, L., Kelley, K.W., and Malarkey, W.B. Prolactin synthesis and secretion by human peripheral blood mononuclear cells; An autocrine growth for lymphoproliferation. PNAS 89: 7713, 1992. 53. Lafuse, W.P., and Zwilling, B.S. Localization of the inhibin βB gene on mouse chromosome 1. Mammalian Genome 4, 399, 1993. 54. Lafuse, W.P., Brown, D., Castle, L., and Zwilling, B.S. IFN-γ increases cathepsin H mRNA levels in mouse macrophages. J. Leukocyte Biol. 57, 663, 1995. 55. Lafuse, W.P., Castle, L., Brown, D. and Zwilling, B.S. Cloning and expression of a gene induced by IFN-γ in mouse macrophages that encodes a protein with GTP binding motifs. J. Leukocyte Biol. 57, 477, 1995 56. Lafuse, W.P., Brown, D., Castle, L., and Zwilling, B.S. The cytotoxic T lymphocyte gene FIBLP with homology to fibrinogen β and γ subunits is also induced in mousemacrophages by IFN- γ. Cellular Immunology 163,187, 1995. 57. Brown, D. H., Lafuse, W.P., and Zwilling, B.S. Cytokine mediated activation of macrophages from BCG resistant and susceptible mice: Differential effects of corticosterone on anti- mycobacterial activity and the expression of the Bcg gene (candidate Nramp). Infection and Immunity 63, 2983, 1995. 58. Miles, B., Lafuse, W.P., and Zwilling, B.S. Binding of α-Adrenergic receptors stimulates the anti- mycobacterial activity of murine peritoneal macrophages. J. Neuroimmunology 71,19,1996. 59. Brown, D.H., Lafuse, W.P., and Zwilling, B.S. The stabilized expression of mRNA is associated with mycobacterial resistance controlled by Nramp1. Infection and Immunity 65, 597, 1997. 60. Lanning, D., and Lafuse, W.P., Localization of the casein kinase II β-subunit gene within the mouse H-2 complex and comparison with Bat genes. Mammalian Genome 8, 519, 1997. 61. Brown,D.H., Lafuse, W.P., and Zwilling, B.S. Host resistance to mycobacteria is compromised by activation of the hypothalamic-pituitary-adrenal axis. Ann. N.Y. Acad. Sci. 840:773-786, 1998. 62. Lanning,D.,and Lafuse, W.P. The Mouse p52 Subunit of the Transcription/DNA Repair Factor TFIIH is located in the Class III Region of the H-2 Complex: Cloning and Sequence Polymorphism. Immunogenetics 49:498-504,1999. 63. Chen, L., Boomershine, C.,Wang, T., Lafuse, W.P., and Zwilling, B.S. Synergistic interaction of Catecholamine hormones and Mycobacterium avium results in the induction of interleukin- 10 mRNA by murine peritoneal macrophages. J. Neuroimmunology 93:149-155,1999. 64. Kuhn, D.E., Baker, B.D., Lafuse, W.P., and Zwilling, B.S. Differential iron transport into phagosomes from the RAW264.7 macrophage cell lines transfected with Nramp1Gly169 or Nramp1Asp169. J. Leukocyte Biol. 66:113-9, 1999. 65. Zwilling, B.S., Kuhn, D.E., Wikoff, L., Brown, D.,, and Lafuse, W.P. The role of iron in Nramp1 mediated inhibition of growth. Infection and Immunity. 67:1386-1392, 1999. 66. Hussain, S., Zwilling, B.S. and Lafuse, W.P. Mycobacterium avium infection of mouse macrophages inhibits IFN-γ JAK-STAT signaling and gene induction by down-regulation of the IFN-γ receptor. J. Immunology 163:2041-9, 1999. 67. Boomershine, C.S., Lafuse, W.P., and Zwilling, B.S. β-2 adrenergic receptor stimulation inhibits nitric oxide generation by Mycobacterium avium infection. J. Neuroimmunology 101:68-75, 1999. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

68. Lafuse, W.P., Alvarez, G.R., and Zwilling, B.S. Regulation of Nramp1 mRNA stability by oxidants and protein kinase C in RAW264.7 macrophages expressing Nramp1Gly169. Biochemical Journal 351:687-696, 2000. 69. Wang, T., Lafuse, W.P., and Zwilling, B.S. Regulation of Toll-like receptor 2 expression by macrophages following Mybacterium avium infection. J. Immunol. 165:6308-6313, 2000. 70. Kuhn, D.E., Lafuse, W.P., and Zwilling, B.S. Iron transport into Mycobacterium avium- containing phagosomes from an Nramp1Gly169-transfected RAW264.7 macrophage cell line. J. Leukocyte Biol. 69:43- 49, 2001.

90 71. Wang, T., Lafuse, W.P., and Zwilling, B.S. NFκβ and Sp-1 elements are necessary for Mycobacterium avium-induced transcription of Toll-like receptor 2. J. Immunol. 167:6924- 6932, 2001. 72. Zhong, W., Lafuse, W.P., and Zwilling, B.S. Infection with Mycobacterium avium differentially regulates the expression of iron transport protein mRNA in murine peritoneal macrophages. Infection and Immunity 69:6618-6624, 2001. 73. Wang, T., Lafuse, W.P., Takeda, K., Akira, S., and Zwilling, B.S. Rapid chromatin remodeling of TLR2 promoter during infection of Macrophages with Mycobacterium avium. J. Immunol. 169:795-801, 2002. 74. Lafuse, W.P., Alvarez, G.R., and Zwilling, B.S. Role of MAP kinase activation in Nramp1 mRNA stability in RAW264.7 macrophages expressing Nramp1Gly169. Cellular Immunology 215: 195-206, 2002. 75. Weatherby, K. E., Zwilling, B.S., and Lafuse, W.P. Resistance of macrophages to Mycobacterium avium is induced by α2-adrenergic stimulation. Infection and Immunity 71:22- 29, 2003. 76. Alvarez, G.R. Zwilling, B.S., and Lafuse, W.P. Mycobacterium avium Inhibition of IFN-γ signaling in mouse macrophages: Toll-like receptor 2 stimulation increases expression of dominant-negative STAT1β by mRNA stabilization. J. Immunol. 171:6766-6773, 2003. 77. Curry, H., Alvarez, G.R., Zwilling, B.S., and Lafuse, W.P. Toll-like receptor 2 stimulation decreases IFN-γ receptor expression in mouse RAW264.7 macrophages. J. Interferon Cytokine Res. 24:699-710, 2004. 78. Bhardwaj, N, Rosas, L.E., Lafuse, W.P., and Satoskar, A. R. Leishmania inhibits STAT1- mediated IFN-γ signaling in macrophages: increased tyrosine phosphorylation of dominant negative STAT1β by Leishmania mexicana. Int. J. Parasitol. 35:75-82, 2005. 79. Wang, Y., Curry, H. M., Zwilling, B. S., and Lafuse, W.P. Mycobacteria inhibition of IFN-γ induced HLA-DR gene expression by up-regulating histone deacetylation at the promoter region in human THP-1 monocytic cells. J. Immunol. 174: 5687-5694, 2005. 80. Studebaker, A.W., Lafuse, W.P., Kloesel, W., and Williams, M.V. Modulation of human dUTPase using small interfering RNA. Biochem. Biophys. Res. Comm. 327:306-310, 2005. 81. Lafuse, W.P., Alvarez, G., Curry, H.M., and Zwilling, B.S. Mycobacterium tuberculosis and Mycobacterium avium inhibit IFN-γ induced gene expression by TLR2-dependent and independent pathways. Journal Interferon Cytokine Research 26:548-561, 2006. 82 Sow, F. B., Florence, W.C., Satoskar, A.R., Schlesinger, L.S., Zwilling, B.S. and Lafuse, W.P. Expression and localization of hepcidin in macrophages: a role in host defense against tuberculosis. J. Leuk. Biol. 82:934-945, 2007. 83. VanZandt, K.E., Sow, F.B., Florence, W.C., Zwilling, B.S., Satoskar, A.R., Schlesinger, L.S., Lafuse, W.P. The iron export protein ferroportin 1 is differentially expressed in mouse macrophage populations and is present in the mycobacterial-containing phagosome. J. Leuk. Biol. 84:689-700, 2008. 84. Barbi, J. Cummings, H.E, Lu, B., Oghumu S., Ruckle, T., Rommel, C., Lafuse, W.P., Whitacre, C.C., and Satoskar, A. R. PI3Kgamma is essential for efficient induction of CXCR3 on activated T cells. Blood 112:304851, 2008. 85. Roth, K.M., Gunn, J.S., Lafuse, W.P., Francisella inhibits STAT1-mediated signaling in macrophages and prevents activation of antigen-specific T cells. Int. Immunol. 21:19-28, 2009. 86. Sow, F.B., Alvarez, G.R., Gross, R.P., Satoskar, A.R., Schlesinger, L.S., Zwilling, B.S., and Lafuse, W.P. Role of STAT1, NF-κβ, and C/EBPβ in the macrophage transcriptional regulation of hepcidin by mycobacterial infection and IFN-γ. J. Leukocyte Biology 86:1247- 1258, 2009. 87. Sow, F.B., Nandakumar, S., Velu, V., Kellar, K.L., Schlesinger, L.S., Amara, R.R., Lafuse, W.P., Shinnick, T.M., Sable, S.B. Mycobacterium tuberculosis components stimulate production of the antimicrobial peptide hepcidin. Tuberculosis 91:314-321, 2011. 88. Allen, R.G., Lafuse, W.P., Galley, J.D. Ali, M.M.,Ahmer, B.M., and Bailey, M.T. The intestinal Microbiota are necessary for stressor-induced enhancement of splenic macrophage microbicidal activity. Brain, Behavior, and Immunity 26:371-82, 2012. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

89. Allen, R.G., Lafuse, W.P., Powell, N.D., Webster Marketon, J.I., Stiner-Jones, L.D., Sheridan, J.F., Bailey, M.T. Stressor-induced increase in microbicidal activity of splenic macrophages is dependent upon peroxynitrite production. Infect. Immun. 80: 3429-37, 2012. 90. Lafuse, WP, Story, R., Mahylis, J., Gupta, G., Varikuti S., Steinkamp, H., Oghumi S, Satoskar, A.R. Leishmania donovani infection induces anemia in hamsters by differentially altering erythropoiesis in bone marrow and spleen. PLOS one, e59509,2013. 91. Moliva,J.I.,Rajaram, M.V.S, Sidiki, S., Guirado, E., Pan, X., Wang, S.H., Ross Jr., P., Lafuse, W.P., Schlesinger, LS, Turner, J., and Torrelles, J.B. Molecular composition of the alveolar lining fluid in the aging

92 lung. AGE .Epup adhead of print, 2014. 92. Canan, C.H., Gokhale, N.S., Carruthers, B., Lafuse, W.P.,Torrelles, J.B., and Turner, J. Characterization of Lung iInflammation and its impact on macrophage function in aging. J. Leuk. Biol. 96:473-80, 2014. 93. Ni B, Rajaram MVS, Lafuse W.P., Landes MB, Schlesinger LS. Mycobacterium tuberculosis decreases human macrophage IFN-γ responsiveness through miR-132 and miR- 26a”. J Immunol. 193:4537-47, 2014. 94. Lafuse WP, Gearinger, R, Fisher S, Nealer C., Mackos, A.R., Bailey, M.T. Exposure to a social Stressor induces translocation of comensal lactobacilli to the spleen and priming of the innate Immune system. J.Immunol. 198:2383-2393, 2017 95. Rajaram MVS, Arnett E, Azad AK, Guirado E, Ni B, Gerberick AD, He LZ, Keler, T, Thomas LJ, Lafuse WP, Schlesinger LS M. tuberculosis-initiated human mannose receptor signaling regulates macrophage Recognition and vesicle trafficking by FcRγ-chain, Grb2, and SHP-1 Cell Reports 21:126-140.2017. 96. Blaszcak AM, Jalilvand A, Liu J, Wright VP. Suzo A, Needleman B, Noria S, Lafuse W, Huseh WA, Bradley D. Human visceral adipose tissue macrophages are not adequately defined by standard methods of characterization. J. Diabetes Res. 2019: 8124563, 2019. 97. Moliva JI, Duncan MA, Olm-Fontanez A, Akhter A, Arnett E, Scordo JM, Ault R, Sasindran SJ, Azad AK, Montoya MJ, Reinhold-Larrsson N, Rajaram MVS, Merrit RE, Lafuse WP, Zhang L, Wang SH, Beamer G, Wang Y, Proud K, Maselli DJ, Peters J, Weintraub ST, Turner J, Schlessinger LS, Torrelles JB. The lung mucosa environment in the elderly increases host susceptibility to Mycobacterium tuberculosis. J. Infect. Dis.2019, jiz138, 2019. 98. Blasczak AM, Wright VP, Anandani K, Liu J, Jalilvand A, Bergin S, Nicoloro SM, Czech MP, Lafuse W, Deng T, Bradley D, Husueh WA. Loss of antigen presentation in adipose tissue macrophages or in adipocytes, but not both, improves glucose metabolism. J. Immunol. 202:2451-2459, 2019. 99. Williams MV, Cox B, Lafuse WP, Ariza ME. 2019. Epstein-Barr virus dUTPase may contribute to Myalgic Encephalomyelitis/Chronic Fatigue Syndrome by inducing neuroinflammatory mediators. Clinical Therapeutics in press. 100. Lafuse WP, Rajaram MVS, Wu Q, Moliva JI, Torrelles JB, Turner J, Schlesinger LS. Identification of an Increased alveolar macrophage subpopulation in old mice that displays unique inflammatory characteristics and is permissive to Mycobacterium tuberculosis infection. J. Immunology 203:2252-2264,2019.

Abstracts of Papers Presented at Meetings 1. Lafuse, W.P., Neely, B., McKean, D., and David, C.S. Biochemical studies of Ia gene products with xenoantisera. Fed Proc. 38:6303, 1979. 2. Long, P.M., Lafuse, W.P., Krco, C.J., and David, C.S. Serological, biochemical, and cellular studies of minor histocompatibility antigens. Fed. Proc, 40: 1159, 1981. 3. Lafuse, W.P., Freed, J.H., and David, C.S. Serological and biochemical studies of mutant Ae(Eβ) Ia polypeptide chain in D.2GD(H-2g2). Fed Proc. 40: 1010, 1981. 4. Singh, S.K., Lafuse, W.P., McKean, D.J., and David, C.S. Evidence for multiple I-E molecules in H-2d tumor cell lines. Fed. Proc. 41:612, 1982. 5. Long, P.M., Lafuse, W.P., Krco, C.J., and David, C.S. Serological, biochemical, and cellular studies of minor histocompatibility antigens. Fed. Proc. 41:427, 1982. 6. Lafuse, W.P., Corser, P., Pierres, M., and David, C.S. Crossreactions of monoclonal anti-I-E antibodies with subsets of I-Ab molecules. Fed. Proc. 41:365, 1982. 7. Lafuse, W.P., Singh, S., Pierres, M., and David, C.S. Isolation and structural characterization of subsets of I-Ab molecules. Fed. Proc. 42:5491, 1983. 8. Levy, N., Singh, S., Lafuse, W.P., and David, C.S. Analysis of I-Eβ polypeptide chains in murine I-A recombinant strains. Fed. Proc. 43:1599, 1984. 9. Lafuse, W.P., Hirose, S., McCormick, J.F., and David, C.S. Serological evidence for gene conversion in mouse Ia genes. Fed. Proc. 44:557,1985. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

10. Levy, N.E, Lafuse, W.P., Savarirayan, S., Duran, L., Pease, L., and David, C.S. Gene order of the H-2D region. Fed. Proc. 44: 553, 1985. 11. David, C.S., Savarirayan, S., McCormick, J.F., Lafuse, W.P. Levy, N., Duran, L. and Pease, L. Genetics of the H-2D Region. Proceedings of the Seventeenth Miami Winter Symposium. Feb, 11-15,1985.

94 12. Lafuse, W.P., Hirose, S., McCormick, J.F., and C.S. David. Serologic evidence for gene conversion like events in mouse Ia genes. Proceedings of the Seventeenth Miami Winter Symposium Feb 11-15, 1985. 13. Lafuse, W.P., Berg, N., Savarirayan, S., and David, C.S. Mapping of a second recombination 'hot spot' within the I region of the mouse H-2 complex. Fed. Proc. 45:988, 1986. 14. Savarirayan, S. Hanson, J., McCormick, J., Lafuse, W.P., and David, C.S. Identification and characterization of new intra-H-2 recombinants. Fed. Proc. 46: 944, 1987. 15. Lafuse, W.P. and David, C.S. Recombination sites within the Mouse H-2 Complex. Conference on H-2 Gene complex: Genes, Molecules, Function. Bar Harbor, Maine, June 5- 9, 1987. 16. Lafuse, W.P. Castle, L.M., and David, C.S. Restriction Fragment Length Polymorphism Analysis of C-2 and Factor B genes in mouse H-2 recombinant mouse strains. FASEB J. 2:A1833, 1988. 17. Lafuse, W.P., Castle, L., and David, C.S. RFLP analysis of HSP70 genes in the mouse H-2 Complex. FASEB Journal 4, A2214, 1990. 18. Lafuse, W., Jung, J., Kaiser, M., and Zwilling, B. Characterization of macrophage IFN-gamma induced genes. FASEB Journal 5, A1346, 1991. 19. Lanning, D., Castle, L., Lafuse, W.P., and David, C.S. RFLP analysis of genes mapping in the S-D Region of the Mouse H-2 complex. FASEB Journal 5, A1682, 1991. 20. Lafuse, W.P., Brown, D., Castle, L., and Zwilling, B.S. Characterization of Novel Genes Induced in Macrophages by IFN-gamma. J. Leukocyte Biol. Supp. 3:53, 1992 21. Lafuse, W.P., Brown,D., Castle, L., and Zwilling, B.S. Characterization of cDNA's Induced in Macrophages by IFN-gamma. FASEB J. 6:A1972, 1992. 22. Jung, J., Lafuse, W.P., Marucha, P.T., and Sheridan,J.F. Patterns of cytokine gene expression in the draining lymph node and lung of Influenza virus infected mice. FASEB J. 8:A780,1994. 23. Lafuse, W.P., Brown, D., Hussain, S., and Zwilling, B. Characterization of a novel gene that is IFN-γ induced in mouse macrophages and encodes a GTP binding proten. J. Leukocyte Biol Suppl. 30, 1994 24. Hussain, S. and Lafuse, W.P. Differential expression of IFn-γ inducible genes in various murine macrophage cell lines. J. Leukocyte Biol. Supp. 32, 1994. 25. Jung, J., Lafuse, W.P., Marucha, P.T., and Sheridan, J.F. Differential Expression of Cytokine Genes in different microenvironments during an influenza viral infection: Effect of restraint stress. 9th Immunology Congress, 156, 1995. 26. Hussain, S. and Lafuse, W.P. Mg21 is an IFN-γ induced gene in mouse macrophages and encodes an intracellular protein with GTP-binding activity. 9th Immunology Congress, 528, 1995. 27. Brown, D.H., Lafuse, W.P., and Zwilling, B.S. Regulation of Nramp expression by GM-CSF activated macrophages: Differential effects of glucocorticoids. J. Leukocyte Biol. Supp., 24, 1995. 28. Zwilling, B,S., Lafuse, W., and Brown, D. Corticosterone regulation of mycobacterial resistance in BCG resistant and susceptible mice. J. Leukocyte Biol. Supp., 17, 1995. 29. Hussain,S., Lafuse, W.P., Brown,D., Castle, L., and Zwilling, B.S. Cloning and characterization of a novel gene induced by IFN-γ in mouse macrophages. FASEB Journal 10: A1339,1996. 30. Hussain, S., Zwilling, B.S., and Lafuse, W.P. Mycobacterium avium infected mouse macrophages show unresponsiveness to IFN-γ and defective JAK-STAT cell signaling pathway. FASEB J. 12: 1733, 1998. 31. Kuhn, D.E. Lafuse, W.P., and Zwilling, B.S. Iron transport by isolated phagolysosomes (PL) obtained from the RAW264.7 macrophage (Mϕ) cell line transfected with resistant or susceptible Nramp1. FASEB J. 12:1611, 1998. 32. Wang, T., Lafuse, W.P., and Zwilling, B.S. regulation of Toll-like receptor 2 expression by murine acrophages following infection with Mycobacterium avium. FASEB J. 14: A1064, 2000. 33. Kuhn, D.E., Lafuse, W.P., and Zwilling, B.S. Regulation of NRAMP1 mediated iron transport. FASEB J. 14:A1231, 2000. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

34. Lafuse, W.P., Alvarez, G.R., and Zwilling, B.S. Regulation of NRAMP1 mRNA stability by oxidants and protein kinase C. FASEB J. 14:A1231, 2000. 35. Lafuse, W.P., Alaverz, G.R., and Zwilling, B.S. Mycobacterium avium infection of macrophages induces protein binding to the 3’UTR of Nramp1 mRNA. J. Leukocyte Biol. Supplement, 2001. 36. Weatherby, K.M., Lafuse, W.P., and Zwilling, B.S. Increased resistance to mycobacterial growth by alpha-2 adrenergic stimulation of macrophages requires both nitric oxide and superoxide production. J.Leukocyte Biology, Supplement 2001. 37. Bishop, C.M., Kuhn, D.E., Lafuse, W.P. and Zwilling, B.S. The consensus transport signature sequence of Nramp1 is required for iron transport. J. Leukocyte Biol. Supplement, 2001.

96 38. Wang, T., Lafuse, W.P., and Zwilling, B.S. NFκβ-mediated TLR2 gene expression requires the transcription factor Sp1. J. Leukocyte Biol. Supplement, 2001. 39. Zhong, W., Lafuse, W.P., and Zwilling, B.S. Infection with Mycobacterium avium differentially regulates the expression of iron transport protein mRNA in murine peritoneal macrophages. J. Leukocyte Biol., Supplement 2001. 40. Lafuse, W.P. Alvarez, G.R., Curry, H.M., and Zwilling, B.S. Inhibition of IFN-γ induced gene expression by M. avium involves both TLR2 dependent and independent pathways. FASEB J. 19: A379, 2005. 41. Van Zandt, K.E., Zwilling, B.S., and Lafuse, W.P. The induction of Nramp1 (SLC11a1) by Francisella tularensis is dependent of TLR2. FASEB J. 19: A377, 2005. 42. Van Zandt, K., Sow, F., Lafuse, W., Satoskar, A., and Zwilling, B. The iron export protein Ferroportin 1 is induced in mycobacterial-infected macrophages and is localized to the mycobacteria-containing phagosome. Tuberculosis: From lab research to field trials. Keystone Symposia. 2007’ 43. Sow, F.B., Schlesinger, L.S., Satoskar, A.R., Zwilling, B.S., and Lafuse, W.P. Hepcidin is synergistically induced in mouse macrophages by mycobacteria and IFN-γ, and is present in the mycobacterial containing phagososome. The macrophage: Homeostasis, Immunoregulation and disease. Keystone Symposia 2007.

44. Cynthia H. Canan, Nandan S Gokhale, Bridget Carruthers, William P Lafuse, Larry S Schlesinger, Jordi B. Torrelles, Joanne Turner. Lung inflammation in old age alters immune function to Mycobacterium tuberculosis. Poster Keystone Symposia 1/25/15

45. Murugesan V.S. Rajaram, William P. Lafuse, Rachel L. Gearinger, Aleksandra I. Adamovich, Russell Ault, Larry S. Schlesinger. Molecular mechanisms of miR125b expression in human macrophages during mycobacterial infection. Poster Keystone Symposia 1/24/15

46. Cynthia H Canan, Nandan S Gokhale, William P Lafuse. Larry S Schlesinger, Jordi B Torrelles, Joanne Turner. Characterization of lung inflammation and its impact on macrophage function and in vivo infection with Mycobacterium tuberculosis in old age. Poster Keystone Symposium, 2/29/16

47. WP Lafuse, R Gearinger, S.E. Fisher, MT Bailey. Social stress-induced priming of innate immunity is associated with translocation of commensal lactobacilli to the spleen. Poster AAI meeting 5/15/16

48. William P Lafuse, Murugesan VS, Rajaram, Heather M Curry, Connor Nealer, Cynthian H Cannan, Juan I Moliva, Jordi B Torrelles, Joanne Turner, Larry S Schlesinger. Influence of aging on the phenotype and function of alveolar macrophages. Poster presentation at Aging and Immunity Symposium May 10-12, 2017 Rockville. MD

49. William P Lafuse, Murugesan VS, Rajaram, Heather M Curry, Connor Nealer, Cynthian H Cannan, Juan I Moliva, Jordi B Torrelles, Joanne Turner, Larry S Schlesinger. Influence of aging on the phenotype and function of alveolar macrophages. Poster presentation at AAI Immunology 2017, May12-16, 2017.

50. Lafuse WP, Rajaram MVS, Wu Q, Moliva JI, Torrelles JB, Turner J, Schlesinger LS. Phenotypic analysis of alveolar macrophage populations in old mice and their response to Mycobaterium tuberculosis infection. Immunology 2019 San Diego, CA. poster and oral presentation Brian M. M. Ahmer, Ph.D. Professor, Department of Microbial Infection and Immunity The Ohio State University

460 West 12th Ave 710 Biomedical Research Tower Columbus OH 43210 USA email: [email protected]

EDUCATION:

1990 B.S., Microbiology, Colorado State University, Fort Collins, Colorado 1994 Ph.D., Genetics and Cell Biology, Washington State University, Pullman, Washington

PROFESSIONAL CAREER:

09/87 - 05/90: Merit based work-study recipient in the lab of Dr. Barry Beaty, Department of Microbiology, Colorado State University, Ft. Collins CO. 09/90 - 05/94: NIH Biotechnology predoctoral training fellow with Dr. Kathleen Postle, Department of Genetics and Cell Biology, Washington State University, Pullman WA. 06/92 - 08/92: Internship with Dr. Richard Darveau, Bristol-Myers Squibb Pharmaceutical Research Institute, Seattle WA. 05/94 - 09/99: Postdoctoral researcher with Dr. Fred Heffron, Department of Molecular Microbiology and Immunology, Oregon Health Sciences University, Portland Oregon. 09/99 - 10/06: Assistant Professor, Department of Microbiology, Ohio State University, Columbus Ohio. 10/06 - 09/12: Associate Professor, Department of Microbiology, Ohio State University, Columbus Ohio. 10/12 - 06/18: Associate Professor, Department of Microbial Infection and Immunity, Ohio State University, Columbus Ohio. 06/18 - present: Professor, Department of Microbial Infection and Immunity, Ohio State University, Columbus Ohio.

HONORS AND PROFESSIONAL AFFILIATIONS:

8/86 - 5/90: Morrison-Knudsen Corporation Scholarship 8/87 - 5/90: Merit-based work-study funding from the State of Colorado 8/90 - 5/94: NIH Biotechnology Training Fellowship 1994 - present: Member of the American Society for Microbiology 1995: N. L. Tartar Research Fellowship, Medical Research Foundation of Oregon 2002 - present: Member of the OSU Center for Microbial Interface Biology 2007 - present: Member of the Targeted Investment in Excellence – Public Health Preparedness for Infectious Diseases 2007 - 2015: Editorial Board for Journal of Bacteriology 2010 - 2016: Editorial Board for Frontiers in Cellular and Infection Microbiology 2016 - present: Associate Editor for Frontiers in Cellular and Infection Microbiology 2013 - present: NIH study section member (BACP) PUBLICATIONS:

As of September 2019, these publications have been cited over 4,000 times and yield an h-index of 31. Search “Brian Ahmer” on Google Scholar for up-to-date citation information. Of the 58 publications listed, 28 are first or last author research articles, 18 are middle author research articles, eight are reviews or book chapters, and four are commentaries or editorials.

1. Jonathan T. Skare, Brian M. M. Ahmer, Carrie L. Seachord, Richard P. Darveau, and Kathleen Postle. 1993. Energy Transduction between Membranes: TonB, a Cytoplasmic Membrane Protein, Can Be Chemically Cross-Linked In Vivo to the Outer Membrane Receptor FepA. The Journal of Biological Chemistry 268: 16,302-16,308.

2. Brian M. M. Ahmer, Michael G. Thomas, Ray A. Larsen, and Kathleen Postle. 1995. Characterization of the exbBD Operon of Escherichia coli and the Role of ExbB and ExbD in TonB Function and Stability. Journal of Bacteriology 177: 4742-4747.

3. Andreas J. Baumler, Andries J. Gilde, Renee M. Tsolis, Adrianus W. M. van der Velden, Brian M. M. Ahmer, and Fred Heffron. 1997. Contribution of Horizontal Gene Transfer and Deletion Events to Development of Distinctive Patterns of Fimbrial Operons during Evolution of Salmonella Serotypes. Journal of Bacteriology 179: 317-322.

4. Brian M. M. Ahmer, Jeroen van Reeuwijk, Cynthia D. Timmers, Peter J. Valentine, and Fred Heffron. 1998. Salmonella typhimurium encodes an SdiA homolog, a putative quorum sensor of the LuxR family, that regulates genes on the virulence plasmid. Journal of Bacteriology 180: 1185- 1193.

5. Brian M. M. Ahmer, Mimi Tran, and Fred Heffron. 1999. The virulence plasmid of Salmonella typhimurium is self-transmissible. Journal of Bacteriology 181: 1364-1368.

6. Brian M. M. Ahmer, Jeroen van Reeuwijk, Patricia R. Watson, Tim S. Wallis, and Fred Heffron. 1999. Salmonella SirA is a global regulator of genes mediating enteropathogenesis. Molecular Microbiology 31: 971-982. (Chosen for commentary by Trends in Microbiology.)

7. Brian M. M. Ahmer and Fred Heffron. 1999. Salmonella typhimurium recognition of intestinal environments: Response. Trends in Microbiology 7: 222-223.

8. Robert I. Goodier and Brian M. M. Ahmer. 2001. SirA orthologs affect both motility and virulence. Journal of Bacteriology 183: 2249-2258. (Chosen for journal cover photo.)

9. Bindhu Michael, Jenee N. Smith, Simon Swift, Fred Heffron, and Brian M. M. Ahmer. 2001. SdiA of Salmonella enterica is a LuxR homolog that detects mixed microbial communities. Journal of Bacteriology 183: 5733-5742.

10. Jenee N. Smith and Brian M. M. Ahmer. 2003. Detection of other microbial species by Salmonella: Expression of the SdiA regulon. Journal of Bacteriology 185: 1357-1366.

11. Yumei Dong, A. Leonardo Iniguez, Brian M. M. Ahmer, and Eric W. Triplett. 2003. Kinetics and strain specificity of rhizosphere and endophytic colonization by enteric bacteria on seedlings of Medicago sativa and Medicago truncatula. Applied and Environmental Microbiology 69: 1783- 1790. 12. Max Teplitski, Robert I. Goodier, and Brian M. M. Ahmer. 2003. Pathways leading from BarA/SirA to motility and virulence gene expression in Salmonella. Journal of Bacteriology 185: 7257-7265.

13. Brian M. M. Ahmer. 2004. Cell to cell signaling in Escherichia coli and Salmonella enterica. Molecular Microbiology 52: 933-945. (2nd most clicked on article of the year for Molecular Microbiology.)

14. A. Leonardo Iniguez, Yuemei Dong, Heather D. Carter, Brian M. M. Ahmer, Julie M. Stone, and Eric W. Triplett. 2005. Regulation of enteric endophytic bacterial colonization by plant defenses. Molecular Plant- Microbe Interactions 18: 169-178.

15. Amber Lindsay and Brian M. M. Ahmer. 2005. The effect of sdiA on biosensors of N- acylhomoserine lactones. Journal of Bacteriology 187: 5054-5058.

16. Max Teplitski and Brian M. M. Ahmer. 2005. The control of secondary metabolism, motility, and virulence by the two-component regulatory system BarA/SirA of Salmonella and other γ- proteobacteria, pp 107-132, In B. M. Pruess (ed.), “Complex regulatory networks in enteric bacteria,” Research Signpost, Trivandrum, India.

17. Jeffrey B. VanDeusen, Manisha H. Shah, Brian Becknell, Bradley W. Blaser, Amy K. Ferketich, Gerard J. Nuovo, Brian M. M. Ahmer, Joan Durbin, Michael A. Caligiuri. 2006. STAT-1-mediated repression of monocyte interleukin-10 gene expression in vivo. European Journal of Immunology 36: 623-630.

18. Max Teplitski, Robert I. Goodier, and Brian M. M. Ahmer. 2006. Catabolite repression of the SirA regulatory cascade in Salmonella. International Journal of Medical Microbiology 296: 449-466.

19. Max Teplitski, Ali Al-Agely, and Brian M. M. Ahmer. 2006. Contribution of the SirA regulon to biofilm formation in Salmonella enterica serovar Typhimurium. Microbiology 152: 3411-3423.

20. Yakhya Dieye, Jessica L. Dyszel, Rebin Kader, and Brian M. M. Ahmer. 2007. Systematic analysis of the regulation of type three secreted effectors in Salmonella enterica serovar Typhimurium. BMC Microbiology 7: 3.

21. Brian M. M. Ahmer, Jenee N. Smith, Jessica L. Dyszel, and Amber Lindsay. 2007. Methods in cell- to-cell signaling in Salmonella. In Schatten, Heide; Eisenstark, Abe (Eds.), Salmonella: Methods and Protocols (pp. 307-322). Humana Press, New Jersey.

22. Jenee N. Smith, Jessica L. Dyszel, Jitesh A. Soares, Craig D. Ellermeier, Craig Altier, Sara D. Lawhon, L. Garry Adams, Vjollca Konjufca, Roy Curtiss III, James M. Slauch, and Brian M. M. Ahmer. 2008. SdiA, an N-Acylhomoserine Lactone Receptor, Becomes Active during the Transit of Salmonella enterica through the Gastrointestinal Tract of Turtles. PLoS One 3(7): e2826.

23. Jessica L. Dyszel, Jenee N. Smith, Darren E. Lucas, Jitesh A. Soares, Matthew C. Swearingen, Mathew A. Vross, Glenn M. Young, and Brian M. M. Ahmer. 2010. Salmonella enterica serovar Typhimurium can detect acyl homoserine lactone production by Yersinia enterocolitica in mice. Journal of Bacteriology 192: 29-37. (Selected for a commentary in Journal of Bacteriology and selected by Faculty of 1000.)

24. Jessica L. Dyszel, Jitesh A. Soares, Matthew C. Swearingen, Amber Lindsay, Jenee N. Smith, and Brian M. M. Ahmer. 2010. E. coli K-12 and EHEC genes regulated by SdiA. PLoS One 5(1): e8946.

25. J. T. Noel, J. Joy, Jenee N. Smith, M. Fatica, K. R. Schneider, Brian M. M. Ahmer, and Max Teplitski. 2010. Salmonella SdiA recognizes N-acyl homoserine lactone signals from Pectobacterium carotovorum in vitro but not in a bacterial soft rot. Molecular Plant-Microbe Interactions 23(3): 273-282.

26. Jitesh A. Soares and Brian M. M. Ahmer. 2011. Detection of acyl-homoserine lactones by Escherichia and Salmonella. Current Opinion in Microbiology 14(2):188-193.

27. Brian M. M. Ahmer and John S. Gunn. 2011. Interaction of Salmonella spp. with the intestinal microbiota. Frontiers in Microbiology 2:101. doi: 10.3389/fmicb.2011.00101

28. Rebecca G. Allen, William P. Lafuse, Jeffrey D. Galley, Mohamed M. Ali, Brian M. M. Ahmer, and Michael T. Bailey. 2012. The intestinal microbiota are necessary for stressor-induced enhancement of splenic macrophage microbicidal activity. Brain, Behavior, and Immunity 26: 371-382.

29. Matthew C. Swearingen, Steffen Porwollik, Prerak T. Desai, Michael McClelland, and Brian M. M. Ahmer. 2012. Virulence of 32 Salmonella strains in mice. PLoS One 7(4): e36043.

30. Anice Sabag-Daigle, Jitesh A. Soares, Jenee N. Smith, Mohamed E. Elmasry, and Brian M. M. Ahmer. 2012. The acyl-homoserine lactone (AHL) receptor, SdiA, of E. coli and Salmonella does not respond to indole. Applied and Environmental Microbiology 78:5424-5431.

31. Rocio Canals, Xiao-Qin Xia, Catrina Fronick, Sandra W. Clifton, Brian M. M. Ahmer, Helene L. Andrews- Polymenis, Steffen Porwollik and Michael McClelland. 2012. High throughput comparison of gene fitness among related bacteria. BMC Genomics 13: 212.

32. Anice Sabag-Daigle and Brian M. M. Ahmer. 2012. ExpI and PhzI are descendants of the long lost cognate signal synthase for SdiA. PLoS One 7(10):e47720.

33. Matthew C. Swearingen, Anice Sabag-Daigle, and Brian M. M. Ahmer. 2013. Are there acyl- homoserine lactones within mammalian intestines? Journal of Bacteriology 195:173-179.

34. Chien-Che Hung, Cherilyn D. Garner, James M. Slauch, Zachary W. Dwyer, Sara D. Lawhon, Jonathan G. Frye, Michael McClelland, Brian M. M. Ahmer, and Craig Altier. 2013. The intestinal fatty acid propionate inhibits Salmonella invasion through the post-translational control of HilD. Molecular Microbiology 87:1045-1060.

35. Steve Oghumu, Ran Dong, Sanjay Varikuti, Todd Shawler, Thomas Kampfrath, Cesar A. Terrazas, Claudio Lezama-Davila, Brian M. M. Ahmer, Caroline C. Whitacre, Sanjay Rajagopalan, Richard Locksley, Arlene H. Sharpe, and Abhay R. Satoskar. 2013. Distinct populations of innate CD8+ T cells revealed in a CXCR3 reporter mouse. The Journal of Immunology 190:2229-2240.

36. Fabien Habyarimana and Brian M. M. Ahmer. 2013. More evidence for secretion signals within the mRNA of Type 3 secreted effectors. Journal of Bacteriology 195:2117-2118. 37. Fabien Habyarimana, Matthew C. Swearingen, Glenn M. Young, Stephanie Seveau, and Brian M. M. Ahmer. 2014. Yersinia enterocolitica inhibits Salmonella enterica and Listeria monocytogenes cellular uptake. Infection and Immunity 82:174-183.

38. Brian M. M. Ahmer. 2014. In this issue of Gut Microbes. Gut Microbes 5:1. (This editorial was the beginning article of a special issue that I organized.)

39. Christopher A. Vakulskas, Archana Pannuri, Diana Cortés-Selva, Tesfalem R. Zere, Brian M. M. Ahmer, Paul Babitzke, and Tony Romeo. 2014. Global effects of the DEAD-box RNA helicase DeaD (CsdA) on gene expression over a broad range of temperatures. Molecular Microbiology 92:945-958.

40. Fabien Habyarimana, Anice Sabag-Daigle, and Brian M. M. Ahmer. 2014. The sdiA-regulated gene srgE encodes a type III secreted effector. Journal of Bacteriology 196:2301-2312.

41. Mohamed M. Ali, David L. Newsom, Juan F. Gonzalez, Anice Sabag-Daigle, Christopher Stahl, Brandi Steidley, Judith Dubena, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, Razvan Arsenescu, Prosper N. Boyaka, Steven Krakowka, Tony Romeo, Edward J. Behrman, Peter White, and Brian M. M. Ahmer. 2014. Fructose-asparagine is a primary nutrient during growth of Salmonella in the inflamed intestine. PLoS Pathogens 10(6): e1004209. (Covered in the press and selected by Faculty of 1000.)

42. Christopher A. Vakulskas, Anastasia H. Potts, Paul Babitzke, Brian M. M. Ahmer, and Tony Romeo. 2015. Regulation of bacterial virulence by Csr (Rsm) systems. Microbiology and Molecular Biology Reviews 79(2):193-224. (Chosen for journal cover photo.)

43. Anice Sabag-Daigle, Jessica L. Dyszel, Juan F. Gonzalez, Mohamed M. Ali, and Brian M. M. Ahmer. 2015. Identification of sdiA-regulated genes in a mouse commensal strain of Enterobacter cloacae. Frontiers in Cellular and Infection Microbiology 5:47.

44. Jason P. Mooney, Kristen L. Lokken, Mariana X. Byndloss, Michael D. George, Eric M. Velazquez, Franziska Faber, Brian P. Butler, Gregory T. Walker, Mohamed M. Ali, Rashaun Potts, Caitlin Tiffany, Brian M. M. Ahmer, Shirley Luckhart, and Renee M. Tsolis. 2015. Inflammation-associated alterations to the intestinal microbiota reduce colonization resistance against non-typhoidal Salmonella during concurrent malaria parasite infection. Scientific Reports 5:14603.

45. Vittorio Venturi and Brian M. M. Ahmer. 2015. Editorial: LuxR solos are becoming major players in cell-cell communication in bacteria. Frontiers in Cellular and Infection Microbiology 5:89. (This editorial was the beginning article of a special issue that I organized with Vittorio Venturi.)

46. Tesfalem R. Zere, Christopher A. Vakulskas, Yuanyuan Leng, Archana Pannuri, Anastasia H. Potts, Raquel Dias, Dongjie Tang, Bryan Kolaczkowski, Dimitris Georgellis, Brian M. M. Ahmer, and Tony Romeo. 2015. Genomic targets and features of BarA-UvrY (-SirA) signal transduction systems. PLoS One 10(12): e0145035.

47. Anice Sabag-Daigle, Henry M. Blunk, Juan F. Gonzalez, Brandi L. Steidley, Prosper N. Boyaka, and Brian M. M. Ahmer. 2016. The use of attenuated but metabolically competent Salmonella as a probiotic to prevent or treat Salmonella infection. Infection and Immunity 84:2131-2141.

48. Anice Sabag-Daigle, Henry M. Blunk, Anindita Sengupta, Jikang Wu, Alexander J. Bogard, Mohamed M. Ali, Christopher Stahl, Vicki H. Wysocki, Venkat Gopalan, Edward J. Behrman, and Brian M. M. Ahmer. 2016. A metabolic intermediate of the fructose-asparagine utilization pathway inhibits growth of a Salmonella fraB mutant. Scientific Reports 6: 28117.

49. Jeffrey D. Galley, Nicola M. Parry, Brian M. M. Ahmer, James G. Fox, Michael T. Bailey. 2017. The commensal microbiota exacerbate infectious colitis in stressor-exposed mice. Brain, Behavior, and Immunity, 60: 44-50.

50. Kristin E. Burnum-Johnson, Jennifer E. Kyle, Amie J. Eisfeld, Cameron P. Casey, Kelly G. Stratton, Juan F. Gonzalez, Fabien Habyarimana, Nicholas M. Negretti, Amy C. Sims, Sadhana Chauhan, Larissa B. Thackray, Peter J. Halfmann, Kevin B. Walters, Young-Mo Kim, Erika M. Zink, Carrie D. Nicora, Karl K. Weitz, Bobbie-Jo M. Webb-Robertson, Ernesto S. Nakayasu, Brian M. M. Ahmer, Michael E. Konkel, Vladimir Motin, Ralph S. Baric, Michael S. Diamond, Yoshihiro Kawaoka, Katrina M. Waters, Richard D. Smith, and Thomas O. Metz. 2017. MPLEx: a method for simultaneous pathogen inactivation and extraction of samples for multi-omics profiling. Analyst, 142: 442-448.

51. Mikayla A. Borton, Anice Sabag-Daigle, Jikang Wu, Lindsey M. Solden, Bridget S. O’Banion, Rebecca A. Daly, Richard A. Wolfe, Juan F. Gonzalez, Vicki H. Wysocki, Brian M. M. Ahmer, Kelly C. Wrighton. 2017. Chemical and pathogen-induced inflammation disrupt the murine intestinal microbiome. Microbiome, 5(1): 47.

52. Anice Sabag-Daigle, Anindita Sengupta, Henry M. Blunk, Pradip K. Biswas, Mary Claire Cron, Alexander Bogard, Edward J. Behrman, Venkat Gopalan, and Brian M. M. Ahmer. 2017. Salmonella FraE, an asparaginase homolog, contributes to fructose-asparagine but not asparagine utilization. Journal of Bacteriology, 199(22): e00330-17. (Chosen for a “spotlight” in the journal.)

53. Vittorio Venturi, Sujatha Subramoni, Anice Sabag-Daigle, and Brian M. M. Ahmer. 2018. Methods to Study Solo/Orphan Quorum-Sensing Receptors. In Livia Leoni and Giordano Rampioni (eds.), Quorum Sensing: Methods and Protocols, Methods in Molecular Biology, vol. 1673: 145–159.

54. Jikang Wu, Anice Sabag-Daigle, Thomas O. Metz, Brooke L. Deatherage Kaiser, Venkat Gopalan, Edward J. Behrman, Vicki H. Wysocki, and Brian M. M. Ahmer. 2018. Measurement of fructose- asparagine concentrations in human and animal foods. Journal of Agricultural and Food Chemistry, 66(1): 212–217.

55. Anice Sabag-Daigle, Jikang Wu, Mikayla A. Borton, Anindita Sengupta, Venkat Gopalan, Kelly C. Wrighton, Vicki H. Wysocki, and Brian M. M. Ahmer. 2018. Identification of bacterial species that can utilize fructose-asparagine. Applied and Environmental Microbiology, 84(5): AEM.01957-17.

56. Jikang Wu, Anice Sabag-Daigle, Mikayla A. Borton, Linnea F. M. Kopp, Blake E. Szkoda, Brooke L. Deatherage Kaiser, Stephen R. Lindemann, Ryan S. Renslow, Siwei Wei, Carrie D. Nicora, Karl K. Weitz, Young-Mo Kim, Joshua N. Adkins, Thomas O. Metz, Prosper Boyaka, Venkat Gopalan, Kelly C. Wrighton, Vicki H. Wysocki, Brian M. M. Ahmer. 2018. Salmonella-mediated inflammation eliminates competitors for fructose-asparagine in the gut. Infection and Immunity, 86(5): IAI.00945–17.

57. Anastasia H. Potts, Yinping Guo, Brian M. M. Ahmer, and Tony Romeo. 2019. Role of CsrA in stress responses and metabolism important for Salmonella virulence revealed by integrated transcriptomics. PLoS ONE, 14(1): e0211430.

58. Anindita Sengupta, Jikang Wu, Justin T. Seffernick, Anice Sabag-Daigle, Nicholas Thomsen, Tien- Hao Chen, Angela DiCapua, Charles E. Bell, Brian M. M. Ahmer, Steffen Lindert, Vicki H. Wysocki, Venkat Gopalan. 2019. Integrated use of biochemical, native mass spectrometry, computational and genome- editing methods to elucidate the mechanism of a Salmonella deglycase. Journal of Molecular Biology, 431: 4497-4513. RESEARCH SUPPORT:

Ongoing research support:

R01 AI143288 Ahmer, Wysocki, Wrighton (multi-PIs) 09/24/18 - 08/31/2023 “Microbial ecology of the inflamed intestine” The goal is to perform a multi-omics analysis of the Salmonella-infected murine intestine (metagenomics, metatranscriptomics, and metabolomics). $3,709,941 (≈$1,500,000 to Ahmer) Role: Contact PI

R01 AI140541 Ahmer, Gopalan (multi-PIs) 08/15/18 - 07/31/2022 NIH/NIAID “Salmonella-specific therapeutics” The goal is to perform high throughput small molecule screening, and in silico screening, to identify inhibitors of the Salmonella FraB enzyme. $2,758,857 (≈$1,000,000 to Ahmer) Role: Contact PI

R01 AI116119 Ahmer, Gopalan, Behrman, Wrighton, Wysocki (multi-PIs) 12/01/14 - 11/30/19 NIH/NIAID “Salmonella, colonization resistance, and fructose-asparagine” The goals are to determine the regulation and enzymology of the Fra proteins, and to use metagenomics to determine which organisms compete with Salmonella for fructose-asparagine and whether these organisms are disrupted by streptomycin or inflammation. $2,485,435 (≈$1,100,000 to Ahmer) Role: Contact PI

Completed research support:

OSU Foods for Health Discovery Theme-Food Innovation Center seed grant 06/01/17 - 04/30/18 “Impacts of Salmonella infection on the chemical and biological landscape of the gut” $25,000 Role: PI

Accelerator Award from the OSU Technology Commercialization Office 07/01/17 - 12/31/17 “A probiotic approach to Salmonella prevention” $50,000 Role: PI

R01 AI097116 Ahmer and Romeo (multi-PIs) 12/01/11 - 11/30/17 NIH/NIAID "Coordination of metabolism and virulence during infection" The goals are to use Salmonella as a model to identify the direct regulatory targets of SirA and CsrA and to study metabolic and virulence gene regulation of Salmonella in vivo. $1,881,875 ($938,000 to Ahmer) Role: Contact PI New investigator grant Habing, Ahmer, Wittum 10/01/14 - 9/30/16 OSU PHPID Stress, the microbiome, and food safety: Quantifying the impact of stress in farm animals on human microbial hazards $100,000 Role: co-PI

Dean’s Discovery Grant Ahmer, Gopalan, Bell 03/01/15 - 9/01/16 OSU College of Medicine “Therapeutics targeting a Salmonella deglycase” $25,000 Role: Contact PI

R01 AI073971 Ahmer (PI) 05/15/08 – 04/30/14 NIH/NIAID "Salmonella polymicrobial interactions" The goal is to develop novel techniques for studying coinfections between Salmonella and other organisms. $2,567,550 (≈$1,567,550 to Ahmer) Role: PI

Supplement to R01 AI073971 Ahmer (PI) 03/08/11 – 04/30/14 NIH/NIAID “Diversity supplement to Salmonella polymicrobial interactions” This supplement supports a postdoctoral researcher for two years. $165,856 Role: PI

Supplement to R01 AI073971 Ahmer (PI) 07/01/09 – 06/30/11 NIH/NIAID “Administrative supplement to Salmonella polymicrobial interactions” This administrative supplement is to purchase gnotobiotic animal isolators and an animal care technician. $177,620 Role: PI

R01 AI075093 McClelland (PI) 07/01/09 – 04/30/11 NIH/NIAID “Salmonella genes associated with colonization of specific hosts” The goal of this grant is to use high throughput methods to identify Salmonella genes required to infect different hosts. $185,000 to OSU Role: Subcontractor

R01 AI052237 McClelland (PI) 04/01/07 - 03/31/10 Subcontract on NIH/NIAID grant administered through Washington University. “A resource of Salmonella genome sequences” The goal of this grant is to sequence the genomes and determine the host range and virulence characteristics of 25 Salmonella serovars. $112,500 to OSU Role: Subcontractor

R56 AI50002 Ahmer (PI) 08/01/06 - 07/31/08 NIH/NIAID "Detection of mixed microbial communities by Salmonella" The goal of this project is to determine the N-acylhomoserine lactones detected by SdiA of Salmonella enterica and the genes regulated in response. $300,000 Role: PI

R21 AI057733 McClelland (PI) 09/01/04 - 08/31/06 NIH/NIAID "Salmonella Gene Expression in Complex Environments" The goal of this project was to develop technologies for studying microbial interactions in the gut. $65,297 to OSU Role: Subcontractor

R01 AI50002 Ahmer (PI) 08/01/01 - 05/31/06 NIH/NIAID "Detection of mixed microbial communities by Salmonella" The goal of this project was to determine the N-acylhomoserine lactones detected by SdiA of Salmonella enterica and the genes regulated in response. $862,138 Role: PI

Institutional Seed Grant Ahmer (PI) 11/01/00 - 10/31/01 American Cancer Society "The conserved function of SirA in gamma-proteobacteria" $15,000 Role: PI

Seed Grant Ahmer (PI) 11/01/99 - 10/31/00 Ohio State University "The conserved function of SirA in gamma-proteobacteria" $10,000 Role: PI

EXTERNAL SERVICE:

Reviewer of manuscripts for 27 different journals: mBio, mSystems, Nature Microbiology, Journal of Bacteriology, Molecular Microbiology, Proceedings of the National Academy of Sciences USA, PLoS Pathogens, PLoS One, Cellular Microbiology, Cell Research, Microbiology, Environmental Microbiology, Frontiers in Microbiology, Canadian Journal of Microbiology, Applied and Environmental Microbiology, Infection and Immunity, In Silico Biology, Trends in Immunology, Trends in Microbiology, Journal of Molecular Microbiology, BMC Microbiology, FEBS Letters, Veterinary Research, ACS Chemical Biology, Nature Structural and Molecular Biology, and Nature Chemical Biology, Environmental Microbiology and Environmental Microbiology Reports Reviewer of grant applications for DFG, BARD, NIH, NSF, and USDA: 2000: Member of an NIH Special Emphasis teleconference review panel 2001: Reviewer for an MBRS (Minority Biomedical Research Support Program) grant panel at NIH 2001: Ad-hoc reviewer for NSF 2002: Ad-hoc reviewer for NSF 2002: Reviewer for an NIH Special Emphasis Panel 2007: Ad-hoc reviewer for NSF 2007: Ad-hoc reviewer for an Israel Science Foundation grant 2007: Ad-hoc reviewer for OARDC Seeds grant 2007: Ad-hoc reviewer of a grant for BARD, the United States - Israel Binational Agricultural Research & Development Fund 2008: Invited member of a grant review panel for a German DFG Priority Program 2009: Ad-hoc reviewer for a DFG Priority Program grant review 2010: Ad-hoc reviewer for a DFG Priority Program grant review 2010: Panel member for a USDA grant review study section 2011: Ad-hoc reviewer for OARDC Seeds grant 2012: Member of an NIH Special Emphasis teleconference review panel 2013: Member of an NIH Special Emphasis teleconference review panel 2013 - present: Permanent member of NIH review panel, BACP.

Miscellaneous external service: 2005: External examiner of undergraduate honors students at Kenyon College, Gambier Ohio 2009: External examiner of undergraduate honors students at Kenyon College, Gambier Ohio 2016: External examiner of undergraduate honors students at Kenyon College, Gambier Ohio 2007: Reviewer of a promotion application for a research professor in Belgium 2008: Reviewer of a promotion application for a research professor in Belgium 2008: Reviewer of a tenure application in the USA 2008: Reviewer of an “Enhancement Grant for Research”, an internal competition at Sam Houston State University 2010: Reviewer of an application for promotion to full professor in the USA 2012: Reviewer of a tenure application in the USA 2013: Pre-review consultant for a grant proposal for University of Nebraska.

INSTITUTIONAL SERVICE:

2004 - 2013: Member of the OSU Institutional Biosafety Committee 2007 - 2014: Reviewer of graduate fellowship applications for the Targeted Investment in Excellence – Public Health Preparedness for Infectious Diseases 2010 - 2011: Presidential Fellowship review panelist (four panels) 2011 - 2014: Steering committee member for the Targeted Investment in Excellence – Public Health Preparedness for Infectious Diseases 2011: Reviewer of graduate fellowships for the Center for Microbial Interface Biology 2012: Reviewer of pilot grants for the OSU Center for Clinical and Translational Science 2016: Reviewer of fellowship applications for the CMIB T32 training grant 2016: Chair of the Organizing Committee for the 7th OSU CMIB Symposium COLLEGE SERVICE:

2000, 2001: Biosci Day 2003, 2004, 2008, 2010, 2012: Judge of the Denman undergraduate research forum 2003 - 2004, 2006 - 2008: Judge of the College undergraduate research colloquium 2014: Judge of the OSUWMC Research Day 2014 - present: Biomedical Sciences Graduate Program Graduate Studies Committee

DEPARTMENTAL SERVICE:

2001: Faculty Search Committee 2002: Faculty Search Committee 2002 - 2005: Undergraduate Research Coordinator 2003: Graduate Studies Committee 2004: Faculty Search Committee 2006: Faculty Search Committee 2007: Faculty Search Committee 2007: Departmental Review Committee 2008 - 2009: Undergraduate Curriculum Committee 2008 - 2009: Department Chair Search Committee 2009: Faculty Search Committee 2010 - 2012: Chair of Graduate Studies Committee 2013 - 2016: Awards committee 2016 - present: Chair of the Professional Development and Mentoring Committee

UNDERGRADUATE STUDENTS:

Rubeal Mann, 2000 Keith Bohman, 2000 Summer Harrish, 2000 Marylou Mandapat, 2001 Amber Lindsay, 2002 Scott Carney, 2003 Gabriela Tomescu, 2003 - 2004 Henaku Yirenkyi, 2006 Emily Bauman 2006 - 2007 James Scott, 2007 Olga Zemska, 2008 - 2009 Omar Mohammed, 2009 Krizia Melendez, 2009 - 2010 Teresa Kuo, 2010 - 2012 Mohamed Elmasry, 2011 - 2012 Christopher Stahl, 2011 - 2013 Jake Schick, 2013 Hunter Spitz, 2013 - 2014 Henry Blunk, 2013 - 2016 (This student wrote an Honor’s thesis) Elina Misicka, 2014 Sherry Zhang, 2014 - 2016 (This student wrote an Honor’s thesis) Alex Bogard, 2014 - 2016 Andrew Dempsey, 2015 - 2016 Bailyn Hogue, 2015 - 2016 (This student wrote an Honor’s thesis) Mary Claire Cron, 2015 - 2017 Tyler Ritchie, 2018 - present Anurup Krishna, 2018

GRADUATE STUDENTS:

Bindhu Michael, 1999 - 2001, M.S., "Detection of heterologous bacterial pheromones by SdiA, a LuxR homolog in Salmonella enterica serovar Typhimurium.” Bindhu published a first author paper in Journal of Bacteriology and presented a poster at the ASM national meeting. She is currently working at Merck.

Jenee Smith, 1999 - 2007, Ph.D., "Characterization of the LuxR homolog, SdiA, a transcriptional regulator activated by N-acylhomoserine lactone produced by other bacterial species." Jenee was an author on nine papers from my lab, two as first author in PLoS One and Journal of Bacteriology. She presented her work at three meetings, the Midwest Microbial Pathogenesis meeting in 2002, the ASM conference on Cell to Cell Signaling in 2004, and the 2006 ASM conference on Salmonella in Victoria, Canada. For this last meeting, Jenee was awarded a Student Travel Award and was chosen to give an oral presentation. She is currently working at the FDA in Washington DC.

Jessica Dyszel, 2001 - 2009, Ph.D., “Phenotypes of the LuxR Homolog, SdiA, in Salmonella and E. coli.” Jessica was an author on seven papers from my lab, with two as first author in PLoS One and Journal of Bacteriology. She presented at four meetings, the Ohio Branch of the ASM meeting in 2003 and 2004, and the ASM national meeting in 2004 and 2008. She is currently the Director of Research at Richter International in Columbus Ohio.

Amber Lindsay, 2002 - 2006, M.S., “Novel bacterial signal molecule biosensors.” Amber published a first author and a middle author research paper, and a first author methods paper. She is currently a Research Scientist at Battelle in Columbus Ohio.

Hunter Davis, 2011, non-thesis M.S., I was the official advisor of this student due to my Graduate Chairperson duties. This student did not perform research in my lab.

Brandi Steidley, 2011 - 2012, non-thesis M.S. I was the official advisor of this student due to my Graduate Chairperson duties. This student did not perform research in my lab.

Laura Schrader, 2011 - 2012, non-thesis M.S. I am the official advisor of this student due to my Graduate Chairperson duties. This student did not perform research in my lab.

Darren Lucas, 2006 - 2013, Ph.D., “Coordinated regulation of Salmonella virulence genes by the BarA/SirA two-component system and the Csr global regulatory system.” Darren was a middle author on one paper and presented a poster at the Midwest Microbial Pathogenesis conference. He went on to be a Molecular Microbiologist at Copernicus Therapeutics in Seattle WA.

Matthew Swearingen, 2008 - 2013, Ph.D., “Phenotypes of Salmonella sdiA in mice and pigs.” Matt presented a poster at the Midwest Microbial Pathogenesis conference, and published five papers, two as first author in PLoS One and Journal of Bacteriology. He is currently a faculty member at Florida Gulf Coast University.

Mohamed Ali, 2008 - 2014, Ph.D., “Studying the genetic determinants of Salmonella fitness in vivo.” Mohamed published a first author paper in PLoS Pathogens and four middle author papers. He also presented posters at six national conferences, and won three travel awards to attend these conferences. One was from ASM, one was from our local Food Innovation Center, and another was for best poster presentation at our local CMIB annual symposium. He is currently a faculty member at Mansoura University in Egypt.

Erin Connors, 2019 - present. Andrew Schwieters, 2019 - present. POSTDOCTORAL RESEARCH ASSOCIATES:

Robert I. Goodier, 1999 - 2001, “SirA control of secondary metabolism and motility in Salmonella.” Rob published a first author paper in Journal of Bacteriology and also had two middle author papers. He then took a position as Study Director at Q-One Biotech Ltd., Glasgow, Scotland.

Yakhya Dieye, 2002 - 2005, “Regulation of two Salmonella Type III secretion systems.” Yakhya published a first author paper in BMC Microbiology and a middle author in PLoS Pathogens. He is currently a faculty member at the Université Cheikh Anta Diop de Dakar, Sénégal.

Max Teplitski, 2002 - 2005, “Genetic and biochemical analysis of SirA function.” Max published three first author papers from my lab, and then went on to rise to Full Professor at the University of Florida.

Jitesh A. Soares, 2008 - 2010, “Identification of CsrA binding sites among Salmonella virulence gene transcripts.” Jay was a middle author on four publications from my lab, and the first author on a review article. He is currently a Managing Editor at ACS Chemical Biology, Washington DC.

Fabien Habyarimana, 2010 - 2016, “Characterization of the SdiA-regulated gene, SrgE, of Salmonella.” Fabien published first author papers in Journal of Bacteriology and Infection and Immunity, and also a first author commentary article for Journal of Bacteriology.

Anice Sabag-Daigle, 2010 - present, “Characterization of the SdiA regulon of Salmonella and Enterobacter, and characterization of the fructose-asparagine utilization pathway with regard to diagnostics and novel therapeutics including probiotics and small molecule inhibitors.” Anice has published eight papers in my laboratory, five as first author.

Juan F. Gonzalez, 2012 - 2015, “Microbiome characterization of Nramp1-/- mice.” Juan published three middle author papers in my lab, and is currently a postdoctoral researcher with Dr. John Gunn at Nationwide Children’s Hospital in Columbus OH.

Katherine Alena Miller, 2015 - 2016. “Characterization of SdiA regulon members of Salmonella.” TEACHING EXPERIENCE (SINCE 2000):

2000, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 44 students. I was the sole instructor of both lecture and laboratory components. In this first year, I developed all of the lectures and laboratory exercises from scratch. 2001, Spring: Microbial Pathogenesis, M724. Enrollment was 10 students. I provided one guest lecture. 2001, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 41 students. I was the sole instructor of both lecture and laboratory components.

2002, Spring: Mechanisms of Microbial Disease, M524.01. Enrollment was 44 students. I provided one two- hour guest lecture.

2002, Autumn: Molecular Basis for Microbial Diversity, M720. Enrollment was 14 students. I provided two two-hour lectures.

2002, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 58 students. I taught 50% of the lecture and 100% of the laboratory components.

2003, Spring: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 39 students. I taught 50% of the lecture and none of the laboratory components.

2003, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 54 students. I was the sole instructor of both lecture and laboratory components.

2004, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 58 students. I was the sole instructor of both lecture and laboratory components.

2005, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 65 students. I was the sole instructor of both lecture and laboratory components.

2006, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 68 students. I was the sole instructor of lecture and supervisor of the laboratory components.

2007, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 69 students. I was the sole instructor of lecture and supervisor of the laboratory components. 2007, Autumn: Microbial Pathogenesis, M724. Enrollment was 15 students. I provided one guest lecture 2008, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 59 students. I was the sole instructor of lecture and supervisor of the laboratory components. 2008, Autumn: Microbial Pathogenesis, M724. Enrollment was 14 students. I provided one guest lecture. 2009, Autumn: Microbial Genetics, M581. This is a ten week, five credit, undergraduate course with three lectures per week and two lab sections that each meet twice per week. This is a total of 30 contact hours for lecture and 80 contact hours for lab. Enrollment was 71 students. I was the sole instructor of lecture and supervisor of the laboratory components. 2009, Autumn: Microbial Pathogenesis, M724. Enrollment was 14 students. I provided one guest lecture. 2010, Spring: Microbial Genetics, M581.01. The course was altered so that the laboratory was a separate course number. It was now 30 contact hours of lecture with no laboratory. Enrollment was 71 students. I was the sole instructor for all lectures.

2011, Autumn: Microbial Genetics, M581.01. The course was altered so that the laboratory was a separate course number. It was now 30 contact hours of lecture with no laboratory. Enrollment was 77 students. I was the sole instructor for all lectures. *OSU switched from quarters to semesters in 2012, so all courses listed after this point are semester- length. 2013, Spring: Microbial Pathogenesis and Immunobiology, M4110. This is a 3 credit undergraduate course with three lectures per week. I taught 13 of the lectures.

2013, Spring: Microbial Genetics, M4130. This is a 3 credit undergraduate course with three lectures per week. I taught 12 of the lectures.

2014, Spring: Microbial Pathogenesis, BSGP7240/Micro7724. This is a 3 credit graduate course. I am the course organizer. I taught the first three lectures and then scheduled about 20 other professors to teach their specialty. The most challenging part is getting the professors scheduled so that the lectures occur in the proper sequence.

2015, Autumn: Microbial Pathogenesis, BSGP7240/Micro7724. This is a 3 credit graduate course. I am the course organizer. I taught the first six lectures and then scheduled about 20 other professors to teach their specialty. 2015, Autumn: Selected Topics in Microbial Pathogenesis, BSGP7400. I provided two 2-hour guest lectures. 2016, Spring: Microbial Pathogenesis and Immunobiology, M4110. I provided one guest lecture. 2017, Autumn: Microbial Pathogenesis, BSGP7240/Micro7724. This is a 3 credit graduate course. I am the course organizer. I taught the first five lectures and then scheduled about 20 other professors to teach their specialty.

2018, Spring: Microbial Pathogenesis and Immunobiology, M4110. I provided one guest lecture.

INVITED SEMINARS (SINCE 2000):

University of North Dakota in Grand Forks North Dakota, January 2000. “Regulatory Networks in Salmonella Pathogenesis.”

Children’s Hospital in Columbus Ohio, May 2000. “Salmonella detection of intestinal environments: Common themes in bacterial pathogenesis.”

Miami University (Ohio) in Oxford Ohio, September 2000. “Detection of normal flora pheromones by Salmonella.”

The 7th Annual Midwest Microbial Pathogenesis meeting in Columbus OH, October 2000. “True pheromone detection by E. coli and S. typhimurium.”

The University of Texas Southwestern Medical Center in Dallas Texas, November 2000. “Detection of normal flora pheromones by Salmonella.”

The American Society for Microbiology Conference on Cell-Cell Communication in Bacteria in Snowbird Utah, July 2001. “SdiA of Salmonella typhimurium is a LuxR homolog that detects mixed microbial communities.”

The Ohio State University Department of Food Science in Columbus Ohio, October 2001. “Pheromone detection and virulence gene regulation in Salmonella.”

Duquesne University in Pittsburgh Pennsylvania, December 2001. “Pheromone detection and virulence gene regulation in Salmonella.”

The Ohio Agricultural Research and Development Center in Wooster Ohio, April 2002. "Regulatory networks and pheromone detection in Salmonella virulence."

Sydney Kimmel Cancer Center in La Jolla California, June 2002. "Regulatory networks and pheromone detection in Salmonella virulence."

The University of Texas Health Science Center at San Antonio in San Antonio Texas, October 2002. "Pheromone detection and regulatory networks in Salmonella pathogenesis." The American Society for Microbiology Conference on Salmonella: Pathogenesis, Epidemiology, and Vaccine Development in Sardinia Italy, September 2003. “Detection of the host environment: SdiA and SirA.”

The Medical College of Ohio in Toledo Ohio, December 2003. “Detection of the host environment by Salmonella.”

Texas A&M University in College Station Texas, February 2004. “Detection of the host environment by Salmonella.”

Kenyon College in Gambier Ohio, February 2004. “Salmonella quorum sensing.”

Oregon Health and Sciences University in Portland Oregon, January 2005. “Use of resolvase IVET to record SdiA activity during transit of Salmonella through animals.”

Oregon State University in Corvallis Oregon, February 2005. “Use of resolvase IVET to record SdiA activity during transit of Salmonella through animals.”

National Institutes of Health Coinfections Workshop, August 2006. “Detection of other pathogens by Salmonella.”

Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, November 2006. “Detection of other pathogens by Salmonella.”

Miami University of Ohio, September 2007. “Salmonella can detect other pathogens in the host.” University of Florida, September 2009. “Salmonella can detect other pathogens in animals.” United Nations International Centre for Genetic Engineering and Biotechnology, Trieste, Italy, May 2011. "Identifying the functions of LuxR solos."

United Nations International Centre for Genetic Engineering and Biotechnology, Trieste, Italy, May 2011. "Identifying new bacterial interaction systems."

Indiana University School of Medicine, March 2012. “Dissecting microbial community interactions: Salmonella in the gut.”

Wright State University, May 2012. “Dissecting microbial community interactions: Salmonella in the gut.”

Indiana University, April 2013. “Use of high-throughput parallel mutant screening to identify interspecies bacterial interactions.”

University of Texas, San Antonio, February 2014. “Use of transposon site hybridization and gnotobiotic mice to Salmonella interactions with other microbes.”

University of Kansas, November 2014. “High-throughput genetic screening identifies the novel nutrient fructose-asparagine as important to Salmonella fitness in the inflamed gut.” University of Pennsylvania, November 2014. “High-throughput genetic screening identifies the novel nutrient fructose-asparagine as important to Salmonella fitness in the inflamed gut.”

University of Georgia, August 2015. “Fructose-asparagine utilization by Salmonella: drug target and diagnostic marker.”

University of Dayton, September 2015. “Fructose-asparagine utilization by Salmonella: drug target and diagnostic marker.”

Carnegie Mellon University, October 2016. “A random walk: genetic screening for Salmonella interactions with gut microbiota leads to the discovery of fructose-asparagine as a nutrient.”

Ohio Mass Spectrometry and Metabolomics Symposium, May 2018, “Impacts of Salmonella infection on the chemical and biological landscape of the gut.”

POSTER PRESENTATIONS AT MEETINGS (SINCE 2000): The presenter is underlined.

P. P. Kumar, K. Fluegal, Brian M. M. Ahmer and Susanne W. Lindgren. “Identification of OmpR/EnvZ Regulated Genes in Salmonella.” The General ASM national meeting, May 2000.

Robert I. Goodier and Brian M. M. Ahmer. “Virulence and flagellar gene expression are inversely affected by Salmonella sirA.” The 7th Annual Midwest Microbial Pathogenesis meeting, October 2000.

Robert I. Goodier and Brian M. M. Ahmer. “SirA orthologs affect both motility and virulence.” The General ASM national meeting, May 2001.

Bindhu Michael, Jenee Smith and Brian M. M. Ahmer. “SdiA of Salmonella typhimurium is a LuxR homolog that detects mixed microbial communities.” The General ASM national meeting, May 2001.

Brian M. M. Ahmer. “SdiA of Salmonella typhimurium is a LuxR homolog that detects mixed microbial communities.” Gordon Research Conference on Molecular Mechanisms of Microbial Adhesion, August 2001.

Jenee Smith and Brian M. M. Ahmer. "Detection of other microbial species by Salmonella: Expression of the SdiA regulon in vitro." The 9th Annual Midwest Microbial Pathogenesis meeting, September 2002.

Jessica L. Dyszel, Heather D. Carter, Jonathon Frye, Michael McClelland and Brian M. M. Ahmer. “Identification of Salmonella genes that are expressed in response to the presence of other bacterial species.” The Ohio Branch of the American Society for Microbiology meeting, April 2003.

Max Teplitski and Brian M. M. Ahmer. “Pathways leading from BarA/SirA to motility and virulence gene expression in Salmonella.” The Ohio Branch of the American Society for Microbiology meeting, April 2003.

Max Teplitski, Robert I. Goodier and Brian M. M. Ahmer. “Pathways leading from BarA/SirA to virulence and motility gene expression in Salmonella.” The Molecular Genetics of Bacteria and Phages meeting, August 2003. Max Teplitski and Brian M. M. Ahmer. “Detection of the host environment: SdiA and SirA.” The ASM Conference on Salmonella: Pathogenesis, Epidemiology, and Vaccine Development, September 2003.

Max Teplitski, Jenee Smith, Robert I. Goodier and Brian M. M. Ahmer. “Detection of the host environment: SdiA and SirA.” The ASM Conference on Polymicrobial Diseases, October 2003.

Jessica L. Dyszel, Heather D. Carter, Jonathan Frye, Michael McClelland and Brian M. M. Ahmer. “Identification of Salmonella genes that are expressed in response to the presence of other bacterial species.” The Ohio branch ASM meeting, April 2004.

Max Teplitski, Jenee Smith, Robert I. Goodier and Brian M. M. Ahmer. “Crp/cya control virulence in Salmonella enterica by transcriptional regulation of sirA.” The General ASM national meeting, May 2004.

Jessica L. Dyszel, Heather D. Carter, Jonathan Frye, Michael McClelland, and Brian M. M. Ahmer. “Identification of Salmonella genes that are expressed in response to the presence of other bacterial species.” The General ASM national meeting, May 2004.

Jenee Smith, Craig D. Ellermeier, James M. Slauch, and Brian M. M. Ahmer. “SdiA activity recorded during transit of Salmonella through animals.” The ASM conference on Cell to Cell Signaling, July 2004.

Jenee N. Smith, Craig D. Ellermeier, Jessica L. Dyszel, Craig Altier, Glenn M. Young, Sara D. Lawhon, L. Garry Adams, Vjollca Konjufca, Roy Curtiss, III, James M. Slauch, Brian M. Ahmer. Detection of other pathogens by Salmonella. Poster abstract selected for a student seminar presentation at the ASM Salmonella conference, Victoria BC, September 2006.

Carlos A. Santiviago, Yakhya Dieye, Steffen Porwollik, Yipeng Wang, Nabil Arrach, Pui Cheng, Jenee N. Smith, Brian M. M. Ahmer, and Michael McClelland. “Global Identification of Genes Required for Salmonella Fitness in Mice by Microarray Hybridization of Transposon-Tagged Mutants.” The General ASM national meeting, May 2007.

Jessica L. Dyszel, Carlos A. Santiviago, Yakhya Dieye, Steffen Porwollik, Yipeng Wang, Pui Cheng, Jenee N. Smith, Michael McClelland, and Brian M. M. Ahmer. “Using ABACUS (Array Based Assay for Cistrons Under Selection) to identify Salmonella genes required for colonization of different host species.” Center for Microbial Interface Biology Retreat, The Ohio State University, June 2007.

Jessica L. Dyszel, Jenee N. Smith, and Brian M. M. Ahmer. “Fitness phenotypes of the Salmonella luxR homolog, sdiA, in animals.” The General ASM national meeting, May 2008.

Cecilia Go, Krystyna Patora, Brian Ahmer, John A. Robinson, Gabriele Varani and Venkat Gopalan. Developing novel antibacterials using cyclic peptide mimics of the protein subunit of bacterial RNAse P. Poster presentation at the annual BioOhio conference, Columbus OH, October 2008.

Jessica L. Dyszel, Jenee N. Smith, Glenn M. Young, and Brian M. M. Ahmer. Salmonella can detect the N- acylhomoserine lactone production of other bacterial pathogens in animals. Poster presentation at the ASM Salmonella conference, Aix en Provence France, October 2009. R. Canals, X. Xiao-Qin, S. Clifton, C. Fronick, Brian M. M. Ahmer, H. Andrews-Polymenis, and Michael McClelland. “High-throughput sequencing of pools of Salmonella mutants after selection in mice.” The General ASM national meeting, May 2010.

Michael McClelland, Steffen Porwollik, Brian M. M. Ahmer, Hee-Jeong Yang, Ping Cui, Lydia Bogomolnaya, and Helene Andrews-Polymenis. “A collection of multigene deletions in the Salmonella genome for efficient hierarchical screening.” The General ASM national meeting, May 2010.

Rocio Canals, X-Q Xia, S. W. Clifton, Brian M. M. Ahmer, Helene Andrews-Polymenis, and Michael McClelland. "Comparison of fitness among shared genes in Salmonella Typhimurium and Typhi grown in rich media." The General ASM national meeting, May 2011.

Mohamed M. Ali, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, David L. Newsom, Steven Krakowka, Peter White, and Brian M. M. Ahmer. “The use of massively parallel screening to identify novel interspecies interactions: Salmonella and Enterobacter in gnotobiotic mice.” The Midwest Microbial Pathogenesis conference, October 2011.

Darren E. Lucas, Jitesh A. Soares, Laura M. Patterson-Fortin, Tony Romeo, and Brian M.M. Ahmer. “Regulation of virulence genes by the carbon storage regulatory system (Csr) in Salmonella enterica.” The Midwest Microbial Pathogenesis conference, October 2011.

Mohamed M. Ali, Matthew C. Swearingen, David L. Newsom, Peter White, and Brian M. M. Ahmer. “The use of massively parallel screening to identify novel interspecies interactions: Salmonella and Yersinia in pigs.” The ASM conference on Cell to Cell signaling, November 2011.

Mohamed M. Ali, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, David L. Newsom, Steven Krakowka, Peter White, and Brian M. M. Ahmer. “The use of massively parallel screening to identify novel interspecies interactions: Salmonella and Enterobacter in gnotobiotic mice.” The ASM conference on Cell to Cell signaling, November 2011.

Anice Sabag-Daigle, Jessica L. Dyszel, and Brian M. M. Ahmer. “Identification of sdiA-dependent AHL- responsive genes in Enterobacter cloacae.” The ASM conference on Cell to Cell signaling, November 2011.

Fabien Habyarimana, Glenn M. Young, Stephanie Seveau, and Brian M. M. Ahmer. “Yersinia enterocolitica prevents Salmonella serovar Typhimurium internalization into non-phagocytic cells.” The ASM conference on Cell to Cell signaling, November 2011.

Mohamed M. Ali, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, David L. Newsom, Steven Krakowka, Peter White, and Brian M. M. Ahmer. “The use of massively parallel screening to identify novel genes required for the fitness of Salmonella in gnotobiotic mice.” The General ASM national meeting, May 2012.

Anice Sabag-Daigle, Jessica L. Dyszel, and Brian M. M. Ahmer. “Characterization of an SdiA regulon in a murine commensal.” The Midwest Microbial Pathogenesis conference, August 2013.

Fabien Habyarimana and Brian M. M. Ahmer. “The SdiA-regulated gene, srgE, encodes a T3SS2 effector.” The Midwest Microbial Pathogenesis conference, August 2013. Matthew C. Swearingen, Anice Sabag-Daigle, Brandi Steidley, Juan F. Gonzalez, Jacob W. Schick, Hunter Spitz, and Brian M. M. Ahmer. “Salmonella enterica serovar Typhimurium detects the N-acyl homoserine lactone production of the proteobacterial bloom during inflammation.” The Midwest Microbial Pathogenesis conference, August 2013.

Mohamed M. Ali, David L. Newsom, Juan F. Gonzalez, Anice Sabag-Daigle, Christopher Stahl, Brandi Steidley, Judith Dubena, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, Razvan Arsenescu, Steven Krakowka, Tony Romeo, Edward J. Behrman, Peter White, and Brian M. M. Ahmer. “A nutrient transporter is crucial to the fitness of Salmonella in the inflamed intestine.” The Midwest Microbial Pathogenesis conference, August 2013.

Fabien Habyarimana and Brian M. M. Ahmer. “The SdiA-regulated gene, srgE, encodes a T3SS2 effector.” The ASM Salmonella conference, October 2013.

Matthew C. Swearingen, Anice Sabag-Daigle, Brandi Steidley, Juan F. Gonzalez, Jacob W. Schick, Hunter Spitz, and Brian M. M. Ahmer. “Salmonella enterica serovar Typhimurium detects the N-acyl homoserine lactone production of the proteobacterial bloom during inflammation.” The ASM Salmonella conference, October 2013.

Mohamed M. Ali, David L. Newsom, Juan F. Gonzalez, Anice Sabag-Daigle, Christopher Stahl, Brandi Steidley, Judith Dubena, Jessica L. Dyszel, Jenee N. Smith, Yakhya Dieye, Razvan Arsenescu, Steven Krakowka, Tony Romeo, Edward J. Behrman, Peter White, and Brian M. M. Ahmer. “A nutrient transporter is crucial to the fitness of Salmonella in the inflamed intestine.” The ASM Salmonella conference, October 2013.

Juan F. González, Hunter Spitz, Brian M. M. Ahmer. “The Effect of Nramp1 on Microbiota.” The ASM Conference on Beneficial Microbes, September 2014.

Mikayla A. Jackson, J.F. Gonzalez, L.M. Solden, A. Sabag-Daigle, R. Wolfe, B.M.M. Ahmer, and K.C. Wrighton. “Battle for Gut Colonization: Identifying Taxa that can Withstand Inflammation Induced by Salmonella.” Poster presented at: Multi-omics for Microbiomes Conference, Pacific Northwest National Laboratory, September 2015.

Katherine A. Miller, Matthew C. Swearingen, Anice Sabag-Daigle, and Brian M. M. Ahmer. "A Salmonella sdiA mutant shows decreased ability to colonize the inflamed mouse gut." Poster presented at the ASM Microbe conference in Boston, May 2016.

Mikayla A. Jackson, Anice Sabag-Daigle, Lindsay M. Solden, Juan F. Gonzalez, Richard Wolfe, Brian M. M. Ahmer, Kelly C. Wrighton. “Community profiling the inflamed intestine.” Poster presented at the ASM Salmonella conference in Potsdam Germany, August 2016.

Anice Sabag-Daigle, Henry M. Blunk, Juan F. Gonzalez, Brandi L. Steidley, Prosper N. Boyaka, and Brian M. M. Ahmer. “The use of attenuated but metabolically competent Salmonella as a probiotic to prevent or treat Salmonella infection.” Poster presented at the ASM Salmonella conference in Potsdam Germany, August 2016.

Anice Sabag-Daigle, Henry M. Blunk, Anindita Sengupta, Jikang Wu, Alexander J. Bogard, Mohamed M. Ali, Christopher Stahl, Vicki H. Wysocki, Venkat Gopalan, Edward J. Behrman, and Brian M. M. Ahmer. “A metabolic intermediate of the fructose-asparagine utilization pathway inhibits growth of a Salmonella fraB mutant.” Poster presented at the ASM Salmonella conference in Potsdam Germany, August 2016. Anindita Sengupta, Nick Thomsen, Anice Sabag-Daigle, Jikang Wu, Vicki H. Wysocki, Brian M. M. Ahmer, Edward J. Behrman, Venkat Gopalan. “Uncovering the Mechanism of Action of a Salmonella Deglycase, a Potential Drug Target.” Chosen for poster presentation at the ASM Microbe conference in New Orleans, June 2017.

Mikayla A. Borton, Anice Sabag-Daigle, Jikang Wu, Lindsey M. Solden, Bridget S. O’Banion, Rebecca A. Daly, Richard A. Wolfe, Juan F. Gonzalez, Vicki H. Wysocki, Brian M. M. Ahmer, and Kelly C. Wrighton. “Winners and losers: Microorganisms impacted by inflammation in the gut.” Chosen for oral presentation at the ASM Microbe conference in New Orleans, June 2017.

Jikang Wu, Anice Sabag-Daigle, Brian M. M. Ahmer, Vicki H. Wysocki. “Quantification of fructose- asparagine, an acrylamide precursor, in human foods, animal foods, and mouse ceca.” Chosen for an oral presentation at the ASMS conference in Indianapolis, June 2017.

CONTINUING EDUCATION ACTIVITIES:

• NIH sponsored course on metabolic modeling at Pacific Northwest National Labs, Richland WA, 2012.

• ASM workshop on “Turning your science into a company”, Washington DC, 2014.

• Attended two day OSU CCTS mentorship workshop, 2015.

• Attended one day OSU Leadership Academy workshop, 2015.

• Ten-day course on metagenomics at the Woods Hole Marine Biological Laboratory, 2015.

• One-week course on starting a company, Concept Academy, by Rev1 Ventures, 2015.

• One week at the ICCB-Longwood small molecule screening facility at Harvard, 2016.

• Completed the University Institute for Teaching and Learning 3-part teaching enhancement program. The Ohio State University

NAME: Daniel J. Wozniak

CURRENT ACADEMIC TITLE: Professor

ADDRESS: Office: Department of Microbial Infection and Immunity, Microbiology Infectious Diseases Institute 704 Biomedical Research Tower 460 West 12th Avenue Columbus, OH, 43210

Office phone number and email address (614) 247-7629, [email protected]

EDUCATION:

Institution Dates Degree Field Aquinas College 1978-1982 B.S. Biology, cum laude The Ohio State 1984-1989 Ph.D. Microbiology University

DOCTORAL DISSERTATION:

Daniel J. Wozniak. 1989. Regulation and Structure-Function Studies of Pseudomonas aeruginosa Exotoxin A. The Ohio State University, Columbus, OH. Advisor: Darrell R. Galloway, Ph.D.

POSTDOCTORAL TRAINING:

Institution Dates Degree Field

University of 1989-1992 Postdoctoral Fellow Microbiology Tennessee

Funded by NIH Training grant (1 year) and Cystic Fibrosis Foundation Fellowship (2 years).

1 ACADEMIC APPOINTMENTS:

May 2019 – Present Professor and Vice Chair Microbial Infection and Immunity and Microbiology Professor, Microbiology The Ohio State University

November 2018 – Jan 2019 Professor and Interim Chair Microbial Infection and Immunity Professor, Microbiology The Ohio State University

October 2008 – Present Professor Microbial Infection and Immunity and Microbiology Professor, Microbiology The Ohio State University

October 2008 – May, 2011 Adjunct Professor Department of Microbiology and Immunology, Wake Forest University School of Medicine

July 2008 to October 2008: Professor Department of Microbiology and Immunology, Wake Forest University School of Medicine

July 2002 to July 2008 Associate Professor with tenure Department of Microbiology and Immunology, Wake Forest University School of Medicine

July 1998 to July 2002: Associate Professor Department of Microbiology and Immunology, Wake Forest University School of Medicine

November 1992 to June 1998: Assistant Professor Department of Microbiology and Immunology, Wake Forest University School of Medicine

November, 1989 to November, 1992: Post Doctoral Research Fellow Department of Microbiology and Immunology, University of Tennessee.

September 1984 to October 1989: Doctoral Student, Department of Microbiology, The Ohio State University

PROFESSIONAL APPOINTMENTS AND ACTIVITIES:

• 2020, Co-Chair CFF Infection Research Initiative Grant Review Panel • 2019-2023, Host Interaction Bacterial Pathogens, HIBP NIH Study Section Member 2 • 2018-Present, Co-Director, Microbial Communities Thematic Area, OSU Infectious Disease Institute. • 2017-2020, Editorial Board of Environmental Microbiology • 2008-2019, Standing member Cystic Fibrosis Foundation Research and Research Training Committee • 2018, NIAID, Drug discovery grant review panel • 2017, NIDDK CFF P30 grant review panel • 2016, NIH COBRE grant review panel • 2015, NIH, SREA grant review panel • 2014, NIH, ZRG1 IDM-S (02) grant review panel (served on two panels) • 2013-Present, NIH, IDM-B (80) grant review panel (served on four panels) • 2013, VA Merit Grant Review Panel, S13 Infectious Diseases-B • 2012-2016, Professional Consultant, Pfizer, Medimmune, Sorrento Therapeutics • 2010 – 2014, NIH College of CSR Reviewers • 2010-2012, Co-chair, chair, AHA peer review committee • 2001-2020, Editorial Board of Journal of Bacteriology • 2008-2010, Editorial Board, Molecular Microbiology • 2005-2008, Ad hoc grant reviewer, Cystic Fibrosis Foundation • 2007-2008, Ad hoc grant reviewer, NIH LCMI Study Section • 2005, Chair ZRG1 IDM NIH Study Section • 2002-2006, Bacteriology and Mycology, BACP Study Section Member, NIH • 2001-2002 Ad hoc grant reviewer, Bacteriology and Mycology (2) Study Section Member, NIH • 2001-2003 Professional Consultant, Abbott Laboratories • 2000 Ad hoc grant reviewer, Cystic Fibrosis Foundation • 2000 Ad hoc grant reviewer, American Cancer Society • 1997-2000 Ad hoc grant reviewer, NIH • 1995-Present ad hoc grant reviewer, National Science Foundation • 2000-Present ad hoc reviewer, Proc. Natl. Acad. Sci. USA • 2005-Present ad hoc reviewer, PLoS Pathogens • 1995-Present ad hoc reviewer, Molecular Microbiology. • 1992-Present ad hoc reviewer, Journal of Bacteriology • 1992-Present ad hoc reviewer, Microbiology

INSTITUTIONAL SERVICE (Since 2004):

2004-2005 2005-2006 Biosafety Biosafety Faculty recruitment committee (4) Faculty recruitment committee (2) Committee for undergraduate Committee for undergraduate medical Phase 2 medical education committee education Phase 2 medical education committee Planning committee, faculty research retreat

2006-2007 2007-2008 Biosafety Biosafety Faculty recruitment committee (2) Committee for undergraduate medical 3 education Committee for undergraduate medical education Recombinant DNA guidelines committee Recombinant DNA guidelines committee RAC subcommittee space guidelines RAC subcommittee space guidelines LCME Basic Science Committee LCME Basic Science Committee LCME Self Study Committee

2009-2011 2011-2013 Faculty recruitment committee (2) Chair faculty recruitment committee Graduate education committee Chair, Promotion and Tenure committee Microbiology Graduate Program committee Microbiology graduate program committee OSU College of Medicine education curriculum committee OSU College of Medicine appointment, promotion and tenure committee

2014-2016 2017-2019 Chair, Department AP&T committee Chair, Department AP&T committee Microbiology graduate program committee Co-director OSU Microbial Communities RAG and OSU Data Analytics Infectious Disease Institute Co-director CFF C3 Research Development Program 2019-Present Microbial Infection and Immunity Chair Department Vice Chair Recruitment Committee Faculty Executive Committee College of Medicine Investigation Chair, Faculty Recruitment Committee Committee Chair, Department AP&T committee Co-director OSU Microbial Communities, Infectious Disease Institute C-director, NIH T32 “Interdisciplinary Program in Microbe-Host Biology” Co-director CFF C3 Research Development Program Director, Department Faculty Development Program

PROFESSIONAL MEMBERSHIPS AND SERVICE:

Memberships: • American Society for Microbiology (Division D; Member since 1986) • American Association for the Advancement of Science • Beta Beta Beta National Biological Honor Society Service: • 2020, CFF RDP site visit team, Case Western Reserve and University of Pittsburgh • 2019, USA-Europe Biofilm Steering committee • 2019, SUNY Binghamton Biofilm Network site visit • 2018, CFF RDP site visit team, Case Western Reserve • 2017, 2019, CFF RDP site visit team, University of Pittsburgh 4 • 2017, Conference Chair, 61st Annual Wind River Conference on Prokaryotic Biology, Estes Park CO. • 2016, Workshop Chair, Pathogenesis of airway infections. North American Cystic Fibrosis Foundation • 2015, 7th International ASM Conference on Biofilms, Chicago, IL, Session chair, “Biofilms and Infection”. • 2015, 15th International ASM Conference on Pseudomonas, Washington, DC, Scientific committee and Session chair, “Biofilms”. • 2014, IWA Conference: The Perfect Slime – Nature, Properties, Regulation and Dynamics of EPS, Essen Germany (Program committee). • 2014, Workshop Co-Chair, North American Cystic Fibrosis Foundation • 2013, Organizing committee, Midwest Microbial Pathogenesis Conference. • 2013, 14th International ASM Conference on Pseudomonas, Lausanne, Switzerland, Symposium session chair, “Biofilms”. • 2012, 6th International ASM Conference on Biofilms, Miami, Symposium session chair, “Biofilm matrix”. • 2011, FASEB Summer Research Conference, Carefree AZ. Symposium session chair. • 2010, Center for Microbial Interface Biology Retreat Meeting Chair, The Ohio State University. • 2010, Mathematical Bioscience Institute at Ohio State, Meeting co-convener, Biofilms and Infectious Disease. • 2009, 5th International ASM Conference on Biofilms. Cancun, Mexico, Session Co- chair, “Cyclic-di-GMP signaling.” • 2009, External thesis appraiser, Carrie-Lynn Keiski, Toronto Hospital For Sick Children. • 2007, North American Cystic Fibrosis Conference. Anaheim, CA, Session Co-chair, “Interactions of Pathogens and the CF Host.” • 2007, 4th International ASM Conference on Biofilms., Quebec City, Canada, Session organizer, The Slime Matrix: What, Where, Why? • 2007, American Society for Microbiology Annual meeting symposium organizer. Toronto, CA, Biofilm vs. Planktonic: Molecular Nature of the Lifestyle Switch • 2007, Williamsburg CFF Conference. Pseudomonas working group participant. • 2007, Chair Mid-Atlantic Microbial Pathogenesis Meeting • 2006, American Association of Pharmaceutical Scientists. San Antonio, TX. Roundtable discussion leader, Bacterial biofilm as a new target in the discovery of new antibiotic drugs. • 2006, FASEB Summer Research Conference, Tucson AZ. Symposium session chair. • 2005, American Society for Microbiology Annual Meeting. Atlanta, GA. Symposium organizer. The matrix of pathogenic bacterial biofilms. • 2005, American Society for Microbiology Annual Meeting. Atlanta, GA. Colloquium organizer. Therapeutics Beyond Antibiotics. • 2005, Vice Chair Mid-Atlantic Microbial Pathogenesis • 2004-2005, American Society for Microbiology Division D Chair • 2003, Abbott Laboratories Biofilm Advisory Group meeting, Chicago, IL. Effects of macrolides on biofilm formation in Pseudomonas aeruginosa.

HONORS AND AWARDS:

• 2020 Nominee, Ohio State University Distinguished Scholar 5 • 2019 XXVII International Prize "Saint Francis and Claire of Assisi" -Science and Research section, Consiglio Nazionale delle Ricerche, Italy • 2016, Fellow American Academy for Microbiology • 2016-2018 ASM Distinguished Lecturer • 2015, ASM Jack Kenney Award for Outstanding Service on the Journal of Bacteriology Editorial Board • 2015, OSU Medical School Faculty Excellence Award • 2012, OSU Microbial Infection and Immunity Excellence in Teaching Award • 1990-1992, Cystic Fibrosis Foundation Post Doctoral Fellow • 1989, National Institutes of Health Training Grant Fellow • 1988, National American Academy of Microbiology Presidents Fellowship Award • 1988, The Ohio State University Deans Fellowship Award • 1987 American Society for Microbiology Raymond W. Sarber Fellowship Award

PROFESSIONAL INTERESTS:

Dr. Wozniak’s research activities are focused on the pathogenesis of several bacteria that cause chronic, devastating infections in humans. In chronic airway infections and wounds. Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter are the most common nosocomial pathogens isolated and are consistently associated with high mortality rates. Resources spent treating such infections in the U.S. are estimated at ~ $25 billion annually. These infections are extremely difficult to control since the bacteria exhibit a biofilm-mode of growth rendering them resistant to antimicrobials and phagocytic cells. Collectively, our research activities are focused on: • Coordinate regulation of the virulence factors that contribute to P. aeruginosa pathogenesis. • Role of biofilm matrix components in promoting recalcitrance to host derived antimicrobials and cells • Experimental therapeutics for combating persistent biofilm infections. • Defining pathoadaptive processes and evolution of bacteria during infection. • Development of chronic animal models of infection that reproduce human disease. • Bacteriophage biology and therapy • Novel bacterial topoisomerase inhibitors

GRANTS:

Current:

• CFF NIH Unfunded award. Novel bacterial topoisomerase inhibitors targeting Gram- negative bacteria. 07/01/2020-06/30/2022. (Mitton-Fry P.I, Wozniak: Co-Investigator). • R01AI077628-10, The pel exopolysaccharide gene cluster of Pseudomonas aeruginosa. 07/01/2009– 03/31/2025. (Parsek, Wozniak, MPI). • R01. The role of CFTR during macrophage-mediated killing of bacteria. 04/01/2020 - 03/31/2025. (Kopp PI, Wozniak Co-Investigator). • Falk Research Foundation. Combating antibiotic resistance with novel bacterial topoisomerase inhibitors. 11/30/2019-11/29/2021 (Mitton-Fry P.I, Wozniak: Co- 6 Investigator) • R01AI146252, Molecular mechanism of cardiac inflammation and dysfunction in Pseudomonas aeruginosa infection. 7/01/2019– 06/30/2024 (Rajaram, P.I., Wozniak co- investigator). • NIH 2R01EB017755-05, Mucin glycans in the regulation of microbial virulence. 05/01/2019 - 01/31/2023 (Ribbeck P.I., Wozniak co-investigator) • POUS001-0000763072. (Prime: BARDA). Development and characterization of the porcine animal model of ventilator-associated bacterial pneumonia caused by Pseudomonas aeruginosa. 11/14/2019-06/22/2020. (Bowman PI, Wozniak, Co- Investigator). • NIH, R01AI143916, The biofilm matrix of P. aeruginosa. 04/01/2019 - 03/31/2024. (Wozniak, Parsek, MPI) • T32AI112542-05, Interdisciplinary Program in Microbe-Host Biology. 08/15/2014 – 07/31/2020. (Wozniak, Justice, MPI). • Cystic Fibrosis Research Development Program. MCCOY19R0. 07/01/2019 - 06/31/2023 Impact of CF immune dysfunction on infection. (MPI: Wozniak, McCoy) • NIH, R01GM124436, The role of Staphylococcus aureus aggregate formation in establishing biofilms in chronic orthopedic periprosthetic joint infection. 09/01/2018 - 08/31/2022. (Stoodley, P.I., Wozniak, Co-I). • CFF, Novel Bacterial Type II Topoisomerase Inhibitors to treat MRSA. 09/01/2017 – 08/31/2019 (Mitton-Fry, PI. Wozniak, Co-I). • NIH, R01AI34895, Revisiting alginate paradigms. 09/01/2017 – 08/31/2022 (Wozniak, PI. Parsek, Co-I). • NIH 2R01AI077628, The pel exopolysaccharide gene cluster of Pseudomonas aeruginosa. 07/01/2014– 06/30/2020 (Parsek, P.I., Wozniak co-investigator). • 1R33AI119116-01. Enzyme therapeutics for biofilm prevention and disruption. 07/01/2017 – 06/30/2020. (Howell P.I., Wozniak, co-investigator).

Pending:

• AHA Rapid research grant, COVID 19 and its cardiovascular impact. Recombinant ACE2: A potential target for cardio protection and therapy for SARS-CoV2 infection. 7/01/2020 – 6/30/2023. Rajaram PI, Wozniak Co-I. • NIH NOT-AI-20-034. NOSI Funding Mechanism. Mechanism of ACEIs and rhACE2 mediated cardio protection during SARS-CoV2 infection. Rajaram PI, Wozniak Co-I • Cystic Fibrosis Foundation Infection Research Initiative. Managing infections in the era of highly effective modulator therapy. 7/01/2020 – 6/30/2023. Wozniak PI • NIH R21. Phage extracellular polysaccharide depolymerases to combat Pseudomonas aeruginosa biofilms. 10/01/2020 – 9/31/2022 Abedon PI, Wozniak Co-I • NIH R21. Analysis and enhancement of bacteriophage antibacterial activity in the presence of antibiotics. 07/01/2020 – 6/30/2022 Abedon PI, Wozniak Co-I • T32AI112542-05, Interdisciplinary Program in Microbe-Host Biology. 08/01/2019 – 07/31/2024. (Wozniak, Deora, MPI). • R01 Subaward (Site PI: Deora, Wozniak Co-I) Bioengineered multi-cell type organoids for airways disease modeling. 04/01/2021 - 03/31/2023. 7 • R01AI151132, Bacteriophage therapy for wound infections, 04/01/2020 - 03/31/2025 (Wozniak, PI).

Past Grant History

• Cystic Fibrosis Foundation, Biofilm-mediated killing of neutrophils by Staphylococcus aureus. 11/01/2016 – 10/31/2018. $186,000 Direct Cost. Wozniak, P.I. • Cystic Fibrosis Foundation, Postdoctoral fellowship GLOAG17FO (Mentor), Role of Pseudomonas aeruginosa pathoadapted variants in chronic infections. 04/01/17 – 03/31/19. $128,426 Direct Cost (Gloag P.I.). • Falk Medical Research Trust Catalyst Award. Novel therapies for MRSA and innovative methods for tackling bacterial resistance. 2/2018-2/2019. (Mitton-Fry, PI. Wozniak, Co- I). • NIH, R01NR0115676, Wound healing endpoint and recurrence, 05/06/2015 – 02/29/2020. (Sen, Gordilla, Roy MPI, Wozniak co-investigator). • NIH R01AI097511-01, Pseudomonas biofilms and immunity, 01/01/2013 – 12/31/2018. $1,336,122 Direct Cost, (Wozniak, P.I.). • NIH, R01NR013898, Biofilms and immunity in chronic wounds. 09/01/12 – 12/31/18. $1,399,765 Direct Cost (Wozniak/Sen, M.P.I.). • Cystic Fibrosis Foundation, Postdoctoral fellowship JONES17FO (Mentor), Systems- level analysis of Pseudomonas aeruginosa initial colonization events. 07/01/17 – 06/30/19. $128,426 Direct Cost (Jones P.I.). • NIH 1R41AI114252-01, Anti-Pseudomonas immunotherapy and targeted drug delivery. 07/01/2014– 06/31/2017 (Kaufmann P.I., Wozniak Co-I.). • Medimmune, Pseudomonas aeruginosa monoclonal antibody therapeutics. 10/01/2013 – 09/30/2017, $180,663 (Wozniak, P.I.). • NIH 1R21AI109448-01, Conserved complement binding to bacterial porins. 07/01/2014– 06/30/2017. (Wozniak, P.I.). • NIH 1R21AI109281 -01, The essential P. aeruginosa pan-genome during chronic infection. 12/01/2013– 11/30/2015 $275,000 Direct Cost (Wozniak, P.I., Whiteley, Sen, co- investigators). • PHPID, Biofilms in human medicine program grant. 11/01/2013-10/31/2015 (Wozniak, P.I., Bakaletz, Gunn, co-investigators). • NIH, 2-R01 HL58334-09, Control of P. aeruginosa algD transcription. 12/01/07 – 11/30/14 $1,250,000 Direct Cost (Wozniak P.I.). • Cystic Fibrosis Foundation, Postdoctoral fellowship Mishra11F0 (Mentor), Role of P. aeruginosa exopolysaccharides in modulating neutrophil function. 4/01/11 – 3/31/14. $80,000 Direct Cost (Mishra P.I.). • American Heart Association 0515325U (Mentor). The Pseudomonas aeruginosa AmrZ regulon. 07/01/12 – 06/30/14. $80,000 Direct Cost (Jones P.I.). • OSU College of Veterinary Medicine (T32 Mentor). Pathogenesis of Pseudomonas aeruginosa biofilm induced chronic skin wounds. 07/01/12 – 06/30/14. $90,000 (Chaney P.I.). • Pfizer, Pseudomonas aeruginosa monoclonal antibody therapeutics. 6/01/2013 – 05/31/2014 8 $77,410 (Wozniak, P.I.). • OSU COM Pilot Grant Program, Molecular interactions between bacterial surface of living Pseudomonas aeruginosa and bacterial exopolysaccharides as first step in biofilm formation. 11/16/2012-11/15/2013. $30,000 Direct Cost (Wozniak, P.I., Lower co- investigator). • DOD W81XWH-11-2-0142, Army Medical Res Acquisition Activity. Alginate oligomers to treat infectious microbial biofilms. 05/01/11 - 05/09/13. $1,398,000 Direct Cost (Sen P.I.). • NIH R56AI061396-01, The matrix of Pseudomonas aeruginosa biofilms, 07/01/10- 05/30/13, $ 473,766 Direct Cost, (Wozniak, P.I). • MAP Pharmaceuticals, Contract, Anti-infective and anti-biofilm properties of gallium. 10/01/2010 – 09/30/2012. $69,670 Direct Cost (Wozniak, P.I.). • NIH, R01AI075081, Bordetella Biofilms and Pathogenesis. 09/15/08 – 08/31/12. $250,000 Direct Cost (Deora P.I.). • NIH F31AI078700, Ruth L. Kirschstein NRSA for Individual Predoctoral Fellowships, The Role of exopolysaccharides in Pseudomonas aeruginosa virulence. 4/01/08 – 3/31/12. $114,560 Direct Cost (Byrd, P.I., Sponsor – Wozniak). • Cystic Fibrosis Foundation Pilot and Feasibility Grant. Vaccines and adjuvant therapies to prevent Pseudomonas biofilm infections. 4/01/08 – 3/31/10. $80,000 Direct Cost, (Mizel, P.I). • American Heart Association Predoctoral Fellowship (Mentor). Structure-function and DNA- binding Properties of Pseudomonas aeruginosa AmrZ. 07/01/2008 - 07/01/2010. $40,000 Direct Cost, (Waligora P.I.). • NIH, 1-R03 AI076561, Pseudomonas Biofilms and Persistence. 06/01/08 – 05/31/10. $100,000 Direct Cost (Wozniak P.I.). • NIH R01AI061396-01, The matrix of Pseudomonas aeruginosa biofilms, 7/01/05 - 6/30/10, $1,786,867 Direct Cost, (Wozniak, P.I). • Cystic Fibrosis Foundation MA06F0 (Mentor). The role of Psl in P. aeruginosa biofilm development and pathogenesis. 5/01/06-4/30/09. $74,100 Direct Cost (Ma P.I.). • WFUSM Venture Fund. Mechanism of Pseudomonas extracellular polysaccharide regulation of neutrophil functional responses. 3/01/08 – 3/31/09. $20,000 Direct Cost, (McPhail, P.I). • WFUSM Venture Fund. Pseudomonas Biofilms and Persistence. 3/01/07 – 3/31/08. $20,000 Direct Cost, (Wozniak, P.I). • USDA CSREES NRI, 2006-01256 (Meetings Grant). Mid Atlantic Microbial Pathogenesis Meeting. $10,000. (Wozniak, P.I.) • NIH R13 AI073025-01 (Meetings Grant). Mid Atlantic Microbial Pathogenesis Meeting. $9,000. (Wozniak, P.I.). • Cystic Fibrosis Foundation WOZNIA06P0. Control of P. aeruginosa algD transcription, 12/01/06 – 11/30/06, $85,000 Direct Cost, (Wozniak, P.I). • American Heart Association 0515325U (Mentor). Negative Regulation of Flagellar Motility in Pseudomonas aeruginosa by the Alternative Sigma Factor AlgT. 07/01/05 – 06/30/06. $40,000 Direct Cost (Tart P.I.). • Cystic Fibrosis Foundation (Co-investigator) Host response to mucoid and motile P. aerguinosa 4/1/04 – 3/31/06, $120,000 Direct Cost (Sanchez P.I.) • NIH 1 R21 AI061396-01, The matrix of Pseudomonas aeruginosa biofilms. 07/01/2004 - 06/30/2005. $255,386 Direct Cost, (Wozniak P.I.). • NIH, 1-R01 AI 45779-01, Regulation of P. aeruginosa motility and virulence. 9/1/99 – 9 8/30/04 $864,603 Direct Cost, (Wozniak P.I.). • American Heart Association, 0215191U (Ramsey), Structural analysis and DNA-binding Properties of the Pseudomonas aeruginosa transcriptional activator AlgZ. 7/1/02 - 6/30/04, $36,400 Direct Cost. (Ramsey P.I.). • Abbott Laboratories, ABT-773 study agreement 200021, Mechanistic basis for non ribosomal effects of macrolide treatment on Pseudomonas aeruginosa infections. 7/1/01 – 6/30/03 $124,440 Direct Cost. (Wozniak P.I.). • Cystic Fibrosis Foundation, 6/01/99 - 5/31/02. Control of P. aeruginosa virulence traits by AlgR. $104,500 Direct Cost. (Erova, P.I.). • NIH, 1-R01-HL 58334, Control of Pseudomonas aeruginosa algD transcription. $508,394 Direct Cost. (Wozniak, P.I.). • NIH R-29 FIRST Award. 7/1/94-6/30/99. Role of AlgB in P. aeruginosa Alginate Gene Expression. $350,000 Direct Cost. (Wozniak, P.I.). • United Way/NIH Biomedical Research Support, 7/1/93-6/30/94. $6,000. (Wozniak, P.I.). • Cystic Fibrosis Foundation, 12/1/93-11/30/94. $73,000, (Wozniak, P.I.). • Cystic Fibrosis Foundation Post Doctoral Fellowship, $104,500, (Wozniak, P.I.).

BIBLIOGRAPHY:

Journal articles (h-index=54; total citations=12,724; Google Scholar)

130. Reichhardt, C., H. Jacobs, M. Matwichuk, C. Wong, D.J. Wozniak, and M.R. Parsek. 2020. The versatile Pseudomonas aeruginosa biofilm matrix protein CdrA promotes aggregate stability through different extracellular EPS interactions. J. Bacteriol. Submitted.

129. Locke, L.W., S. Kothandaraman, L. Gong, P.S. Stoodley, S. Vozar, S.L. Cole, M.F. Tweedle, and D.J. Wozniak. 2020. Evaluation of peptide-based probes towards in vivo diagnostic imaging of bacterial biofilm-associated infections. ACS Infect. Dis. Submitted

128. Sindeldecker, D., K. Moore, D.J. Wozniak, M. Anderson, D.H. Dusane, and P.S. Stoodley. 2020. Novel aminoglycoside tolerant “Phoenix” colony variants of Pseudomonas aeruginosa. Antimicrobial Agents and Chemotherapy. Submitted.

127. Pestrak, M.J., N.P. Murphy, Q. Wu, E.R. Lubbers, S. Dellos-Nolan, N. Saloughian, S. Patel, T. Thushara, C. Kashef, L. Yu, W.P. Lafuse, L.P. Ganesan, P.J. Mohler, D.J. Wozniak, and M.V.S Rajaram. 2020. MicroRNA-155 promotes cardiac inflammation, fibrosis and impair heart function during Pseudomonas aeruginosa infection. JCI Insight, Submitted

126. Harrison, J.J., H. Almblad, Y. Irie , D.J. Wolter, H.C. Eggleston, T.E. Randall, J.O. Kitzman, B. Stackhouse, J.C. Emerson, S. Mcnamara, T.J. Larsen, J. Shendure, L.R. Hoffman, D.J. Wozniak, and M.R. Parsek. 2020. Pseudomonas aeruginosa flagellar mutants exhibit elevated exopolysaccharide production: Implications for biofilm growth and cystic fibrosis infections. PLoS Genetics. Revised submitted.

125. Bhattacharya, M., T.M. Berends, R. Chan, V.J. Torres, and D.J. Wozniak. 2020. Leukocidins act in concert with the nuclease Nuc to prevent neutrophil mediated killing of Staphylococcus aureus 10 biofilms. Infect. Immun. In revision.

124. Okuma, A., Y. Lu, S. Dellos-Nolan, J. Papa, B. Koci, N.T. Cockroft, J. Gallucci, D.J. Wozniak, J.C Yalowich, and M. J. Mitton-Fry. 2020. Novel bacterial topoisomerase inhibitors derived from isomannide. In Press.

123. Deng, B., Ghatak, S., Sarkar, S., Singh, K., Das Ghatak, P., Matthew-Steiner, S., Roy, S., Khanna, S., Wozniak, D.J., McComb, D.W., and C.K. Sen. 2020. Novel bacterial diversity and fragmented eDNA identified in hyper-biofilm forming Pseudomonas aeruginosa small colony variant. iScience, 23(2):100827. doi: 10.1016/j.isci.2020.100827.

122. Tang, J., Tu, S., Lin, G., Guo, H., Yan, C., Liu, Huang, L., Tang, N., Xiao, Y., Pope, R.M, Rajaram, M.V.S, Amer, A.O., Ahmer, B.M., Gunn, J.S., Wozniak, D.J., Tao, L., Coppola, V., Zhang, L., Langdon, W.Y., Torrelles, J.B, Lipkowitz, S., and J. Zhang. 2020. Sequential ubiquitination of NLRP3 by RNF125 and Cbl-b limits inflammasome activation and endotoxemia. J. Exp. Med., 217 (4):e20182091.

121. Coenye, T., B. Kjellerup, P.S. Stoodley, T. Bjarnsholt, the 2019 Biofilm Bash Participants. 2020. The future of biofilm research – report on the ‘2019 Biofilm Bash’. doi.org/10.1016/j.biofilm.2019.100012

120. Gloag, E.S., S. Fabbri, D.J. Wozniak, and P.S. Stoodley. 2020. Biofilm mechanics: implications in infection and survival. Biofilm. 2 (2020), 100017.

119. Wu, Q., A. Hossfeld, A. Gerberick, N.S. Esfahani, C. Tiwari, S. Mehra, L.P. Ganesan, D.J. Wozniak, and M. Rajaram. 2019. Mycobacterium tuberculosis enhances macrophage P- glycoproteins (MDR-1) expression and activity to promote intracellular survival. J. Infect. Dis. 10.1093/infdis/jiz405. 220(12): 1989-1998.

118. Wheeler, K.M., G. Carcamo-Oyarce, B.S. Turner, J.Y. Co, S. Lehoux, R.D. Cummings, D.J. Wozniak, and K. Ribbeck. 2019. Mucin glycans attenuate the virulence of Pseudomonas aeruginosa in infection. Nat. Microbiol. doi: 10.1038/s41564-019-0581-8. Epub 2019 Oct 14.

117. Gloag, E.S., C.W. Marshall, D. Snyder, G.R. Lewin, J.S. Harris, S.B. Chaney, M. Whiteley, V.S. Cooper, and D.J. Wozniak. 2019. Pseudomonas aeruginosa interstrain dynamics and selection of hyperbiofilm mutants during a chronic infection. mBio. 10.1128/mBio.01698-19.

116. Pestrak, M.J., P. Baker, S. Dellos-Nolan, P.J. Hill, D. Passos da Silva, H. Silver, I. Lacdao, D. Raju, M.R. Parsek, D.J. Wozniak, and P. Lynne Howell. 2019. Treatment with the Pseudomonas aeruginosa glycosyl hydrolase PslG combats wound infection by improving antibiotic efficacy and host innate immune activity. Antimicrob Agents Chemother. 10.1128/AAC.00234-19.

115. Li, L., A. Okumu, S. Dellos-Nolan, A. English, S. Vibhute, Y. Lu, K.L. Thomas, J.T. Seffernick, L. Azap, S. L. Cole, D. Shinabarger, L.M. Koeth, S. Lindert, J. C. Yalowich, D.J.

11 Wozniak, and M. J. Mitton-Fry. 2019. 1,3-dioxane-linked bacterial topoisomerase inhibitors with enhanced antibacterial activity and reduced hERG inhibition. Jul 12;5(7):1115-1128. doi: 10.1021/acsinfecdis.8b00375.

114. Malhotra, S., D. Hayes, Jr., and D.J. Wozniak. 2019. Mucoid Pseudomonas aeruginosa and regional inflammation in the cystic fibrosis lung. J. Cystic Fibrosis. 10.1016/j.jcf.2019.04.009.

113. Malhotra, S., D. Hayes, Jr., and D.J. Wozniak. 2019. Cystic Fibrosis and Pseudomonas aeruginosa: The host-microbial interface. Clin. Micro. Rev. 10.1128/CMR.00138-18.

112. Passos da Silva, D., M.L. Matwichuk, D.O. Townsend, C. Reichardt, D. Lamba, D.J. Wozniak, and M.R. Parsek. 2019. The Pseudomonas aeruginosa lectin LecB binds to Psl and stabilizes the biofilm matrix. Nat. Comm. 10.1038/s41467-019-10201-4.

111. Dusane, D., V Lochab, T. Jones, C. Peters, A. Das, S. Roy, C.K. Sen, V. Subramaniam, D.J. Wozniak, S. Prakash, and P.S. Stoodley. 2018. Electroceutical treatment of Pseudomonas aeruginosa biofilms. Nature Sci Rep. doi: 10.1038/s41598-018-37891-y.

110. Chandler, C.E., A.M. Horspool, P.J. Hill, D.J. Wozniak, J.W. Schertzer, D.A. Rasko, and R. K. Ernst. 2018. Genomic and phenotypic diversity among ten laboratory isolates of Pseudomonas aeruginosa PAO1. J. Bacteriol. 201(5) doi 10.1128/JB.00595-18.

109. Roy, S., S. Santra, A. Das, S. Dixith, M. Sinha, S. Ghatak, N. Ghosh, P. Banerjee, S. Khanna, S. Mathew-Steiner, P. Ghatak, B. Blackstone, H.M. Powell, V.K. Bergdall, D. J. Wozniak, and C.K. Sen. 2018. Staphylococcus aureus biofilm infection compromises wound healing by causing deficiencies in granulation tissue collagen. Ann. Surg. doi: 10.1097/SLA.0000000000003053.

108. Reichhardt, C., C. Wong, D. Passos da Silva, D.J. Wozniak, and M.R. Parsek. 2018. CdrA interactions within the Pseudomonas aeruginosa biofilm matrix can promote cellular packing and protect it from proteolysis. mBio. 9(5):e01137-18.

107. Li, Linsen, A. Okumu, S. Dellos-Nolan, Z. Li, K. Soumendrakrishna, J.C. Yalowich, D.J. Wozniak, and M.J. Mitton-Fry. 2018. Synthesis and anti-Staphylococcal activity of novel bacterial topoisomerase inhibitors with a 5-amino-1,3-dioxane linker moiety. Bio. Med. Chem. Lett. 28(14): 2477-2480.

106. Bhattacharya, M., E.T.M. Berends, R. Chan, E. Schwab, S. Roy, C.K. Sen, V.J. Torres, and D.J. Wozniak. 2017. Staphylococcus aureus biofilms escape neutrophil mediated killing by releasing leukocidins to elicit extracellular trap formation. Proc. Natl. Acad. Sci. USA. 115(28):7416-7421.

105. Gloag, E.S., G.K. German, P. Stoodley, and D.J. Wozniak. 2017. Viscoelasticity of Pseudomonas aeruginosa biofilms: implications as a virulence factor resisting clearance from the lung. Nature Sci Rep. doi: 10.1038/s41598-018-28009-5.

12 104. Malhotra, S., D.H. Limoli, A.E. English, and D.J. Wozniak. 2018. Mixed communities of mucoid and nonmucoid Pseudomonas aeruginosa exhibit enhanced resistance to host antimicrobials. mBio. mBio. 2018 Mar 27;9(2). pii: e00275-18. doi: 10.1128/mBio.00275-18.

103. Locke, L.W., S.B. Chaney, S. Kothandaraman, M. Tweedle, D.J. Wozniak, and L.S. Schlesinger, 2017. Assessment of a leukocyte-targeted peptide probe as a potential tracer for imaging the tuberculosis granuloma. Tuberculosis. Jan;108:201-210. doi: 10.1016/j.tube.2018.01.001. Epub 2018 Jan 8.

102. Pestrak, M.J., S.B. Chaney, H.C. Eggleston, S.S. Mathew-Steiner, K. Ganesh, S. Roy, M.R. Parsek, C.K. Sen, and D.J. Wozniak. 2018. Pseudomonas aeruginosa rugose small colony variants inhibit host immunity, are hyper-inflammatory, and persist in multiple host environments. PLoS Pathogens, 2;14(2):e1006842. doi: 10.1371/journal.ppat.1006842. eCollection 2018 Feb.

101. Ray, V.A., P.J. Hill, C.K. Stover, S. Roy, C.K. Sen, D.J. Wozniak, and A. DiGiandomenico. 2017. Disruption of Pseudomonas aeruginosa biofilms by novel immunotherapeutics targeting Psl exopolysaccharide. Nat. Sci. Rep. Nov 22; 7(1): 16065. Doi:10.1038/s41958-017-16245-6.

100. Stella, N.A., K. Brothers, J. Callaghan, A. Pesserini, C. Sigindere, P.J. Hill, D. J. Wozniak, and R.M.Q. Shanks. An Iga/UmoB-family protein from Serratia marcescens regulates motility, capsular polysaccharide, and secondary metabolite production. Appl. Env. Microbiol. 2018 Mar 1;84(6). pii: e02575- 17. doi: 10.1128/AEM.02575-17. Print 2018 Mar 15.

100. Jones, C.J., and D.J. Wozniak. 2017. Congo Red stain identifies matrix overproduction and is an indirect measurement for c-di-GMP in many species of bacteria. Methods Mol. Biol. 1657:147- 156. doi 10:1007/978-1-4939-7240-1_12.

99. Barki, K.G., Das, A., Dixith, S., Ghatak, P.D., Matthew-Steiner, S., Schwab, E., Khanna, S., D.J. Wozniak, Roy, S., and C.K. Sen. 2017. Electric field based dressing disrupts mixed-species bacterial biofilm infection and restores functional wound healing. Ann. . doi: 10.1097/SLA.0000000000002504.

98. Jones, C.J., and D.J. Wozniak. 2017. Psl produced by mucoid Pseudomonas aeruginosa contributes to the establishment of biofilms and immune evasion. mBio. Doi:10.1128/mBio.00864- 17.

97. Hill, P.J., J.M. Scordo, J. Arcos, S.E. Kirkby, M.D. Wewers, D.J. Wozniak, and J.B. Torrelles. 2017. Modifications of the Pseudomonas aeruginosa cell envelope in the cystic fibrosis airway alters interactions with immune cells. Nat. Sci. Rep. doi:1038/s41598-017-05256

96. Chaney, S.B., K. Ganesh, S. Matthew-Steiner, P. Stromberg, S. Roy, C.K. Sen, and D.J. Wozniak. 2016. Histopathological comparisons of Staphylococcus aureus and Pseudomonas aeruginosa experimental infected porcine wounds. Wound Rep. Reg. 25(3): 541-549.

13 (DOI:10.1111/wrr.12527).

95. Xu, B., R.J. Soukup, C.J. Jones, R. Fishel, and D.J. Wozniak. 2016. Pseudomonas aeruginosa AmrZ binds to four sites in the algD promoter, inducing DNA-AmrZ complex formation and transcriptional activation. J. Bacteriol. 198:2673-2681.

94. Armbruster, C., D. Wolter, M. Mishra, H. Hayden, M. Radey, G. Merrihew, M. MacCoss, J. Burns, D.J. Wozniak, M.R. Parsek, and L. Hoffman. 2016. Staphylococcus aureus protein A mediates interspecies interactions at the cell surface of Pseudomonas aeruginosa. mBio. e00538- 16. doi: 10.1128/mBio.00538- 16.

93. Baker, P., P.J. Hill, B.D. Snarr, N. Alnabelseya, M.J. Pestrak, M.J. Lee, L.K. Jennings, J. Tam, R. Melnyk, M.R. Parsek, D.C. Sheppard, D.J. Wozniak, and P. Lynne Howell. 2016. Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms. Science Advances 2(5):e1501632.

92. Gunn, J.S., L.O. Bakaletz, and D.J. Wozniak. 2016. What’s on the outside matters: The role of the extracellular polymeric substance of Gram-negative biofilms in evading host immunity and as a target for therapeutic intervention. J. Biol. Chem. 291(24):12538-12546.

91. Xu, B., Y. Ju, R.J. Soukup, D.M. Ramsey, R. Fischel, V.H. Wysocki, and D.J. Wozniak. 2016. The Pseudomonas aeruginosa AmrZ C-terminal domain mediates tetramerization and is required for its activator and repressor functions. Env. Microbiol. Reports. 8:85-90.

90. Cooley, B., S. Dellos-Nolan, N. Dhamani, T. Ross, W. Waller, D.J. Wozniak, and V. Gordon. 2016. Asymmetry and inequity in the inheritance of a bacterial adhesive. N. J. Physics. 18(2016) 045019.

89. Swearingen, M., A. Mehta, A. Mehta, L. Nistico, P.J. Hill, A. Falzarano, D.J. Wozniak, L. Hall- Stoodley, and P. Stoodley. 2016. A novel technique using potassium permanganate and reflectance confocal microscopy to image biofilm extracellular polymeric matrix reveals non eDNA networks in Pseudomonas aeruginosa biofilms. Pathogens and Disease. Feb;74(1):ftv104. doi: 10.1093/femspd/ftv104. Epub 2015 Nov 3.

88. Baker, P., G.B. Whitfield, P.J. Hill, D.J. Little, M.J. Pestrak, H. Robinson, D.J. Wozniak, and P. Lynne Howell. 2015. Characterization of the Pseudomonas aeruginosa glycoside hydrolase PslG reveals that its levels are critical for Psl polysaccharide biosynthesis and biofilm formation. J. Biol. Chem. 290(47):28374- 28387.

87. Bhattacharya, M., D.J. Wozniak, P. Stoodley, and L.H. Stoodley. 2015. Prevention and treatment of S. aureus biofilms. Expert Rev. Anti-Infect. Ther. 13:1499-1516.

86. Xu, B., and D.J. Wozniak. 2015. Development of a novel method for analyses of Pseudomonas aeruginosa twitching motility and its application to define the AmrZ regulon. PLoS One.

14 10(8):e0136426.

85. Jennings, L.K., K.M. Storek, H.E. Ledvina, C. Coulon, L.S. Marmont, I. Sadovskaya, P.R. Secor, B.S. Tseng, M. Scian, A. Filloux, D.J. Wozniak, P.L. Howell, and M.R. Parsek. 2015. Pel is a cationic exopolysaccharide that cross-links extracellular DNA in the Pseudomonas aeruginosa biofilm matrix. Proc. Natl. Acad. Sci. U.S.A. 112(36):11353-11358.

84. Limoli, D.H., C.J. Jones, D.J. Wozniak. 2015. Bacterial extracellular polysaccharides in biofilm formation and function. Microbiology Spectrum. doi: 10.1128/microbiolspec.MB-0011- 2014.

83. Mishra, M., A. Ressler, L.S. Schlesinger, and D. J. Wozniak. 2015. Identification of OprF as a complement component C3 binding acceptor molecule on the surface of Pseudomonas aeruginosa. Infect. Immun. 83:3006-3014.

82. Silo-Suh, L.A., S.J. Suh, D.E. Ohman, D.J. Wozniak, and J.W. Pridgeon. 2015. Complete genome sequence of Pseudomonas aeruginosa mucoid strain FRD1 isolated from a cystic fibrosis patient. Genome Announcements. 19;3(2). pii: e00153-15.

81. Banerjee, J., P. Das Ghatak, S. Roy, S. Khanna, C. Hemann, B. Deng, A. Das, J.L. Zweier, D.J. Wozniak, and C.K. Sen. 2015. Silver-zinc redox-coupled electroceutical wound dressing disrupts bacterial biofilm. PLoS One. 10(3):e0119531.

80. Wang, S., X. Liu, L. Hongsheng, L. Zhang, Y. Guo, S. Yu, D.J. Wozniak, and L.Z. Ma. 2014. The exopolysaccharide Psl-eDNA interaction enables the formation of a biofilm skeleton in Pseudomonas aeruginosa. Env. Microbiol. Rep. 7(2): 330-340.

79. Limoli, D.H.W. and D.J. Wozniak. Mutagenesis by host antimicrobial peptides: insights into microbial evolution during chronic infections. 2014. Microbial Cell. 1 (7): 247-249.

78. Wozniak, D.J. and M.R. Parsek. Surface-associated microbes continue to surprise us in their sophisticated strategies for assembling biofilm communities. 2014. F1000 Prime Reports. 6:26; eCollection2014.

77. Roy, S., H. Elgharably, M. Sinha, K. Ganesh, E. Mann, S. Chaney, C. Miller, S. Khanna, V.K. Bergdall, H.M. Powell, C.H. Cook, G.M. Gordillo, D.J. Wozniak, and C.K. Sen. 2014. Mixed- species biofilm compromises burn wound healing by disrupting epidermal barrier function. J. Pathology. 233(4):331-343.

76. Limoli, D.H.W, A. Rockel, K. Host, A. Jha, B. Kopp, T. Hollis, and D.J. Wozniak. 2014. Cationic antimicrobial peptides promote microbial mutagenesis and pathoadaptation in chronic infections. PLoS Pathogens. 10(4): e1004083.

75. Jones, C.J., D. Newsom, B. Kelly, Y. Irie, L.K. Jennings, B. Xu, D. Limoli, J.J. Harrison, M.R.

15 Parsek, P. White, and D.J. Wozniak. 2014. ChIP-Seq and RNA-Seq reveal an AmrZ-mediated mechanism for cyclic-di-GMP synthesis and biofilm formation by Pseudomonas aeruginosa. PLoS Pathogens. 10(3): e1003984.

74. Elgharably, H., Mann, E., Awad, H., Ganesh, K., Das Ghatak, P., Gordillo, G., Sai-Sudhakar, C.B., Roy, S., Wozniak, D.J., and C.K. Sen. 2013. First evidence of sternal would biofilm following . PLoS One. 8(8):e70360

73. L. Ma, S. Wang, M.R. Parsek, and D.J. Wozniak. 2013. A spider-web mechanism of type IV pili- mediated migration to build a fiber-like Psl polysaccharide matrix in Pseudomonas aeruginosa biofilms. Env. Microbiol. 18(8):2238-2253.

72. Jones, C.J., C.R. Ryder, E.E. Mann, and D. J. Wozniak. 2013. AmrZ modulates Pseudomonas aeruginosa biofilm architecture by directly repressing transcription of the psl operon. J. Bacteriol. 195:1637-1644.

71. Irie, Y. Borlee, B.R., O’Connor, J.R. Hill, P.J., Harwood, C.S., D.J. Wozniak, and M.R. Parsek. 2012. Self-produced exopolysaccharide is a signal that stimulates biofilm formation in Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. U.S.A. 109:20632-20636.

70. E. Pryor, D.J. Wozniak, and T. Hollis. 2012. Crystallization of Pseudomonas aeruginosa AmrZ protein: Development of a comprehensive method for obtaining and optimization of protein – DNA crystals. Acta Crystallographica Section F. 1;68(Pt 8):985-93.

69. Zegans, M.E., D.J. Wozniak, E. Griffin, C.M. Toutain-Kidd, J.H. Hammond, A. Garfoot, and J.S. Lam. 2012. Pseudomonas aeruginosa exopolysaccharide Psl promotes resistance to the biofilm inhibitor polysorbate 80. Antimicrob Agents Chem. 56: 4112-4122.

68. L. Ma, J. Wang, S. Wang, C. Ryder, E.M. Anderson, J.S. Lam, M.R. Parsek, and D.J. Wozniak. 2012. Synthesis of multiple Pseudomonas aeruginosa biofilm matrix expolysaccharides is post- transcriptionally regulated. Env. Microbiol. 14(8):1995-2005.

67. E.E. Pryor, E.A. Waligora, B. Xu, S. Dellos-Nolan, D.J. Wozniak, and T. Hollis. 2012. The transcription factor AmrZ uses multiple DNA binding modes to recognize activator and repressor sequences of Pseudomonas aeruginosa virulence genes. PLoS Pathogens. 8(4): e1002648.

66. L. Ma, Wang, S., Wang, D., M.R. Parsek, and D.J. Wozniak. 2012. The roles of biofilm matrix polysaccharide Psl in mucoid Pseudomonas aeruginosa biofilms. FEMS Immunol. Med. Micro. 65(2):377- 380.

65. M. Mishra, M.S. Byrd, S. Sergeant, A. Azad, M.R. Parsek, L. McPhail, L.S. Schlesinger, and D.J. Wozniak. 2012. Pseudomonas aeruginosa Psl polysaccharide reduces neutrophil phagocytosis and the oxidative response by limiting complement-mediated opsonization. Cellular Micro. 14:95- 106.

16 64. K.M. Colvin, Y. Irie, C. S. Tart, R. Urbano, J. C. Whitney, C. Ryder, P. L. Howell, D. J. Wozniak, and M. R. Parsek. 2012. The Pel and Psl polysaccharides provide Pseudomonas aeruginosa structural redundancy within the biofilm matrix. Environ Microbiol. 4(8):1913-1928.

63. M. Alhede, K. Qvortup, M. Allesen-Holm, M. van Gennip, L.D. Christensen, P. Jensen, A.K. Nielsen, M.R. Parsek, D.J. Wozniak, S. Molin, T. Tolker-Nielsen, N. Hoiby, M. Givskov, and T. Bjarnsholt. 2011. Phenotypes of non-attached Pseudomonas aeruginosa aggregates resemble surface attached biofilm. PLoS One. 6(11):e27943.

62. E. Mann and D.J. Wozniak. 2011. Pseudomonas biofilm matrix composition and function. FEMS Microbiol. Rev: 36(4): 893-916

61. C. S. Lee, K. Wetzel, Buckley, T., D.J. Wozniak, and J. Lee. 2011. Rapid and sensitive detection of Pseudomonas aeruginosa in chlorinated water and aerosols targeting gyrB gene using real-time PCR. J. Appl. Microbiol. 111:893-903.

60. N.G. Cogan, J.S. Gunn, and D.J. Wozniak. 2011. Biofilms and infectious diseases: Biology to mathematics and back again. FEMS Microbiol. Lett. 322:1-7.

59. M.S. Byrd, B. Pang, W. Hong, E.A. Waligora, R.A. Juneau, C.E. Armbruster, K.E.D. Weimer, K. Murrah, E.E. Mann, H. Lu, A. Sprinkle, N.D. Kock, M.R. Parsek, D.J. Wozniak, and W.E. Swords. 2011. Direct evaluation of Pseudomonas aeruginosa biofilm mediators in a chronic infection model. Infect. Immun. 79:3087-3095.

58. L. H. Sanders, B. Devadoss, G.V. Raja, J. O’Connor, S. Su, D.J. Wozniak, D.J. Hassett, A.J. Berdis, and M. D. Sutton. 2011. Epistatic roles for Pseudomonas aeruginosa MutS and DinB (DNA Pol IV) in coping with reactive oxygen species-induced DNA damage. PLoS One. 2011;6(4):e18824.

57. K. Colvin, V. Gordon, K. Murakami, B. R. Borlee, D.J. Wozniak, G. Wong, and M.R. Parsek. 2011. The Pel polysaccharide can serve a structural and protective role in the biofilm matrix of Pseudomonas aeruginosa. PLoS Pathogens, 7(1): e10001264. Doi:10.1371/journal.ppat.10001264.

56. E.A. Waligora, D.M. Ramsey, E.E. Pryor, H. Lu, G.P. Sloan, T. Hollis, R. Deora, and D.J. Wozniak. 2010. AmrZ beta-sheet residues are essential for DNA-binding and transcriptional control of Pseudomonas aeruginosa virulence genes. J. Bacteriol., 192:5390-5401.

55. Y. Irie, M. Starkey, A. Edwards, D.J. Wozniak, T. Romeo, and M.R. Parsek. 2010. Pseudomonas aeruginosa biofilm matrix polysaccharide, Psl, is regulated transcriptionally by RpoS and post- transcriptionally by RsmA. Mol. Microbiol. 78:158-172.

54. M. S. Byrd, B. Pang, M. Mishra, W.E. Swords, and D.J. Wozniak. 2010. The Pseudomonas aeruginosa exopolysaccharide Psl facilitates surface adherence and NF-κB activation in A549 cells. mBio. 1(2):e00140-10.doi10.1128/mBio.00140-10.

17 53. Bazire, A., K. Shioya, E.S-Soutera, E. Bouffartigues, C. Ryder, L. G-Dombrowsky, G. Hemery, I. Linossier, S. Chevalier, D.J. Wozniak, O. Lesouhaitier, and A. Dufour. 2010. The sigma factor AlgU plays a key role in formation of robust biofilms by nonmucoid Pseudomonas aeruginosa. J. Bacteriol. 192:3001- 3010

52. B. Borlee, A. Goldman, Keiji Murakami, R. Samudrala, D.J. Wozniak, and M.R. Parsek. 2010. Pseudomonas aeruginosa uses a cyclic-di-GMP-regulated adhesin to reinforce the biofilm extracellular matrix. Mol. Microbiol. 75:827-842.

51. E. T. Weimer, S. Ervin, D.J. Wozniak, and S.B. Mizel. 2009. Immunization of young African green monkeys with OprF epitope 8-OprI-type A- and B-flagellin fusion proteins promotes the production of protective antibodies against nonmucoid Pseudomonas aeruginosa. Vaccine. 27:6762-6769.

50. M. S. Byrd, I. Sadovskaya, E. Vinogradov, H. Lu, A. B. Sprinkle, S. H. Richardson, L. Ma, B. Ralston, M. R. Parsek, E. M. Anderson, J. S. Lam, and D. J. Wozniak. 2009. Genetic and biochemical analyses of the Pseudomonas aeruginosa Psl exopolysaccharide reveal overlapping roles for polysaccharide synthesis enzymes in Psl and LPS production. Mol. Microbiol. 73:622-638.

49. E. T. Weimer, H. Lu, Kock, N.D., D.J. Wozniak, and S.B. Mizel. 2009. A fusion protein vaccine containing OprF epitope 8, OprI, and type A and B flagellins promotes enhanced clearance of nonmucoid Pseudomonas aeruginosa. Infect. Immun. 77:2356-2366.

48. M. Starkey, J. H. Hickman, L. Ma, N. Zhang, S. De Long, A. Hinz, S. Palacios, C. Manoil, M. J. Kirisits, T. Starner, D. J. Wozniak, C. S. Harwood, and M. R. Parsek. 2009. Pseudomonas aeruginosa rugose small colony variants have adaptations that likely promote persistence in the cystic fibrosis lung. J. Bacteriol. 191:3942-3503.

47. L. Ma, M. Conover, H. Lu, M. R. Parsek, K. Bayles, and D.J. Wozniak. 2009. Assembly and development of the Pseudomonas aeruginosa biofilm matrix. PLoS Pathogens. 5:e1000354.

46. Tart, A. H., and D. J. Wozniak. 2008. Shifting paradigms in Pseudomonas aeruginosa biofilm research. Curr. Top. Microbiol. Immunol. 322: 193-206.

45. R. W. Huigens, L. Ma, C. Gambino, P.D.R. Moeller, A. Basso, J. Cavanagh, D.J. Wozniak, and C. Melander. 2008. Control of bacterial biofilms with marine alkaloid derivatives. Mol. BioSyst. 4(6):614-621.

44. Belete, B., H. Lu, and D.J. Wozniak. 2008. Pseudomonas aeruginosa AlgR regulates type IV pilus biosynthesis by activating expression of the fimU-pilVWXY1Y2E operon. J. Bacteriol. 190:2023-2030.

43. Andrew J. Leech, A. Sprinkle, L. Wood, D.J. Wozniak, and D.E. Ohman. 2008. The NtrC- family regulator AlgB, which controls alginate biosynthesis in mucoid Pseudomonas aeruginosa, 18 binds directly to the algD promoter. J. Bacteriol. 190:581-589.

42. Hans-Curt Flemming, T, R. Neu, and D.J. Wozniak. 2007. The EPS matrix: The “house of biofilm cells.” J. Bacteriol. 189:7945-7947.

41. L. Ma, H. Lu, A. Sprinkle, M. R. Parsek, and D.J. Wozniak. 2007. Pseudomonas aeruginosa Psl is a galactose- and mannose-rich exopolysaccharide. J. Bacteriol. 189:8353-8356.

40. Ryder, C., M. Byrd, and D.J. Wozniak. 2007. Role of polysaccharides in Pseudomonas aeruginosa biofilm development. Current Opinion in Microbiology. 10:1-5.

39. Sanders, L.H., A. Rockel, H. Lu, D. J. Wozniak, and M.D. Sutton. 2006 Role of Pseudomonas aeruginosa dinB-encoded DNA polymerase IV in mutagenesis. J. Bacteriol. 188: 8573-8585.

38. Ma, L., K.D. Jackson, R.M. Landry, M.R. Parsek, and D.J. Wozniak. 2006. Analysis of Pseudomonas aeruginosa conditional Psl variants reveals roles for the Psl polysaccharide in adhesion and maintaining biofilm structure post-attachment. J. Bacteriol. 188: 8213-8221.

37. Tart, A.H., M.J. Blanks, and D. J. Wozniak. 2006. The AlgT-dependent transcriptional regulator AmrZ (AlgZ) inhibits flagellum biosynthesis in mucoid, non-motile Pseudomonas aeruginosa cystic fibrosis isolates. J. Bacteriol. 188. 6843-6849.

36. P. J. Baynham, D. M. Ramsey, B. V. Gvozdyev, E. M. Cordonnier, and D. J. Wozniak. 2006. The Pseudomonas aeruginosa ribbon-helix-helix DNA-binding protein AlgZ (AmrZ) controls twitching motility and biogenesis of type IV pili. J. Bacteriol. 188:132-140.

35. Anne H. Tart, M.C. Wolfgang, and D.J. Wozniak. 2005. The alternative aigma factor AlgT represses Pseudomonas aeruginosa flagellum biosynthesis by inhibiting expression of fleQ. J. Bacteriol. 187:7955- 7962.

34. D. J. Wozniak, K.D. Jackson, L. Ma, Y. Sanchez, M. Starkey, R. Landry, J. Hupp, and M.R. Parsek. 2005. The psl-encoded exopolysaccharide is required for cell-cell and cell-surface interactions: implications for understanding early events of P. aeruginosa biofilm development in CF. Pediatric , S6.3, 117-118.

33. D. M. Ramsey and D. J. Wozniak. 2005. Understanding the control of Pseudomonas aeruginosa alginate synthesis and the prospects for management of chronic infections in cystic fibrosis. Mol. Microbiol., 56:309-322.

32. D. M. Ramsey, P. J. Baynham, and D. J. Wozniak. 2005. Binding of Pseudomonas aeruginosa AlgZ to sites upstream of the algZ promoter leads to repression of transcription. J. Bacteriol. 187:4430-4443.

31. M. Mishra, G. Parise, K. Jackson, D.J. Wozniak, and R. Deora. 2005. The BvgAS signal 19 transduction system regulates biofilm development in Bordetella. J. Bacteriol., 187:1474-1484.

30. L. M. Cobb, J.C. Mychaleckyj, D.J. Wozniak, and Y.S. Lopez-Boado. 2004. Pseudomonas aeruginosa flagellin and alginate elicit very distinct gene expression patterns in airway epithelial cells: Implications for cystic fibrosis disease. J. Immunol. 173:5659-5670.

29. K. Jackson, M. Starkey, S. Kremer, M.R. Parsek, and D.J. Wozniak. 2004. Identification of psl, a locus encoding a potential exopolysaccharide that is essential for Pseudomonas aeruginosa PAO1 biofilm formation. J. Bacteriol. 186:4466-4475.

28. R. Keyser and D.J. Wozniak. 2004. Effects of sub-inhibitory concentrations of macrolide antibiotics on Pseudomonas aeruginosa. Chest. 125:62S-69S.

27. K. Jackson, R. Keyser, and D.J. Wozniak, 2003. The role of biofilms in airway disease. Sem. Resp. Crit. Care Med. 24:663-670.

26. D. J. Wozniak, A. Sprinkle, and P.J. Baynham, 2003. Control of Pseudomonas aeruginosa algZ expression by the alternative sigma factor AlgT. J. Bacteriol. 185:7297-7300.

25. D.J. Wozniak., T.J.O.Wyckoff, M. Starkey, R. Keyser, P. Azadi, G. O’Toole and. M. Parsek. 2003 Alginate is not a significant component of the extracellular polysaccharide matrix of Pseudomonas aeruginosa PA14 and PAO1 biofilms. Proc. Natl. Acad. Sci. U.S.A. 100:7907-7912.

24. Yoon, S.S, R. F. Hennigan, G. M. Hilliard, U. A. Ochsner, K. Parvatiyar, M. C. Kamani, H. Allen, T. DeKievit, P. R. Gardner, U. Schwab, J. J. Rowe, B. H. Iglewski, T. R. McDermott, T. Livinghouse, R. P. Mason, D. J. Wozniak, R. E.W. Hancock, R. C. Boucher and D. J. Hassett, 2002. Pseudomonas aeruginosa anaerobic respiration in biofilms: Relationships to cystic fibrosis pathogenesis. Dev. Cell. 3: 593-603.

23. Wyckoff, T.J.O., B. Thomas, D. J. Hassett, and D.J. Wozniak. 2002. Static growth of mucoid Pseudomonas aeruginosa selects for nonmucoid variants that have acquired flagella-dependent motility. Microbiology. 148: 3423-3430.

22. D. J. Hassett, J. Cuppoletti, B. Trapnell, J. J. Rowe, S. S. Yoon, G. M. Hilliard, K. Parvatiyar, M. C. Kamani , D. J. Wozniak, S.-H. Hwang, T. R. McDermott, and U. A. Ochsner. 2002. Anaerobic metabolism and quorum sensing by Pseudomonas aeruginosa biofilms in chronically infected cystic Fibrosis airways: Rethinking antibiotic treatment strategies and drug targets. Adv. Drug Del. Rev. 54:1425- 1443.

21. Whitchurch, C., T. Erova, S. Beatson, J. Sargent, J. Emery, J. Harris, R. Levesque, J. S. Mattick, and D. J. Wozniak. 2002. Phosphorylation of the Pseudomonas aeruginosa response regulator AlgR is essential for type IV fimbriae-mediated twitching motility. J. Bacteriol. 184: 4544-4554.

20. Woolwine, S., Sprinkle, AB, and D.J. Wozniak. 2001. Loss of Pseudomonas aeruginosa PhpA 20 aminopeptidase activity results in increased algD transcription. J. Bacteriol. 183: 4674-4679.

19. Wyckoff, T.J.O., and D.J. Wozniak. 2001. Transcriptional analysis of genes involved in Pseudomonas aeruginosa biofilms. Meth. Enzymology. 336:144-151.

18. Garrett, E.S., D. Perlegas, and D.J. Wozniak. 1999. Negative control of flagellin synthesis in Pseudomonas aeruginosa is modulated by the alternative sigma factor AlgT (AlgU). J. Bacteriol. 181: 7401-7404.

17. Baynham, P.J., A.L. Brown, L.L. Hall, and D.J. Wozniak. 1999. Pseudomonas aeruginosa AlgZ, a ribbon-helix-helix DNA binding protein, is essential for alginate synthesis and algD transcriptional activation. Mol. Microbiol. 33: 1069-1080.

16. Woolwine, S., and D.J. Wozniak. 1999. Identification of an Escherichia coli pepA homolog and its involvement in suppression of the algB phenotype in mucoid Pseudomonas aeruginosa. J. Bacteriol. 181: 107-116.

15. Ma, S., U. Selvaraj, R. Quarless, D.J. Hassett, D. E. Ohman, and D.J. Wozniak. 1998. Phosphorylation-Independent activity of the Pseudomonas aeruginosa response regulators AlgB and AlgR in promoting alginate biosynthesis. J. Bacteriol. 180: 956-968.

14. S. Ma, D.J. Wozniak, and D.E. Ohman. 1997. Identification of the histidine protein kinase KinB in Pseudomonas aeruginosa and its phosphorylation of the alginate regulator AlgB. J. Biol. Chem. 272: 17952-17960.

13. Richardson, S.H., and D.J. Wozniak. 1996. An ace up the sleeve of the cholera bacterium: A bacteriophage mediates the horizontal transfer of virulence genes between Vibrio cholerae with broad implications for the emergence of pathogenic strains. Nature Medicine (News and Views). 2: 853-855.

12. Baynham, P.J., and D.J. Wozniak. 1996. Identification and characterization of AlgZ, an AlgT- dependent DNA binding protein required for Pseudomonas aeruginosa algD transcription. Mol Microbiol. 22: 97-108.

11. Wozniak, D.J., X.Y. Han, and D.R. Galloway. 1995. Construction of a nontoxigenic strain of Pseudomonas aeruginosa for the production of recombinant exotoxin A. Appl. Env. Microbiol. 61:1739- 1744.

10. Walker, S.L., L.S. Hiremath, D.J. Wozniak, and D.R. Galloway. 1994. ToxR-mediated in vitro transcription of Pseudomonas aeruginosa toxA. Gene. 150: 87-92.

9. Wozniak, D.J., and D.E. Ohman. 1994. Transcriptional analysis of the Pseudomonas aeruginosa genes algR, algB, and algD reveals a hierarchy of alginate gene expression which is modulated by algT. J. Bacteriol. 176: 6007-6014. 21 8. Wozniak, D.J. 1994. Integration host factor and sequences 3’ of the Pseudomonas aeruginosa algD transcriptional start site are required for expression. J. Bacteriol. 176:5068-5076.

7. Hassett, D.J., W. Woodruff, D.J. Wozniak, M. L. Vasil, M.S. Cohen, and D.E. Ohman. 1993. Cloning and characterization of the Pseudomonas aeruginosa sodA and sodB genes encoding iron- and manganese cofactored superoxide dismutase: Demonstration of increased manganese superoxide dismutase activity in alginate-producing bacteria. J. Bacteriol. 175:7658-7665.

6. Ofek, I., K. Kabha, A. Athamna, G.Frankel, D.J. Wozniak, D.L. Hasty, and D.E. Ohman. 1993. Conjugal transfer of genetic determinants for capsular polysaccharide biosynthesis between Klebsiella pneumoniae strains of serotypes K2 and K21a. Infect. Immun. 61:4208-4216

5. Wozniak, D.J., and D.E. Ohman. 1993. Involvement of the alginate algT gene and integration host factor in the regulation of the Pseudomonas aeruginosa algB gene. J. Bacteriol. 175:4145- 4153.

4. Wozniak, D.J., and D.E. Ohman. 1991. Pseudomonas aeruginosa AlgB, a two-component response regulator of the NtrC-family, is required for algD transcription. J. Bacteriol. 173:1406- 1413.

3. Galloway, D.R., R.C. Hedstrom, J.L. McGowan, S.P. Kessler, and D.J. Wozniak. 1989. Biochemical analysis of CRM 66. J. Biol. Chem. 264:14869-14873.

2. Wozniak, D.J., L.H. Hsu, and D.R. Galloway. 1988. His 426 of the Pseudomonas aeruginosa exotoxin A is required for ADP-Ribosylation of elongation factor II. Proc. Natl. Acad. Sci. USA. 85:8880-8884.

1. Wozniak, D.J., D.C. Cram, C.J. Daniels, and D.R. Galloway. 1987. Nucleotide sequence and characterization of toxR: a gene involved in exotoxin A regulation in Pseudomonas aeruginosa. Nucleic Acids Research. 15:2123-2135.

Medical Education Publications:

Edmunds S, Givner L, and D.J. Wozniak. 2013. Lotta Coughlin: A case for problem based learning (PBL) in medical education. MedEdPORTAL, AAMC Publications. (http://www.mededportal.org/publication/9443.).

Book Chapters:

Pestrak, M.J. and D.J. Wozniak. 2020. Regulation of cyclic di-GMP signaling in Pseudomonas aeruginosa. In Microbial Cyclic Di-Nucleotide Signaling. Chou, Guiliani, Lee, and Romling eds. Springer Publishing, pp. 471-486.

22 Pestrak, M.J., H.C. Eggleston, and Daniel J. Wozniak. 2016. Genetics and regulation of ePS formation in Pseudomonas aeruginosa. In: “The Perfect Slime”. Hans-Curt Flemming, T. R. Neu, J. Wingender eds. IWA Publishing, pp. 79-112.

Limoli, D.H.W., C.J. Jones, and D.J. Wozniak. 2015. Bacterial extracellular polysaccharides in biofilm formation. Microbiol Spectr. 2015 Jun;3(3). doi: 10.1128/microbiolspec.MB-0011-2014. Review.

Ohman, D.E., K.M. Mathee, C.J. McPherson, C.A. Devries, S. Ma, D.J. Wozniak, and M.J. Franklin. Regulation of the alginate (algD) operon in Pseudomonas aeruginosa. In: “Molecular Biology of Pseudomonas”. T. Nakazawa, K. Furukawa, D. Haas, and S. Silver, eds. ASM Press, Washington, DC, p. 472-483.

Shumard, C.M., D.J. Wozniak, and D.R. Galloway. 1993. Regulation of Toxin A Synthesis in Pseudomonas aeruginosa. In: “Pseudomonas aeruginosa as an Opportunistic Pathogen”. H. Freidman, M. Bendinelli and M. Campa, eds. Plenum Press, New York, p. 107-121.

PATENTS:

US 20120064106 # 9,096,659; Flagellin fusion proteins and use thereof to induce immune responses against Pseudomonas aeruginosa.

10336-726PV1 Electroceutical dressing for wound care

INVITED PRESENTATIONS (Since 1998):

• 1998, University of Queensland, Brisbane Australia. The response regulator AlgR controls both twitching motility and alginate synthesis in Pseudomonas aeruginosa. • 1999, American Society for Microbiology National Meeting. "Signal Dependent Control of Bacterial Virulence." Coordinate control of motility and alginate synthesis in Pseudomonas aeruginosa. Chicago, IL. • 2000, International conference on biological response modification of macrolides. The clinical applications of macrolides in the new millenium. Macrolide treatment for the control of Pseudomonas aeruginosa biofilm formation in cystic fibrosis and diffuse panbronchiolitis. Seville, Spain. Sponsored by Dainabot Co., LTD. • 2000, Montana State University and Center for Biofilm Research. Coordinate control of motility and alginate synthesis in Pseudomonas aeruginosa: implications for understanding biofilm development. • 2000, University of Kent at Canterbury, UK. Motility and alginate production in Pseudomonas aeruginosa. • 2000, Florida International University. Inverse relationship of motility and alginate production in mucoid Pseudomonas aeruginosa. 23 • 2001, Abbott Laboratories. Non ribosomal effects of clarithromycin treatment on Pseudomonas aeruginosa infections. • 2001, American Society for Microbiology North Carolina State Meeting, Coordinate control of motility and polysaccharide synthesis in the cystic fibrosis pathogen Pseudomonas aeruginosa. • 2001, North Carolina State University, Genetics and regulation of Pseudomonas aeruginosa polysaccharides. • 2002, East Carolina University School Medicine. Genetics and regulation of Pseudomonas aeruginosa polysaccharides. • 2002, Ohio State University. Pseudomonas aeruginosa polysaccharide genetics and regulation in biofilms and CF airway infections • 2002, American College of Chest Annual Meeting. San Diego, CA. Effects of macrolides on biofilm formation in Pseudomonas aeruginosa. • 2003, State University of New York, Buffalo, Buffalo, NY. Regulation of alginate production in mucoid Pseudomonas aeruginosa. • 2003, Northwestern University, Chicago, IL. Polysaccharide genetics and regulation in P. aeruginosa biofilms. • 2003, University of Iowa, Iowa City, IA. Role of exopolysacchardies in Pseudomonas aeruginosa biofilm development. • 2003, American College of Chest Physicians Annual Meeting. Orlando, FL. Effects of macrolides on biofilm formation and bacterial pathogenesis. • 2003, University of Kentucky, Lexington, KY. Role of exopolysacchardies in Pseudomonas aeruginosa biofilm development. • 2004, North Carolina State University, Raleigh, NC. The matrix of Pseudomonas aeruginosa biofilms. • 2004, Cystic Fibrosis Foundation’s annual Williamsburg Conference. Pseudomonas aeruginosa biofilms. • 2004, 12th Biennial UA-UC Conference on Infectious Diseases, Banff, Alberta Canada. The matrix of Pseudomonas aeruginosa biofilms. • 2004, Wake Forest University, Department of Biology. Pseudomonas aeruginosa infections in cystic fibrosis: an unusual host-pathogen interaction. • 2005, North American Cystic Fibrosis Conference. Baltimore, MD. The psl-encoded exopolysaccharide is required for cell-cell and cell-surface interactions: Implications for understanding early events of P. aeruginosa biofilm development in CF. • 2006, North Carolina State University. Raleigh, NC. The psl-encoded exopolysaccharide is required for cell-cell and cell-surface interactions: Implications for understanding early events of P. aeruginosa biofilm development in CF. • 2006, FASEB Summer Research Conference, Tucson AZ. Symposium organizer and speaker. The matrix of Pseudomonas aeruginosa biofilms. • 2006, Ohio State University Seminar Series. Polysaccharides of Pseudomonas aeruginosa biofilms. • 2006, Marshall University Seminar Series. Coordinate control of virulence gene expression in Pseudomonas aeruginosa. • 2006, North Carolina bacterial pathogenesis Meeting. Chapel Hill, NC. Pseudomonas aeruginosa biofilms. Matrix Revisited. • 2006, Medical College of Virginia Seminar Series. Richmond, VA. Pseudomonas aeruginosa biofilms. Matrix Revisited.

24 • 2006, West Virginia University College of Medicine Seminar Series. Morgantown, WV. Polysaccharides of Pseudomonas aeruginosa biofilms. • 2007, University of Washington Microbiology Seminar Series. Seattle, WA, The Pseudomonas aeruginosa biofilm matrix. • 2007, University of Iowa College of Medicine Seminar Series. Iowa City, IA. The matrix of Pseudomonas aeruginosa biofilms. • 2007, Pseudomonas 2007. Seattle, WA. Invited plenary speaker. Polysaccharides of Pseudomonas aeruginosa biofilms. • 2007, Appalachian State University Seminar Series. Boone, NC. Biofilms of Pseudomonas aeruginosa. • 2007, Virginia Polytechnic Institute Seminar Series. Blacksburg, VA. Polysaccharides of Pseudomonas aeruginosa biofilms • 2008, ASM Committee for Undergraduate Education Meeting. Beverly, MA. P. aeruginosa biofilms in cystic fibrosis • 2008, FASEB Summer Research Conference, Carefree AZ. Invited speaker. The matrix of Pseudomonas aeruginosa biofilms. • 2008, 5th Annual Center for Microbial Interface Biology Retreat, The Ohio State University. Evolution of Pseudomonas aeruginosa in the CF airway. • 2008, Ohio State University Division of Infectious Disease, Bad bugs, no drugs. Are vaccines a potential solution? • 2009, Kenyon College Seminar Series. Gambier, OH. The anatomy of a bacterial biofilm. • 2009, University of Texas, Houston Seminar Series. Evolution of Pseudomonas aeruginosa in the CF airway. • 2009, University of Nebraska Seminar Series. Lincoln, NE. The matrix of Pseudomonas aeruginosa biofilms. • 2009, Midwest Microbial Pathogenesis Meeting. Invited speaker. Purdue, IN. Pseudomonas aeruginosa polysaccharides in biofilms. • 2009, The Hospital for Sick Children Seminar Series. Toronto, CA. Pseudomonas aeruginosa polysaccharides in biofilms. • 2009, ASM Biofilms 2009. Invited speaker. The matrix of Pseudomonas aeruginosa biofilms. • 2010, Miami University, Miami OH. Microbiology Seminar Series. The anatomy of a bacterial biofilm. • 2010, University of Maryland, College Park, MD., Cell Biology and Molecular Genetics Seminar Series. Biofilms and persistence of P. aeruginosa biofilms in cystic fibrosis. • 2010, University of Texas, Austin. Cell and Molecular Biology Seminar series. Biofilms and persistence of P. aeruginosa in cystic fibrosis • 2011, University of Arizona, Tucson, AZ. College of Public Health Seminar Series. The anatomy of a bacterial biofilm. • 2011, Pseudomonas 2011, Sydney Australia. Roles of Pseudomonas polysaccharides in biofilm formation and interactions with phagocytes. • 2011, Life Sciences Summit, New York, NY. Polysaccharides and Biofilms. • 2012, Indiana University, Bloomington IN, Department of Biology, Microbiology seminar series. Polysaccharides and biofilms of Pseudomonas aeruginosa. 25 • 2012, Dartmouth University, Dartmouth NH, Department of Microbiology and Immunology. • 2012, Medimmune, Gaithersburg, MD, Roles of Pseudomonas aeruginosa polysaccharides in biofilm development and pathogenesis. • 2012, Institute of Microbiology, Chinese Academy of Science, Beijing, China. Polysaccharides and biofilms of Pseudomonas aeruginosa. • 2012, OSU College of /College of Veterinary Medicine. Intersection of bacterial biofilms and host immunity. • 2012, Pfizer, Boston, MA, Roles of Pseudomonas aeruginosa polysaccharides in biofilm development and pathogenesis. • 2013, Hannover, Germany, Pseudomonas aeruginosa biofilm matrix exopolysaccharides. • 2013, Ohio Branch ASM Opening Plenary Lecture, Ashland University, Intersection of bacterial biofilms and host immunity. • 2013, Duquesne University, Department of Biology. Pseudomonas biofilms and immunity • 2014, FASEB Summer Research Conference in Microbial Glycobiology. Biofilms and immunity in chronic infections. Itasca, Ill. • 2014, International Symposium on Staphylococci and Staphylococcal Infections (ISSI 2014), Pseudomonas aeruginosa and Staphylococcus aureus in chronic wound infections. Chicago, Ill • 2014, Translation to Clinical, Regenerative Medicine Wound Care Conference. Ohio State University, Podium talk. • 2014, 20IWA Conference: The Perfect Slime – Nature, Properties, Regulation and Dynamics of EPS, Essen Germany • 2014, Center for Regenerative Medicine and Cell-Based Therapies. Mohican Resort, Podium talk. • 2014, 3rd Annual Microbiology Retreat, Plenary speaker, Indiana University, Bloomington. • 2014, University of Toledo Health Sciences seminar series speaker. Pathoadaptation in chronic infection. • 2014, Regeneron Inc., Tarrytown, NY, Biofilms and immunity in chronic infection • 2015, Keynote Speaker, Mid Atlantic Microbial Pathogenesis Meeting, Pathoadaptation in chronic infection. • 2015, Keynote Speaker, North American Veterinary Forum, Nashville, TN. Biofilms, wounds, and chronic infections in animals and humans. • 2015, Antimicrobial Peptide Gordon Research Conference. La Barga, Italy. Mutagenesis by host antimicrobial peptides: insights into microbial evolution during chronic infections. • 2015, University of Michigan, Seminar series speaker, Division of pulmonary and critical care. Biofilms and immunity in chronic infection. • 2015, 7th ASM Biofilms conference. Chicago, IL, Biofilms and immunity in chronic infection. • 2015, Keynote speaker, Conference of Research Workers in Animal Diseases (CRWAD), Chicago, IL. Biofilms, wounds, and chronic infections in animals and humans. • 2016, SUNY Binghamton seminar series speaker. Biofilms and immunity in chronic infection • 2016, West Virginia University seminar series speaker. Biofilms and immunity in chronic 26 infection. • 2017, Plenary speaker, Eastern Pennsylvania ASM Branch meeting, Thomas Jefferson University, Biofilms and immunity in chronic infection, March 27, 2017. • 2017, University of Pittsburgh Department of Microbiology and Molecular Genetics, Pathoadaptation in chronic infection, May 3, 2017. • 2017, Institute for Biological and Medical Engineering, Pontifica Universidad Catolica de Chile, Biofilms and immunity in chronic infection, October 11-13, 2017. • 2017, OSU Pulmonary Division, Biofilms and immunity in chronic infection, October 23. • 2017, Keynote speaker, Wound symposium, Toledo, OH, Biofilms, wounds, and chronic infections in animals and humans. November 16, 2017. • 2017, Plenary speaker, Kentucky-Tennessee ASM Branch meeting, Biofilms and immunity in chronic infection, November 10-11, 2017 • 2017, Rensselaer Polytechnic Institute, Troy NY, Biofilms and immunity in chronic infection. December 11, 2017. • 2018, Michigan State University seminar series speaker, Biofilms and immunity in chronic infection, February 13, 2018. • 2018, Plenary speaker, Indiana ASM Branch meeting, Biofilms and immunity in chronic infection, April 6-7, 2018. • 2018, Plenary speaker, Miami University Cell, Molecular, and Structural Biology summer retreat, June 1, 2018. • 2018, Invited speaker, NACFC Annual meeting, Pseudomonas aeruginosa pathoadaptation during chronic infection. October, 2018. • 2018, Plenary speaker, International symposium Frontiers in Microbial Pathogenesis, Signaling, and Intercellular Communication. Guangzhou, China, November, 2018 • 2018, Emory University seminar series speaker (trainee invitee). Biofilms and immunity in chronic infection. December, 2018 • 2019, Texas Biomedical Research Institute Research Symposium, Plenary Speaker. March 2019 • 2019, University of Minnesota, Annual Biofilm Symposium (trainee invitee), Plenary Speaker. May, 2019. • 2019, 3M, St. Paul, MN. Pseudomonas aeruginosa pathoadaptation during chronic infection. May, 2019. • 2019, Indiana University School of Medicine. Biofilms in chronic wound infections. October, 2019. • 2020, University of Toledo Medicinal and Biological Chemistry. Biofilms and therapeutics in chronic infections. February, 2020 • 2020, University of Cincinnati, Cincinnati Children’s. Biofilms and immunity in chronic infection. May, 2020

GRADUATE STUDENTS/RESIDENTS/FELLOWS ADVISED (Wozniak laboratory):

Current

• Landon Locke, Visiting Assistant Professor; Source of support ATS grant 27 • Erin Gloag, Postdoctoral research fellow; Source of support AHA Career Development Award • Katarzyna Danis-Wlodarczyk, Postdoctoral research fellow; Source of support OSU President’s Postdoctoral Fellowship • Yiwei Liu, Graduate student; Source of support; R01AI34895 • Danielle Ferguson, Graduate student; Source of support: R01AI34895 • Pranav Rana, Graduate student; Source of support: R01AI143916 • Sabrina Lamont, Graduate student; Source of support R01AI46262 Previous • Matthew Pestrak, Graduate student; Source of stipend support R01AI097511-01, Current: Postdoctoral fellow • Mohini Bhattacharya, Graduate student; Source of stipend support R01NR013898, Current: Postdoctoral fellow • Sankalp Malhorta, MSTP graduate student; Source of stipend support CCTS TL-1, Current: Medical Student • Christopher Jones, Postdoctoral research fellow; Source of support CFF fellowship, Current: Sharklet, Inc. • Heather Eggleston, Graduate student; Source of stipend support SIBS training grant, Current: Research Associate, Henry Jackson Foundation • Sarah Chaney, DVM, Graduate student; Source of stipend support T32 Veterinary Bioscience training grant, Current: Hugh Trumbull Adams Resident in Zoological Medicine and Surgery, Bronx Zoo • Valerie Ray, Postdoctoral research fellow; Source of support CFF fellowship, Current: Baxter International • Binjie Xu, Graduate student, Source of stipend support, R01HL58334, Current: Sharklet, Technologies, Denver CO. • Meenu Mishra, Ph.D., Postdoctoral research fellow; Source of support CFF fellowship, Current: Assistant Professor, University Services University. • Dominique Limoli, Graduate student; Source of stipend support R01AI061396, Current: Assistant Professor, Iowa University. • Christopher Jones, Source of stipend support, SIBS and AHA fellowships, Current: Sharklet, Technologies, Denver CO. • Ethan Mann, Ph.D., Postdoctoral research fellow; Source of support T32 Pulmonary training grant. Current: Sharklet Technologies, Denver CO • Elizabeth Waligora, Ph.D. candidate; Source of stipend support: AHA predoctoral fellowship. Current: Program Manager, Syneos Health. • Cynthia Ryder, Graduate student; Source of stipend support R01AI061396. Current: Associate Professor, Lincoln Memorial University. • Matthew Byrd, Graduate student; Source of stipend support NIH F31AI078700, Ruth L. Kirschstein NRSA. Research Fellow, Feed the Hunger. • Luyan Ma, Ph.D., Research scientist; Source of support: CFF fellowship grant MA06F0. Current: Professor, Chinese National Academy of Sciences • Andrea Rockel, Graduate student, funding by NIH grant R01HL58334. Current: Associate Professor, Mars Hill College, Mars Hill, NC. • Belen Belete, Graduate student, funding by NIH grant R01HL58334. Current: Assistant Professor, Winston Salem State University, Winston Salem, NC. • Deborah Ramsey, Graduate student and postdoctoral fellow, funding by NIH grant R01HL58334 and American Heart Association Fellowship 0215191U. Current: Group Leader, CFD Research. • Kara Jackson, Graduate student, funded by Lucille & David Packard Research Grant. Current: 28 Research Edit Specialist, NC A&T University. • Anne Tart, Ph.D. Graduate student, funded by AHA 0515325U fellowship and NIH grant R01HL58334. Current: Research scientist, Methodist Hospital Research Institute. • Tanya Erova, Ph.D. (postdoctoral fellow), funded by a Cystic Fibrosis Foundation postdoctoral fellowship. Current: Research Associate, University of Texas, Galveston. • Patricia Baynham, Ph.D. (graduate student/postdoctoral fellow), funded by NIH grant R01 HL59334 and R29 AI35177, Current: Professor of Biology/Interim Dean, St. Edward's University, Austin, TX. • Samuel Woolwine, Ph.D. (graduate student), funded by NIH grant R01 HL59334 and R29 AI35177, Current: Biotechnology Patent Specialist, Baltimore, MD • Timna Wyckoff, Ph.D. (postdoctoral fellow), funded by NIH R01grant AI25779, Current: Assistant Director, Institute Bioscience and Biotechnology Research.

OSU GRADUATE STUDENTS THESIS COMMITTEE SERVICE:

• Amelia Staats, Microbiology, Stoodley laboratory • Audra Fullen, BSGP, Deora laboratory • Mariella Mestes-Villanueva, MCDB, Rikihisa laboratory • Marissa Gittrich, Microbiology, Sullivan laboratory • Oscar Mejia, BSGP, Robinson laboratory • Danielle Ferguson, BSGP, Wozniak laboratory • Pranav Rana, Microbiology, Wozniak laboratory • Yiwei Liu, Microbiology, Wozniak laboratory • Yunxiao Liu, Microbiology, Sullivan laboratory • Mark Hahn, BSGP, Gunn laboratory • Matt Dunn, Microbiology, Anderson laboratory • Devin Sindeldecker, BSGP, Stoodley laboratory • Rebecca Steiner, Microbiology, Ibba laboratory • Sankalp Malhotra, BSGP, Wozniak laboratory • Michelle Gerst, Microbiology, Yousef laboratory • Lindsey Solden, Microbiology, Wrighton laboratory • Heather Eggleston, BSGP, Wozniak laboratory • Matthew Pestrak, BSGP, Wozniak laboratory • Claire Dodd, Microbiology, Schlesinger laboratory • Juan Moliva, BSGP, Torrelles laboratory • Jonathan Lam, Microbiology, Seveau laboratory • Nicki Limoli, IBGP, Wozniak laboratory • Bingie Xu, Microbiology, Wozniak laboratory • Sarah Chaney, Veterinary Biosciences, Wozniak laboratory • Mohini Bhattacharya, Microbiology, Wozniak laboratory • Geoffrey Gonzalez-Escobedo, Microbiology, Gunn laboratory • Ran Dong, Microbiology, Satoskar laboratory • Mohamed Ali, Microbiology, Ahmer laboratory • Forrest Raffel, IBGP, Mason laboratory • Matt Swearingen, Microbiology, Ahmer laboratory 29 • Joanna Marshall, IBGP, Gunn laboratory • Olga Zemska, Microbiology, Rappleye laboratory • Estevan Santana, IBGP, Munson laboratory • Hussam Salhi, IBGP, King laboratory • Mia Tazi, IBGP, Amer laboratory • Christian Harding, IBGP, Munson laboratory • Dominique Olds, Microbiology, Gunn laboratory

OSU FACULTY MENTORING/GRANT REVIEW SERVICE:

• Stephanie Seveau, Ph.D., Assistant Professor, Microbiology • Samantha King, Ph.D., Associate Professor, Center for Microbial Pathogenesis, NCH • Jennifer Edwards, Ph.D., Associate Professor, Center for Microbial Pathogenesis, NCH • Kevin Mason, Ph.D., Assistant Professor, Center for Microbial Pathogenesis, NCH • Sheryl Justice, Ph.D., Assistant Professor, Center for Microbial Pathogenesis, NCH • John Gunn, Ph.D., Professor, Microbial Infection and Immunity • Chad Rappleye, Ph.D., Associate Professor, Microbiology • Larry Schlesinger, MD, Professor, Microbial Infection and Immunity • Amal Amer, MD, Ph.D., Associate Professor, Microbial Infection and Immunity • Mark Drew, Ph.D., Assistant Professor, Microbial Infection and Immunity • Jordi Torrelles, Ph.D., Assistant Professor, Microbial Infection and Immunity • Benjamin Kopp, MD, Assistant Professor, Center for Microbial Pathogenesis, NCH • Steven Lower, Ph.D., Associate Professor, Earth Sciences • Luanne Hall-Stoodley, Ph.D., Associate Professor, Microbial Infection and Immunity • Paul Stoodley, Ph.D., Professor, Microbial Infection and Immunity • Murugesan Rajaram, Ph.D., Research Assistant Professor, Microbial Infection and Immunity • Raj Deora, Ph.D., Associate Professor, Microbial Infection and Immunity • Haitao Wen, Ph.D. Assistant Professor, Microbial Infection and Immunity • Namal Liyanage, Assistant Professor, Microbial Infection and Immunity • Amit Sharma, Assistant Professor, Veterinary Biosciences

30 Curriculum Vitae

Haitao Wen 2255 Concord Village Dr Columbus, OH 43220 919-370-1151 Email: [email protected]

Education

Ph.D. Graduate Program in Molecular and Cellular Pathology (MCP), December 2007 University of Michigan Medical School, Ann Arbor MI

M.S. Pathophysiology, July 2001 Shan-Dong University School of Medicine, Jinan, P.R. China

M.D. Medicine, July 1998 Shan-Dong University School of Medicine, Jinan, P.R. China

Research Experience

03/2018 – Present Assistant Professor Department of Microbial Infection and Immunity Infectious Disease Institute The “James” Comprehensive Cancer Center The Ohio State University, Columbus, OH

09/2015 – 02/2018 Assistant Professor Department of Pathology and Microbiology Holland Regenerative Medicine Program University of Nebraska Medicine Center, Omaha, NE

10/2013 – 08/2015 Assistant Professor Department of Surgery Lineberger Comprehensive Cancer Center University of North Carolina at Chapel Hill, NC

01/2008 - 09/2013 Postdoctoral Research Associate Laboratory of Jenny P-Y. Ting, Ph.D. University of North Carolina at Chapel Hill, NC

07/2003 - 12/2007 Graduate Research Assistant Laboratory of Steven L. Kunkel, Ph.D. University of Michigan Medical School, Ann Arbor, MI

09/1998-07/2001 Master’s Degree Research Assistant Laboratory of Hu, Wei-Cheng, M.D. Shan-Dong University School of Medicine, Jinan, China

Academic and Professional Honours:

• The 2020 AAI Annual Meeting Early-Career Faculty Travel Award • 2019 Dean’s List, The Ohio State University Wexner Medical Center

• The 2015 AAI Annual Meeting Early-Career Faculty Travel Award

• The 2012 UNC-CH Postdoctoral Award for Research Excellence

• The 2012 Keystone Symposia (Innate Immunity: Sensing the Microbes and Damage Signals) Postdoctoral Scholarship

• The 2010 Lineberger Comprehensive Cancer Center Joseph S. Pagano Award at UNC-Chapel Hill

• The 2006 Experimental Biology Conference Travel Award

• The 2005 Experimental Biology Conference Travel Award

Professional Affiliations

1. The American Association of Immunologists (AAI): 2013-Present. 2. Shock Society. 2014-Present

Publications

1. Li T*, Kong L*, Li X, Wu S, Attri KS, Li Y, Herring LE, Asara JM, Lei Y, Cheng X, Singh PK, Green DR, Wang H, Wen H. Mitochondrial calcium signaling facilitates bacterial survival by restraining LC3- associated phagocytosis. Nat Microbiol. Resubmitted. (*Equal contribution)

2. Luo X, Donnelly CR, Gong W, Heath BR, Hao Y, Donnelly LA, Moghbeli T, Tan YS, Lin X, Bellile E, Kansy BA, Carey TE, Brenner JC, Cheng L, Polverini PJ, Morgan MA, Wen H, Prince ME, Ferris RL, Xie Y, Young S, Wolf GT, Chen Q, Lei YL. HPV16 drives cancer immune escapes via NLRX1- mediated degradation of STING. J Clin Invest. 2020 Feb 24 [Epub ahead of print]. PMCID:

3. Liu X, Clemens DL, Grunkemeyer JA, Price JD, O'Connell K, Chapman NM, Storz P, Wen H, Cox JL, Reid WL, Hollingsworth MA, Thayer S. Mucin-1 is required for Coxsackie Virus B3-induced inflammation in pancreatitis. Sci Rep. 2019 Jul 23;9(1):10656. PMCID: PMC6650496.

4. Gong W, Wen H. Sepsis induced by cecal ligation and puncture. Methods Mol Biol. 2019;1960:249- 255.

5. Li X*, Gong W*, Wang H, Li T, Attri KS, Lewis RE, Kalil AC, Bhinderwala F, Powers R, Yin G, Herring LE, Asara JM, Lei Y, Yang X, Mocarski ES, Rodriguez DA, Yang M, Green DR, Singh PK, Wen H. O- GlcNAc transferase suppresses inflammation and necroptosis by targeting receptor- interacting serine/threonine-protein kinase 3. Immunity. 2019 Mar 19;50(3):576-590. PMCID: PMC6426684. (*Equal contribution)

6. Li T, Li X, Attri KS, Liu C, Li L, Herring LE, Asara JM, Lei YL, Singh PK, Gao C, Wen H. O-GlcNAc transferase links glucose metabolism to MAVS-mediated antiviral innate immunity. Cell Host Microbe. 2018 Dec 12;24(6):791-803. PMCID: PMC6296827. (Highlighted by Nat Immunol. 2019 Feb;20(2): 111)

7. Cheng L, Chen L, Wei X, Wang Y, Ren Z, Zeng S, Zhang X, Wen H, Gao C, Liu H. NOD2 promotes dopaminergic degeneration regulated by NAPDH oxidase 2 in 6-hydroxydopamine model of Parkinson’s disease. J Neuroinflammation. 2018 Aug 29;15(1):243. PMCID: PMC5379975. 8. Li X, Zhang Z, Li L, Gong W, Herring LE, Asara JM, Singer JD, Wen H. Myeloid-derived cullin 3 promotes STAT3 phosphorylation by inhibiting OGT expression and protects against intestinal inflammation. J Exp Med. 2017 Apr 3;214(4):1093-1109. PMCID: PMC5379975.

9. Lei Y, Kansy BA, Li J, Cong L, Liu Y, Trivedi S, Wen H, Ting JP, Ouyang H, Ferris RL. EGFR- targeted mAb therapy modulates autophagy in head and neck squamous cell carcinoma through NLRX1-TUFM protein complex. Oncogene. 2016 Sep 8;35(36):4698-707. PMCID: PMC5257174.

10. Guo H, König R, Deng M, Riess M, Mo J, Zhang L, Petrucelli A, Yoh SM, Barefoot B, Samo M, Sempowski GD, Zhang A, Colberg-Poley AM, Feng H, Lemon SM, Liu Y, Zhang Y, Wen H, Zhang Z, Damania B, Tsao LC, Wang Q, Su L, Duncan JA, Chanda SK, Ting JP. NLRX1 Sequesters STING to Negatively Regulate the Interferon Response, Thereby Facilitating the Replication of HIV-1 and DNA Viruses. Cell Host Microbe. 2016 Apr 13;19(4):515-28. PMCID: PMC4833116.

11. Zhang J, Wang C, Chen X, Takada M, Fan C, Zheng X, Wen H, Liu Y, Wang C, Pestell RG, Aird KM, Kaelin WG Jr, Liu XS, Zhang Q. EglN2 Associates with the NRF1-PGC1α Complex and Controls Mitochondrial Function in Breast Cancer. EMBO J. 2015 Dec 2;34(23):2953-70. PMCID: PMC4687683.

12. Zhao C, Gillette DD, Li X, Zhang Z, Wen H. Nuclear factor E2-related factor-2 (Nrf2) is required for NLRP3 and AIM2 inflammasome activation. J Biol Chem. 2014 Jun 13;289(24):17020-9. PMCID: PMC4059144.

13. Robbins GR, Wen H, Ting JP. Inflammasomes and metabolic disorders: old genes in modern diseases. Mol cell. 2014 Apr 24; 54(2):297-308. PMCID: PMC4084585.

14. Zhang L, Mo J, Swanson KV, Wen H, Petrucelli A, Gregory SM, Zhang Z, Schneider M, Jiang Y, Fitzgerald KA, Ouyang S, Liu ZJ, Damania B, Shu HB, Duncan JA, Ting JP. NLRC3, a member of the NLR family of proteins, is a negative regulator of innate immune signaling induced by the DNA sensor STING. Immunity. 2014 Mar 20; 40(3):329-41. PMCID: PMC4011014.

15. Eitas TK, Chou WC, Wen H, Gris D, Robbins GR, Brickey J, Oyama Y, Ting JP. The NLR family member, NLRX1, mediates protection against experimental autoimmune encephalomyelitis and represses macrophage/microglial-induced inflammation. J Biol Chem. 2014 Feb 14;289(7):4173- 9. PMCID: PMC3924282.

16. Wen H, Miao EA, Ting JP. Mechanisms of NLR-associated inflammasome activation. Immunity. 2013 Sep 19;39(3):432-41. PMCID: PMC3835203.

17. Wen H. Sepsis induced by cecal ligation and puncture. Methods Mol Biol. 2013;1031 :117-24.

18. Wen H, Ting JP. Agitation by suffocation: how hypoxia activates innate immunity via the Warburg effect. Cell Metab. 2013 Jun 4;17(6):814-5.

19. Lei Y, Wen H, Ting JP. The NLR protein, NLRX1, and its partner TUFM reduces type I interferon and enhances autophagy. Autophagy. 2013 Jan 15;9(3). PMCID: PMC3590269.

20. Taxman DJ, Swanson KV, Broglie PM, Wen H, Holley-Guthrie E, Huang MT, Callaway JB, Eitas TK, Duncan JA, Ting JP. Porphyromonas gingivalis Mediates Inflammasome Repression in Polymicrobial Cultures through a Novel Mechanism Involving Reduced Endocytosis. J Biol Chem. 2012 Sep 21;287(39):32791-9. PMCID: PMC3463344.

21. Schneider M, Zimmermann AG, Roberts RA, Zhang L, Swanson KV, Wen H, Davis BK, Allen IC, Holl EK, Ye Z, Rahman AH, Conti BJ, Eitas TK, Koller BH, Ting JP. NLRC3 Attenuates TLR Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Signaling via Modification of TRAF6 and NF-κB. Nat Immunol. 2012 Sep;13(9):823-31. PMCID: PMC3721195.

153 22. Lei Y*, Wen H*, Yu Y, Taxman DJ, Zhang L, Widman DG, Swanson KV, Wen KW, Damania BA, Moore CB, Giguère PM, Siderovski DP, Hiscott J, Razani B, Semenkovich CF, Chen X, Ting JP. NLRX1 and TUFM form a mitochondrial complex that regulates type 1 interferon and autophagy. Immunity. 2012 June 29;36(6):933-46. (*Equal contribution). PMCID: PMC3397828.

23. Razani B, Feng C, Coleman T, Emanuel R, Wen H, Hwang S, Ting JP, Virgin HW, Kastan MB, Semenkovich CF. Autophagy links inflammasomes to atherosclerotic progression. Cell Metab. 2012 Apr 4;15(4):534-44. PMCID: PMC3322320.

24. Wen H, Ting JP, O'Neill LA. A role for the NLRP3 inflammasome in metabolic diseases - did Warburg miss inflammation? Nat Immunol. 2012 Apr;13(4):352-7. PMCID: PMC4090390.

25. Roney KE, O'Connor BP, Wen H, Holl EK, Guthrie EH, Davis BK, Jones SW, Jha S, Sharek L, Garcia- Mata R, Bear JE, Ting JP. Plexin-B2 negatively regulates macrophage motility, Rac, and Cdc42 activation. PLoS One. 2011;6(9):e24795. PMCID: PMC3179467.

26. Wen H*, Gris D*, Lei Y, Jha S, Zhang L, Huang MT, Brickey WJ, Ting JP. Fatty acid-induced NLRP3/ PYCARD inflammasome activation interferes with insulin signaling. Nat Immunol. 2011 May;12(5):408- 15 (Equal contribution). PMCID: PMC4090391.

27. Beckley KD, Wen H, Ting JP. The inflammasome NLRs in immunity, inflammation and associated diseases. Annu Rev Immunol. 2011;29;707-35. PMCID: PMC4067317.

28. Wen H, Lei Y, Eun So-Young, Ting JP. Plexin-A4-semaphorin 3A signaling is required for Toll like receptor- and sepsis-induced cytokine storm. J Exp Med. 2010 Dec 20;207(13):2943-57. PMCID: PMC3005237

29. Huang MT, Mortensen BL, Taxman DJ, Craven RR, Taft-Benz S, Kijek TM, Fuller JR, Davis BK, Allen IC, Brickey WJ, Gris D, Wen H, Kawula TH, Ting JP. Deletion of ripA alleviates suppression of the inflammasome and MAPK by Francisella tularensis. J Immunol. 2010 Nov 1;185(9):5476- 85. PMCID: PMC4671501

30. Gris D, Ye Z, Iocca HA, Wen H, Craven RR, Gris P, Huang M, Schneider M, Miller SD, Ting JP. NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses. J. Immunol. 2010 Jul 15;185(2):974-81. PMCID: PMC3593010

31. Lyn-Kew K, Rich E, Zeng X, Wen H, Kunkel SL, Newstead MW, Bhan U, Standiford TJ. IRAK-M regulates chromatin remodeling in lung macrophages during experimental sepsis. PLoS One. 2010 Jun 16;5(6):e11145. PMCID: PMC2886833

32. Cavassani KA, Wen H, Moreira AP, Schaller MA, Carson WF 4th, Ishii M, Lindell DM, Lukacs NW, Keshamouni VG, Hogaboam CM, Kunkel SL. The post sepsis-induced expansion and enhanced function of regulatory T cells creates an environment to potentiate tumor growth. Blood. 2010 Jun 3;115(22):4403- 11. PMCID: PMC2881495

33. Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, Carson WF 4th, Cavassani KA, Li X, Lukacs NW, Hogaboam CM, Dou Y, Kunkel SL. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009 Oct 8;114(15):3244-54. PMCID: PMC2759649

34. Cavassani KA, Ishii M, Wen H, Schaller MA, Lincoln PM, Lukacs NW, Hogaboam CM, Kunkel SL. TLR3 is an endogenous sensor of tissue necrosis during acute inflammatory events. J Exp Med. 2008 Oct 27;205(11):2609-21. PMCID: PMC2571935

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

35. Davis J, Wen H, Edwards T, Metzger JM. Allele and species dependent contractile defects by restrictive and hypertrophic cardiomyopathy-linked troponin I mutants. J Mol Cell Cardiol. 2008 May;44(5):891-904. PMCID: PMC2443058

155 36. Wen H, Schaller MA, Dou Y, Hogaboam CM, Kunkel SL. Dendritic cells at the interface of innate and acquired immunity: the role for epigenetic changes. J Leukoc Biol. 2008 Mar;83(3):439-46.

37. Wen H, Dou Y, Hogaboam CM, Kunkel SL. Epigenetic regulation of dendritic cell-derived interleukin-12 facilitates immunosuppression following a severe innate immune response. Blood. 2008 Feb;111(4):1797-804. PMCID: PMC2234040

38. Wen H, Hogaboam CM, Lira SA, Cook DN, Kunkel SL. The chemokine receptor CCR6 is an important component of the innate immune response. Eur J Immunol. 2007 Sep;37(9):2487-98.

39. Davis J, Wen H, Edwards T, Metzger JM. Thin filament disinhibition by restrictive cardiomyopathy mutant R193H troponin I induces Ca2+-independent mechanical tone and acute myocyte remodeling. Circ Res. 2007 May;100(10):1494-502

40. Wen H, Hogaboam CM, Gauldie J, Kunkel SL. Severe sepsis exacerbates cell-mediated immunity in the lung due to an altered dendritic cell cytokine profile. Am J Pathol. 2006 Jun;168(6):1940- 50. PMCID: PMC1606615

41. Jakubzick C, Wen H, Matsukawa A, Keller M, Kunkel SL, Hogaboam CM. Role of CCR4 ligands, CCL17 and CCL22, during Schistosoma mansoni egg-induced pulmonary granuloma formation in mice. Am J Pathol. 2004 Oct;165(4):1211-21. PMCID: PMC1618636

Patents

Ting JP, Wen H. Methods and compositions for the treatment of immune disorders. U.S. Patent 20120251539, filled on November 23, 2010, and issued on October 4, 2012.

Presentations at Scientific Conferences

NLRX1 and TUFM form a mitochondrial complex that regulates type 1 interferon and autophagy. (Poster presentation). Keystone Symposia (Innate Immunity: Sensing the Microbes and Damage Signals), March 4-9, 2012 Keysone, CO

Plexin-A4/semaphorin 3A signaling is required for TLR- and sepsis-induced cytokine storm. (Poster presentation). Federation of Clinical Immunology Societies, June 24-27, 2010 Boston, MA

Dysfunction of dendritic cells during CLP-induced experimental sepsis. (Poster presentation). Society for Leukocyte Biology, November 8-12, 2006 San Antonio, TX

Role of CCR6 in the dysregulated systemic immune response during severe experimental sepsis. (Oral presentation). Experimental Biology 2006, April 1-5, 2006 San Francisco, CA

Severe sepsis impairs pulmonary dendritic cell IL-12 production: implications for the long-term pulmonary immune response. (Oral presentation). Experimental Biology 2005, April 2-6, 2005 San Diego, CA

Grant Support:

Active

1R01GM135234 (PI: Wen) 09/15/19 – 07/31/23 NIH-NIGMS Mitochondrial Metabolism in Microbial Sepsis Award Amount: $322,920, 35% effort The goal of this project is to investigate the role and mechanism of mitochondrial calcium signaling- mediated acetyl coenzyme A metabolism in the innate immune function during microbial sepsis.

1R01GM120496 (PI: Wen) 8/15/17 – 06/30/22 NIH-NIGMS Immunometabolism in Microbial Sepsis Award Amount: $321,180, 30% effort The goal of this project is to investigate the role and mechanism of two glucose metabolism pathways, the pentose phosphate pathway and the hexosamine biosynthesis pathway, in the innate immune function during microbial sepsis.

Pending

3/5/2020 Preoperative exercise therapy for surgery-triggered inflammation NIH/NIDDK R01 grant (Resubmission) PI: Hai Huang Co-investigator: Haitao Wen (5% effort)

2/5/2020 The role of innate immunity in progression of nonalcoholic fatty liver disease NIH/NIDDK R01 grant PI: Allan Tsung Co-investigator: Haitao Wen (5% effort), Zihai Li, Chengli Shen

Chan-Zuckerberg Initiative (PI: Wen) 07/01/20 – 06/30/22 Single-cell analysis of septic inflammation Award Amount: $175,000, 10% effort Submitted on 11/19/2019

Completed

32675-Y3 (PI: Wen) 07/01/16 – 06/30/17 Nebraska Stem Cell Research Project Award O-GlcNAc Signaling in Intestinal Stem Cell Mediated Epithelial Regeneration The goal of this project is to investigate the role of OGT-mediated protein O-GlcNAcylation in intestinal stem cell function and epithelial regeneration after tissue damage.

1K01DK098307 (PI: Wen) 7/1/2013 – 5/30/2016 NIH-NIDDK The Role and Mechanism of NLRX1-mediated Cell Stress Response in Insulin Resistance Award Amount: $150,036, 100% effort

Cancer Research Institute (PI: Wen) 07/01/10 – 06/30/13 Postdoctoral Fellowship Award The Role and Mechanism of Plexin-A4 in Colitis-Associated Cancer The goal of this project was to evaluate disease outcome in Plxna4-/- mice chemically induced colitis and colitis-associated cancer.

American Heart Association Mid Atlantic Affiliate (PI: Wen) 07/01/08 – 06/30/10 Postdoctoral Fellowship Award The Role of Plexin-A1 Mediated T cell Activation in Atherosclerosis The goal of this project was to analyze the atherosclerotic lesion burden and phenotype in Apoe-/- mice after in vivo treatment with Sema6D-lg. Professional Services Ad hoc reviewer for Cell Death & Disease Communications Biology Endocrine Research Infection and Immunity International Journal of Biochemistry & Cell Biology Journal of Critical Care Journal of Experimental Medicine Journal of Immunology Journal of Lipid Research Nature Nature Chemical Biology Nature Communications Nature Immunology Oncotarget Oral Oncology PLOS ONE PNAS Trends in Immunology Ad Hoc Grant reviewer for NIAID NIH Support for Conferences and Scientific Meetings (R13) (ZAI1 MFH-D (M2) 1), 2020 NIGMS Maximizing Investigators’ Research Award (R35) (ZRG1 CB-T (55) R), 2020 NIAID NIH Support for Conferences and Scientific Meetings (R13) (ZAI1 AZ-D (S1) 1), 2019 NIH Cellular and Molecular Immunology - A Study Section, 2016 HAZEM E. GHONEIM, Ph.D.

Assistant Professor Department of Microbial Infection and Immunity - College of Medicine – The Ohio State University Lab Website: https://u.osu.edu/ghoneim.2/ 460 W 12th Ave., E-mail: [email protected] Office: +1 794 Biomedical Research (614) 293-9608 Tower, Columbus, OH 43210 Fax: +1 (614) 292-9616 USA

ACADEMIC APPOINTMENTS Apr 2019–Present Assistant Professor, Department of Microbial Infection and Immunity, College of Medicine, the Ohio State University, Columbus, OH, USA Apr 2019–Present Member, Cancer Biology Program, the Ohio State University Comprehensive Cancer Center, Columbus, OH, USA

Apr 2019–Present Member, T Cell Biology Program, the Pelotonia Institute for Immuno-Oncology, the Ohio State University, Columbus, OH, USA

Apr 2019–Present Member, Host Defense and Microbial Biology Program, the Infectious Diseases Institute, the Ohio State University, Columbus, OH, USA

EDUCATION AND TRAINING Mar 2014–Mar 2019 Postdoctoral Fellowship, St. Jude Children’s Research Hospital, Memphis, TN, USA Dec 2013 Ph.D., Microbiology and Immunology, University of Tennessee Health Science Center, Memphis, TN, USA May 2005 B.Sc.Pharm., Distinction with Highest Honors, Cairo University, Cairo, Egypt

RESEARCH INTERESTS The focus of my research program is to understand how T cell immune responses are generated and maintained during infections and cancer. I am particularly interested in elucidating the mechanisms of epigenetic programming that regulate T cell exhaustion during chronic virus infections and cancer. My ultimate goal is to develop novel T cell-targeted epigenetic therapies to enhance the efficacy of cancer immunotherapies.

HONORS AND AWARDS Apr 2018 Young Scientist Award, The World Academy of Sciences for the advancement of science in developing countries (TWAS)/BioVision Alexandria.NXT 2018 Conference, Alexandria, Egypt 1 Hazem E. Ghoneim, PhD 04/2020

Oct 2017 Scholar-in-Training Award, American Association for Cancer Research (AACR) Special Conference on Tumor Immunology and Immunotherapy, Boston, MA, USA Jun 2017 Travel Award, the Federation of Clinical Immunology Societies Annual Meeting, Chicago, IL, USA Oct 2016 Keynote Speaker, Statewide Communicable and Environmental Diseases and Emergency Preparedness (CEDEP) Meeting, Nashville, TN, USA Jun 2014 Young Scientist Travel Award, the Fifth Influenza Conference of the European Scientific Working group on Influenza (ESWI), Riga, Latvia Jun 2013 Travel Award and Poster of Merit, the Federation of Clinical Immunology Societies Annual Meeting, Boston, MA, USA Apr 2012 Travel Award, First International Graduate Student Immunology Conference, Harvard Medical School, Harvard University, Boston, MA, USA Feb 2011 Global Health Travel Award, Keystone Symposia Conference in Vancouver, BC, Canada

PROFESSIONAL MEMBERSHIP 2018–Present Elected Member to the Association for Cancer Immunotherapy (CIMT) 2017–Present Elected Regular Member to the American Association of Immunologists (AAI) 2016–Present The American Association for Cancer Research (AACR) 2013–Present The American Society for Virology (ASV) 2013–Present The Federation of Clinical Immunology Societies (FOCIS) 2013–Present The Federation of American Societies for Experimental Biology (FASEB) 2013–2017 The American Association for the Advancement of Science (AAAS)

PUBLICATIONS 1. Ghoneim, H.E.; Fan, Y.; Crawford, J.C.; Boi, S.K.; Li, J.; Alli, S.; Abdelsamed, H. A.; Feng, Y.; and Youngblood, B. “De novo DNA methylation regulates CD8 T cell exhaustion- associated chromatin accessibility” (In Prep.).

2. Abdelsamed, H. A.; Zebley, C.C.; Nguyen, h.; Rutishauser, R.L.; Fan, Y.; Ghoneim, H. E.; Crawford, J.C.; Alfei, F.; Alli, S.; Ribeiro, S.P.;, Castellaw, A.; McGargill, M.A.; Jin, H.; Boi, S.K.; Speake, C.; Serti, E.; Turka, L.A.; Busch, M.E.; Stone, M.; Deeks, S.G.; Sekaly, R.P.; Zehn, D.; James, E.; Nepom; Youngblood, B. “Beta cell-specific CD8 T cells acquire stem- cell memory-associated epigenetic programs during development of type 1 diabetes” (In Press-Nature Immunology).

3. Fonseca, R.; Beura, L.K.; Quarnstrom, C.F.; Ghoneim, H.E.; Fan, Y.; Zebley, C.C.; Scott, M.C.; Fares- Frederickson, N.J.; Wijeyesinghe, S.; Thompson, E.A.; Borges da Silva, H.; Vezys, V.; Youngblood, B.; Masopust, D. (2020) “Developmental plasticity allows outside-in immune responses by resident memory T cells.” Nature Immunology; 21(4):412-421. 2 Hazem E. Ghoneim, PhD 04/2020

4. LeMessurier, K.S.; Rooney, R.; Ghoneim, H.E.; Liu, B.; Li, K.; Smallwood, H.; and Samarasinghe, A. “Influenza A Virus Infection Modulates Eosinophils in vitro and in vivo” (In Press-Journal of Leukocyte Biology)

5. Alfei, F.; Kanev, K.; Hofmann, M.; Wu, M.; Ghoneim, H. E.; Roelli, P.; Utzschneider, D.T.; von Hö sslin, M.; Cullen, J.; Fan, Y.; Eisenberg, V.; Wohlleber, D.; Steiger, K.; Merkler, D.; Delorenzi, M.; Knolle, P.A.; Cohen, C. J.; Thimme, R.; Youngblood, B.; and Zehn, D. (2019) “Tox reinforces the phenotype and longevity of exhausted T-cells in chronic viral infection” Nature; 571(7764): 265-269.

6. Ghoneim, H. E.; Fan, Y.; Moustaki, A.; Abdelsamed, H. A.; Dash, P.; Dogra, P.; Carter, R.; Awad, W.; Neale, G.; Thomas, P. G.; and Youngblood, B. (2017) “De Novo Epigenetic Programs Inhibit PD- 1 Blockade-Mediated T-Cell Rejuvenation” Cell; 170(1): 142-157. v Featured in Science Magazine and Faculty of 1000 Prime for special significance

7. Youngblood, B.; Hale, J. S.; Kissick, H. T.; Ahn, E.; Xu, X.; Wieland, A.; Araki, K; West, E. E.; Ghoneim, H. E.; Fan, Y.; Dogra, P.; Davis, C. W.; Konieczny, B. T.; Antia, R.; Cheng, X; and Ahmed, R. (2017) “Effector CD8 T Cells Dedifferentiate into Long-Lived Memory Cells” Nature; 552(7685): 404- 409

8. Abdelsamed, H. A.; Moustaki, A.; Fan, Y.; Dogra. P.; Ghoneim, H. E.; Zebley, C.; Triplett, B.; Sekaly, R. P.; Youngblood, B. (2017) “Human Memory CD8 T-Cell Effector-Potential is Epigenetically Preserved during In Vivo Homeostasis” Journal of Experimental Medicine; 214(6): 1593- 1606

9. Ghoneim, H. E.; Zamora, A.; Thomas, P. G.; and Youngblood, B. (2016) “Cell-Intrinsic Barriers of T Cell-Based Immunotherapy” Trends in Molecular Medicine; 22(12): 1000- 1011. v Selected among Trends in Molecular Medicine Best of 2016

10. Dogra, P.; Ghoneim, H. E.; Abdelsamed, H.A.; Youngblood, B. (2016) “Generating Long- lived CD8+ T-Cell Memory: Insights from Epigenetic Programs.” European Journal of Immunology; 46(7): 1548- 62.

11. Ghoneim, H. E.; and McCullers, J. A. (2014) “Adjunctive Corticosteroid Therapy Improves Lung Immunopathology and Survival during Severe Secondary Pneumococcal Pneumonia in Mice.” Journal of Infectious Diseases; 209(9): 1459-68. v Highlighted in the St. Jude Scientific Report 2014 for special significance

12. Ghoneim, H. E.; Thomas, P. G.; and McCullers, J. A. (2013). “Depletion of Alveolar Macrophages during Influenza Infection Facilitates Bacterial Superinfections.” Journal of Immunology; 191(3): 1250-59. v Highlighted in the St. Jude Scientific Report 2014 for special significance

13. Alymova, I. V.; Green, A. M.; Van De Velde, N.; McAuley, J. L.; Boyd, K. L.; Ghoneim, H. E.; and McCullers, J. A. (2011). “Immunopathogenic and Antibacterial Effects of H3N2 Influenza A Virus PB1-F2 Map to Amino Acid Residues 62, 75, 79, and 82.” Journal of Virology; 85(23):12324-33.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

14. Marcelin, G.; Aldridge, J. R.; Duan, S.; Ghoneim, H. E.; Rehg, J.; Marjuki, H.; Boon, A. C.; McCullers, J. A.; and Webby, R. J. (2011). “Fatal Outcome of Pandemic H1N1 2009 Influenza Virus Infection is Associated with Immunopathology and Impaired Lung Repair, Not Enhanced Viral Burden, in Pregnant Mice.” Journal of Virology; 85(21):11208-19.

3

164 Hazem E. Ghoneim, PhD 04/2020

15. Ellebedy, A. H.; Lupfer, C.; Ghoneim, H. E.; DeBeauchamp, J.; Kanneganti, T. D.; and Webby, R. J. (2011). “Inflammasome-Independent Role of the Apoptosis-associated Speck- like Protein Containing CARD (ASC) in the Adjuvant Effect of MF59.”, Proc Natl Acad Sci USA; 108(7):2927-32.

RESEARCH EXPERIENCE Mar 2014–Mar 2019 Postdoctoral Fellow, Departments of Infectious Diseases and Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA - Dr. Benjamin Youngblood’s lab Jan 2010– Graduate Research Assistant, Dec 2013 Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, USA - Prof. Dr. Jonathan A. McCullers’ lab Aug–Dec 2009 Graduate Research Assistant, Molecular Sciences Department, University of Tennessee Health Science Center, Memphis, TN, USA Oct 2005– Research Assistant, Jul 2009 Department of Microbiology and Immunology, Faculty of , Cairo University, Egypt

INVITED TALKS Mar 2020 The 23rd Annual Meeting of the Translational Research Cancer Centers Consortium, Seven Spring, PA, USA Nov 2019 The Inaugural Symposium of the Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center, Columbus, OH, USA Aug 2019 Work-in-Progress Seminar, The Infectious Diseases Institute, the Ohio State University, Columbus, OH, USA Aug 2019 Research-in-Progress Seminar, Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center, Columbus, OH, USA May 2019 The 20th Annual Scientific Meeting, the Ohio State University Comprehensive Cancer Center, Columbus, OH, USA May 2019 The Everett D. Reese Immunology and Research Seminar Series, College of Medicine, the Ohio State University, Columbus, OH, USA Nov 2018 Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA Oct 2018 UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA Oct 2018 Department of Microbial Infection and Immunity, College of Medicine, the Ohio State University, Columbus, OH, USA Sep 2018 Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA

4 Hazem E. Ghoneim, PhD 04/2020

Aug 2018 Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA May 2018 Research Department, the Children’s Cancer Hospital 57357, Cairo, Egypt Apr 2018 The World Academy of Sciences/BioVision Alexandria.NXT 2018 Conference, Alexandria, Egypt Feb 2018 Department of Biological Sciences, University of Memphis, Memphis, TN, USA Jan 2018 Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA Jan 2018 Keystone Symposium: “T Cell Dysfunction, Cancer and Infection”, Breckenridge, CO, USA Jun 2017 The Federation of Clinical Immunology Societies Annual Meeting, Chicago, IL, USA (Thematic Talk) Feb 2017 EPIC meeting, St. Jude Children’s Research Hospital, Memphis, TN, USA Oct 2016 Statewide Communicable and Environmental Diseases and Emergency Preparedness Meeting, Nashville, TN, USA (Keynote Speaker) Feb 2015 Responsible Conduct of Research “Mentoring” Workshop, St. Jude Children’s Research Hospital, Memphis, TN, USA (Panel Discussion) Sep 2014 The Fifth European Scientific Working group on Influenza Conference, Riga, Latvia Jul 2014 Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA May 2014 Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA Feb 2014 Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Egypt Dec 2013 Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA Nov 2012 Infectious Diseases Research Conference, St. Jude Children’s Research Hospital, Memphis, TN, USA Apr 2012 The First International Graduate Student Immunology Conference, Harvard Medical School, Harvard University, Boston, MA, USA

POSTER PRESENTATIONS Oct 2018 The Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference, New York City, NY, USA (Peer-reviewed) Jan 2018 Keystone Symposium: “T Cell Dysfunction, Cancer and Infection”, Breckenridge, CO, USA (Peer-reviewed)

5 Hazem E. Ghoneim, PhD 04/2020

Oct 2017 The AACR Special Conference on Tumor Immunology and Immunotherapy in Boston, MA, USA (Peer-reviewed) May 2016 The Annual Meeting of the American Association of Immunologists, Seattle, WA, USA (Peer- reviewed) Nov 2015 The 2015 Faculty/Postdoctoral Poster Session, St. Jude Children’s Research Hospital, Memphis, TN, USA (Invited) Apr 2015 Keystone Symposium: “DNA Methylation”, Keystone, CO, USA (Peer- reviewed) Jun 2014 The Federation of Clinical Immunology Societies Annual Meeting, Chicago, IL, USA (Peer- reviewed) May 2014 Postdoctoral Professional Development Week, St. Jude Children’s Research Hospital, Memphis, TN, USA (Peer-reviewed) Jul 2013 The Seventh Annual Network Meeting of the NIAID Centers of Excellence for Influenza Research and Surveillance, Memphis, TN, USA (Peer-reviewed) Jun 2013 The Federation of Clinical Immunology Societies Annual Meeting, Boston, MA, USA (Peer- reviewed) Apr 2013 Global Health and Infectious Disease Conference, School of Medicine, Washington University, St. Louis, MO, USA (Peer-reviewed) Nov 2012 The Fifth Annual Pediatric Research Day, The Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, USA (Peer-reviewed) Nov 2012 The 2012 Faculty/Postdoctoral Poster Session, St. Jude Children’s Research Hospital, Memphis, TN, USA (Invited) Apr 2012 The First International Graduate Student Immunology Conference, Harvard Medical School, Harvard University, Boston, MA, USA (Peer-reviewed) Feb 2011 Keystone Symposium: “A Fine Balance between Tolerance and Immunity”, Vancouver, BC, Canada (Peer-reviewed)

ACADEMIC AND COMMUNITY SERVICE Mar 2020 Chair of “Tumor Microenvironment” session, the 23rd Annual Meeting of the Translational Research Cancer Centers Consortium, Seven Spring, PA, USA May 2019–Present Faculty Search Committee, Department of Microbial Infection and Immunity, the Ohio State University, College of Medicine; Chair: Daniel Wozniak, PhD May 2019–Present Space and Resources Committee, Department of Microbial Infection and Immunity, the Ohio State University, College of Medicine; Chair: Eugene Oltz, PhD—Flow Cytometry/Sorting, and Omics Data Storage sub-committees. Apr 2019–Apr 2020 Scientific Judge, Annual College of Medicine Trainee Research Day, the Ohio State University, Columbus, OH, USA

6 Hazem E. Ghoneim, PhD 04/2020

2016–2018 St. Jude Science Ambassador for Public Outreach: Apr 2018 Chair of a session entitled “The Role of Science Academies and Intergovernmental Organizations in Science Diplomacy”, the World Academy of Sciences/BioVision Alexandria.NXT 2018 Conference, Alexandria, Egypt Nov 2017 Presented a lecture “The Future of Cancer Immunotherapy”, White Station High School, Memphis, TN, USA Sep 2017 Presented a lecture “How to Be a Future Scientist”, Grahamwood Elementary School, Memphis, TN, USA Dec 2016 Presented a lecture “Human Genetics and Cancer” and organized a practical session on DNA extraction, Memphis Central Library, Memphis, TN, USA Oct 2016 Taught preparation strategies for the Science section of the American College Test (ACT), Memphis, TN, USA May 2016 Participated in a vaccination campaign organized by Shelby County Health Department for protection against measles, Memphis, TN, USA May 2016 Organized sessions on public health education and protection strategies during the measles outbreak in Memphis, TN, USA 2014–Present Ad Hoc Reviewer: Scientific Reports; Journal; Journal of Cellular and Molecular Medicine; Molecular Immunology Journal; International Immunopharmacology Journal; BMC Infectious Diseases Journal; PLOS ONE Journal.

ADVISING AND MENTORING Dec 2019–Present Kaitlin Read, PhD Student in the Biomedical Sciences Graduate Program, the Ohio State University, Columbus, OH (Thesis and Mentorship Committees)

Dec 2019–Present Anqi Li, PhD Student in the Biomedical Sciences Graduate Program, the Ohio State University, Columbus, OH (Thesis Committee)

Aug 2019–Present Parker Bauman, Undergraduate Student, Honors Neuroscience Program-Pre- Medical Track, the Ohio State University, Columbus, OH

Aug 2019–Present Nicole Osborne, Undergraduate Student, Microbiology Program, the Ohio State University, Columbus, OH

TEACHING EXPERIENCE 2019–Present Assistant Professor, The Ohio State University, College of Medicine, Columbus, OH, USA - “Immunology and Inflammation” course, the Signature Program Translational Science- BSGP 8800.01 (Fall 2019) - “Cellular and Molecular Immunology”- MEDMCIM 7010 (Spring 2020)

Spring 2018 Adjunct Lecturer, The St. Jude Graduate School of Biomedical Sciences, Memphis, TN, USA 7 Hazem E. Ghoneim, PhD 04/2020

- Participated in teaching Immunology course Spring 2017 Visiting Lecturer, Rhodes College, Memphis, TN, USA - Participated in teaching Senior Biology Seminars course Fall 2010 and Teaching Assistant, Spring 2011 University of Tennessee, College of Medicine, Memphis, TN, USA - Taught the practical methodology of Medical and Diagnostic Microbiology. Oct 2005– Teaching Assistant, May 2009 Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Egypt - Taught the practical sections, demonstrated the laboratory techniques, and assisted in grading and E-learning of the following courses: • “Medical and Diagnostic Microbiology” for the Medical and Diagnostic Microbiology Diploma students • “Microbiological Quality Control and Assurance” for the Pharmaceutical Cosmetics Diploma students • “Pharmaceutical and Medical Microbiology” for the undergraduate Pharmacy students

ADDITIONAL TRAINING Apr 2017 Clinical Shadowing Program, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA Apr 2015 Workshop on Epigenomics, Keystone Symposium, Keystone, CO, USA Jun 2014 Flow Cytometry for Immune Monitoring Course, the Federation of Clinical Immunology Societies Annual Meeting, Chicago, IL, USA Feb 2011 Global Health Travel Awardees Pre-meeting Workshop, Vancouver, BC, Canada Apr 2007 Viral Hepatitis Workshop, the American University, Cairo, Egypt Dec 2006 Bioinformatics and Systems Biology, Faculty of Pharmacy, Cairo University, Egypt Jul 2006 Protein Crystallography and Drug Design, Faculty of Sciences, Cairo University, Egypt

8 JACOB S. YOUNT, PhD The Ohio State University Email: [email protected] 460 W 12th Avenue Phone: (614)688-1639 Biomedical Research Tower Room 790 Google Scholar: http://goo.gl/7QIC5y Columbus, OH 43210 Twitter: @YountLabOSU www.YountLab.org

ACADEMIC POSITIONS The Ohio State University College of Medicine 2012 – 2019 Assistant Professor, Department of Microbial Infection and Immunity 2019 – present Associate Professor (tenured), Department of Microbial Infection and Immunity Additional OSU Appointments: - Department of Microbiology - Comprehensive Cancer Center 2019 – present Co-Director, Viruses and Emerging Pathogens Program of the Ohio State University Infectious Diseases Institute

City University of New York, Sophie Davis School of Biomedical Education 2004 – 2012 Adjunct Instructor (while an advanced graduate student and postdoctoral fellow)

ACADEMIC TRAINING The Rockefeller University 2007 – 2011 Postdoctoral Fellow 2011 – 2012 Research Associate Mentor: Howard C. Hang Co-mentor 2011-2012: Charles M. Rice Studied post-translational regulation of innate antiviral immunity using novel chemical tools and mass spectrometry.

Mount Sinai School of Medicine of New York University 2002 – 2007 PhD in Biomedical Science Mentor: Thomas M. Moran Co-mentor: Carolina B. López Studied viral triggers of dendritic cell maturation and cellular proteins that detect virus infections.

Grove City College, Pennsylvania 1998 – 2002 BS in Biochemistry, magna cum laude Completed two summer undergraduate research fellowships at the University of Pittsburgh studying 1) bacterial decontamination strategies using foam polymers (Mentor: Alan Russell) and 2) mass spectrometry-based detection of neuropeptides (Mentor: Joseph Grabowski).

AWARDS, DISTINCTIONS, AND HONORS 2019 Milstein Travel Award – International Cytokine and Interferon Society 2018,19 American Association of Immunologists Careers in Immunology Fellowship Grant (Declined twice due to other funding) 2014,15,16,18 American Association of Immunologists Early Career Faculty Travel Grant 2017 OSU FAME Career Development Award for Researchers 2011 – 2015 NIH/NIAID K99/R00 Pathway to Independence Career Award 2010 Rockefeller University Postdoctoral Association Career Development Award 2008 – 2010 Cancer Research Institute Postdoctoral Fellowship in Immunology 2005, 06, 07 Mount Sinai Graduate Student Travel Award 2004 – 2005 Mount Sinai Virology Training Grant Predoctoral Fellowship 2003, 04 Mount Sinai Excellence in Teaching Award 2001 Howard Hughes Medical Institute Undergraduate Research Fellowship 2000 University of Pittsburgh Department of Chemical and Petroleum Engineering Undergraduate Research Fellowship

PUBLICATIONS (45 total; Google Scholar h-index = 28)

1. Chemudupati M, Smith AC, Fillinger RJ, Kenney AD, Zhang L, Zani A, Liu SL, Anderson MZ, Sharma A, and Yount JS. Short chain fatty acid butyrate promotes virus infection by repressing interferon stimulated genes. BioRxiv Preprint. 2020. 2. Qin Z, Bonifati S, St Gelais C, Li TW, Kim SH, Antonucci JM, Mahboubi B, Yount JS, Xiong Y, Kim B, and Wu L. The dNTPase activity of SAMDH1 is important for its suppression of innate immune responses in differentiated monocytic cells. Journal of Biological Chemistry. 2020. [abstract] 3. Zani A, Zhang L, Kenney A, McMichael TM, Chemudupati M, Kwiek JJ, Liu SL, and Yount JS. Interferon-Induced Transmembrane Proteins Inhibit Cell Fusion Mediated by Trophoblast Syncytins. Journal of Biological Chemistry. 2019. [abstract] 4. Kenney AD, McMichael TM, Imas A, Chesarino NM, Zhang L, Dorn LE, Wu Q, Alfaour O, Amari F, Chen M, Zani A, Chemudupati M, Accornero F, Coppola V, Rajaram MVS#, and Yount JS#. IFITM3 protects the heart during influenza virus infection. Proceedings of the National Academy of Sciences. 2019. [abstract] #Corresponding authors. 5. Sharma A, McLaughlin RN, Basom RS, Kikawa C, OhAinle M, Yount JS, Emerman M, and Overbaugh J. Macaque interferon-induced transmembrane proteins limit replication of SHIV strains in an envelope-dependent manner. PLOS Pathogens. 2019. [abstract] 6. Chemudupati M, Kenney AD, Bonifati S, Zani A, McMichael TM, Wu L, and Yount JS. From APOBEC to ZAP: Diverse Mechanisms Used by Cellular Restriction Factors to Inhibit Virus Infections. Biochimica et Biophysica Acta – Molecular Cell Research. 2019. [abstract] 7. McMichael TM, Zhang Y, Kenney AD, Zhang L, Zani A, Lu M, Chemudupati M, Li J, and Yount JS. IFITM3 restricts human metapneumovirus infection. Journal of Infectious Diseases. 2018. [abstract] 8. Zani A and Yount JS. Antiviral protection by IFITM3 in vivo. Current Clinical Microbiology Reports. 2018. Review. [abstract] 9. Chen S, Bonifati S, Qin Z, St. Gelais C, Kodigepalli KM, Barrett BS, Kim SH, Antonucci JM, Ladner KJ, Vuzovetsky O, Knecht KM, Xiong Y, Yount JS, Guttridge DC, Santiago ML, and Wu L. SAMHD1 suppresses innate immune responses to viral infections and inflammatory stimuli by inhibiting the NF- kB and interferon pathways. Proceedings of the National Academy of Sciences. 2018. [abstract] 10. McMichael TM, Zhang L, Chemudupati M, Hach JC, Kenney AD, Hang HC, and Yount JS. Palmitoyltransferase ZDHHC20 Enhances IFITM3 S-Palmitoylation and Antiviral Activity. Journal of Biological Chemistry. 2017. [abstract] 11. Chesarino NM, Compton A, McMichael TM, Zhang L, Kenney A, Soewarna V, Doering R, Davis M, Schwartz O, and Yount JS. IFITM3 requires an amphipathic helix for restriction of virus infections. EMBO Reports. 2017. [abstract] 12. Seveau S, Turner J, Gavrilin M, Torrelles JB, Hall-Stoodley L, Yount JS, and Amer AO. Checks and balances between the inflammasome and the autophagosome. Accepted at Journal of Molecular Biology. 2017. Review. [abstract] 13. Kenney A, Dowdle JA, McMichael TM, Bozacca L, St. Gelais C, Panfill A, Schlesinger LS, Anderson MZ, Greene P, Lopez CB, Rosenberg B, Wu L, and Yount JS. Human Genetic Determinants of Viral Diseases. Annual Review of Genetics. 2017. Invited Review. [abstract] 14. McMichael TM, Chemudupati M, and Yount JS. A balancing act between IFITM3 and IRF3. Cellular and Molecular Immunology. 2017. Invited Commentary. [abstract] 15. Tsou LK, Yount JS, and Hang HC. Epigallocatechin-3-gallate inhibits bacterial virulence and invasion of host cells. Bioorganic and Medicinal Chemistry. 2017. [abstract] 16. St. Gelais C, Kim SH, Ding L, Yount JS, Ivanov D, Spearman P, and Wu L. A putative cyclin- binding motif in human SAMHD1 contributes to protein phosphorylation, localization and stability. Journal of Biological Chemistry. 2016. [abstract] 17. Compton A, Roy N, Porrot F, Billet A, Casartelli N, Yount JS, Liang C, and Schwartz O. Natural mutations in IFITM3 modulate post-translational regulation and toggle antiviral specificity. EMBO Reports. 2016. [abstract] 18. Antonucci J, St. Gelais C, de Silva S, Yount JS, Tang C, Ji X, Xiong Y, Kim B, and Wu L. RNase activity of SAMHD1 is not essential for HIV-1 restriction in cells. Nature Medicine. 2016. [abstract] 19. Percher A, Ramakrishnan S, Yuan X, Thinon E, Yount JS#, and Hang HC#. Mass-tag labeling reveals site-specific and quantitative levels of protein S-fatty acylation. Proceedings of the National Academy of Science. 2016. [abstract] #Corresponding authors. *Highlighted in ACS Chemical Biology, May 2016, Spotlight section. 20. Tsou LK, Lara-Tejero M, RoseFigura J, Zhang Z, Wang YC, Yount JS, Lefefre M, Dossa P, Kata J, Guan F, Lam W, Cheng YC, Galan J, and Hang HC. Anti-bacterial flavonoids from medicinal plants covalently inactivate type III protein secretion substrates. Journal of the American Chemical Society. 2016. [abstract] 21. Wang F, St. Gelais C, de Silva S, Zhang H, Geng Y, Shepard C, Kim B, Yount JS, Wu L. Phosphorylation of mouse SAMHD1 regulates its restriction of human immunodeficiency virus type 1 infection, but not murine leukemia virus infection. Virology. 2016. [abstract] 22. Chesarino NC, McMichael TM, and Yount JS. E3 ubiquitin ligase NEDD4 promotes influenza virus infection by decreasing levels of the antiviral protein IFITM3. PLOS Pathogens. 2015. [abstract] 23. Melvin, WJ, McMichael TM, Chesarino NC Hach JC, and Yount JS. IFITMs from mycobacteria confer resistance to influenza virus when expressed in human cells. Viruses. 2015. [abstract] 24. Chesarino NC, McMichael TM, and Yount JS. Regulation of the trafficking and antiviral activity of IFITM3 by post-translational modifications. Future Microbiology. 2014. Invited Review. [abstract] 25. Chesarino NM, Hach JC, Chen JL, Zaro BW, Rajaram M, Turner J, Schlesinger LS, Pratt M, Hang HC, and Yount JS. Chemoproteomics reveals Toll-like receptor fatty acylation. BMC Biology. 2014. [abstract] 26. Chesarino NC, McMichael TM, Hach JC, and Yount JS. Phosphorylation of the antiviral protein IFITM3 dually regulates its endocytosis and ubiquitination. Journal of Biological Chemistry. 2014. [abstract] *Related artwork chosen for the journal cover. 27. St. Gelais C, Hach JC, DeSilva S, Yount JS, and Wu L. Identification of cell cycle proteins interacting with human and mouse anti-retroviral protein SAMHD1. Journal of Virology. 2014. [abstract] 28. Hach JC, McMichael T, Chesarino N, and Yount JS. Palmitoylation on conserved and non- conserved cysteines of murine IFITM1 regulates its stability and anti-influenza A virus activity. Journal of Virology. 2013. [abstract] *Chosen by the editors for inclusion in “Spotlight,” a feature in the Journal of Virology that highlights research articles of significant interest. 29. Yount JS, Zhang MM, and Hang HC. Emerging roles for protein S-palmitoylation in immunity from chemical proteomics. Current Opinion in Chemical Biology. 2013. Review. [abstract] 30. Yount JS, Karssmeijer RA, and Hang HC. S-palmitoylation and ubiquitination differentially regulate interferon-induced transmembrane protein 3 (IFITM3)-mediated resistance to influenza virus. Journal of Biological Chemistry. 2012. [abstract] 31. Moltedo B, Li W, Yount JS, and Moran TM. Unique type I interferon responses determine the functional fate of migratory lung dendritic cells during influenza virus infection. PLOS Pathogens. 2011. [abstract] 32. Cotter C, Kim W-C, Nguyen M, Yount JS, López CB, Blaho J, and Moran TM. The virion host shut- off (vhs) protein of HSV-1 blocks the replication-independent activation of NF-κB in dendritic cells in the absence of type I interferon signaling. Journal of Virology. 2011. [abstract] 33. Yount JS, Zhang M, and Hang HC. Visualization and identification of fatty acylated proteins using chemical reporters. Current Protocols in Chemical Biology. 2011. [abstract] 34. Yount JS, Charron G, and Hang HC. Bioorthogonal proteomics of 15-hexadecynyloxyacetic acid chemical reporter reveals preferential targeting of fatty acid modified proteins and biosynthetic enzymes. Bioorganic & Medicinal Chemistry. 2011. [abstract] 35. Charron G, Tsou LK, Maguire W, Yount JS, and Hang HC. Alkynyl-farnesol reporters for detection of protein S-prenylation in cells. Molecular BioSystems. 2011. [abstract] 36. Yount JS, Moltedo B, Yang YY, Charron G, Moran TM, Lopez CB, and Hang HC. Palmitoylome profiling reveals S-palmitoylation-dependent antiviral activity of IFITM3. Nature Chemical Biology. 2010. [abstract] *Reviewed by Faculty of 1000 Biology as “Must Read.” 37. Yount JS, Tsou LK, Dossa PD, Kullas AL, van der Velden AW, and Hang HC. Visible fluorescence detection of type III protein secretion from bacterial pathogens. Journal of the American Chemical Society. 2010. [abstract] 38. Cotter C, Nguyen ML, Yount JS, Lopez CB, Blaho JA, and Moran TM. The virion host shut- off (vhs) protein blocks a TLR-independent pathway of Herpes Simplex Virus type 1 recognition in dendritic cells. PLOS One. 2010. [abstract] 39. Charron G, Zhang MM, Yount JS, Wilson J, Raghavan AS, Shamir E, and Hang HC. Robust fluorescent detection of protein fatty-acylation with chemical reporters. Journal of the American Chemical Society. 2009. [abstract]

40. Friedman CS, O’Donnel MA, Legarda-Addison D, Cárdenas WB, Yount JS, Basler CF, Komuro A, Horvath CM, Sun S-C, Xavier R, Ting AT. The tumor suppressor CYLD is a negative regulator of RIG-I-mediated anti-viral response. EMBO Reports. 2008. [abstract]

41. Yount JS, Gitlin L, Moran TM, and Lopez CB. MDA5 participates in the detection of paramyxovirus infection and is essential for the early activation of dendritic cells in response to Sendai Virus defective interfering particles. Journal of Immunology. 2008. [abstract] 42. Yount JS, Moran TM, and Lopez CB. Cytokine-Independent Upregulation of MDA5 in Viral Infection. Journal of Virology. 2007. [abstract] 43. Yount JS, Kraus TA, Horvath CM, Moran TM, and Lopez CB. A Novel Role for Viral Defective Interfering Particles in Enhancing Dendritic Cell Maturation. Journal of Immunology. 2006. [abstract] 44. Lopez CB, Yount JS, Hermesh T, and Moran TM. Sendai Virus Infection Induces Efficient Adaptive Immunity Independently of Type I Interferons. Journal of Virology. 2006. [abstract] 45. Lopez CB, Yount JS, and Moran TM. Toll-Like Receptor-Independent Triggering of Dendritic Cell Maturation by Viruses. Journal of Virology. 2006. Review. [abstract]

FUNDING AS PRINCIPAL INVESTIGATOR CURRENT 2019 – 2020 OSU Infectious Diseases Institute Seed Grant (MPI w/Federica Accornero )“Role of Bex1 in mediating cardiac viral responses” ($25,000 total award) 2019 – 2021 NIH/NIAID R21 (MPI w/ Murugesan Rajaram) “Mechanisms of cardioprotection by the innate immune system during influenza virus infections” ($414,961 total award) 2019 – 2021 NIH/NIAID R21 “Inhibition of cell-to-cell fusion by interferons and interferon-induced proteins” ($429,000 total award) 2017 – 2022 NIH/NIAID R01 “Mechanisms of innate resistance to virus infections” ($1,872,000 total award) PAST 2015 – 2016 NIH/NIAID R56 “Molecular control of IFITM3 in restricting influenza virus infection” ($366,224 total award) 2013 – 2015 OSU Public Health Preparedness for Infectious Diseases Pilot Grant “Regulation of Toll-like receptor activity by fatty acylation” ($100,000 direct) 2013 – 2015 NIH/NIAID R00 “Mechanistic analysis of a posttranslationally modified innate antiviral effector”($493,543 total award) 2013 – 2014 ARNO Therapeutics Pilot Grant “Does AR-12 inhibit influenza virus?” ($17,000 direct) 2011 – 2012 NIH/NIAID K99 “Mechanistic analysis of a posttranslationally modified innate antiviral effector” ($139,995 total award) 2008 – 2010 Cancer Research Institute Postdoctoral Fellowship in Immunology “Uncaging of small molecules to control dendritic cell activity” ($124,500 direct)

NATIONAL AND INTERNATIONAL SERVICE Manuscript Reviewer: Cell Chemical Biology, Journal of Clinical Investigation, PLOS Pathogens, Journal of Virology, Journal of Molecular Biology, EMBO Journal, EMBO Reports, EMBO Molecular Medicine, Cell Reports, Critical Reviews in Microbiology, Future Virology, Future Microbiology, Virulence, Scientific Reports, Infection and Immunity, Journal of the American Chemical Society, Cellular and Molecular Immunology, PLOS One, Molecular and Cellular Biology, BMC Medical Genetics, BMC Research Notes, Viruses, Tuberculosis, Journal of Biological Chemistry, Cancers, Veterinary Microbiology, Journal of Biochemistry, Cytokine, Vaccines, Science Advances, Molecular Therapy, Emerging Microbes and Infections, Frontiers in Microbiology, Virus Research

Editor: PLOS Pathogens (Guest Editor, 2016, 2018, 2020) Grant Reviewer (ad hoc): 2020 European Research Council, Advanced Grant – Immunity and Infection 2020 American Heart Association - Transformative Research Award, Immunol/Microbiol 2015, National Institutes of Health - Virology B 19 2019 Medical Research Council UK - Research Grant reviewer 2019 Pasteur Institute Italy - internal grant competition 2019 National Institute of Health - Virology SEP 2018, American Heart Association - Microbiology Fellowship Bsc 19 2018 National Institutes of Health CSR “Anonymization Project” 2015,1 American Heart Association - Microbiology Bsc1/2 6,17,18 2017 Wellcome Trust-DBT India Alliance 2016,1 Wellcome Trust UK 7

Co-chair: Block Symposium "Pathogen control and evasion strategies." American Association of Immunologists 2014 Annual Meeting. Pittsburgh, PA. 2014. Convener: “Innate Immunity: Effector Mechanisms” session. American Society for Virology 2019 Annual Meeting. Minneapolis, MN. 2019. ASV Travel Award Committee: American Society for Virology, appointed 2018 – 2020 Co-Organizer: 2019 Midwest Virology Symposium. Columbus, OH. Inaugural meeting, 250 registrants from 30 institutions. MEMBERSHIPS Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

American Association of Immunologists American Society for Virology

175 American Society for Microbiology American Society for Biochemistry and Molecular Biology International Cytokine & Interferon Society Sigma Xi

UNIVERSITY SERVICE Committees 2014 – present OSU Institutional Biosafety Committee, Member 2017 – present Department of Microbial Infection and Immunity Seminar Committee, Chair 2018 – present OSU Infectious Diseases Institute (IDI) Education Committee, Member 2018—2019 OSU IDI Viruses and Emerging Pathogens Seminar Series, Co-Director 2014—present OSUMC NIH T32 Systems and Integrated Biology Training Grant, Review and Interview Committee 2019 – present Department of Microbial Infection and Immunity Executive Committee, Member 2018 OSU Mouse Genetic Engineering Core Facility Oversight Committee, Member 2014 – 2016 Center for Microbial Interface Biology Symposium, Planning Committee member 2014, 15, 16 Public Health Preparedness for Infectious Disease Pilot Grant, Proposal reviewer

Education and Advising

2014 – present Biomedical Sciences Graduate Program, Microbial Pathogenesis Emphasis Area, Co-Director 2018 – present Ohio Virology Association (OVA), Student group faculty advisor 2013 – 2019 Buckeyes for Ronald McDonald House, Student group faculty advisor 2017, 2018 OSUMC Bench 2 Bedside Seminar, Faculty advisor for trainee presenters

Reviewing/Judging 2015, 16, 17, 18 Hayes Graduate Research Forum, Oral Presentation Judge, Biological sciences 2013, 14, 15, 17 OSUMC Trainee Research Day, Poster Judge 2013, 14, 17, 20 Roessler Medical Student Research Scholarship, Proposal reviewer

INVITED PRESENTATIONS AT UNIVERSITIES

• University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA. 2020. • Johns Hopkins University, Immunopathology Seminar Series, Baltimore, MD. 2020. • Indiana University, Microbiology Seminar Series, Bloomington, IN. 2019. • Washington State University, Department of Veterinary Microbiology and Pathology, Pullman, WA. 2018 • University of Chicago, Department of Microbiology, Chicago, IL. 2018. • Medical College of Wisconsin, Department of Microbiology and Molecular Genetics, Milwaukee, WI. 2017. • Northwestern University, Department of Microbiology-Immunology, Chicago, IL. 2017. • University of Toledo, Department of Chemistry and Biochemistry, Toledo, OH. 2017. • West Virginia University, Immunology and Microbial Pathogenesis Seminar, Morgantown, WV. 2017. • Icahn School of Medicine at Mount Sinai, Department of Microbiology, New York, NY. 2017. • University of Toledo, Department of Medical Microbiology, Toledo, OH. 2016. • Oklahoma State University, Center for Respiratory and Infectious Diseases, Oklahoma City, OK. 2016.

ORAL PRESENTATIONS AT CONFERENCES

• Cytokines 2019 – Annual Meeting of the International Cytokine and Interferon Society. Vienna, Austria. 2019. (Short Talk) • American Society for Virology 36th Annual Meeting. Madison, WI. 2017. (Workshop Presentation) • Viruses and Cells Gordon Research Conference. Lucca, Italy. May, 2017. (Short Talk) • Midwest Microbial Pathogenesis Conference. Indianapolis, IN. 2015. (Invited Presentation) • Innate Immunity and Determinants of Microbial Pathogenesis Keystone Meeting. Olympic Valley, CA. 2015. (Systems Biology Workshop) • American Association of Immunologists Annual Meeting. Pittsburgh, PA. 2014. (Block Symposia) • International Congress of Immunology. Kobe, Japan. 2010. (Minisymposia) • International Congress of Immunology. Rio de Janeiro, Brazil. 2007. (Minisymposia)

INVITED LOCAL PRESENTATIONS

• MSTP Roundtable Presentation. 2020. • Davis Heart and Lung Research Institute Seminar Series. 2019. • OSU Biomedical Sciences Graduate Program Retreat. Internal Keynote. 2019. • RNA Virus Seminar. OSU College of Veterinary Medicine. 2018. • OSU College of Medicine Immunology Seminar Series. 2015. • Ohio Virology Association Monthly Meeting. 2015. • Bench to Outcomes Seminar Series. Nationwide Children’s Hospital. 2015. • OSU Department of Microbiology Seminar Series. 2014. • OSU Center for Microbial Interface Biology Symposium. 2014. • RNA Virus Seminar. OSU College of Veterinary Medicine. 2014. • Child Health Research Seminar. Nationwide Children’s Hospital. 2013.

ORAL CONFERENCE PRESENTATIONS BY YOUNT LABORATORY MEMBERS

• American Society for Virology 38th Annual Meeting. Minneapolis, MN. 2019. (Two Workshop Presentations) • American Society for Virology 37th Annual Meeting. College Park, MD. 2018. (Workshop Presentation) • American Society for Virology 36th Annual Meeting. Madison, WI. 2017. (Workshop Presentation) • American Association of Immunologists Annual Meeting. Seattle, WA. 2016. (Block Symposia) • American Association of Immunologists Annual Meeting. New Orleans, LA. 2015. (Block Symposia) • Ohio Branch American Society for Microbiology Annual Meeting. Elyria, OH. 2015. (Podium Presentation) • Ohio Branch American Society for Microbiology Annual Meeting. Columbus, OH. 2014. (Podium Presentation)

NOTEWORTHY TRAINEE ACCOMPLISHMENTS

• American Society for Microbiology Predoctoral Fellowship (McMichael) • Howard Hughes Medical Institute Gilliam Predoctoral Fellowship (McMichael) • Ohio State University Systems and Integrated Biology T32 Fellowship (Chesarino) • Ohio State University Presidential Fellowship (Chesarino) • N. Paul Hudson Award for Research Excellence from the American Society for Microbiology Ohio Branch (Chesarino) • Ohio State University Systems and Integrated Biology T32 Fellowship (Kenney) • AAI Careers in Immunology Fellowship (Chemudupati, selected twice, declined due to other funding) • Ohio State University Infectious Diseases T32 Fellowship (Chemudupati) • Ohio State University Infectious Diseases T32 Fellowship (Zani) • NSF Graduate Research Fellowship (Zani)

COMPLETE LIST OF TRAINEE MENTORING AND ACCOMPLISHMENTS

GRADUATE STUDENTS 1. Nicholas Chesarino – Graduate Student, 2013 – 2016 (completed PhD program) a. Awarded an Ohio State University Systems and Integrated Biology two-year fellowship sponsored by the NIH based on a research proposal and panel interview of the student and mentor. b. Received an American Association of Immunologists Trainee Abstract Award in 2014, 2015, and 2016. c. Received the N. Paul Hudson Award for Research Excellence at the 2014 Ohio Branch American Society for Microbiology Annual Meeting for his poster presentation as chosen by faculty judges. d. Awarded a Keystone Symposia Scholarship for travel to the 2015 Innate Immunity and Determinants of Microbial Pathogenesis Keystone conference. e. Received the Ohio State University Presidential Fellowship. “The Presidential Fellowship is the most prestigious award given by the Graduate School to recognize the outstanding scholarly accomplishments and potential of graduate students entering the final phase of their dissertation research or terminal degree project.” f. Selected as the 2015 OSU Biomedical Sciences Graduate Program nominee for the Harold M. Weintraub Graduate Student Award (Fred Hutchinson Cancer Research Center). g. After graduating, moved on to a postdoctoral fellowship in Michael Emerman’s laboratory at the Fred Hutchinson Cancer Research Center. 2. Temet McMichael – Graduate Student, 2013 – 2018 (completed PhD program) a. Awarded a 2014 FASEB MARC Travel Award. b. Received an Honorable Mention from the 2014 Ford Foundation Predoctoral Fellowship Competition. c. Chosen for a podium presentation at the Ohio Branch American Society for Microbiology 2014 Annual Meeting. d. Awarded a $1000 travel award for an “exceptional” research poster presentation at the OSU Wexner Medical Center 2014 Trainee Research Day as determined by faculty judges. e. Awarded the ASM Watkins Predoctoral Fellowship (2014-2015, declined additional two years of funding, but remained an ASM Fellow). f. Awarded a SACNAS 2014 Travel Scholarship. g. Awarded the HHMI Gilliam Predoctoral Fellowship (2015-2018). h. After graduating, moved on to the US Centers for Disease Control Epidemic Intelligence Service Fellowship Program. 3. Adam Kenney – Graduate Student, 2016 – present a. Awarded an Ohio State University Systems and Integrated Biology two-year fellowship sponsored by the NIH based on a research proposal and panel interview. b. Received an American Association of Immunologists Trainee Poster Award in 2018. c. Chosen for an oral presentation at the 2018 American Society for Virology annual meeting. d. Received an American Society for Virology Graduate Student Travel Award in 2018. e. Awarded 1st place oral presentation at the 2019 OSU Hayes Graduate Research Forum, Biological Sciences category. f. Chosen for an oral presentation at the 2019 American Society for Virology annual meeting. g. Awarded the Procter and Gamble Research Award at the 2019 OSU Biomedical Sciences Graduate Program annual retreat. 4. Ashley Zani – Graduate Student, 2018 – present a. Awarded an Ohio State University Infectious Diseases Institute Microbial Pathogenesis fellowship sponsored by the NIH in 2018 b. Received a 2018 Hodges Family Legacy Trainee Travel Award from the OSU Department of Microbial Infection and Immunity c. Received an American Society for Virology Graduate Student Travel Award in 2019. d. Awarded a National Science Foundation Graduate Research Fellowship (2019- 2022).

POSTDOCTORAL RESEARCHERS 1. Mahesh Chemudupati – Postdoctoral Fellow, 2017 – present a. Awarded a 2018 and 2019 American Association Careers in Immunology Fellowship (declined both times due to other funding) b. Awarded an Ohio State University Infectious Diseases Institute Microbial Pathogenesis fellowship sponsored by the NIH and the OSU College of Medicine in 2018 and successfully recompeted for a 2nd year of funding in 2019 c. Chosen for an oral presentation at the 2019 American Society for Virology annual meeting. d. Received a 2019 OSU Postdoctoral Association Travel Grant for attendance at the American Society for Virology annual meeting.

MEDICAL STUDENTS 1. William Melvin – Medical Student, 2013 - 2014 a. Awarded a 2014 Roessler Medical Student Research Scholarship by the OSU College of Medicine. b. Awarded a $1000 travel award for an “exceptional” research poster presentation at the OSU Wexner Medical Center 2015 Trainee Research Day as determined by faculty judges. c. Awarded the 2016 Jack H. Henry Medical Student Research Award from the OSU College of Medicine.

UNDERGRADUATE STUDENTS 1. Victoria Soewarna – Undergraduate, 2014 - 2017 a. Awarded a 2014 SOLAR Undergraduate Research Scholarship by the OSU Office of Undergraduate Research. b. Awarded a 2015 Summer Undergraduate Research Fellowship by the OSU Office of Undergraduate Education c. Awarded an Undergraduate Research Scholarship for the 2015-2016 school year by the OSU Arts and Sciences Honors Committee. 2. Rachel Doering – Undergraduate, Research for credit during summer 2015 3. Matthew Davis – Undergraduate, Research for credit in Biomedical Sciences Program, 2015 - 2017 4. Alexander Imas – Undergraduate, 2017 - 2018 5. Omar Alfaour – Undergraduate, Research for credit, 2017 - present 6. Eric Prescott – Undergraduate, 2019 - present

THESIS COMMITTEES 1. Matthew Sermersheim (BSGP, Jianjie Ma laboratory) - Graduated 2019 2. Lauren Doolittle (BSGP, Ian Davis laboratory) 3. Joel Lee (BSGP, Kevin Cassady laboratory) 4. Aaren Kettelhut (MSTP, Nicholas Funderberg laboratory) 5. Krithika Karthigeyan (Microbiology, Jesse Kwiek laboratory) 6. Don Caulkins (BSGP, Kristine Yoder laboratory) 7. Kacy Yount (BSGP Rajendar Deora and Purnima Dubey laboratories) 8. Taiwei Li (MCDB, Li Wu laboratory) 9. Victoria Maksimova (BSGP, Li Wu laboratory) 10. Peter Brechting (Microbiology, Chad Rappleye laboratory) 11. Pranav Rana (Microbiology, Daniel Wozniak laboratory) 12. Dillon McBride (Microbiology, Chad Rappleye laboratory) 13. Anna Smith (Veterinary Biosciences, Amit Sharma laboratory) 14. Zhihua Qin (Veterinary Biosciences, Li Wu laboratory)

ROTATION STUDENTS (who did not join the lab) 1. Brandon Murphy, BSGP 2. Kelsey Crossen, Microbiology 3. Ilse Hernandez, MSTP 4. Kyle Spencer, BSGP 5. Sergio Corrales Guerrero, BSGP 6. Rachel Woodfint, BSGP 7. Sabrina Lamont, Microbiology February 2020 Curriculum Vitae KEN J. OESTREICH, Ph.D.

Contact Information Address: The Ohio State University College of Medicine Department of Microbial Infection and Immunity 798 Biomedical Research Tower 460 W. 12th Ave. Columbus, OH 43210 E-mail: [email protected] Telephone: (614) 685-3549 Cell: (770) 238-8244

Current Appointments September 2019-Present Associate Professor Department of Microbial Infection and Immunity, The Ohio State University College of Medicine Columbus, OH November 2019-Present Member The Ohio State University Comprehensive Cancer Center (OSUCCC) Columbus, OH

Past Appointments June 2019-August 2019 Associate Professor Fralin Biomedical Research Institute at VTC (formerly VTCRI), Roanoke, VA Department of Biomedical Sciences and Pathobiology (DBSP), Virginia-Maryland College of Veterinary Medicine Virginia Tech, Blacksburg, VA July 2014-2019 Assistant Professor Faculty of Health Sciences, Virginia Tech, Blacksburg, VA July 2014-Present Assistant Professor Virginia Tech Carilion School of Medicine (VTCSOM), Roanoke, VA October 2013-2019 Assistant Professor Department of Biomedical Sciences and Pathobiology (DBSP), Virginia-Maryland College of Veterinary Medicine Virginia Tech, Blacksburg, VA October 2013-2019 Assistant Professor Virginia Tech Carilion Research Institute (VTCRI), Roanoke, VA

Education and Training 2008-2013 Senior Postdoctoral Fellow University of Washington, Seattle, WA Mentor: Dr. Amy S. Weinmann Area: Molecular mechanisms regulating T helper cell differentiation

1 2006-2008 Postdoctoral Fellow Emory University, Atlanta, GA Mentor: Dr. Jeremy M. Boss Area: Transcriptional regulation of PD-1 2006 Ph.D. - Microbiology and Immunology Vanderbilt University, Nashville, TN Advisor: Dr. Eugene M. Oltz Area: Mechanisms regulating TCRb rearrangement and early T cell development 2000 Bachelor of Science, Chemistry College of Arts and Sciences Tennessee Tech University, Cookeville, TN

Research and Educational Interests ▪ I am an immunologist with research and teaching interests in the areas of molecular and cellular immunology as they relate to the pathogenesis of human disease. The mission of my laboratory is to acquire fundamental knowledge regarding the regulatory mechanisms that govern T cell differentiation and function during the immune response so that it can be applied to the development of novel, targeted immunotherapies to prevent or treat human disease. My primary research interests include the role of transcriptional and environmental factors, such as cytokines, responsible for promoting these regulatory events. As an educator, I particularly enjoy sharing cutting-edge concepts and fundamental principles of immunology with students at both the graduate and undergraduate levels.

Publications 1. Powell, M.D., K.A. Read, B.K. Sreekumar, D.M. Jones, and K.J. Oestreich. IL-12 signaling drives the differentiation and function of a TH1-derived TFH1-like cell population. Scientific Reports. 9: 13991 (2019). 2. Papillion, A., M.D. Powell, D.A. Chisolm, H. Bachus, M.J. Fuller, A.S. Weinmann, A. Villarino, J.J. O’Shea, B. Leon, K.J. Oestreich, and A. Ballesteros-Tato. Inhibition of IL-2 responsiveness by IL-6 is required for the generation of GC-TFH cells. Science Immunology. 4(39): eaaw7636 (2019). 3. Powell, M.D., K.A. Read, B.K. Sreekumar, and K.J. Oestreich. Ikaros zinc finger transcription factors: Regulators of cytokine signaling pathways and CD4+ T helper cell differentiation. Frontiers in Immunology. 10:1299 (2019). 4. Ringel-Scaia, V.M., N.B. White, M.F. Lorenzo, R.M. Brock, K.E. Huie, S. Coutermarsh-Ott, K. Eden, D.K. McDaniel, S.S. Verbridge, J.H. Rossmeisl, K.J. Oestreich, R.V. Davalos, and I.C. Allen. High-frequency irreversible electroporation is an effective tumor ablation strategy that induces immunologic cell death and promotes systemic anti-tumor immunity. EBioMedicine. 44: 112-125 (2019). PMID: 31130474. 5. Read, K.A., M.D. Powell, B.K. Sreekumar, and K.J. Oestreich. In vitro differentiation of effector CD4+ T helper cell subsets. Methods in Molecular Biology. 1960: 75-84 (2019). PMID: 30798522. 6. Ringel-Scaia, V.M., M.D. Powell, K.A. Read, I.C. Allen, and K.J. Oestreich. Systemic Listeria monocytogenes infection as a model to study T helper cell immune responses. Methods in Molecular Biology. 1960: 149-160 (2019). PMID: 30798529. 7. Cesewski, E., A.P. Haring, Y. Tong, M. Singh, R. Thakur, S. Laheri, K.A. Read, M.D. Powell, K.J. Oestreich, and B.N. Johnson. Additive manufacturing of three-dimensional (3D) microfluidic- based microelectromechanical systems (MEMS) for acoustofluidic applications. Lab on a Chip. 18: 2087-2098 (2018). PMID: PMC6077993. 2 8. Read, K.A., M.D. Powell, C.E. Baker, B.K. Sreekumar, V.M. Ringel-Scaia, H. Bacchus, R.E. Martin, I.D. Cooley, I.C. Allen, A. Ballesteros-Tato, and K.J. Oestreich. Integrated STAT3 and Ikaros Zinc Finger transcription factor activities regulate Bcl-6 expression in CD4+ T helper cells. Journal of Immunology. 199: 2377-2387 (2017). PMCID: PMC5657606. 9. Mu, Q., H. Zhang, X. Liao, K. Lin, H. Liu, M.R. Edwards, S.A. Ahmed, R. Yuan, L. Li, T.E. Cecere, D.B. Branson, J.L. Kirby, P. Goswami, C.M. Leeth, K.A. Read, K.J. Oestreich, M.D. Vieson, C.M. Reilly, and X.M. Luo. Control of lupus nephritis by changes of gut microbiota. Microbiome. 5: 73 (2017). PMCID: PMC5505136. 10. McDaniel, D.K., A. Jo, V.M. Ringel-Scaia, S. Coutermarsh-Ott, D.E. Rothschild, M.D. Powell, R. Zhang, T.E. Long, K.J. Oestreich, J.S. Riffle, R.M. Davis, and I.C. Allen. TIPS pentacene loaded PE)-PDLLA core-shell nanoparticles have similar cellular uptake dynamics in M1 and M2 macrophages and in corresponding in vivo microenvironments. . 13: 1255-1266 (2017). PMCID: PMC5392431. 11. McDonald, P.W., K.A. Read, C.E. Baker, A.E. Anderson, M.D. Powell, A. Ballesteros-Tato, and K.J. Oestreich. IL-7 signaling represses Bcl-6 and the TFH gene program. Nature Communications. 7: 10285 (2016). PMCID: PMC4729877. 12. Read, K.A., M.D. Powell, and K.J. Oestreich. T follicular helper (TFH) cell programming by cytokine- mediated events. Immunology. 149: 253-261 (2016). PMCID: PMC5046059. 13. Read, K.A., M.D. Powell, P.W. McDonald, and K.J. Oestreich. IL-2, IL-7, and IL-15: Multistage regulators of CD4+ T helper cell differentiation. Experimental Hematology. 44: 799-808 (2016). PMID: 27423815. 14. Cooley, I.D., K.A. Read, and K.J. Oestreich. Trans-presentation of IL-15 modulates STAT5 activation and Bcl-6 expression in TH1 cells. Scientific Reports. 5: 15722 (2015). PMCID: PMC4620557. 15. Oestreich, K.J.*, K.A. Read, S.E. Gilbertson, K.P. Hough, P.W. McDonald, V. Krishnamoorthy, and A.S. Weinmann*. Bcl-6 directly represses the glycolysis pathway gene program. Nature Immunology. 15: 957-964 (2014). *Co-Corresponding authors PMCID: PMC4226759. Featured in “News and Views”: Man, K. and Kallies, A. Bcl-6 gets T cells off the sugar. Nat. Immunol. 15: 904-905 (2014). Featured in “Nature Reviews Immunology”: Leavy, O. BCL-6 curbs glycolysis. Nat. Rev. Immunol. 14: 650-651 (2014). 16. Oestreich, K.J., S.E. Mohn, and A.S. Weinmann. Molecular mechanisms that control the expression and activity of Bcl-6 in TH1 cells to regulate flexibility with a TFH-like gene profile. Nature Immunology. 13: 405-411 (2012). PMCID: PMC3561768. 17. Oestreich, K.J. and A.S. Weinmann. Master regulators or lineage-specifying? Changing views on CD4+ T cell transcription factors. Nature Reviews Immunology. 12: 799-804 (2012). PMCID: PMC3584691. 18. Oestreich, K.J. and A.S. Weinmann. Transcriptional mechanisms that regulate T helper 1 cell differentiation. Current Opinions in Immunology. 24: 191-195 (2012). PMCID: PMC3319510. 19. Oestreich, K.J. and A.S. Weinmann. T-bet employs diverse regulatory mechanisms to repress transcription. Trends in Immunology. 33: 78-83 (2012). PMCID: PMC3273642. 20. Oestreich, K.J. and A.S. Weinmann. Encoding stability versus flexibility: Lessons learned from examining epigenetics in T helper cell differentiation. Curr. Top. Microbiol. Immunol. 356: 145- 164 (2012). 3 PMID: 21748629. 21. Oestreich, K.J. and A.S. Weinmann. Ikaros changes the face of NuRD remodeling. Nature Immunology. 13: 16-18 (2012). PMID: 22179274. 22. Youngblood, B*, K.J. Oestreich*, S.J. Ha, J. Duraiswamy, R.S. Akondy, E.E. West, Z. Wei, P. Lu, J.W. Austin, J.L. Riley, J.M. Boss, and R. Ahmed. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity. 35: 400-412 (2011). PMCID: PMC3183460 *These authors contributed equally to this manuscript. 23. Oestreich, K.J., A.C. Huang, and A.S. Weinmann. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate TH1 gene expression patterns. Journal of Experimental Medicine. 208: 1001- 1013 (2011). PMCID: 3092354. 24. Kao, C., K.J. Oestreich, M.A. Paley, A. Crawford, J.M. Angelosanto, M.A. Ali, A.M. Intlekofer, J.M. Boss, S.L. Reiner, A.S. Weinmann, E.J. Wherry. T-bet represses expression of PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nature Immunology. 12: 663-671 (2011). PMCID: PMC3306165. 25. Clauson, C.L., K.J. Oestreich, J.W. Austin, and P.W. Doetsch. Abasic sites and strand breaks in DNA cause transcriptional mutagenesis in Escherichia coli. Proc. Natl. Acad. Sci. 107: 3657-3662 (2010). PMCID: PMC2840506. 26. Oestreich, K.J., H. Yoon, R. Ahmed, and J.M. Boss. Regulation of programmed death-1 by NFATc1 upon T cell activation. Journal of Immunology. 181: 4832-4839 (2008). PMCID: PMC2645436. 27. Osipovich, O., R. Cobb, K.J. Oestreich, S. Pierce, P. Ferrier, J. Chen, and E. Oltz. Essential function for SWI/SNF chromatin-remodeling complexes in the promoter-directed assembly of TCRb genes. Nature Immunology. 8: 809-16 (2007). PMID: 17589511. 28. Cobb, R., K.J. Oestreich, O. Osipovich, and E. Oltz. Accessibility control of V(D)J recombination. Advances in Immunology. 91: 91:45-109 (2006). PMID: 16938538. 29. Oestreich, K.J., R. Cobb, S. Pierce, J. Chen, P. Ferrier, and E. Oltz. Regulation of TCRb gene assembly by a promoter/enhancer holocomplex. Immunity. 24: 381-91 (2006). PMID: 16618597

Selected Research Conference Presentations Read KA, Sreekumar BK, Powell MD, Zafar SJ, Jones D, Baker CE, Carrillo GL, Fox MA, Allen IC, and K.J. Oestreich. Ikaros zinc finger transcription factors as novel regulators of T helper cell differentiation and STAT factor activity. Southeast Immunology Symposium, Atlanta, GA, 2019. Powell MD, Read KA, Sreekumar BK, K.J. Oestreich. IL-12 signaling drives the differentiation and function of a TH1-derived TFH1-like cell population. American Association of Immunologists Annual Meeting, San Diego, CA, 2019. Sreekumar BK, Read KA, Powell MD, Zafar SJ, K.J. Oestreich. The Ikaros zinc finger transcription factor + + Eos regulates CD4 TH1 and CD8 TC1 differentiation programs. American Association of Immunologists Annual Meeting, San Diego, CA, 2019. Read KA, Sreekumar BK, Powell MD, Jones D, K.J. Oestreich. Ikaros zinc finger transcription factors as novel regulators of STAT factor activity. American Association of Immunologists Annual Meeting, San Diego, CA, 2019. Zafar SJ, Sreekumar BK, Read KA, Powell MD, Baker CE, K.J. Oestreich. Eos and STAT5 cooperatively promote T helper 1 cell development. Autumn Immunology Conference, Chicago, IL, 2018.

4 Sreekumar BK, Read KA, Powell MD, Baker CE, Zafar J, Martin RE, Allen IC, K.J. Oestreich. An IL- 2/STAT5/Eos regulatory axis promotes TH1 cell differentiation. Southeast Immunology Symposium, Birmingham, AL, 2018. Powell MD, Read KA, Sreekumar BK, Carrillo GL, Ringel-Scaia VM, Bachus HR, Allen IC, Ballesteros- Tato A, K.J. Oestreich. Local and long-range transcriptional regulation of the Bcl6 locus by Ikaros zinc finger factors. Southeast Immunology Symposium, Birmingham, AL, 2018. Sreekumar BK, Read KA, Powell MD, Baker CE, Zafar J, Martin RE, Allen IC, K.J. Oestreich. An IL- 2/STAT5/Eos regulatory axis promotes TH1 cell differentiation. American Association of Immunologists Annual Meeting, Austin, TX, 2018. Powell MD, Read KA, Sreekumar BK, Carrillo GL, Ringel-Scaia VM, Bachus HR, Allen IC, Ballesteros- Tato A, K.J. Oestreich. Local and long-range transcriptional regulation of the Bcl6 locus by Ikaros zinc finger factors. American Association of Immunologists Annual Meeting, Austin, TX, 2018. Powell MD, Read KA, Baker CE, Sreekumar BK, Martin RE, Cooley ID, Ringel-Scaia VM, Allen IC, K.J. Oestreich. Cooperative activities between STAT3 and Ikaros Zinc Finger transcription factors regulate Bcl-6 expression in CD4+ T helper cells. American Association of Immunologists Annual Meeting, Washington D.C., 2017. Cooley ID, Read KA, Powell MD, Baker CE, Sreekumar B, Martin RE, Vaughn JE, K.J. Oestreich. The Ikaros family of transcription factors regulates the expression of the transcriptional repressor Bcl-6 in CD4+ T cells. American Society of Hematology Annual Meeting, San Diego, CA, 2016. McDonald PW, Read KA, Baker CE, Anderson AE, Powell MD, Ballesteros-Tato A, and K.J. Oestreich. Differential regulation of TH1, TFH, and TCM gene programs by cytokine-mediated events. American Association of Immunologists Annual Meeting, Seattle, WA, 2016. McDonald P.W., C.E. Baker, K.A. Read, A.E. Anderson, and K.J. Oestreich. Identification of extrinsic and + intrinsic factors that regulate CD4 TFH and TCM gene programs. American Association of Immunologists Annual Meeting, New Orleans, LA, 2015. Read, K.A., P.W. McDonald, C.E. Baker, I.D. Cooley, and K.J. Oestreich. Identification of molecular mechanisms that regulate T helper 1 cell plasticity. American Association of Immunologists Annual Meeting, Pittsburgh, PA, 2014. Read, K.A., McDonald, P.W., and K.J. Oestreich. How do lineage-defining transcription factors regulate immune cell development and function? Southwest Virginia Life Sciences Forum, Roanoke, VA, 2014. Oestreich, K.J., S.E. Mohn, and A.S. Weinmann. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate TH1 gene expression patterns. Gene Expression and Signaling in the Immune System FASEB Meeting, Aspen, CO, 2011. Oestreich, K.J., R. Ahmed, J.M. Boss. Regulation of programmed death-1 by a novel NFAT-binding regulatory element. Gene Expression and Signaling in the Immune System, Cold Spring Harbor, NY, 2008. Oestreich, K.J., R. Ahmed, J.M. Boss. Transcriptional regulation of programmed death-1, Mechanisms of Eukaryotic Transcription, Cold Spring Harbor, NY, 2007. Oestreich, K.J., S. Pierce, E. Oltz. Promoter-directed remodeling of local chromatin regulates V(D)J recombination. International FOCIS meeting, Montreal, Canada, 2004.

Invited Seminars 2020 Invited speaker, Center for Vaccines and Immunity, Nationwide Children’s Hospital (OH) 2020 Invited speaker, Department of Microbiology, University of Alabama-Birmingham (AL) 2019 Invited speaker, Department of Microbiology and Immunology, Scripps Research Institute (FL) 2019 Invited speaker, Department of Veterinary Medicine, University of Maryland

5 2018 Invited speaker, Pathology, Microbiology, and Immunology seminar series, Vanderbilt University 2018 Invited speaker, Department of Microbial Infection and Immunity, The Ohio State University 2018 Invited speaker, Department of Biological Sciences, Wake Forest University 2018 Invited speaker, Center for Immunity and Immunotherapies, Seattle Children’s Research Institute 2018 Invited speaker, Southeast Immunology Symposium, UAB School of Medicine – 2017 Invited speaker, Department of Microbiology and Immunology, Indiana University School of Medicine 2017 Invited speaker, Department of Veterinary Medicine, University of Maryland 2016 Invited speaker, Pioneers in Biomedical Research Seminar Series, Virginia Tech Carilion Research Institute 2015 Invited speaker, Block Symposium, AAI annual meeting, New Orleans 2015 Invited speaker, Department of Biological Sciences, Roanoke College 2015 Invited speaker, Grand rounds, Department of Gastroenterology, Carilion Clinic 2014 Invited speaker, Virginia-Maryland Regional College of Veterinary Medicine 2014 Invited speaker, Modeling Mucosal Immunity Symposium, Virginia Bioinformatics Institute 2012 Invited speaker, Earl Stadtman Immunology Symposium, National Institutes of Health

Academic and Professional Activities 2020 Invited Chair, T helper cell differentiation and responses: Molecular mechanisms, American Association of Immunologists annual meeting, Honolulu, HI 2019-Present Review Editor, Frontiers in Immunology 2019-Present Review Editor, Frontiers in Microbiology 2018-Present Associate Editor, The Journal of Immunology 2018-Present Member, The American Association for Cancer Research (AACR) 2016 Invited Chair, Regulating the outcomes of T helper cell differentiation, American Association of Immunologists annual meeting, Seattle, WA 2013-Present Member, The American Association of Immunologists (AAI) 2013-Present Ad hoc Reviewer: Science Immunology, Nature Communications, The Journal of Immunology, Frontiers in Immunology, European Journal of Immunology, Immunohorizons, Genes and Immunity, Scientific Reports, Human Immunology, Viral Immunology, Immunology BioMed Central, Immunology Letters, Journal of Cell Science, Cell Transplantation, PLoS One, Experimental Medicine and Biology, American Journal of Pathology, DNA and Cell Biology, Diabetes UK

Courses Taught (selected) 2016-2019 Co-Director, Immunity and Infectious Disease Track, TBMH Graduate program 2016-2019 TBMH 5004 Introduction to Immunity and Infectious Disease (Graduate level) 2015-2019 TBMH 5054 Fundamentals of Immunity and Infectious Disease (Graduate level) 2014-2019 BMVS 6714 Immunology in Health and Disease (Graduate level) 2014-2019 VTCSOM/TBMH Methods in Logic (Graduate/Medical school level)

Trainees 2019-Present Devin Jones, Graduate Student, Biomedical Sciences Graduate Program (OSU) 6 2018-Present Kaitlin Read, Graduate Student, Biomedical Sciences Graduate Program (OSU) 2017- Present Mustafa Rasheed, Medical Student, Virginia Tech Carilion School of Medicine 2017-Present Simran “Nikki” Sandhu, Medical Student, Virginia Tech Carilion School of Medicine 2017-Present Sol Moon, Medical Student, Virginia Tech Carilion School of Medicine 2015-2019 Bharath Sreekumar, Graduate Student, Translational Biology Medicine and Health graduate program (Current position: Graduated Fall 2019, currently looking for post-doctoral position) 2015-2019 Shah “Jawad” Zafar, Medical Student, Virginia Tech Carilion School of Medicine (Current position: Medical resident, University of Maryland-Baltimore) 2014-2019 Michael Powell, Graduate Student, Tranlational Biology Medicine and Health graduate program (Current position: Postdoctoral Fellow, Emory University) 2014-2017 Emily Martin, Undergraduate Student, Virginia Tech (Current position: Graduate student, Cellular and Molecular Biosciences, Wake Forest University) 2013-2017 Ian Cooley, Medical Student, Virginia Tech Carilion School of Medicine (Current position: Medical resident, University of North Carolina) 2014-2016 Ashlyn Anderson, Undergraduate Student, Roanoke College (Current position: Graduate student, Dept. of Microbiology and Immunology, UAB School of Medicine) 2013-2016 Paul McDonald, Senior Post-doctoral fellow (Current position: Senior Analyst, Proactive worldwide) 2019-Present Member, Graduate Thesis Committee, Margaret Nagai (Advisor: Dr. Irving Allen) 2019- Present Member, Graduate Thesis Committee, Devin Jones (Advisor: Dr. Ken Oestreich) 2018-2019 Member, Graduate Thesis Committee, Valentina Stevenson (Advisor: Dr. Bill Huckle) 2018-Present Member, Graduate Thesis Committee, Gabriella Carrillo (Advisor: Dr. Mike Fox) 2018-Present Member, Graduate Thesis Committee, Kaitlin Read (Advisor: Dr. Ken Oestreich) 2018-2019 Member, Graduate Thesis Committee, Kijana George (Advisor: Dr. Stefanie Robel) 2018-2019 Member, Graduate Thesis Committee, Alyssa Hendricks (Advisor: Dr. Irving Allen) 2018-Present Member, Graduate Thesis Committee, Holly Morrison (Advisor: Dr. Irving Allen) 2017-Present Member, Graduate Thesis Committee, Rebecca Brock (Advisor: Dr. Irving Allen) 2016- 2018 Member, Graduate Thesis Committee, Poorna Goswami (Advisor: Dr. Tom Inzana) 2016-2018 Member, Graduate Thesis Committee, Vanessa Brayman (Advisor: Dr. Greg Valdez) 2016-2019 Member, Graduate Thesis Committee, Bharath Sreekumar (Adv: Dr. Ken Oestreich) 2015-2019 Member, Graduate Thesis Committee, Jingjing Ren (Advisor: Dr. Xin Luo) 2015-2019 Member, Graduate Thesis Committee, Michael Powell (Advisor: Dr. Ken Oestreich) 2015- Present Member, Graduate Thesis Committee, Xiguang Xu (Advisor: Dr. David Xie) 2015-2019 Member, Graduate Thesis Committee, Veronica Ringel (Advisor: Dr. Irving Allen) 2015-2018 Member, Graduate Thesis Committee, Kristin Eden (Advisor: Dr. Irving Allen) 2014-2018 Member, Graduate Thesis Committee, Daniel Rothschild (Advisor: Dr. Irving Allen) 2014-2017 Member, Graduate Thesis Committee, Backiya Venkatachalam (Advisor: Dr. Subbiah)

Academic and Professional Honors

2019 American Association of Immunologist Early Career Faculty Travel Award Recipient 2018 American Association of Immunologist Early Career Faculty Travel Award Recipient 2017 American Association of Immunologist Early Career Faculty Travel Award Recipient 2017 “Faculty of 1000” designation for: McDonald, P.W., K.A. Read, C.E. Baker, A.E. Anderson, M.D. Powell, A. Ballesteros-Tato, and K.J. Oestreich. IL-7 signaling represses Bcl-6 and the TFH gene program. Nat. Commun. 7: 10285 (2016). 7 2016 American Association of Immunologist Early Career Faculty Travel Award Recipient 2016 Teaching Excellence Award, Translational Biology Medicine and Health graduate program (Co-Director of Immunology and Infectious Disease course) 2015 American Association of Immunologists Pfizer-Showell Award Recipient (presented annually by AAI to recognize the professional promise of an early career investigator) 2012 Article of the Month designation for: Oestreich, K.J., S.E. Mohn, and A.S. Weinmann. Molecular mechanisms that control the expression and activity of Bcl-6 in

TH1 cells to regulate flexibility with a TFH-like gene profile. Nat. Immunol. 13: 405- 411 (2012). 2011 “Faculty of 1000” designation for: Oestreich, K.J., A.C. Huang, and A.S. Weinmann. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate TH1 gene expression patterns. J. Exp. Med. 208: 1001-1013 (2011). 2011 “Faculty of 1000” designation for: Youngblood, B, K.J. Oestreich, S.J. Ha, J. Duraiswamy, R.S. Akondy, E.E. West, Z. Wei, P. Lu, J.W. Austin, J.L. Riley, J.M. Boss, and R. Ahmed. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity. 35: 400-412 (2011). 2008-2009 Developmental Immunology institutional training grant recipient 2006-2008 Ruth Kirschstein institutional postdoctoral training grant recipient 2006 “Faculty of 1000” designation for: Oestreich, K.J., R. Cobb, S. Pierce, J. Chen, P. Ferrier, and E. Oltz. Regulation of TCRb gene assembly by a promoter/enhancer holocomplex. Immunity 24: 381-391 (2006).

Grant Support Active Support: R01 AI134972 (Oestreich, PI) 2018-2023 NIH/NIAID; $1,250,000 direct ($1,975,506 total) Identifying novel regulatory pathways underlying T helper 1 cell immune responses Start-up funds (Oestreich, PI) 2019 Department of Microbial Infection and Immunity The Ohio State University College of Medicine Completed Support: Jeffress Trust Award in Interdisciplinary Research (Oestreich/Childs, Co-PIs) 2018-2019 Virginia Tech; $100,000 direct (~$80,000 to Oestreich) Modeling immunological memory: A combined biological and mathematical approach to understanding T helper cell differentiation Vet-Med IRC (Oestreich, PI) 2018-2019 Virginia-Maryland College of Veterinary Medicine; $20,000 direct Novel roles for a lymphocyte-associated transcriptional complex in promoting breast cancer metastasis ICTAS Junior Faculty Award (Oestreich, PI) 2017-2019 Virginia Tech; $80,000 direct A novel bioengineering approach to manufacture CAR T cells for immunotherapy R56 AI127800 (Oestreich, PI) 2017-2018 NIH/NIAID; $172,444 direct ($262,535 total) Regulation of T helper cell differentiation by integrated STAT and Ikaros zinc finger transcription factor mechanisms Vet-Med IRC (Oestreich, PI) 2017-2018 Virginia-Maryland College of Veterinary Medicine; $20,000 direct Defining the role of a novel Aiolos/STAT3 transcriptional complex in TFH cell differentiation 8 AAI Careers in Immunology Fellowship (Oestreich, PI) 2016-2017 American Association of Immunologists; $23,376 direct Identification of novel transcription factors that regulate the expression of Bcl-6 and the TFH and TCM gene programs Vet-Med IRC (Oestreich, PI) 2016-2017 Virginia-Maryland College of Veterinary Medicine; $20,000 direct Defining the roles of Ikaros zinc finger (IkZF) transcription factors in central memory (TCM) and T follicular helper (TFH) cell development

Vet-Med IRC (Oestreich, PI) 2015-2016 Virginia-Maryland College of Veterinary Medicine; $20,000 direct Identification of the transcriptional network governing T follicular helper cell development VTCRI/Vet-Med Pilot Grant (Oestreich/Allen, Co-PIs) 2015-2016 Virginia-Maryland College of Veterinary Medicine; $20,000 direct Intrinsic and extrinsic determinants of + CD4 TCM cell fate

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

9

192 Microbial Infection and Immunity The Ohio State University 714 BRT, 460 West 12th Avenue Columbus, OH, 43210 Phone: 614-292-7851 (Office) Curriculum Vitae Email: Luanne.Hall- [email protected]

Luanne Hall- Citizenship: US and Stoodley, PhD British Associate Professor PERSONAL STATEMENT Expertise and Research. My expertise focuses on bacteria that opportunistically infect the human respiratory tract. This includes Streptococcus pneumoniae, nontypeable Haemophilus influenzae (NTHi), Staphylococcus aureus, Pseudomonas aeruginosa, and nontuberculous mycobacteria (NTM). Pathogenic bacteria can develop biofilm aggregates in the human airway when mucociliary clearance is compromised, such as in children with chronic otitis media, or in the lungs of people with primary ciliary dyskinesia (PCD) or cystic fibrosis (CF). I have more than 20 years of research experience in bacterial/host interactions, biofilm infections, and anti- biofilm/infective therapies. I have given numerous invited seminars in the field of biofilm infections and co- authored several reviews. The impact of my contributions involves four major findings: ◆ Discoveries linking biofilm development to chronic infection. I developed specific molecular tools and novel protocols to identify aggregates of pathogenic bacteria in clinical samples from children with chronic otitis media. This approach addressed a critical problem in the field: how to determine if biofilms observed in clinical samples by microscopy were comprised of pathogenic or nonpathogenic commensal bacteria. This study was published in JAMA in 2006 and has been cited over 900 times. The impact of this work led to numerous other studies that used this approach to identify bacteria in situ in the context of human infections. I was invited to be part of a European Society of Clinical Microbiology and Infectious Diseases panel of experts, which led to the publication of the first “Guideline for the Diagnosis and treatment of Biofilm Infections”. ◆ Interrogating bacterial pathogens in human airway diseases. I have established human cell infection models to interrogate underlying factors that contribute to persistent infection. Using primary airway epithelial cells differentiated at air-liquid interface (ALI) from people with PCD, these studies were the first to establish that NTHi is more likely to form biofilm-like aggregates on PCD than on non-PCD airway mucosal epithelia and that PCD was associated with biofilm infections. These human cell models are key to understanding the cellular mechanisms that contribute to persistent infection. ◆ Mycobacterial aggregate development and biofilms. With funding from the Wellcome Trust, I hypothesized that pathogenic NTM attached to surfaces and formed biofilm-like aggregates under environmental conditions (e.g. flow and low nutrient conditions). A novel hypothesis at the time, it is proving to be relevant in NTM infections in the context of CF and chronic obstructive pulmonary disease (COPD). ◆ Translational platforms that assess antimicrobial treatment efficacy on bacterial aggregates. Bacterial biofilms are difficult to eradicate because aggregated bacteria tolerate concentrations of antimicrobial effectors that kill single cell bacteria. While in vitro studies of antibiotic efficacy typically use single cell bacteria to define minimum inhibitory or bactericidal concentrations these are not mirrored in vivo. I have established model systems of biofilm bacteria associated with human cells that recapitulate antibiotic tolerance and used these models to evaluate strategies that abrogate antibiotic tolerance. My current research focuses on the evasion of host innate immunity by pathogenic NTM. Biofilm development (aggregated bacteria associated with mucosal epithelial cells) and intracellular survival in host macrophages can each contribute to persistent respiratory tract infections. Mycobacterium abscessus survives in human macrophages, but also develops biofilm-like aggregates that are highly tolerant of antimicrobial host defenses and antibiotic therapies. We are focusing on this understudied NTM, which causes life-threatening chronic infections in people with lung diseases, such as CF or COPD. Understanding mechanisms that allow respiratory pathogens to evade host control in the human airway and, in turn, lead to persistent chronic infection is crucial to developing strategies that target persistent reservoirs of pathogenic bacteria in respiratory tract infections. Curriculum Vitae: Luanne Hall-Stoodley, PhD.

EDUCATION AND TRAINING Undergraduate 1981 BA. Philosophy and History (dual major, with honors), St. Olaf College, Northfield, MN Junior year, University of Washington, Seattle, WA. Semester Abroad, Anthropology, Cambridge University, Cambridge, United Kingdom. 1987 BSc. Microbiology with honors, Montana State University, Bozeman, MT Graduate 1995 PhD. Immunology and Microbiology, Montana State University, Bozeman, MT Advisor: N.D. Reed Post-graduate 1997 – 1999 Wellcome Trust Post-doctoral Research Fellowship, Biological Sciences, University of Exeter, United Kingdom Advisor: H.M. Lappin-Scott, OBE 1999 – 2000 Post-Doctoral Fellow, Center for Biofilm Engineering, Montana State University, Bozeman, MT Advisor: J.W. Costerton APPOINTMENTS AND POSITIONS 1995 Adjunct faculty, WWAMI Medical Sciences, Montana State University-Bozeman, MT 1997-1999 Lecturer, Immunology, Department of Biological Sciences, University of Exeter, United Kingdom 2000-2003 Assistant Research Professor, Veterinary Molecular Biology – Pulmonary Immunology and Immunopathology, and Department of Microbiology, Montana State University- Bozeman, MT (Mentor: Prof. Allan Harmsen) 2004-2009 Assistant Professor, Center for Genomic Sciences, Allegheny-Singer Research Institute, Pittsburgh; Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 2009-2013 Translational Scientist, National Institute for Health Research, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom ** Manager, Wellcome Trust Clinical Research Facility (WTCRF) Laboratories, National Institute for Health Research, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom** 2010-2017 Honorary Lecturer, Faculty of Medicine, Division of Inflammation, Infection and Immunity, University of Southampton, United Kingdom 2013-present Associate Research Professor, Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH HONORS/AWARDS • Departmental Distinction (History), St. Olaf College, Northfield, MN (1981) • National Science Foundation Undergraduate Research Experience (REU) Fellow (Biochemistry), Montana State University, Bozeman, MT (1986) • Lackman Award for Outstanding Graduate Student in Microbiology, Microbiology Department, Montana State University, Bozeman, MT (1995) • Wellcome Trust Post-doctoral Fellowship (Sir Henry Wellcome Showcase Award) (1997) • Carnegie Science Emerging Female Scientist Award Nominee (2007) • Who’s Who in America (2008) • Who’s Who of American Women (2008) PROFESSIONAL MEMBERSHIPS/AFFILIATIONS

1990- American Association of Immunology 1995 1997- Society of General Microbiology, United Kingdom 2003 1997- American Society of Microbiology present 2003- Association of Research in Otolaryngology 2008 2010- Infectious Disease Research Network, United Kingdom 2013 2 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Wessex Immunology Group, United Kingdom University Southampton Clinical and Exp Sciences Infection & Immunity Board University Southampton Biofilms and Microbial Communities Research Group University Southampton Strategic Research Group 2013-present OSU Institute for Infectious Diseases (formerly Center for Microbial Interface Biology), The Ohio State University 2014-present European Society of Clinical Microbiology and Infectious Diseases (ESCMID) 2014-present ESCMID Study Group for Biofilms 2014-2018 European Cooperation in Scientific and Technology (COST) International Partner Working Group Member –Translational research in primary ciliary dyskinesia: bench, bedside, and population perspectives. Expertise: Microbiology, Biofilms 2015-present OSU/Nationwide Children’s Hospital Cystic Fibrosis Translational Research Group 2018- present FAME, Women in Medicine and Science (WIMS), WIMS Data Subcommittee 2019-present OSU/Nationwide Children’s Hospital Cure CF Columbus Cystic Fibrosis Foundation Research Development Program (http://www.curecfcolumbus.org) PROFESSIONAL ACTIVITIES 2012 Panelist, European Society of Clinical Microbiology and Infectious Disease Study Group for Biofilms, Consensus Conference on Biofilm Guidelines, November 2012, Copenhagen, DK 2014-present - Basic Science Research Collaborator, Center for Respiratory Health, Nationwide Children’s Hospital, Columbus, OH - Executive Committee member, European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Biofilms 2017 - Advancing Translational Science for Pulmonary Nontuberculous mycobacterial Infections Workshop, NIH-NIAID, September 25, 2017. Rockville, MD - Panelist, Many Hosts of Mycobacteria VII Meeting, Fort Collins, CO, August 9-11, 2017 2018 - Invited Faculty, International Course on Antibiotic Resistance (ICARe), Institut Pasteur, Les Pensières, 6-14 Oct 2018. Annecy, France - Panelist, Clinical Manifestations of Antimicrobial Resistant Biofilms, Institute for Infectious Diseases-Centers of Disease Control Joint Program, June 27, 2018. The Ohio State University. 2019 - Invited Speaker, Macrophages from CF Patients Are Susceptible to Rough Mycobacterium abscessus, Pathogenesis of Infection Workshop, North American CF Conference, October 31, 2019. Nashville, TN - Invited Discussion Leader, The Future of Biofilm Research Colloquium, May 9-11, 2019. Leavenworth, WA - Invited Panelist/Panel Leader, Nontuberculous mycobacteria and M. ulcerans/Buruli ulcer and M. abscessus, Many Hosts of Mycobacteria VIII, Albert Einstein College of Medicine, Mar 3-6, 2019, New York, NY 2020 - Invited Discussion Leader, NTM Physiology, Colorado Mycobacteria Conference 2020: Focus on Nontuberculous Mycobacteria, May 31-June 3, 2020. Colorado State University, Fort Collins, CO (postponed due to Covid-19) - Core Faculty, International Course on Antibiotic Resistance (ICARe), Targeting microbial - biofilms, Institut Pasteur, Les Pensières, 17-25 Oct 2020. Annecy, France, (https://www.pasteur.fr/en/international-course-antibiotics-and-resistance-icare) International Conferences/Scientific meetings Conference Organizing Committee: Biofilms 4: Communities Bridging Disciplines, 4th International European Biofilm Conference, September 2010. Winchester, United Kingdom

3 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Session Chair/Keynote speaker/Invited speaker: § Chair, Biofilms in Infections, 5th American Society of Microbiology Conference on Biofilms, November 2009. Cancun, Mexico § Plenary Speaker, New Diagnostics for an Old Problem – When Culture Is Not the Gold Standard: Ibis T5000 Biosensor System in the Detection of Microbial Biofilms. Abbott Diagnostics 3rd Asia Pacific & Japan Scientific Symposium, November 2010, Ho Chi Minh City, Vietnam § Chair, Clinical and Applied Biofilms, European Society of Clinical Microbiology and Infectious Diseases (ESCMID) EUROBIOFILMS 2nd European Congress on Microbial Biofilms – Basic Science and Clinical Aspects, 6-8 July 2011, Copenhagen, Denmark § Expert Faculty, Infectious Disease Workshop – ESCMID Guideline for the diagnosis and treatment of biofilm infections, 25th European Congress of Clinical Microbiology and Infectious Diseases (ECCMID 2015), 25-28 April 2015, Copenhagen, Denmark § Session Chair/Invited speaker, Detection & Diagnosis of Biofilm Infections, ESCMID Study Group for Biofilms 4th European Congress on Biofilms, EUROBIOFILMS 2015, June 2015, Brno, Czech Republic § Invited speaker, Biofilm Infections, International Biofilm Symposium 2015, September 2015, Chongqing, China § Keynote speaker, Bacterial Biofilms in Chronic Otitis Media, International Conference on Cholesteatoma & Middle Ear Surgery, June 2016, Edinburgh, Scotland § Invited Panelist, An Adenoid Reservoir, Biofilms in Otitis, International Conference on Cholesteatoma & Middle Ear Surgery, June 2016, Edinburgh, Scotland § Keynote speaker, Biofilms: Past, Present and Future Symposium, European Commission Horizon 2020 Research and Innovation Project, November 2016, Barcelona, Spain § Invited speaker, Diagnosis and treatment of biofilm-related infections, 28th European Congress of Clinical Microbiology and Infectious Diseases (ECCMID 2018), 21-24 April 2018, Madrid, Spain § Invited speaker, NTM Workshop, International Rare Lung Diseases Research Conference, RLDC•2018•Cincinnati, September 6-9, 2018, Covington, Kentucky, USA SERVICE Laboratory oversight and regulatory management 1999-2003 Biosafety Officer, Center for Biofilm Engineering, Montana State University—Bozeman, MT 2000-2003 Set up BSL Laboratory for work with Mycobacterium tuberculosis at Montana State University to implement NAIAD-funded grant 2001-2003 Montana State University Human Subjects Committee (Institutional Review Board) 2004-2009 Oversight of BSL3 laboratory development/construction Allegheny-Singer Research Institute. Consulted with contractors for BSL3 construction at Allegheny General Hospital and with State of Pennsylvania regulators for commissioning. 2009-2013 Manager WTCRF Laboratories, Southampton Wellcome Trust-National Institute for Health Research Clinical Research Facility, University Hospital Southampton NHS Foundation Trust. Supervised five laboratory technicians. Duties included training of all CRF medical, technical and student users. Regulatory oversight, consulting with United Kingdom regulatory bodies, and Healthcare products Regulatory Agency for accreditation, and Health and Safety Executive (HSE) to coordinate and update the BSL3 facility (primarily used for hepatitis viruses) Education and Public Outreach

2005 Presenter, Germs and Immunity, Marshall Elementary School, Wexford, Pennsylvania 2007 Panelist, Pittsburgh Science and Technology Learning Community Focus Group 2007- Coordinator, Carnegie Science Center-Allegheny-Singer Research Institute Corporate 2008 Sponsorship of SciTech Exhibit, Pittsburgh, PA 2007- Biofilm/Infection-based exhibit coordinator, SciTech Carnegie Center Science Fair 2009 Pittsburgh, PA. Exhibits were targeted to tours of school children in middle school (ages 8-12) and high school (ages 13-18) 4 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

2008 - Education advisor, Engineer’s Society of Western Pennsylvania - Invited Career Day Presenter, Marshall Middle School, Wexford, PA 2009-2011 Student work experience intern coordinator, Wellcome Trust Clinical Research Facility (WTCRF) labs 2009-2012 Coordinator WTCRF Laboratories, WTCRF and Trust Open days for Clinical Research Facility Open Days (Semi-annual) and Trust Open Days (Annual) 2016 TEDx Discovery Talk, IDI Infectious Disease Speakers Bureau, “Biofilm Infections: the Bacterial Scrum”. OSU Alumni Association, 8 Aug 2016 2017 – present MI&I Education and Curriculum Committee 2020 Cure Cystic Fibrosis Columbus Research Development Program Trainee Committee Diversity 1986-1989 Founder, Student Director “Women in Science” Program through the Women’s Resource Center, Montana State University. This program was expanded by the Dean of the Montana State University College of Letters and Sciences to the Women in Science and Engineering (WISE) 2002 National Institutes of Health Biomedical Research Infrastructure Network – Outreach to Little Bighorn College, Crow Agency, Montana 2019 – present - Speaker, Women in Math and Science Club at Ohio State, Ohio Union - OSU College of Medicine Women in Medicine and Science Data Subcommittee - College of Medicine Women in Medicine Annual Celebration of Women Faculty, Presentation on behalf of WIMS Data Subcommittee, Oct. 16, 2019 International and Federal Grant Review • National Science Foundation, Institutional Graduate Education and Research Training Grant (IGERT) Panelist (2001) Ad Hoc Review • Food and Drug Administration (2001) • National Science Foundation, Biocomplexity in the Environment (2001) • US Civilian Research and Development Foundation (2004) • National Institutes of Health-NIAID, Biodefense and Emerging Infectious Diseases Research Opportunities, (2004) • Garnett Passé and Rodney Williams Memorial Foundation, Australia (2005) • Biomedical Research Council (BMRC), Agency for Science, Technology and Research (A*STAR), Singapore (2006) • Israel Science Foundation (2012) • University of Ghent, Belgium (2014) • Cystic Fibrosis Foundation Research Development Program Training Awards (2019) • European Society of Clinical Medicine and Infectious Diseases Research Grants (2014-present) • The Wellcome Trust (2015) • Cystic Fibrosis Canada (2017) • British Lung Foundation (2018) • National Science Foundation, Molecular and Cellular Biosciences (2018) • British Society for Antimicrobial Chemotherapy (BSAC) (2018) • Cystic Fibrosis Foundation Research Development Program Training Awards (2019) Peer Review Editorial Boards 2009-2013 Editorial Board Biofouling: The Journal of Bioadhesion and Biofilm Research (IF 4.4) 2018-present Associate Editor Frontiers in Cellular and Infection Microbiology Journal Review

5 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Acta Biomaterialia (IF= 6.38); Antimicrob Agents Chemother (IF=4.25); Applied Environ Microbiol; BMC Infectious Diseases; BMC Microbiology; BMC ; Clinical Infectious Disease(IF= 8.2); Clinical Microbiol & Infection (IF=5.39); Clinical Microbiol Reviews (IF=17.4); Expert Opin Ther Targets; Expert Rev Anti-Infective Ther; FEMS Microbiol Rev (IF = 11.39); Front Microbiol (IF=4.07); Infection & Immunity; International J Oral Science; Int J Ped Otorhinolaryngol; ISME J (IF=9.52); J Bacteriology; JCI Insights (IF=6.01); J Immunology; J Infection (IF=4.6); J Internal Med (IF = 6.06); Microbial Drug Resistance; Microbial Pathogenesis; mSphere; Nature Protocols (IF=12.2); Pathogens Dis; PLoS Pathogens (IF=6.15); Scientific Reports (IF=4.6); Respiration; Science Adv (IF=12. ); Vaccine

PUBLICATIONS Total = 62 (23 first or last author) [Total citations: 13027; h-index=36, i10-index=60, Google Scholar] The following URL link shows a full list of publications: http://www.ncbi.nlm.nih.gov/pubmed/?term=Hall- Stoodley Intellectual Property 1. Patent US7871791. Hall-Stoodley L, Stoodley P. 2007. A Biofilm Preparation using Potassium Permanganate: “Biofilm EPS Contrast Enhancer” (Issue date Jan 18, 2011). Original Peer-Reviewed Articles *denotes equal contribution 1. Krause K, Caution K, Badr A, Hamilton K, Saleh A, Patel K, Seveau S, Hall-Stoodley L, Hegazi R, Zhang X, Gavrilin MA & AO Amer. 2018. CASP4/caspase-11 promotes autophagosome formation in response to bacterial infection. Autophagy. 14(11):1928-1942. PMID: 30165781 2. Clary G, Sasindran S, Nesbitt N, Mason L, Azad A, Cole S, McCoy K, Schlesinger LS and L Hall- Stoodley. Mycobacterium abscessus Smooth And Rough Morphotypes Form Antimicrobial- Tolerant Biofilm Phenotypes But Are Killed by Acetic Acid. Antimicrob Agents Chemother 62(3). pii:e01782-17. PMID: 29311080 3. Krause K, Kopp BT, Tazi MF, Caution K, Hamilton K, Badr A, Shrestha C, Tumin D, Hayes D Jr, Robledo F, Hall-Stoodley L, Klamer BG, Zhang X, Partida-Sanchez S, Parinandi NL, Kirkby SE, Dakhlallah D, McCoy KS, Cormet-Boyaka E, Amer AO. 2017. The expression of Mirc1/Mir17-92 cluster in sputum samples correlates with pulmonary exacerbations in cystic fibrosis patients. J Cyst Fibros. S1569- 1993(17)30962-1. PMID: 29241629 4. Walker WT, Jackson CL, Allan RN, Collins SA, Kelso MJ, Rineh A, Yepuri NR, Nicholas B, Lau L, Johnston D, Lackie P, Faust SN, Lucas JSA, Hall-Stoodley L. 2017. Primary ciliary dyskinesia ciliated airway cells show increased susceptibility to Haemophilus influenzae biofilm formation. Eur Respir J. 50(3). pii: 1700612. PMID: 28890436 – Featured Editorial: Shoemark A. Haemophilus influenzae biofilms in primary ciliary dyskinesia: a moving story. Eur Respir J. 2017 50(3) pii: 1701369. 5. Vicetti Miguela RD, Quispe Calla NE, Dixon D, Foster RA, Gambotto A, Pavelko SD, Hall-Stoodley L, TL Cherpes. 2017. IL-4-secreting eosinophils stimulate endometrial stromal cell proliferation and prevent Chlamydia-induced upper genital tract damage. Proc Natl Acad Sci U S A. 114(33):E6892- E6901 PMID: 28765368 6. Howlin R, Cathie K, Hall-Stoodley L, et al. 2017. Low dose Nitric Oxide Targeted Anti-biofilm Adjunctive Therapy to Treat Chronic Pseudomonas aeruginosa Infection in Cystic Fibrosis. Molecular Therapy. 25(9):2104-2116. PMID: 28750737 – Featured “In this issue Molecular Therapy” Vol. 25, No 9 September 2017: Low-Dose Nitric Oxide Fights Biofilms in CF 7. Dawe H, Berger E, Sihlbom C, Angus EM, Howlin RP, Laver JR, Tebruegge M, Hall-Stoodley L, Stoodley P, Faust SN, RN Allan. 2017. D-methionine interferes with non-typeable Haemophilus influenzae peptidoglycan synthesis during biofilm formation. Microbiology. 163(7):1093-1104. PMID: 28699879. 8. Grande R, Celia C, Mincione G, Stringaro A, Di Marzio L, Colone M, Carmela Di Marcantonio M, Savino L, Puca V, Santoliquido R, Locatelli M, Muraro R, Hall-Stoodley L, P Stoodley. 2017. Detection and Physicochemical Characterization of Membrane Vesicles (MVs) of Lactobacillus reuteri DSM17938. Front Microbiol. 8:1040. PMID: 28659878

6 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

9. Allan RN, Kelso MJ, Rineh A, Yepuri NR, Feelisch M, Soren O, Brito-Mutunayagam S, Salib RJ, Stoodley P, Clarke SC, Webb JS, Hall-Stoodley L*, Faust SN*. 2017. Cephalosporin-NO-donor prodrug PYRRO- C3D shows β-lactam-mediated activity against Streptococcus pneumoniae biofilms. Nitric Oxide. 65:43-49. PMID: 28235635 10. SA Collins, MJ Kelso, A Rineh, NR Yepuri, M Feelisch, J Coles, C Jackson, GD Halladay, JS Webb, L Hall-Stoodley, Connett G, M Feelisch, SN Faust, JS Lucas, RN Allan. 2017. Cephalosporin-3’- diazeniumdiolate NO-donor prodrug PYRRO-C3D enhances azithromycin susceptibility of Non- typeable Haemophilus influenzae biofilms. Antimicrob Agents Chemother. 61(2) pii:e02086-16. PMID: 27919896 11. Quispe Calla NE, Vicetti Miguel RD, Boyaka PN, Hall-Stoodley L, Kaur B, Trout W, Pavelko SD, Cherpes TL. 2016. Medroxyprogesterone acetate and levonorgestrel increase genital mucosal permeability and enhance susceptibility to genital herpes simplex virus type 2 infection. Mucosal Immunol. 9(6):1571-1583. PMID: 27007679 12. Allan RN, Morgan S, Brito-Mutunayagam S, Skipp P, Feelisch M, Hayes SM, Hellier W, Clarke SC, Stoodley P, Burgess A, Ismail-Koch H, Salib RJ, Webb JS, Faust SN, Hall-Stoodley L. 2016. Low concentrations of nitric oxide modulate Streptococcus pneumoniae biofilm metabolism and antibiotic tolerance. Antimicrob Agents Chemother. 60(4):2456-66. PMID: 26856845 13. Grande R, Di Marcantonio MC, Robuffo I, Pompilio A, Celia C, Di Marzio L, Paolino D, Codagnone M, Muraro R, Stoodley P, Hall-Stoodley L, Mincione G. 2015. Helicobacter pylori ATCC 43629/NCTC 11639 Outer Membrane Vesicles (OMVs) from Biofilm and Planktonic Phase Associated with Extracellular DNA (eDNA). Front Microbiol. 6:1369. PMID: 26733944 14. Swearingen MC, Mehta A, Mehta A, Nisticò L, Hill PJ, Falzarano AR, Wozniak DJ, Hall-Stoodley L, Stoodley P. 2016. A novel technique using potassium permanganate and reflectance confocal microscopy to image biofilm extracellular polymeric matrix reveals non eDNA networks in Pseudomonas aeruginosa biofilms. Pathog Dis. 74(1) pii: ftv104. PMID: 26536894 15. Hayes SM, Howlin R, Johnston DA, Webb JS, Clarke SC, Stoodley P, Harries PG, Wilson SJ, Pender SL, Faust SN, Hall-Stoodley L*, Salib RJ*. 2015. Intracellular residency of Staphylococcus aureus within mast cells in nasal polyps: A novel observation. J Allergy Clin Immunol. 135(6):1648- 51. PMID: 25680455 16. Høiby N, Bjarnsholt T, Moser C, Bassi GL, Coenye T, Donelli G, Hall-Stoodley L, Holá V, Imbert C, Kirketerp-Møller K, Lebeaux D, Oliver A, Ullmann AJ, Williams C, for the European Society of Clinical Microbiology and Infectious Diseases Study Group for Biofilms and Consulting External Expert Werner Zimmerli. 2015. ESCMID guideline for the diagnosis and treatment of biofilm infections 2014. Clin Microbiol Infect. May 21; Suppl 1:S1-S25. PMID: 25596784 – First Clinical Guideline for Diagnosis and Treatment of Biofilm Infections. 17. Allan R, Skipp P, Jefferies J, Clarke S, Faust S, *Hall-Stoodley L and *Webb J. 2014. Pronounced Metabolic Changes in Adaptation to Biofilm Growth by Streptococcus pneumonia. PLoS One. 9(9):e107015. PMID: 25188255 18. Schaudinn C, Stoodley P, Hall-Stoodley L, Gorur A, Remis J, Wu S, Auer M, Hertwig S, Guerrero- Given D, Hu FZ, Ehrlich G, Costerton J, Robinson D and Webster P. 2014. Death and transfiguration in static Staphylococcus epidermidis Cultures. PLoS One 9(6):e100002. PMID: 24964210 19. Nisticò L, Hall-Stoodley L, Stoodley P. 2014. Imaging bacteria and biofilms on hardware and periprosthetic tissue in orthopedic infections. Methods Mol Biol. 1147:105-26. PMID: 24664829 20. Walker WT, Jackson CL, Coles J, Lackie P, Faust SN, Hall-Stoodley L, JS Lucas. 2014. Ciliated Cultures From Patients With Primary Ciliary Dyskinesia Produce Nitric Oxide in Response to Haemophilus influenzae Infection and Proinflammatory Cytokines. Chest. 145(3): 668-669. PMID: 24590042 21. Grande R, Nisticò L, Sambanthamoorthy K, Longwell M, Iannitelli A, Cellini L, Di Stefano A, *Hall- Stoodley L and *Stoodley P. 2014. Temporal expression of agrB, cidA and alsS in the early development of Staphylococcus aureus UAMS-1 biofilm formation and the structural role of extracellular DNA and carbohydrates. Pathog Dis. 70(3):414-22. PMID: 24535842 22. Kruszewski KM, Nisticò L, Longwell MJ, Hynes MJ, Maurer JA, *Hall-Stoodley L and *Gawalt ES. 2013. Reducing Staphylococcus aureus biofilm formation on stainless steel 316L using Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

functionalized self- assembled monolayers. Journal of Materials Science and Engineering C – Biol Appl. 33(4):2059-69. PMID: 23498233

7

203 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

23. Limbert G, Bryan R, Cotton R, Young P, Hall-Stoodley L, Kathju S and P Stoodley. 2013. On the mechanics of bacterial biofilms on non-dissolvable surgical sutures: A laser scanning confocal microscopy- based finite element study. Acta Biomater. 9(5): 6641-52. PMID: 23376125 24. Stoodley P, Sidhu S, Nisticò L, Mather M, Boucek A, Hall-Stoodley L, S Kathju. 2012. Kinetics and Morphology of Polymicrobial Biofilm Formation on Polypropylene Mesh. FEMS Immunology & Medical Microbiology 65(2): 283-90. PMID: 22364207 25. Nisticò L, Kreft R, Gieseke A, Coticchia JM, Burrows A, Khampang P, Liu Y, Kerschner JE, Post JC, Lonergan S, Sampath R, Hu FZ, Ehrlich GD, Stoodley P, Hall-Stoodley L. 2011. An Adenoid Reservoir for Pathogenic Biofilm Bacteria. J Clin Microbiol. 49(4):1411-20. PMID: 21307211 – Chosen for press release: News from The Journals of The American Society for Microbiology; Science Daily; Medical News Daily 26. Kizhner V, Krespi YP, Hall-Stoodley L, Stoodley P. 2011. Laser-Generated Shockwave for Clearing Medical Device Biofilms. Photomed Laser Surg. 29(4): 277-282. PMID: 21182450 27. Krespi YP, Kizhner V, Nistico L, Hall-Stoodley L, Stoodley P. 2011. Laser disruption and killing of methicillin-resistant Staphylococcus aureus biofilms. Am J Otolaryngology—Head and Neck Medicine and Surgery. 32(3):198-202. PMID: 20434806 28. Stoodley P, Braxton EE Jr, Nistico L, Hall-Stoodley L, Johnson S, Quigley M, Post JC, Ehrlich GD, Kathju S. 2010. Direct demonstration of Staphylococcus biofilm in an external ventricular drain in a patient with a history of recurrent ventriculoperitoneal shunt failure. Pediatr Neurosurg. 46(2): 127-32. PMID: 20664301 29. Pittet LA, Hall-Stoodley L, Rutkowski MR, Harmsen AG. 2010. Influenza Virus Infection Decreases Tracheal Mucociliary Velocity and Clearance of Streptococcus pneumoniae. Am J Respir Cell Mol Biol. 42:1-11. PMID: 19520922 30. Kathju S, Nistico L, Hall-Stoodley L, Post JC, Ehrlich GD, Stoodley P. 2009. Chronic surgical site infection due to suture-associated polymicrobial biofilm. Surgical Infections. (Larchmt) 10(5):457- 61. PMID: 19811056 31. Stoodley P, De Beer D, Longwell M, Nistico L, Hall-Stoodley L, Wenig B, Krespi YP. 2009. Tonsillolith: Not just a stone but a living biofilm. Otolaryngol Head Neck Surg. 141(3): 316-21. PMID: 19716006 32. Hoa M, Tomovic S, Nistico L, Hall-Stoodley L, Stoodley P, Sachdeva L, Berk R, Coticchia JM. 2009. Identification of adenoid biofilms with middle ear pathogens in otitis-prone children utilizing SEM and FISH. Int J Pediatr Otorhinolaryngol. 73(9): 1242-1248. PMID: 19525016 33. Nistico L, Gieseke A, Stoodley P, Hall-Stoodley L, Kerschner JE, Ehrlich GD. 2009. Fluorescence in situ hybridization for the detection of biofilm in the middle ear and upper respiratory tract mucosa. Methods Mol Biol. 493:191-213. PMID:18839349 34. Hall-Stoodley L, L Nistico, D Nguyen B Dice, WJ Mershon, C Johnson, FZ Hu, P Stoodley, GD Ehrlich, JC Post. 2008. Characterization of Biofilm Matrix, Degradation by DNase Treatment and Evidence of Capsule Downregulation in Streptococcus pneumoniae Clinical Isolates. BMC Microbiology 8:173. PMID: 18842140 – Highly accessed article 35. Krespi J, Stoodley P, L Hall-Stoodley. 2008. Laser Disruption of Biofilm. Laryngoscope. 118(7):1168-73. PMID: 18401277. 36. Hall-Stoodley L, Hu F, Gieseke A, Nisticò L, Nguyen D, Hayes J, Forbes M, Greenberg D, Dice B, Burrows A, Stoodley P, Post JC, GD Ehrlich, J Kerschner. 2006. Direct Detection of Bacterial Biofilms on the Middle-Ear Mucosa of Children with Chronic Otitis Media. JAMA. 296(2):202-211. Rapid Publication. PMID: 16835426 (>900 citations) This paper was the first to definitively show that bacteria presented as biofilms in the middle ear of children with chronic otitis media. – Faculty of 1000 recommended paper; Google Scholar Classic in Otolaryngology (3rd most cited paper for 2006); Featured: MIT Tech Rev; Nature News. 37. Hall-Stoodley L, Watts G, Crowther J, Balagopal A, Torrelles JB, Robison-Cox J, Bargatze R, Harmsen AG, Crouch EC, LS Schlesinger. 2006. Mycobacterium tuberculosis Binding to Human Surfactant Proteins A and D, Fibronectin and Small Airway Epithelial Cells Under Shear. Infect. Immun. 74(6):3587-96. PMID: 16714591 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

38. Hall-Stoodley L, Brun OS, Polshyna GP and LP Barker. 2006. Mycobacterium marinum Biofilm Formation Reveals Cording Morphology. FEMS Microbiology Letters. 257:43-49. PMID: 16553830

8

205 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

39. Stoodley P, Kathju S, Ze Hu F, Erdos G, Levenson JE, Mehta NS, Dice BL, Johnson SL, Hall- Stoodley L, Nisticò L, Sotereanos NG, Sewecke JJ, Post JC, GD Ehrlich. 2005. Molecular and Imaging Techniques for Bacterial Biofilms in Arthroplastic Joint Infections. Clinical Orthopedics and Related Research. 437:31-40. PMID: 16056023 40. Dunsmore BC, Jacobsen A, Hall-Stoodley L, Bass CJ, Lappin-Scott HM, Stoodley P. 2002. The influence of fluid shear on the structure and material properties of sulphate-reducing bacterial biofilms. J. Industrial Microbiol. Biotech. 29(6):347-353. PMID: 12483477 41. Stoodley P, Wilson S, Hall-Stoodley L, Boyle JD, Lappin-Scott HM, JW Costerton. 2001. Growth and detachment of cell clusters from mature mixed species biofilms. Appl. Env. Microbiol. 67(12): 5608-13. PMID: 11722913 42. Stoodley P, Hall-Stoodley L, Lappin-Scott HM. 2001. Detachment, surface migration and other dynamic behavior in bacterial biofilms revealed by digital time-lapse imaging. In: Methods Enzymology: Microbial Growth in Biofilms. R.J. Doyle (Ed). 337:306-319. PMID: 11398439 43. Hall-Stoodley L, CW Keevil and HM Lappin-Scott. 1999. Mycobacterium fortuitum and Mycobacterium chelonae form biofilms under high and low nutrient conditions. J. Appl. Microbiol. 85:60S-69S. PMID: 21182694 44. Hall-Stoodley L and HM Lappin-Scott. 1998. Biofilm formation by the rapidly growing non- tuberculous mycobacteria species Mycobacterium fortuitum. FEMS Microbiological Letters 168:79- 84. PMID: 9812366 Review Articles (Peer reviewed) 1. Larsen MH, Lacourciere K, Parker TM, Kraigsley A, Achkar JM, Adams LB,Dupnik KM, Hall- Stoodley L, Hartman T, Kanipe C, Kurtz SL, Miller MA, Salvador LCM, Spencer JS, Robinson RT. 2020. The many hosts of mycobacteria(MHM8): A conference report, Tuberculosis. doi: https://doi.org/10.1016/j.tube.2020.101914. In the Press. 2. Seveau S, Turner J, Gavrilin M, Torelles JB, Hall-Stoodley L, Yount J, AO Amer. 2018. Checks and Balances between the Inflammasome and the Autophagosome during Infection. J Mol Biol. 430(2):174- 192. PMID: 29162504 3. Koo H, Allan RN, Howlin RP, Stoodley P, Hall-Stoodley L. 2017. Targeting microbial biofilms: current and prospective therapeutic strategies. Nature Reviews Microbiology 12:740-755. 28944770. – Featured in Medical News Today 24 October 2017 “Strength in numbers: How biofilms outfox antibiotics”. 97th percentile (ranked 7,839th) of the 274,106 tracked articles of a similar age in all journals and the 92nd percentile (ranked 4th) of the 51 tracked articles of a similar age in Nature Reviews Microbiology. 4. Xu Y, Larsen LH, Lorenzen J, Hall-Stoodley L, Kikhney J, Moter A, Rolighed Thomsen T. 2017. Microbiological diagnosis of device-related biofilm infections. APMIS 125(4):289-303. PMID: 28407422 5. Bhattacharya M, Wozniak DJ, Stoodley P, L Hall-Stoodley. 2015. Prevention and Treatment of Staphylococcus aureus biofilms. Expert Rev Anti Infect Ther.12:1499-516. PMID: 26646248 6. Wilkins M, Hall-Stoodley L, Allan RN, Faust SN. 2014. New approaches to the treatment of biofilm- related infections. J Infect. 69 Suppl 1:S47-52. PMID: 25240819 7. Hall-Stoodley L, Stoodley P, Kathju S, Høiby N, Moser C, Costerton JW, Moter, A, Bjarnsholt, T. 2012. Towards Diagnostic Guidelines for Biofilm-Associated Infections. FEMS Immunol Med Microbiol 65(2):127- 45. PMID: 22469292 8. Stoodley P, Ehrlich GD, Sedghizadeh PP, Hall-Stoodley L, Baratz ME, Altman DT, Sotereanos NG, Costerton JW, DeMeo P. 2011. Orthopaedic biofilm infections. Current Orthopaedic Practice 22(6):558- 563. PMID: 22323927 9. Costerton JW, Post JC, Ehrlich GD, Hu FZ, Kreft R, Nisticò L, Kathju S, Stoodley P, Hall-Stoodley L, Maale G, James G, Sotereanos N and P DeMeo. 2011. New, rapid, and accurate methods for the detection of orthopedic biofilm infections. FEMS Immunol Med Microbiol 61:133-140. PMID: 21204998 10. Hall-Stoodley L and P Stoodley. 2009. Evolving Concepts in Biofilm Infections. Modeling Pathogenesis. Cellular Microbiology 11(7):1034. PMID: 19374653 (>1000 citations) Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

11. Snyder KN, Compliment JM, Buchinsky FJ, Hall-Stoodley L, Stoodley P and Post JC. 2009. Biofilms: a new enemy. Nurse Pract. 34(9):35-9. PMID: 19713821 12. Marcus RJ, Post JC, Stoodley P, Hall-Stoodley L, McGill RL, Sureshkumar KK, Gahlot V. 2008. Biofilms in . Expert Opinion on Biological Therapy 8:1159-66. PMID: 18613767

9

207 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

13. Braxton EE Jr, Ehrlich GD, Hall-Stoodley L, Stoodley P, Veeh R, Fux C, Hu FZ, Quigley M, Post JC. 2005. Role of biofilms in neurosurgical device-related infections. Neurosurg Rev. 28(4):249- 55. PMID: 15991051 14. Hall-Stoodley L and P Stoodley. 2005. Biofilm formation and dispersal and the transmission of human pathogens. Trends Microbiol. 13(1): 7-10. PMID: 15639625 (>600 citations) – Science Direct Top 25 Hottest Articles 2006 15. Post JC, Stoodley P, Hall-Stoodley L, GD Ehrlich. 2004. The Role of Biofilms in Otolaryngologic Infections. Current Opinion in Otolaryngology and Head & Neck Surgery 12(3):185-190. PMID: 15167027 16. Hall-Stoodley L, Costerton JW, and Stoodley P. 2004. Bacterial biofilms: from the natural environment to infectious diseases. Nature Reviews Microbiology 2(2):95-108. PMID: 15040259 (>5500 citations) – 95th percentile (top 5%, ranked 12,646th) of the 281,841 tracked articles of a similar age in all journals and the 89th percentile (ranked 5th) of 48 tracked articles of a similar age in Nature Reviews Microbiology 17. Fux CA, Stoodley P, Hall-Stoodley L, Costerton WJ. 2003. Bacterial biofilms - a diagnostic and therapeutic challenge. Expert Rev Anti-Infective Ther. 1(4):667-683. PMID: 15482163 18. Hall-Stoodley L and Stoodley P. 2002. Developmental regulation of microbial biofilms. Curr Opin Biotech. 13:228-233. PMID: 12180097 Book Chapters 1. Francolini I, Hall-Stoodley L, P Stoodley. 2020. Biofilms, Biomaterials and Device-Related Infections. In: Biomaterials Science Fourth Edition: An Introduction to Materials in Medicine. Ratner BD, Hoffman AS, Schoen FJ, and Lemons JE. (Eds.) Elsevier Academic Press. (Textbook) 2. Hall-Stoodley L, Stoodley P & JE Kerschner. 2015. An Adenoid Reservoir for Otitis Media Pathogens. In: The Biofilm in Otitis. Ed. R. Kania and B. Ars. Kugler Publications. Amsterdam, The Netherlands. 3. Stoodley P, Hall-Stoodley L, Costerton JW, DeMeo P, Shirtliff M, Gawalt E, Kathju S. 2012. Biofilms, Biomaterials and Device-Related Infections. In: Biomaterials Science Third Edition: An Introduction to Materials in Medicine. Ratner BD, Hoffman AS, Schoen FJ, and Lemons JE. (Eds.) Elsevier Academic Press. (Textbook) 4. Ehrlich GD, DeMeo P, Palmer M, Sauber TJ, Altman D, Altman G, Sotereanos N, Conti S, Baratz M, Maale G, Hu FZ, Post JC, Nisticò L, Kreft R, Hall-Stoodley L, Costerton JW, Stoodley P. 2012. Culture Negative Orthopedic Biofilm Infections. pp. 17-27. In: Culture-negative infections in . GD Ehrlich, PJ DeMeo, JW Costerton, H Winkler (eds). Springer Series on Biofilms 7, Springer-Verlag Berlin Heidelberg. DOI 10.1007/978-3-642-29554-6_2 5. Rolighed Thomsen T, Hall-Stoodley L, Moser C and P Stoodley. 2011. The Role of Bacterial Biofilms in Infections of Catheters and Shunts. In: Biofilm Infections. T. Bjarnsholt, P. Østrup Jensen, C. Moser and N. Høiby (Eds). pp. 91-109. ISBN 978-1-4419-6083-2. DOI: 10.1007/978-1- 4419-6084-9_6. Springer New York 6. Stoodley P, Hall-Stoodley L, Boyle JD, Jørgensen F and Lappin-Scott HM. 2000. Environmental and genetic factors influencing biofilm structure. In: Community Structure and Cooperation in Biofilms. pp. 53- 64. D. Allison, P. Gilbert, H.M. Lappin-Scott and M. Wilson (Eds.). SGM Symposium Series 59. Cambridge University Press, Cambridge. ISBN 0521793025. 7. Hall-Stoodley L, Rayner JC, Stoodley P, Lappin-Scott HM. 1999. Establishment of Experimental Biofilms Using the Modified Robbins Device and Flow Cells. In: Methods in Biotechnology Series: Environmental Monitoring of Bacteria. Edwards, C (Ed). 12:307-318. Humana Press, Totowa, N.J. DOI: 10.1385/0-89603- 566-2:307 8. Reed ND, Hall-Stoodley, Schultz L. 1989. Mast cell production by scid/scid mice: in vivo and in vitro studies. pp. 63-67. In: Wu B. and Zheng J (Eds). Immune-deficient Animals in Experimental Medicine. Karger, Basel. Refereed Conference Contributions Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

1. Hall-Stoodley. Macrophages From CF Patients Are Susceptible to Rough Mycobacterium abscessus. 2019. Pediatric Pulmonology. 54:S268-S268. 2. Hall-Stoodley, L. Bacterial Biofilms and Chronic Otitis Media: Stuck in the middle. J. Laryngology Otology. 130:S3 pp S80-S80. 2016. DOI: http://dx.doi.org/10.1017/S0022215116003339

10

209 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

3. Cathie K, Howlin R, Carroll M, Clarke S, Connett G, Cornelius V, Daniels T, Duignan C, Hall- Stoodley L, et al. 2014. G385 RATNO–Reducing Antibiotic Tolerance using Nitric Oxide in Cystic Fibrosis: report of a proof of concept clinical trial. Archives Dis Child. 99 Suppl 1:A159-A159. 7 April 2014. doi:10.1136/archdischild-2014-306237.367 4. Cathie K, Howlin R, Barraud N, Carroll M, Clarke S, Connett G, Cornelius V, Daniels T, Duignan C, Feelisch M, Fernandez B, Hall-Stoodley L, et al. 2014. Low Dose Nitric Oxide As Adjunctive Therapy To Reduce Antimicrobial Tolerance of Pseudomonas aeruginosa biofilms In The Treatment Of Patients With Cystic Fibrosis: Report Of A Proof Of Concept Clinical Trial. 2014.189.1_MeetingAbstracts.A2843 ATS International Conference published in AJRCCM. 5. Walker W, Jackson C, Lackie P, Howlin R, Allan R, Faust S, Lucas J, L Hall-Stoodley. 2012. Is primary ciliary dyskinesia a "biofilm" disease? Eur Respir J (40) Suppl 56: P2949 6. Kruszewski K, Hall-Stoodley L, Gawalt ES. 2011. Formation of rigid and flexible self-assembled monolayers for use in retarding biofilm formation on stainless steel. 85th ACS Colloid and Surface Science Symposium, Montreal, Quebec, June 2011, COLLSYMP-242. 7. Krespi YP, Kizhner V, Nisticò L, Hall-Stoodley L, Stoodley P. 2009. Laser disruption and killing of MRSA biofilms. Triological Society Poster Presentation. Laryngoscope 119(S1):42. doi.10.1002/lary.20367 Media My research • TEDx Discovery Talk, Bacterial Biofilms: “Why Some Infections Just Won’t Go Away”, OSU Infectious Disease Speakers Bureau-Infectious Diseases Institute. https://idi.osu.edu/resources/discovery- talks/discovery-talks-faculty • “Strength in numbers: How biofilms outfox antibiotics” Medical News Today, 24 October 2017 https://www.medicalnewstoday.com/articles/319858.php • Inside Science News Service. Study of bacterial persistence in biofilms in otitis media: “Research May Reveal Why Some Ear Infections Refuse To Go Away.” Jul 18 2014. https://www.insidescience.org/news/research-may-reveal-why-some-ear-infections-refuse-go- away • News from The Journals of The American Society for Microbiology; Science Daily; Medical News Daily. May 20, 2011 http://www.sciencedaily.com/releases/2011/05/110516175342.htm • MIT Tech Rev; Nature News 11 July 2006 | Nature | doi:10.1038/news060710-6 “Bulky biofilms found in kids' ears” http://www.nature.com/news/2006/060710/full/news060710-6.html Interviewed for comments/opinion on others’ research • OZY The Daily Dose. Oct 30, 2016. Dr. Dianne Newman - MacArthur “Genius” Has Found a Connection Between Geology and Genetics: Pseudomonas aeruginosa metabolites – impact on cystic fibrosis https://www.ozy.com/rising-stars/this-macarthur-genius-has-found-a-connection- between-geology-and- genetics/72507/ • NPR Health News “Shots”. June 27 2014. http://www.npr.org/sections/health- shots/2014/06/27/325502998/sticky-streamers-of-staph-bacteria-may-clog-up-medical-devices Selected Oral Presentations – see page 2 for recent invited presentations • Mycobacterial biofilm formation in high and low nutrient conditions. Society for Applied Microbiology Conference on Aquatic Microbiology, Lancaster, United Kingdom, April 1998. Oral presentation. • Mycobacterial Biofilms: a Sticky Wicket. American Society of Microbiology Biofilms 2003 Conference. Victoria Conference Centre, 1-6 November 2003. Victoria, BC, Canada. Oral presentation. • Polymicrobial, pathogenic biofilms associated with pediatric adenoids. Microbial communities in disease. Biofilms IV: Communities Bridging Disciplines, 4th International European Biofilm Conference, 1-3 September 2010. Winchester, United Kingdom. Oral presentation. • Invited Speaker, Biofilms in Medicine, University of Southampton Community Microbiology Symposium, 26 March, 2010. (peer reviewed) • Invited Speaker, Taking off the blinders: PLEX-ID/Ibis system—a culture independent, non-targeted approach to evaluating biofilm-associated infections. Microbial Community Profiling Workshop, Infectious Disease Research Network, Wellcome Trust, 20 June 2011, London, UK.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

11

211 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

• Biofilm-associated infections: does it matter what is there? Institute for Life Sciences Biofilms and Microbial Communities Conference. Inflammation, Infection and Immunity, 12 September 2011. University of Southampton. Oral Presentation. • Invited Speaker, New Diagnostics for an Old Problem: PlexID – Abbott’s Ibis T5000 Biosensor System in the Detection of Microbial Biofilms. Workshop on PLEX-ID technology, SVA, 9-10 May 2012. Uppsala, Sweden. • Invited Speaker, Chronic Otitis Media: evidence for bacterial biofilms in a multifactorial disease. Otitis Media Annual Meeting, Wolfson College, 24 April 2012. Oxford, United Kingdom. • Invited Expert Faculty, ECCMID, ESCMID guideline for the diagnosis and treatment of biofilm infections, 25th ECCMID, 25-28 April 2015, Copenhagen, DK. • Invited Speaker and Symposium Chair: Methods of Detection & Diagnosis of Biofilm Infections; ESCMID Study Group for Biofilms 4th European Congress on Biofilms, the EUROBIOFILMS 2015. June 23-26 2015. Brno, Czech Republic, • Low Dose Nitric Oxide Modulates Streptococcus pneumoniae Biofilm Metabolism and Antibiotic Tolerance. 7th ASM Conference on Biofilms, October 24-29, 2015. Chicago Hotel in Chicago, Illinois. Abstract S6:4 Oral Presentation. Selected Invited Non-conference, departmental seminars • Mycobacterial Biofilms. 1999. Center for Biofilm Engineering Departmental Seminar, Montana State University, Bozeman, MT. • Biofilm Imaging. 2000. Workshop on Microbial Biofilms. Proctor and Gamble, Cincinnati, OH. • Mycobacterial Biofilms of Potential Medical Significance. 2001. LigoCyte Pharmaceuticals. Bozeman, Montana. Invited Seminar. • Mycobacterium tuberculosis Binding to Human Proteins Involved in Innate Immunity: Using Shear to Select for High Affinity Binding for Vaccine Design. 2002. Department of Immunology and Microbiology, University of Iowa Medical School, Iowa City. • Mycobacteria: adhere to stay. 2003. Microbiology Departmental Seminar Series, Montana State University, Bozeman, MT. • Mycobacteria, biofilms and infection. 2003. Center for Biofilm Engineering Seminar, Montana State University, Bozeman, MT. • Biofilms: from the natural environment to infectious disease. 2004. Biomedical and Environmental Engineering Departments Seminar Series. Carnegie Mellon University, Pittsburgh, PA. Invited Seminar • Biofilm Infections and Pathogenesis. 2005. Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA. • Bacterial Biofilms: Multicellular Strategies for Survival. 2005. Medical Microbiology and Immunology Departmental Seminar, University of Minnesota Medical School, Duluth, MN. • Biofilms in Nature and Disease. 2007. Department of Chemistry and Biochemistry, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA. • Mycobacterial biofilms: a Sticky Wicket. Joint Seminar – School of Chemistry/School of Biological Science, University of Bangor. 8 October 2010. Wales, United Kingdom. • Biofilms in ear, nose and throat disease. GlaxoSmithKline Respiratory Infection Group, Belgium/IfLS Microbial Communities Research Group Meeting, 9 March 2011. University of Southampton, UK. • Biofilms, inflammation and infection: which came first the chicken or the egg and does it matter? University of Southampton School of Medicine, Division of Inflammation, Infection and Immunity, Academic Seminar Series, 6 May 2011. Southampton, UK. • Biofilm development by respiratory pathogens: implications for mucosal pathogenesis. The Ohio State University, Center for Microbial Interface Biology, 18 August 2011. Columbus, OH. • Biofilm development by respiratory pathogens: implications for pathogenesis. Department of Biotechnology, Chemistry and Environmental Engineering, 9 November 2012. Aalborg University, Aalborg, Denmark. • Bacterial biofilms in the respiratory tract: growing against the flow. Department of Infection and Immunity, 14 Feb 2013. University of Sheffield, United Kingdom. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

• Host factors inducing selection and persistence of the emerging CF pathogen, Mycobacterium abscessus 12

213 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Cystic Fibrosis Translational Research Group – OSU-NCH, August 13, 2015. CF Translational Meeting. • Mycobacterium abscessus: the TB of cystic fibrosis. MII/CMIB World TB Day Mini-symposium. March 25, 2016. The Ohio State University College of Medicine, Columbus, OH. • Streptococcus pneumoniae biofilms, metabolism and antibiotic tolerance. Pittsburgh Area Microbial Pathogenesis Seminar Series, May 6, 2016. Carnegie Mellon University, Pittsburgh, PA. Invited Seminar. • Streptococcus pneumoniae pathogenesis: a mystery encased in an enigma. State University of New York (SUNY- Binghamton) Harpur College Dean’s Speaker Series for Biological Sciences. December 2, 2016. Binghamton, New York. • “Story telling without a beat – Primary ciliary dyskinesia host factors contributing to biofilm infection with nontypeable Haemophilus influenzae”, Microbial Infection & Immunity, Biofilms in Human Medicine Interest Group, September 6, 2017. The Ohio State University. • Mycobacterium abscessus in cystic fibrosis lung disease, C3 Cystic Fibrosis Translational Research Group and CF Foundation Advisory Board Meeting, January 17, 2018. Ohio State University, Columbus, OH. • Two persistence strategies of the emerging pathogen Mycobacterium abscessus. Infectious Diseases Institute (IDI) Work in Progress, February 5, 2018. The Ohio State University, • Bacterial biofilms: from the natural environment to infectious diseases – the case of Mycobacterium abscessus, National Jewish Hospital, Oct 17, 2018. Denver, CO. • The Role of Autophagy in Defective Mycobacterium abscessus Clearance in CF Macrophages. Cure CF Columbus (C3) Translational Research Group External Advisory Board Meeting, April 4, 2018. Nationwide Children’s Hospital-Ohio State University, Selected peer-reviewed Posters, Abstracts, and post-doctoral, student Presentations (Student, Trainee, research associate, post-doc fellow or clinical fellow presenter underlined) • Hall-Stoodley L, Lappin-Scott HM. Mycobacterium fortuitum biofilm formation under flow conditions. American Society of Microbiology General Meeting. 1998. Atlanta, GA • Hall-Stoodley L, Watkins AP, Lappin-Scott HM. Development of mycobacterial biofilms: implications for nosocomial infections. Society for General Microbiology General Meeting. 1999. Edinburgh, Scotland. • Hall-Stoodley L, Stoodley P, Lappin-Scott HM. Monitoring detachment and motility of environmental mycobacteria. American Society of Microbiology, 2nd International Conference on Biofilms. Session II – Detachment. 16-20 July 2000. Big Sky, MT. • Janzen J, L Hall-Stoodley. 2002. In situ lectin staining of Gram-positive and Gram-negative bacterial glycoconjugates. National Science Foundation, Research Education for Undergraduates (REU). Center for Biofilm Engineering, Montana State University, Bozeman, MT. Selected Student Oral Presentation for National Technical Advisory Committee Meeting • Star W, L Hall-Stoodley. 2003. Mycobacterial adherence to human proteins. Minority Leadership Alliance Conference, Chantilly, VA. Student oral presentation. Award for outstanding student research • Fux CA, McNamee LA, Hall-Stoodley L. Host factors are required for Streptococcus pneumoniae biofilm formation. American Society of Microbiology Biofilms 2003 Conference. 1-6 November 2003. Victoria, BC, Canada. • McNamee LA, Hall-Stoodley L and Harmsen AG. 2005. Development of an ex vivo murine tracheal explant system to model influenza-induced tissue damage and subsequent Streptococcus pneumoniae adherence. The Society for Experimental Biology and Medicine, San Diego, CA. • Hall-Stoodley L et al. 2005. Pseudomonas aeruginosa Fluorescent Protein Reporter Vectors for the Investigation of Biofilm Formation, Abstract 1308, Session D13. Association for Research in Otolaryngology Midwinter Meeting. New Orleans, LA. • Duberstein DJ, Coticchia JM, Kerschner JE, Hall-Stoodley L, et al. Biofilms in Recurrent Acute Otitis Media in Children. Otolaryngology-Head and Neck Surgery Annual Meeting. 135(2) P160. 2006. Toronto, Canada. • Johnson C, Nisticò L, L Hall-Stoodley. Antibiotic Susceptibility of Streptococcus pneumoniae During Planktonic and Biofilm Growth #C78. Annual Biomedical Research Conference for Minority Students (ABRCMS). 8-11 November 2006. Anaheim, CA. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

• Hall-Stoodley L, Nisticò L, et al. Clinically Relevant Strains of Streptococcus pneumoniae Form Biofilms and Exhibit Variability in the Extent of Polysaccharide Matrix Production and Resistance to Antibiotics. 13

215 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

ASM Session Title: 138/D. Streptococci, Enterococci and Staphylococci. Poster Presentation: #D-123. American Society of Microbiology 107th General Meeting. 21-25 May 2007. Toronto, Canada. • Nisticò L, Gieseke A, Coticchia JM, Burrows A, Khampang P, Kerschner JE, Post JC, Ehrlich GD, Stoodley, P, Hall-Stoodley L. Localization of upper respiratory pathogens in pediatric adenoids exhibits biofilm formation Poster Presentation: #D-087. ASM Session: 069/D. Oral and upper respiratory bacterial pathogens. American Society of Microbiology 107th General Meeting. 21-25 May 2007. Toronto, Canada. • Nisticò L, Hall-Stoodley L, et al. Imaging Biofilms in Clinical Samples. 4th American Society of Microbiology Conference on Biofilms. 25-29 March 2007. Quebec City, Canada. • Krespi YP, Arora A, Longwell M, Nisticò L, L Hall-Stoodley, et al. 2008. Tonsillolith: Not Just a Stone But a Living Biofilm. Otolaryngology - Head and Neck Surgery. 139:P94-P95. Chicago, IL. USA *Featured in Meeting Daily Newsletter of the AAO-HNS* • Hoa M, Kingsley E, Christensen L, Hall-Stoodley L, et al. 2008. Identification of Nasopharyngeal Biofilms with Middle Ear Pathogens in Otitis-Prone Children Utilizing Scanning Electron Microscopy and Fluorescent In-Situ Hybridization Confocal Microscopy. Association for Research in Otolaryngology Midwinter Research Meeting, 16-21 February, Phoenix, AZ. • Stoodley P, Nisticò L, Gieseke A, Kreft R, Coticchia JM, Burrows A, Khampang P, Kerschner JE, Post JC, Ehrlich GD and L Hall-Stoodley. Adenoids: A Reservoir for Pathogenic Biofilms. 5th American Society of Microbiology Conference on Biofilms. 15-19 November 2009. Cancun, Mexico. • Nisticò L, Gieseke A, Kreft R, Coticchia JM, Kerschner JE, Post JC, Ehrlich GD, Stoodley P and L Hall- Stoodley. Pediatric adenoids ecology and otitis media: is biofilm formation the linkage? First European Congress on Microbial Biofilms, 2-5 September 2009. Rome, Italy. • Grande R, Nisticò L, Sambanthamoorthy K, Iannitelli A, Cellini L, Di Stefano A, L Hall Stoodley, Stoodley P. Staphylococcus aureus UAMS-1 biofilm development over time: the role of extracellular DNA and carbohydrates in the biofilm structure. First European Congress on Microbial Biofilms, 2-5 September 2009. Rome, Italy. • Gawalt ES, Palchesko RN, Kruszewski K, Nisticò L, L Hall-Stoodley. Interfacial Control of Alloy Implants by Self-Assembled Monolayers and Surface-Initiated Polymerization. Biofilms IV, European Biofilm Conference, Winchester, UK, Sept 1-3, 2010 • Howlin RP, Hall-Stoodley L, et al. Nitric oxide induces dispersal of multispecies cystic fibrosis biofilms and enhances antibiotic sensitivity of Pseudomonas aeruginosa. European Society of Paediatric Infectious Diseases, The Hague. Netherlands, 2010. Oral ePoster. • Kruszewski K, Gawalt ES, Hall-Stoodley L. Effect of the surface wettability of adhered monolayer and polymer coatings on protein and bacterial adhesion to metal biomaterial surfaces. UPMC McGowan Institute Retreat for Regenerative Medicine. 7-10 March 2010. Pittsburgh, PA. • Howlin R, Cathie K, Hall-Stoodley L, et al. Nitric oxide-mediated dispersal and enhanced antibiotic sensitivity in Pseudomonas aeruginosa biofilms from the cystic fibrosis lung. British Society Paediatric Dermatology/British Paediatric Allergy Immunology and Infection Group. Arch Dis Child (2011) 96:A45 doi:10.1136/adc.2011.212563.100. RCPCH Annual Conference, Warwick, UK, April 2010 Oral presentation. • Howlin R, Hall-Stoodley L, et al. Nitric Oxide-Mediated Dispersal and Enhanced Antibiotic Sensitivity in Pseudomonas aeruginosa Biofilms from the Cystic Fibrosis Lung. European Society Clinical Microbiology and Infectious Diseases (ESCMID) Eurobiofilms, 2nd European Congress on Microbial Biofilms – Basic and Clinical Aspects, 6-8 July 2011, Copenhagen, Denmark. • Howlin R, Hall-Stoodley L, et al. Breaking Antibiotic Resistance in Biofilms. Royal College of Paediatrics and Child Health Conference, April 2011. Coventry, United Kingdom. Oral Presentation. • Hayes S, Hall-Stoodley L, et al. Bacterial biofilms in chronic rhinosinusitis. 2011. British Rhinological Society 10th Annual Meeting. 13 May 2011. Royal College of Surgeons of Edinburg, Scotland. Oral Presentation. • Walker W, Jackson C, Hall-Stoodley L, Faust S, Lucas J. The relationship between Haemophilus influenzae biofilms and respiratory epithelia. 2011. European Society of Pediatric Infectious Disease Conference, 7-11 June 2011. The Hague, The Netherlands, Oral Presentation. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

• Kruszewski K, Hall-Stoodley L, Gawalt ES. Rigid versus flexible self-assembled monolayers (SAMs) for interfacial control of biomaterial implant devices. UPMC McGowan Institute Retreat for Regenerative 14

217 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Medicine, March 6-8, 2011. University of Pittsburgh, Pittsburgh, PA. First Prize Poster, Biomedical Devices Category • SM Hayes, S Faust, *RJ Salib & *L Hall-Stoodley. Characterisation of bacterial biofilms in nasal polyps. 14th British Academic Conference in Otolaryngology. July 4-7, 2012. Glasgow, UK • SM Hayes, J Webb, S Clarke, P Stoodley, S Faust, PG Harries, RJ Salib & L Hall-Stoodley. Characterization of Bacterial Biofilms in Nasal Polyps. Biofilms 5, 10-12 December 2012. Paris, France. • Leir S, Allan R, Webb J, Faust SN and L Hall-Stoodley. Investigating the use of sodium nitroprusside (SNP) in the treatment of paediatric otitis media. British Pharmacological Society Winter Meeting, 15-19 December 2012. London, UK. First prize poster • Barbara R, Allan R, Webb J, Faust SN, and L Hall-Stoodley. Investigating the effect of enzymatic treatment on Streptococcus pneumoniae Biofilms. Antimicrobial Strategies for Biofilm Control. University of Winchester, IBBS Conference 10 - 11 September 2012. Winchester, UK. • Walker WT, Jackson CL, Lackie P, Howlin R, Allan R, Faust SN, Lucas JS, Hall-Stoodley L. Is Primary Ciliary Dyskinesia a “Biofilm” Disease? Poster presentation. European Respiratory Society Annual Congress, 1-5 September 2012. Vienna, Austria • Walker WT, Jackson CL, Lackie P, Howlin R, Allan R, Faust SN, Lucas JS, Hall-Stoodley L. Developing a primary ciliated airway epithelial cell model to assess the effect of ciliary function on Haemophilus influenzae biofilm formation. Translational Medicine Conference, 2012. University of Southampton, UK. Poster. • Cathie K, Howlin RP, Sukhtankar P, Duignan C, Pink S, Smith C, Jefferies J, Salib R, Carroll M, Daniels T, Connett G, Legg J, Barraud N, Rice S, Kjelleberg S, Kelso M, Stoodley P, Hall-Stoodley L, Webb, J and S Faust. Low dose nitric oxide as adjunctive therapy to treat chronic Pseudomonas aeruginosa infection in cystic fibrosis. ID Week, IDSA, SHEA, HIVMA, PIDS. 2-6 October, 2013. San Francisco, CA. • Howlin RP, Cathie K, Sukhtankar P, Duignan C, Pink S, Smith, C, Jefferies J, Salib R, Carroll, M, Daniels, T, Connett, G, Legg, J, Barraud, N, Rice, S, Kjelleberg, S, Kelso, M, Stoodley P, Hall- Stoodley L, Webb, J, S Faust. Nitric oxide adjunctive therapy to treat chronic Pseudomonas aeruginosa infection in cystic fibrosis. Eurobiofilms 2013, 9-12 September, 2013. Ghent, Belgium • Sasindran S, Clary G, Hall-Stoodley L. Host factors Inducing Selection and Persistence of Mycobacterium abscessus. CMIB Symposium, March 31-April 1, 2016. The Ohio State University. • Nathan Nesbitt, Clary G, Sasindran S, Luanne Hall-Stoodley. Mycobacterium abscessus Smooth and Rough Morphotypes Form Antimicrobial-Tolerant Biofilm Phenotypes But are Killed By Acetic Acid, 2018 OSUWMC College of Medicine Trainee Research Day, April 12, 2018. The Ohio State University. • Allan RN, Walker W, Collins SA, Jackson CL, Hall-Stoodley L, JSA Lucas. A moving story: How epithelium with impaired ciliary function responds to biofilm infection. Biofilms 8, 27-29 May, 2018. Arhus, Denmark.

TEACHING University Courses/Lectures/International Courses 1. Montana State University, Bozeman, Montana, United States 1996 – 2003 Undergraduate • Microbes in the Environment (UNIV 125CS – Undergraduate CORE course, College of Letters and Sciences) • Human Pathophysiology (BIOH 395) • Medical Bacteriology (MB 466) • Microbiology Graduate Seminar – Department of Immunology and Microbiology (MB500) Graduate – Department Medical Sciences, Alaska, Montana, Idaho and Wyoming (WAMI) Medical Program, U Washington • Histology (MEDS 510 Micro Anatomy) • Pathology (MEDS 520 Molecular & Cellular Basis of Disease) • Medical Microbiology (MEDS 520 Molecular & Cellular Basis of Disease) 2. University of Exeter, Department of Biological Sciences, United Kingdom 1997 – 1999 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

15

219 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

• Immunology, Lecturer 1997 - 1999 • Medical Microbiology, Instructor 1998 - 1999 • Methods in Environmental Microbiology, Graduate Research Seminar 1998 3. Allegheny Singer Research Institute, Center for Genomic Science, Pittsburgh, PA 2005 – 2009 • Science Curriculum Director: National Center for Education Research (NCER) Institute of Education Sciences, Effective Teachers and Effective Teaching Development and Innovation Program, “Mentoring Teachers through Pedagogical Content Knowledge” o Directed a continuing education model for high school biology teachers to bring updated leading-edge scientific content into the classroom by providing basic research-based concepts and laboratory skills that fundamentally explore new technology and research approaches within the context of health-related topics • 2007 “From Mendel to Molecular Genetics and Functional Genomics: Where no one has gone before…” Developed curriculum and coordinated scientific lectures and bioethical seminars on genetics, stem cell and genomic research • 2008 “Microbial Communities and Human Communities: close encounters”. Developed curriculum and coordinated scientific lectures, bioethical seminars and laboratory sessions on infection and immunity, infectious disease, bacterial genomics, biofilm infections and bioinformatics, including timely discussions on MRSA, commensal v pathogenic bacteria, the human microbiome and the vaccine debate 4. University of Southampton, United Kingdom 2010 – 2013 • Faculty of Biological Sciences, Lecturer, Biofilms and Microbial Communities - 29860 “Biofilm- associated Infections; Biofilm Approaches for Clinically Relevant Biofilms” 5. The Ohio State University (2013 – present) • Microbiology 7724/IBGP 7240 (Team instructed course), Molecular Pathogenesis, Microbiology – (Biofilm Infections; Mycobacteria) • Microbiology 4110, Microbial Pathogenesis and Immunobiology (Attachment to Host Cells and Tissues) International Courses 1. University of Minho, Braga, Portugal (2011) Faculty, Theoretical Class: Medical Biofilms: 3rd International Course in Biofilm Science – Biofilms: From Basic To Emergent; 9 -13 May, “Medically Important Biofilms”, Centre for Biological Engineering 2. University of Copenhagen, Copenhagen, DK (2011 – present) Faculty, Online Course: Bacterial Biofilms and Their Role in Chronic Infections, Diagnosis of Bacterial Biofilms. Department of International Health, Immunology and Microbiology, University of Copenhagen and European Society of Clinical Microbiology and Infectious Disease (ESCMID), https://biofilmcourse.ku.dk 3. Institut Pasteur, Les Pensières, Annecy, France (2018 – present) Core Faculty, International Course on Antibiotic Resistance (ICARe), Targeting microbial biofilms, https://www.pasteur.fr/en/international-course-antibiotics-and-resistance-icare MENTORSHIP Sabbatical Faculty /visiting scientists 1. Center for Biofilm Engineering, Montana State University, Bozeman, MT. Dr. Christoph Fux, Swiss National Science Foundation, Berne, Switzerland. 2003 2. Allegheny Singer Research Institute, Center for Genomic Sciences, Sabbatical Research: Dr Rosella Grande, Department of Biological Sciences, University G. D’Annunzio, Chieti, Italy, Staphylococcus aureus biofilm development: role of DNA and carbohydrates in biofilm structure, University G. D’Annunzio, Chieti, Italy (2011) 3. University of Southampton, UK Sabbatical Research: Dr. Ellen Gawalt, Duquesne University, Dept of Biochemistry and Chemistry: “Immobilized antimicrobial compounds for the prevention of orthopaedic implant biofilms” 2012

16 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

4. OSU Visiting Scholar/Scientist: Dr. Ray Allan, University of Southampton, UK “Effect of mutations in carbohydrate transport genes on biofilm development by Streptococcus pneumoniae” 2015 Mentorship of Postdoctoral Fellows 1. Duc Nguyen, Ph.D, Carnegie Mellon University, Pittsburgh, PA, (Post-doctoral Fellow) (2004- 2005) 2. Armin Gieseke, PhD, Max Planck Institute for Marine Microbiology, Bremen, Germany, Visiting Post- doctoral Fellow. The application of fluorescence in situ hybridization to clinical samples (2005) 3. Karthik Sambanthamoorthy, PhD, Center for Genomic Sciences, Senior Research Fellow, Gram positive biofilms (2007-2008) 4. Rosella Grande, PhD, Department of Biological Sciences, University G. D’Annunzio, Chieti, Italy, Visiting Post-doctoral Fellow, Role of DNA and carbohydrates in biofilm structure (2008) 5. Laura Nistico, PhD, Center for Genomic Sciences, Biofilms in chronic infections – Senior Research Fellow (2004-2009) 6. Rob Howlin, PhD, School of Biological Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, “Role of pathogenic bacteria and Nitric Oxide Mediated effects in CF”— National Institutes of Health Research Respiratory BRU Fellow (2009 – 2013) 7. Ray Allen, PhD, School of Biological Sciences, University of Southampton, University Hospital Southampton NHS Foundation Trust, Post-doctoral Fellow, New whole-species pneumococcal vaccines, Gates Foundation Grand Challenges; Investigation of novel adjunctive therapies to improve treatment success in paediatric otitis media – SPARKS The Children’s Trust (2010 – 2017) Mentorship of Medical Residents 1. Earnest Braxton, MD (), Allegheny-General Hospital resident research program, “Biofilm infections in brain shunts” (2005) 2. Vikram Gahlot, MD, Allegheny-General Hospital resident research program, “Biofilm infection- resistant implants for otologic application” (2007-2008) 3. Laura Mejía MD, Allegheny General Hospital, Pittsburgh, PA. “Modeling biofilm EPS” (2008) 4. Anurin Young, MD, Academic Foundation: General Practice Research, Southampton University Hospitals NHS Trust “Growth response of planktonic and biofilm Haemophilus influenzae to adjunctive therapies” (2012) RESEARCH ADVISOR Ph.D. Candidate Dissertation Committees 1. Lynnelle Pittet, PhD, “Interactions of Streptococcus pneumoniae with murine tracheal explants after influenza infection” Department of Veterinary Molecular Biology, Montana State University- Bozeman, MT (Committee, research supervisor) (2003 – 2006) 2. Kristen Kruszewski, PhD, “Prevention of biofilm growth on orthopedic implant materials through surface chemical modification” Chemistry and Biochemistry, Duquesne University, Pittsburgh, PA, (External Reader) (2008 – 2012) 3. Dr. Stephen Hayes, Royal College of Surgeons Clinical Fellow, PhD, “Investigation of novel adjunctive therapies in the eradication of bacterial biofilms in chronic rhinosinusitis”, University of Southampton, School of Medicine, Division Inflammation, Infection and Immunity, UK (Co- Supervisor) (2010 – 2013) 4. Dr. Woolf Walker, Clinical Fellow PhD, “Cilia, Nitric Oxide and Bacterial Biofilm Infections” University of Southampton, School of Medicine, Division Inflammation, Infection and Immunity, UK (Co-supervisor) (2011 – 2014) 5. Dr. Sarah Chaney, DVM, PhD. Department of Veterinary Medicine, Microbial Infection and Immunity, The Ohio State University College of Medicine (Dissertation Committee) (2014 – 2016) 6. Matt Wilkins, PhD. University of Southampton, School of Medicine, Clinical and Experimental Sciences, UK, “Investigation of the role of host immunity to bacterial biofilms in the pathogenesis Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

of upper respiratory tract infection & bacterial carriage in the normal host” (Co-supervisor) (2013 – 2018) International PhD examination 1. Ruth Thornton, PhD candidate, “Biofilm and intracellular infection: Persistence mechanisms of bacterial otopathogens in chronic and recurrent otitis media” Paediatrics and Child Health, University of Western Australia, Australia (External Examiner) (2010) 17

222 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

2. Vibeke Rudkjobing, PhD candidate, Diversity and distribution of microorganisms in cystic fibrosis lungs, Teknologisk Institut, Life Science Division, Aalborg University, Denmark (External Examiner) (2012) Medical Science/Medical students Supervisor The Ohio State University 1. Nathan Nesbitt – Roessler Medical Student Research Scholar, Project: Antibiotic Susceptibility in Smooth and Rough Morphotypes of Mycobacterium abscessus, an Emerging Cystic Fibrosis Pathogen (2018) Published in: Antimicrob Agents Chemother 62(3). pii:e01782-17. PMID: 29311080, Clary G, Sasindran S, Nesbitt N, Mason L, Azad A, Cole S, McCoy K, Schlesinger LS and L Hall-Stoodley. Mycobacterium abscessus Smooth And Rough Morphotypes Form Antimicrobial-Tolerant Biofilm Phenotypes But Are Killed by Acetic Acid. University of Southampton, UK (2010 – 2013) Faculty of Medicine, Supervisor Medical Science research projects. I have mentored 4 MedSci Students: all students received distinction (1st) for their project. (1st indicates Distinction) 1. Kerrie Burnett, Masters MedSci (2012 - 2013) 2. Sarah Leir, Novel therapies for microbial biofilms in paediatric otitis media. Awarded 2012 British Pharmacological Society for Undergraduate Pharmacology Research Prize (£500) 3. Raquel Barbara, Enzymatic treatment of Streptococcus pneumoniae Biofilms 4. Samantha Morgan, Pneumococcal biofilms in pediatric adenoids. Published in: Antimicrob Agents Chemother. 60(4):2456-66. PMID: 26856845: Allan RN, Morgan S, … Faust SN, Hall- Stoodley L. 2016. Low concentrations of nitric oxide modulate Streptococcus pneumoniae biofilm metabolism and antibiotic tolerance. Undergraduate, Masters and PhD students (Microbiology, Cellular Biology) 1. University of Exeter, UK (1998 – 1999) I advised three PhD candidates and one BSc student during my Post-doctoral Fellowship. 2. Montana State University—Bozeman, Montana (2000 – 2003) BSc Students (3): 1. National Institutes of Health IMSD Student Fellow. 2. National Science Foundation Research Education for Undergraduates (REU) Fellow. 3. Chemical Engineering Undergraduate Scholar. 3. Allegheny-Singer Research Institute (2004 – 2009) Research Internships, Center for Genomic Science, Pittsburgh, PA: PhD (1) Department of Cell and Molecular Biology, Pennsylvania State University; MSc (2) Department of Biochemistry & Chemistry, Duquesne University, Pittsburgh, PA; BSc (4): Forensic Sciences, Duquesne University, Pittsburgh, PA; Honors Thesis; NIH IMSD Student Fellow – Department of Biology, University of Maryland-Eastern Shore, MD; (2) CGS Summer Internships, Honors Thesis; Mathematics/Undergraduate Medical Program, University of Pittsburgh, PA 4. University of Southampton (2010 – 2013) PhD student research projects: PCR and FISH”, Southeast Regional Health Protection Agency, U Southampton, School of Medicine, Division Inflammation, Infection and Immunity (Ahmed N, 2012-2013) Phenotype-Switching and Pleomorphic-Changes in Haemophilus influenzae biofilms” U Southampton, School of Medicine, Division Inflammation, Infection and Immunity, School of Medicine (Devine V, 2011) 5. The Ohio State University Emma Rico (Honors); Devon Leahy (Biology) 2019 Abdulsalam Bah Honors Thesis Committee, (Ohio State University and Icahn School of Medicine at Mount Sinai) 2018; Austin Maloney (Biology) 2018 Laurel Mason (Microbiology) 2017 (Published in Antimicrob Agents Chemother 62(3)pii:e01782-17. PMID: 29311080, Clary G, Sasindran S, Nesbitt N, Mason L, Azad A, Cole S, McCoy K, Schlesinger LS and L Hall-Stoodley. Mycobacterium abscessus Smooth And Rough Morphotypes Form Antimicrobial- Tolerant Biofilm Phenotypes But Are Killed by Acetic Acid. Mentorship – diverse students

18 Curriculum Vitae: Luanne Hall-Stoodley, PhD.

Undergra duates 1. Woodrow Star, National Institutes of Health Institutional Minorities Student Development (IMSD) Student Fellow, Center for Biofilm Engineering, Montana State University. Award for outstanding student research § Star, W and L Hall-Stoodley. Mycobacterial adherence to human proteins. Minority Leadership Alliance Conference, Chantilly, VA http://www.montana.edu/news/1306/native-american- students-bring-home- conference-awards 2. Candace Johnson, Honors Thesis Project, NIH Institutional Minorities Student Development (IMSD) Student Fellow – Department of Biology, University of Maryland. § Johnson C, Nistico L and L Hall-Stoodley. Antibiotic Susceptibility of Streptococcus pneumoniae During Planktonic and Biofilm Growth. Annual Biomedical Research Conference for Minority Students (ABRCMS). 8 - 11 November 2006. Anaheim, California. USA #C78. § Published in BMC Microbiology, 8:173. PMID: 18842140. Hall-Stoodley L, L Nistico, D Nguyen B Dice, WJ Mershon, C Johnson, et al. 2008. Characterization of Biofilm Matrix, Degradation by DNase Treatment and Evidence of Capsule Downregulation in Streptococcus pneumoniae Clinical Isolates. Medical Resident/Intern 1. Earnest Braxton, MD. (Neurosurgery), Allegheny-General Hospital Resident Research Program, “Biofilm infections in brain shunts” § Published in Neurosurg Rev. 28(4):249-55. Braxton EE Jr, Ehrlich GD, Hall-Stoodley L, et al. 2005. Role of biofilms in neurosurgical device-related infections. § Published in Pediatr Neurosurg. 46(2): 127-32. PMID: 206643. Stoodley P, Braxton EE Jr, Nistico L, Hall-Stoodley L, et al. 2010. Direct demonstration of Staphylococcus biofilm in an external ventricular drain in a patient with a history of recurrent ventriculoperitoneal shunt failure.

FUNDING Current ◆ C3 Research Pilot and Feasibility Award “T cell dysfunction in CF patients with nontuberculous mycobacteria infection” Role: Co-I 2019 ◆ Cystic Fibrosis Foundation Research Grant “Xenophagy and clearance of nontuberculous mycobacteria in CF macrophages” Role: PI 2018-2020 ◆ Cystic Fibrosis Foundation Pilot & Feasibility Research Grant “Macrophage responses to Mycobacterium abscessus infection in cystic fibrosis” Role: PI 2018-2020 ◆ College of Medicine Bridge Funding Program “Novel virulence determinant discovery in Mycobacterium abscessus” Role: PI 2018-2019 ◆ CF Foundation RDP Pilot and Feasibility Grant “T cell dysfunction in CF patients with nontuberculous mycobacteria infection” Role: (Co-I) 2019-2021 Pending Cystic Fibrosis Foundation Infection Research Initiative Team Science Grant “Managing Infections in the Era of Highly Effective Modulator Therapy” Role: Co-PI 07/01/2020 – 06/31/2023 Past ◊ The Wellcome Trust. Sir Henry Wellcome Award for Innovative Research (Showcase Award) Post- doctoral Fellowship Trust Ref. 050950/MJM/APHAward (One of 20 applications funded of 507 - 4% success rate) Role: PI 1997-1998 ◊ National Institutes of Health - National Institute of Allergy and Infectious Diseases “Tubercle bacilli binding to host cells: vaccine design.” Role: PI 2003-2004 ◊ Department of Education Institute of Education Sciences- Effective Teaching Development and Innovation Program “Mentoring Teachers through Pedagogical Content Knowledge.” Role: Acting PI 2006-2009 ◊ Bill and Melinda Gates Foundation. Grand Challenges Award “New whole-species pneumococcal vaccines.” Identification of uniquely expressed proteins in Streptococcus pneumoniae biofilms Role: Co- I 2010-2011 ◊ SPARKS The Children’s Medical Research Charity, Innovation and Patient Benefit Grant (UK) “Low dose

19 Curriculum Vitae: Luanne Hall-Stoodley, PhD. nitric oxide (NO) as adjunctive therapy with antibiotics using a novel NO donor and localized NO release.” Role: PI 2012-2014 ◊ Sir Jules Thorn Trust (UK) “Investigation of the role of host immunity to bacterial biofilms in the pathogenesis of upper respiratory tract infection & bacterial carriage in the normal host.” Role: Co- PI, supervisor 2012-2017 ◊ Cystic Fibrosis Foundation Sub-award (PI: McCoy, K) “Mycobacterium abscessus selection and persistence” Role: PI Subaward 2016-2017 ◊ C3 Research Pilot and Feasibility Award “The role of autophagy in defective Mycobacterium abscessus clearance in CF macrophages” Role: PI 2018 Consultancy – Industry Projects § Novel vaccine approaches to medically significant mycobacteria. LigoCyte Pharmaceuticals. Bozeman, Montana (2001-2003) Project PI § Orthopedic Materials for Inhibition of Biofilm Development by Clinically Relevant Pathogenic Bacteria. Stryker Orthopedics, Pittsburgh, PA (2007) Project Advisor § Disruption of clinically relevant biofilms in the ear, nose and throat using a Q-switched Nd:YAG laser. Valam Corp, New York (2008-2009) Project Advisor § Plex-ID – a non-targeted, high resolution and high throughput, rapid molecular approach to the investigation of bacteria, viruses, vector borne pathogens and fungi in disease. Abbott Molecular /Ibis Biosciences Europe, Wiesbaden, Germany (2010-2012) Project Advisor § Catabasis Pharmaceuticals, Cambridge MA “Study of CAT-5571 in the clearance of nontuberculous mycobacteria” (2018) Project PI

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

20

227 Mark E. Drew, PhD Curriculum Vitae and Bibliography May 19, 2020

Personal Data Address and telephone numbers Office: Home: 460 W 12th Avenue, 2547 Wickliffe Rd 792 Biomedical Research Tower Upper Arlington, Ohio 43221 Columbus, Ohio 43210 614-859-2309 614-292-4337; 292-9616 (fax) [email protected]

Date of Birth: January 18, 1967 Place of Birth: Bremerton, Washington Citizenship: USA Married: Joanne Drew Children: Fred and Matilda

Educ ation Year Degree Institution 1991 BA (Chemistry) Seattle Pacific University, Seattle, WA 2002 PhD Johns Hopkins University School of Medicine, Baltimore, MD

Train ing Year Position Institution 1991 Research Assistant Seattle Biomedical Research Institute, Seattle, WA - 1994 1994 Research Assistant Oregon Health Sciences Univeristy, Portland, OR - 1996 2002 Postdoctoral Molecular Microbiology and Immunology Howard - Researcher Hughes Medical Institutes 2008 Washington University School of Medicine, St. Louis, MO

Academic Appointments

2008- Assistant Professor Ohio State University College of Medicine, 2014 Columbus, OH 200114- Research Assistant Ohio State University College of Medicine, present Professor Columbus, OH

Professional Society Memberships 2010-Present American Society for Microbiology

Professional Activities 1. Antimicrobial Agents and Chemotherapy, Ad hoc Reviewer, 2009-present 2. Journal of Autophagy, Ad hoc Reviewer, 2009-present 3. PLOSOne, Ad hoc Reviewer, 2010-present 4. Microbiology, Ad hoc Reviewer, 2010-present 5. The Wellcome Trust/DBT India Alliance, Ad hoc Reviewer, Jan 2011 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

6. Proceedings of the National Academy of Sciences, Ad hoc Reviewer, 2012 7. PLOS Neglected Pathogens, Ad hoc Reviewer, 2016 8. Current Medicinal Chemistry, Ad hoc Reviewer, 2016

Other Professional/Public Services

1. Invited Teaching Faculty: Biology of Parasitism, MBL, Woods Hole, MA. 2006, 2007, 2008

229 Mark E. Drew, PhD 2

2. OSU Representative: Pharmaceutical Assets Portal: Matching Academia and Industry for Drug Repositioning, NIH CTSA, Bethesda, MD. Dec 2009. 3. Invited Moderator: International Molecular Parasitology Meeting, International Molecular Parasitology Meeting, Woods Hole, MA. Sep 2010. 4. Invited Reviewer, NIH Special Emphasis Review Panel, “Discovery/Development of Novel Therapeutics for Eukaryotic Pathogens (R21/R33)”, May 2016 5. Invited Reviewer, NIH Special Emphasis Review Panel, “Pre-Clinical Models of Infectious Diseases”, December 2016

University Committees

Service to Student Affairs (Honors and Scholars): Maximus Essay Evaluator, 2010. OSU College of Medicine, CMIB Host Pathogen Seminar Series, Acting Organizer, 2011-2012. OSU College of Medicine, CMIB Retreat Organizing Committee, Member, 2011. OSU College of Medicine, Faculty Search Committee, PHPID Clinical Trials, Member, 2009-2011. OSU College of Pharmacy, BSPS Course Restructuring, Member, 2010. OSU Undergraduate Research Office, Faculty Representative, 2015-2018

Research Focus The research of the Drew lab focuses on Plasmodium falciparum, the protozoan parasite that causes the most pathogenic form of human malaria. There are more than 200 million new clinical cases of malaria each year with well over 1 million fatalities; it is the leading cause of childhood death by any single infectious agent in the world. Treating this disease requires anti-malarial drugs that are affordable and accessible, and a strong effort toward drug development is crucial in order to combat the ongoing threat of drug resistance. Our efforts for discovery and development of novel therapeutics to treat infections with P. falciparum are divided into two strategies: First, to identify novel structures for potential development of therapeutic leads. In this regard we are screening chemical compounds for their ability to kill malaria parasites. Toxicity screens are performed on in vitro cell cultures of P. falciparum in 96- or 384-well plate formats using a FACSCantoII flow cytometer housed in our lab. Promising compounds are prioritized based on a scoring matrix using criteria such as potency, death-phenotype, and selectivity. These are further characterized through structure-activity analysis and target identification. This work is done in close collaboration with members of the Division of Medicinal Chemistry and Pharmacognosy in the OSU College of Pharmacy. Second, we are evaluating a number of parasite-expressed proteases as viable drug targets. Our studies involve the characterization of these enzymes through biochemical, genetic, and cell biological methods. We have completed a screen of a library of mechanism-based cysteine protease inhibitors against P. falciparum, and we have identified compounds that kill parasites at low micromolar concentrations. This approach has allowed us to elucidate the mechanism of activation of the aspartic protease plasmepsins and has uncovered a protease homologous to the yeast autophagy protein, ATG4, which appears to be involved in regulating nutrient acquisition via autophagy in the parasite. We are in the process of conducting experiments to assess the essentiality and cellular function of this putative protease.

Past Areas of Research Interest 1. Transcriptional regulation of mitochondrial ribosomal RNA of Plasmodium falciparum 2. Facilitative nucleoside transporters of Leishmania donovani 3. Mitochondrial DNA replication mechanisms in Trypanosoma brucei 4. Characterization of RNAi pathways in Trypanosoma brucei 5. Catabolic proteases of the Plasmodium falciparum digestive vacuole

Current Projects 1. Whole-cell screening of novel small molecules for antimalarial activity 2. Identification of putative small molecule kinase inhibitors with antimalarial activity 3. Characterization of the autophagy pathway in Plasmodium falciparum Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Awards and Honors

231 Mark E. Drew, PhD 3

2010, 2012, 2015, 2016 Nominated as Distinguished Undergraduate Mentor, The Ohio State University Honors & Scholars Center Mark E. Drew, PhD 4

Thesis Advisor

Name Role D Tra Researc Awards/Ho Current e ini h Topic nors Position g ng re Per e iod at E nt ry Shansh College M 20 Anti- Post- an He of S 08- doctoral Pharma 20 Trypanos Researcher, cy, 12 omal NIH, Graduat drug Bethesda, e discovery MD Student Dawn College B 20 Autopha Post- Walker of S 09- gy doctoral Medicin 20 pathway Researcher, e, 13 in IBGP, Plasmodi Battelle, um Columbus, Graduat falciparu OH e m Student

PhD and Masters Advisory Committees • Trupti Pandharkar, PhD candidate, College of Pharmacy, OSU (PhD awarded 2011) • Basant Abdulrahman, PhD candidate, College of Medicine, OSU (PhD awarded 2011) Undergraduate Students (partial updates) Past: • Sanandan Maholtra (Johns Hopkins University, PhD candidate) • Katie Kemme (University of Cincinnati, School of Nursing) • Caitlin Blaut (Research Assistant, Industry) • Emily Cason (Emory University, Master’s candidate, MPH) • Benoit Mapa (Lake Erie College of Osteopathic Medicine, DO) • Elizabeth Butler (OSU BMS) • Kieran Tebben (Microbiology) • Basil Jafri (Microbiology)

Current: • Abu Rogers (Biochemistry) • Maria Barreras (Biology)

Ohio State University Undergraduate / Graduate / Professional Courses or Lectures 1. Chemotherapy of Infectious Diseases. Jan 2008. (Graduate) 2. Molecular Biology of Microbial Pathogenesis. Jan 2009. (Graduate) 3. Microbial Pathogenesis. Jan 2009. (Graduate) 4. Microbial Pathogenesis. Jan 2009. (Graduate) 5. Integrated Biology I Journal Club. Jan 2009. (Undergraduate) Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

6. Chemotherapy of Infectious Diseases. Jan 2010. (Graduate) 7. Medicinal Chemistry Laboratory. Jan 2010. (Undergraduate) 8. Medicinal Chemistry Laboratory. Jan 2011. (Undergraduate) 9. Biology of Human Disease. Jan 2011. (Graduate) 10. Biology of Human Disease. Jan 2012. (Graduate) 11. Host Defense. Jan 2012. (Graduate) 12. Infection & Host/Pathogen Science, Fall 2015 13. Cellular and Molecular Biology of Pathogenic Eukaryotes, Fall 2015 14. Infection & Host Pathogen Science, Spring 2016 15. Concepts in Biomedical Science, Fall 2016

Publications–Peer-reviewed Journal Articles

234 Mark E. Drew, PhD 5

1. Feagin JE, Drew ME. "Plasmodium falciparum: alterations in organelle transcript abundance during the erythrocytic cycle." Experimental Parasitology. Vol. 80, no. 3. (Jan 1995): 430-40. 2. Langford CK, Kavanaugh MP, Stenberg PE, Drew ME, Zhang W, Landfear SM. "Functional expression of a myo-inositol/H+ symporter from Leishmania donovani." Molecular and Cellular Biology. Vol. 15, no. 10. (Jan 1995): 5508-15. 3. Drew ME, Langford CK, Klamo EM, Russell DG, Kavanaugh MP, Landfear SM. "Functional expression and subcellular localization of a high-Km hexose transporter from Leishmania donovani." Biochemistry. Vol. 34, no. 37. (Jan 1995): 11814-21. 4. Klamo EM, Drew ME, Landfear SM, Kavanaugh MP. "Kinetics and stoichiometry of a proton/myo-inositol cotransporter." The Journal of Biological Chemistry. Vol. 271, no. 25. (Jan 1996): 14937-43. 5. Vasudevan G, Carter NS, Drew ME, Beverley SM, Sanchez MA, Seyfang A, Ullman B, Landfear SM. "Cloning of Leishmania nucleoside transporter genes by rescue of a transport- deficient mutant." Proceedings of the National Academy of Sciences of the United States of America. Vol. 95, no. 17. (Jan 1998): 9873-8. 6. Carter NS, Drew ME, Sanchez M, Vasudevan G, Landfear SM, Ullman B. "Cloning of a novel inosine- guanosine transporter gene from Leishmania donovani by functional rescue of a transport-deficient mutant." The Journal of Biological Chemistry. Vol. 275, no. 27.(Jan 2000): 20935-41. 7. Wang Z, Morris JC, Drew ME (Co-first Author), Englund PT. "Inhibition of Trypanosoma brucei gene expression by RNA interference using an integratable vector with opposing T7 promoters." The Journal of Biological Chemistry. Vol. 275, no. 51. (Jan 2000):40174-9. 8. Abu-Elneel K, Robinson DR, Drew ME, Englund PT, Shlomai J. "Intramitochondrial localization of universal minicircle sequence binding protein, a trypanosomatid protein that binds kinetoplast minicircle replication origins." The Journal of Cell Biology. Vol. 153, no. 4. (Jan 2001): 725-34. 9. Drew, M, E; Englund, P T. "Intramitochondrial location and dynamics of Crithidia fasciculata kinetoplast minicircle replication intermediates." The Journal of Cell Biology. Vol. 153, no. 4. (Jan 2001): 735-44. 10. Klingbeil MM, Drew ME, Liu Y, Morris JC, Motyka SA, Saxowsky TT, Wang Z, Englund PT. "Unlocking the secrets of trypanosome kinetoplast DNA network replication." Protist. Vol. 152, no. 4. (Jan 2001): 255-62. 11. Morris JC, Drew ME, Klingbeil MM, Motyka SA, Saxowsky TT, Wang Z, Englund PT. "Replication of kinetoplast DNA: an update for the new millennium." International Journal for Parasitology. Vol. 31, no. 5-6. (Jan 2001): 453-8. 12. Morris JC, Wang Z, Drew ME, Paul KS, Englund PT. "Inhibition of bloodstream form Trypanosoma brucei gene expression by RNA interference using the pZJM dual T7 vector." Molecular and Biochemical Parasitology. Vol. 117, no. 1. (Jan 2001): 111-3. 13. Grams J, Morris JC, Drew ME, Wang Z, Englund PT, Hajduk SL. "A trypanosome mitochondrial RNA polymerase is required for transcription and replication." The Journal of Biological Chemistry. Vol. 277, no. 19. (Jan 2002): 16952-9. 14. Morris JC, Wang Z, Drew ME (Co-first Author), Englund PT. "Glycolysis modulates trypanosome glycoprotein expression as revealed by an RNAi library." The EMBO Journal. Vol. 21, no. 17. (Jan 2002): 4429-38. 15. Wang Z, Drew ME (Co-first Author), Morris JC, Englund PT. "Asymmetrical division of the kinetoplast DNA network of the trypanosome." The EMBO Journal. Vol. 21, no. 18. (Jan 2002): 4998-5005. (Published). 16. Drew ME, Morris JC, Wang Z, Wells L, Sanchez M, Landfear SM, Englund PT. "The adenosine analog tubercidin inhibits glycolysis in Trypanosoma brucei as revealed by an RNA interference library." The Journal of Biological Chemistry. Vol. 278, no. 47. (Jan 2003): 46596-600. 17. Liu J, Gluzman IY, Drew ME, Goldberg DE. "The role of Plasmodium falciparum food vacuole plasmepsins." The Journal of Biological Chemistry. Vol. 280, no. 2. (Jan 2005): 1432-7. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

18. Motyka SA, Drew ME, Yildirir G, Englund PT. "Overexpression of a cytochrome b5 reductase- like protein causes kinetoplast DNA loss in Trypanosoma brucei." The Journal of Biological Chemistry. Vol. 281, no. 27. (Jan 2006): 18499-506. 19. Drew ME, Banerjee R, Uffman EW, Gilbertson S, Rosenthal PJ, Goldberg DE. "Plasmodium food vacuole plasmepsins are activated by falcipains." The Journal of Biological Chemistry. Vol. 283, no. 19. (Jan 2008): 12870-6. 20. Bahar M, Deng Y, Zhu X, He S, Pandharkar T, Drew ME, Navarro-Vázquez A, Anklin C, Gil RR, Doskotch RW, Werbovetz KA, Kinghorn AD. "Potent antiprotozoal activity of a novel semi- synthetic berberine derivative." Bioorganic and Medicinal Chemistry Letters. Vol. 9, no. 21. (May 2011): 2606-2610.

236 Mark E. Drew, PhD 6

21. Abandeh FI, Drew ME, Sopirala MM. "Carbapenem-Hydrolyzing Gram-Negative Bacteria: Current Options for Treatment and Review of Drugs in Development." Recent Patents on Anti- Infective Drug Discovery. (Jan 2012): 22. Klionsky DJ, Abdalla FC, Drew ME, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012 Apr;8(4):445-544. 23. He S, Dayton A, Kuppusamy P, Werbovetz KA, Drew ME. Induction of oxidative stress in Trypanosoma brucei by the antitrypanosomal dihydroquinoline OSU-40. Antimicrobial Agents and Chemotherapy. 2012 May;56(5):2428-34. 24. Harris, MT, Walker, DM, Drew, ME, Mitchell, WG, Dao, K, Schroeder, CE, Flaherty, DP, Weiner, WS, Golden, JE, Morris, JC. "Interrogating a Hexokinase-Selected Small-Molecule Library for Inhibitors of Plasmodium falciparum Hexokinase." Antimicrobial Agents and Chemotherapy. Vol. 57, (May 2013): 3731-7. 25. Endeshaw M, Zhu X, He S, Pandharkar T, Cason E, Mahasenan KV, Agarwal H, Li C, Munde M, Wilson WD, Bahar M, Doskotch RW, Kinghorn AD, Kaiser M, Brun R, Drew ME, Werbovetz KA."8,8- Dialkyldihydroberberines with Potent Antiprotozoal Activity." Journal of Natural Products. Vol. 76, no. 3. (Mar 2013): 311-315. 26. Walker DM, Mahfooz N, Kemme KA, Patel VC, Spangler M, Drew ME. "Plasmodium falciparum Erythrocytic Stage Parasites Require the Putative Autophagy Protein PfAtg7 for Normal Growth." PLoS ONE. Vol. 8, (Jun 2013) 27. Walker DM, Oghumu S, Gupta G, McGwire BS, Drew ME, Satoskar AR. “Mechanisms of cellular invasion by intracellular parasites” Cellular and Molecular Life Sciences. Vol. 71, no. 7 (April 2014):1245-63. 28. Davis MI, Patrick S, Blanding WM, Dwivedi V, Suryadi J, Golden JE, Coussens NP, Shen M, Hall MD, Boxer M, Sharlow ER, Drew ME, Morris JC. “Identification of Novel Plasmodium falciparum Hexokinase Inhibitors with Anti-parasitic Activity” Antimicrobial Agents and Chemotherapy. Vol. 60, no. 11 (Sept 2016): 6023-33.

Chapters in Edited Books 1. Mark E. Drew, Shawn A. Motyka, James C. Morris, Zefeng Wang, Paul T. Englund. "Inducible RNAi as a forward genetic tool in Trypanosoma brucei." In RNA Interference Technology. Edited by K. Appasani. 247- 256. Cambridge: Cambridge University Press, 2005. 2. James C. Morris, Zefeng Wang, Shawn A. Motyka, Mark E. Drew, Paul T. Englund. "An RNAi- based genomic library for forward genetics in the African trypanosome." In Gene Silencing by RNA Interference: Technology and Application. Edited by M. Sohail. Boca Raton: CRC Press, 2004.

Funding

Current Funding Support

OSU Public Health Preparedness for Infectious Diseases (PHPID) 2008-present PHPID provides 100% of salary support

Department of Microbial Infection and Immunity, OSU College of Medicine 2013-present MI&I provides partial Research Assistant salary support

Division of Medicinal Chemistry and Pharmacognosy, OSU College of Pharmacy 2013-present Division provides $5,000/year toward lab expenses Grant Funding Funded Grant Support Mark E. Drew, PhD 7

Werbovetz K, Drew M (co-investigators) “Antiparasitic Berberine Derivatives – Synthesis, Evaluation, and Mechanistic Studies” 10/2010 – 10/2012 OSU PHPID Pilot Grant, Total Award Amount - $100,000

2014/09/17-2016/08/31 1R03HD081723-01, National Institute Of Child Health & Human Development (NICHD) DREW, ME (MULTI-PI, 3% salary recovery) “Probe identification by HTS for Plasmodium Falciparum Hexokinase”

2015/11/01-2017/11/01 Public Health Preparedness for Infectious Disease – Ohio State University DREW, ME (MULTI-PI). “Identification of new transmission blocking drugs for malaria”

Pending

In Preparation

Drew, M (MULTI-PI W/Clemson University, salary recovery TBD), “Hit-to-lead development of chemical probes that inhibit Plasmodium falciparum hexokinase as lead therapeutics), Drug Discovery and Mechanisms of Antimicrobial Resistance Study Section, expected submission date, October 2018

Not Funded

Drew, M (MULTI-PI W/Clemson University, salary recovery TBD), “Hit-to-lead development of chemical probes that inhibit Plasmodium falciparum hexokinase as lead therapeutics”, National Institute Of Child Health & Human Development (NICHD), Submitted June 6, 2017, impact score: 56, 29th percentile

DREW, M (CO-I) and Badu-Tawiah, A. “Malaria Management through an On-demand Diagnostic Approach using Novel Ionic Probes”, NIH Review Panel: Enabling Bioanalytical and Imaging Technologies, NIH R01-A1, Submitted December 2016

DREW, M (CO-I) and Badu-Tawiah, A. “Malaria Management through an On-demand Diagnostic Approach using Novel Ionic Probes”, NIH Review Panel: Enabling Bioanalytical and Imaging Technologies, NIH R01, 2016

Drew, M (PI). “Exploring a non-enzymatic role for Plasmodium falciparum hexokinase” NIH Review Panel: Pathogenic Eukaryotes, NIH R21, 2016

Drew, M (CO-I) and Subrimaniam, V. “Continuous, non-invasive detection for a deadly form of malaria: Plasmodium Falciparum” Explorations NIH National Center for Accelerated Innovations (LOI selected for full submission, full application not selected), 2016

Drew M (CO-I), Subrimaniam V “A Non-Invasive Method for Detection of Malaria Parasites.” Gates Grand Challenges Explorations. 2015

Werbovetz K, Drew M, Green-Church K, Gunn J, Kwiek J, Li C, Rappleye C, Wozniak D (co- investigators) “Kinome Targets in Biofilm-forming Prokaryotes and Drug-resistant Eukaryotes.” NIH, R01. 2012.

Drew M. (PI) “ATG8 and its activation pathway in Plasmodium falciparum erythrocytic stages.” NIH RO1-A1, 2012. Mark E. Drew, PhD 8

Werbovetz K, Drew M. (co-investigators) “Potent antiprotozoal berberine derivatives-SAR and RNAi studies.” NIH R21. 2011.

Drew M. (PI) “ATG8 and its activation pathway in Plasmodium falciparum erythrocytic stages.” NIH. 2010 (unscored)

Christensen K, Drew M, Morris J, Paul K, Marcus K, Temesvari L, Smith K. (co-investigators) “Parallel Multi-Pathogen Diagnosis Using Capillary-Channeled Polymers.” Bill and Melinda Gates Foundation. 2010.

Drew M (CO-I), Wang Z “A genetic approach to characterize secreted antigens in human malaria parasites.” Bill and Melinda Gates Foundation. 2009.

Patents

International Patent Application. “Non-invasive method for detecting a deadly form of malaria: Plasmodium falciparum” Vishwanath V. Subramaniam, Mark Drew, Brad Smith, and Joseph West (11/9/2015, Provisional, Patent Pending, Application # PCT/US16/61019)

U.S. Patent Application. “Non-invasive method for detecting a deadly form of malaria: Plasmodium falciparum”, Vishwanath V. Subramaniam, Mark Drew, Brad Smith, and Joseph West (11/9/2015, Provisional, Patent Pending; Non-Provisional filed 11/5/2016)

U. S. Patent Application. “Use of Sesquiterpens and their Analogs as Green Insecticides for Controlling Disease Vectors and Plant Pests”, Piermarini P, Rakotondriabe L, and Drew M (4/6/2016, Provisional, Patent Pending).

U.S. Patent Application. “8,8-dialkyldihydroberberines with potent antiprotozoal activity”, filed 2011 by Douglas A Kinghorn, Karl Werbovetz, Mark Drew. (Provisional, expired) Mark E. Drew, PhD 9

Invited Presentations 1. “Characterization of essential proteases involved in nutrient acquisition and protein turnover in the malarial parasite, Plasmodium falciparum.” University of Michigan. Ann Arbor, MI. Mar 2009.

2. “Proteases as therapeutic drug targets in the human malarial parasite Plasmodium falciparum.” The Research Institute, Nationwide Children’s Hospital, Columbus, OH. Aug 2009.

3. “Proteases as therapeutic drug targets in the human malarial parasite Plasmodium falciparum.” Clemson University, Clemson, SC. Nov 2009.

4. “The protein catabolic pathways of Plasmodium falciparum as antimalarial drug targets.” Case Western Reserve University. Cleveland, OH. Oct 2010.

5. “The protein catabolic pathways of Plasmodium falciparum as antimalarial drug targets.” Millennium Pharmaceuticals, Boston, MA, Oct 2011.

6. “The autophagy pathway of Plasmodium falciparum as an antimalarial drug target.” University of Washington, WA. May 2013

7. “Finding the Right Gear: Shifting Between Forward And Reverse Efforts in Antimalarial Drug Discovery.” The Infectious Disease Research Institute. Seattle, WA, March 2015 Murugesan Rajaram Ph.D Department of Microbial Infection and Immunity, The Ohio State University 460 West 12th Ave, Room 712 Biomedical Research Tower Columbus, OH 43210 [email protected]

Current Employment

2012 -Present Research Assistant Professor

Education

1995-2001 Ph.D., University of Madras, Chennai, India 1993-1995 M.Phill, Madurai Kamaraj University, India 1991-1993 M.Sc., Madurai Kamaraj University, India 1987-1991 B.Sc., Madurai Kamaraj University, India

Research Experience

2008- Research Scientist, Center for Microbial Interface Biology, 2012 Department of Internal Medicine, College of Medicine, The Ohio State University. 2004- Research Associate 2 2008 Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University. 2002- Post-Doctoral Researcher, Department of Microbiology, The Ohio State University. 2004 2000- Post-Doctoral Researcher 2001 Institute De Molecular Biology & genetics, University of Paris XI, France.

Funding Active: R01 AI146252-01 Murugesan Rajaram (PI), $2,270,838.00 07-01-2019 to 06-30-2024 Molecular Mechanism of cardiac inflammation and dysfunction in Pseudomonas aeruginosa infection. R21 AI146690-01 Murugesan Rajaram (MPI), $429,000.00 06-01-2019 to 05-30-2021 Mechanisms of Cardioprotection by the Innate Immune System during Influenza Virus Infections. Murugesan Rajaram – Contact PI. Sponsored project: Oxford Immunotec, UK Dr. Shu Wang and Murugesan VS Rajaram (MPI) 01-12-2020 to 07-12-2020 Cost: $56,500.00 Clinical Concordance evaluation of the T-SPOT®.TB assay performance using positive selection with magnetic beads and density gradient cell isolation methods

Funded Projects as Co-Investigator: P01 AG044298 Joanne Turner (PI); Murugesan Rajaram (Co-I, Project 2) 03/15/2016 to 02/28/2021 Exploring the impact of inflammaging on immune function during Mycobacterium tuberculosis infection. R01 AI121196 Jian Zhang (PI); Murugesan Rajaram (Co-I) 03/15/2016 to 02/28/2021 Role of Protein Ubiquitination in Sepsis

Pending Research grants: 1. R21 AI142430-01A1 Murugesan Rajaram (PI) Role of the macrophage P-glycoprotein in enhancing emergence of antibiotic resistant Mycobacterium tuberculosis. Received 40 Impact score. 2. R21 AI140015-01- Murugesan Rajaram (PI) The Role of HSF1 in regulating macrophage functions during Mycobacterium tuberculosis infection. Resubmission process. Completed: 1. New Investigator Grant Murugesan Rajaram (PI) 11/01/2016 to 10/30/2018 Role of miR155 and HSF-1 on cardiac inflammation and dysfunction in sepsis. The Ohio State University.

2. R01 AI059639 Schlesinger (PI); Murugesan Rajaram (Co-I) 08/01/2012 to 07/31/2017 TB and Innate Immune Regulation of Lung Macrophages.

3. Sponsored project: Oxford Immunotec, UK Dr. Shu Wang and Murugesan VS Rajaram (MPI) 3-12-2018 to 09-11-2018 Cost: $43,887.00 Clinical Concordance evaluation of the T-SPOT®.TB assay performance using positive selection with magnetic beads and density gradient cell isolation methods

4. The American Heart Association, 17GRNT33420101, Murugesan Rajaram (PI), $144,000.00, 2017 to 2019. Effect of miR155 and HSF-1 on cardiac dysfunction during bacterial infection (No Cost extension).

5. AAI Carriers-In-Immunology Fellowship, Murugesan Rajaram (PI), $56,000.00 2018-2019

Awards and Honors

2020 Fellow of American heart Association- BCVS 2019 American Association of Immunologist Early Career Faculty Travel Award 2017 American Association of Immunologist Early Career Faculty Travel Award 2016 American Association of Immunologist Early Career Faculty Travel Award 2015 American Association of Immunologist Early Career Faculty Travel Award 2014 CMIB 2014 Young Faculty Mentor Award, Department of Internal medicine, OSU 2014 American Association of Immunologist Early Career Faculty Travel Award 2012 Second Prize: Poster presentation, 7th Annual Davis Heart and Lung Research Institute Research Day, 10-09-2012, Category- Research Scientist, College of Medicine, OSU. 2011 First Prize: Poster Presentation, 6th Annual Davis Heart and Lung Research Institute Research Day, Category- Research Scientist, College of Medicine, OSU. 2010 First Prize: Poster Presentation, 5th Annual Davis Heart and Lung Research Institute Research Day, Category- Research Scientist, College of Medicine, OSU. 1998- Senior Research Associate Fellowship, Council of Scientific and Industrial Research, 2000 Government of India, India 1996- Junior Research 1995-1998 Department of Science and Technology, India. 1998 Invited Talks 1. Invited Talk in University of Tennessee Health Science Center, “ Pneumonia and Cardiac dysfunction” March 5th, 2020. 2. Invited Talk in Publish Health Research Institute, New Jersey School of Medicine, “ Microbial Infections and Cardiac dysfunction” February 24th, 2020. 3. Invited Talk in International Symposia, Advances in Immunology and Theraphy of Infectious Diseases. Havana, Cuba. October 29-31st, 2019. Title: Bacterial Infection and cardiac dysfunction. 4. Invited talk in International Emerging Infectious Disease Conference in Mumbai, India December 2-4, 2019. Title: New and Emerging Infections-Bio-terrorism and Public Health 5. Abstract is selected for Oral Presentation in CMIB- Research Retreat-2010, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210. 6. Oral Presentation in American Association of Immunologist, Annual meeting held at New Orleans, May 8th to 12th, 2015. Title: FcRγ-chain, Grb2 and SHP-1 are involved in mannose receptor (CD206)-mediated signaling in relation to tuberculosis.

7. Oral Presentation in American Association of Immunologist Annual meeting held at Seattle, WA, May 13 to 17th, 2016. Title: Cardiac dysfunction during Francisella novicida bacterial infection; an instructive sepsis model to study heart function.

Professional Memberships 2008-present The American Association of Immunologist 2012-present International Endotoxin & Innate Immunity Society 2013-present The American Heart Association 2017-present American Society of Microbiology

Teaching Experience

20 Faculty Lecturer: IMM 7010; Cellular and Molecular Immunology (3 lecturers) 20 20 Faculty Lecturer: BSGP 7070 Fundamentals in Grant writing course for Graduate s 19 Students (10 x 3 h Lectures), College of Medicine, The Ohio State University. 20 Faculty Lecturer: IMM 7010; Cellular and Molecular Immunology (4 lecturers) 19 20 Faculty Lecturer: IMM 7010; Cellular and Molecular Immunology (4 lecturers) 18 20 Faculty Lecturer: IMM 7010; Cellular and Molecular Immunology (3 lecturers) 17 20 Faculty Lecturer: IMM 7010; Cellular and Molecular Immunology (3 lecturers) 16

Academic Services Scientific Sessions Co-Chair: 2015 Session Chair for Microbial, Parasitic and Fungal Immunology II American Association of Immunology, 2015 Annual meeting held at New Orleans, May 2015. 2017 Session Chair for Microbial, Parasitic and Fungal Immunology II American Association of Immunology, Annual meeting, Washington DC, May, 2017. 2018 Session Chair, International Emerging Infectious Disease Conference, India. Editorial Boards: 2013-2018 Editorial Board Member: American Journal of Current Immunology. 2015-present Editorial Board Member: PPAR Research 2019-present Topic Editor The Role of Small RNAs in Host Cell Immune Functions' Regulation. Frontiers in Cellular and Infection Microbiology. AdHoc Reviewer: Journal of Immunology PLoS One Bio-Medical Central Group of Journals Journal of Stem Cell Research & Therapy Nature Scientific Reports PLOS Pathogens Vaccine

Trainees Mentored Post-Doctoral Researchers and graduate students 1. Michelle N. Brooks- Graduate Student, 2008- 2011, Role Co-Advisor 2. Bin Ni- MD, Ph.D student, 2008- 2012, Role Co-Advisor 3. Jessica Morris- Research Associate, 2008-2009, Role Co-Advisor 4. Bret Betz- MD Student Summer Training, 2009, Role Co-Advisor 5. Huy Nguyen- Research Associate, 2009-2011, Role Co-Advisor 6. Daniel House – Graduate Student, 2011- 2013, Role Co-Advisor 7. Alaric W. D’Souza- MSTP Summer Training Student, 2012, Advisor 8. Ky Hoang- Post Doctoral Researcher, 2013- 2018, Mentor and Advisor 9. Claire Dood- Graduate student, 2013- 2016, Role Co-Advisor 10. Eusondia Arnett- Post Doctoral Researcher, 2014- 2016, present 11. Abigail Gerberick- Undergraduate Student, 2015-2017, Role Advisor 12. Sumiran Mehta- Undergraduate Student, 2015-2017, Role Advisor 13. Sonali Patel- Undergraduate student 2018- present- Role Advisor 14. Cameron Kashef- Undergraduate student 2018- present- Role Advisor 15. Noushin Saljoughian- Post Doctoral Researcher- 2018- Present, Role Advisor 16. Naresh Kumar- Post Doctoral Researcher, 2019- present, Advisor 17. Whitestone Elizabeth- Undergraduate Student, 2019- present, Advisor

Additional Training 2008- 2020 Animal Biosafety level 3/ Select Agent Certification

Publications *Shared 1st Author; $Shared Corresponding Author h-Index:29; i10-Index:38 https://scholar.google.com/citations?user=tpzBYocAAAAJ&hl=en https://www.ncbi.nlm.nih.gov/pubmed/?term=Murugesan+Rajaram

1. Juan Tang, Hui Guo, Qingjun Liu, Guoxin Lin, Rajaram MVS, Amal O. Amer, Brian M. Ahmer, John S. Gunn, Daniel J. Wozniak Lijian Tao, Coppola Vincenzo, Liwen Zhang, Wallace Y. Langdon, Jordi B. Torrelles, and Jian Zhang. Inhibition of Septic Shock by Sequential K63- and K48-linked Ubiquitination of NLRP3 Mediated by E3 Ubiquitin Ligases RNF125 and Cbl-b. Accepted in Journal of Experimental Medicine, 11/18/2019. 2. Lafuse WP, *$Rajaram MVS, Wu Q, Moliva JI, Torrelles JB, Turner J, Schlesinger LS. Identification of an Increased Alveolar Macrophage Subpopulation in Old Mice That Displays Unique Inflammatory Characteristics and Is Permissive to Mycobacterium tuberculosis Infection. J Immunol. 2019 Oct 15; 203(8):2252-2264. doi: 10.4049/jimmunol.1900495. Epub 2019 Sep 11. 3. Qian Wu, Austin Hossfeld, Abigail Gerberick, Noushin Saljoughian , Charu Tiwari, Smriti Mehra, Latha Prabha Ganesan, Daniel J. Wozniak, and Rajaram MVS. Mycobacterium tuberculosis enhances macrophage P-glycoprotein (MDR-1) expression and activity to promote intracellular survival. The Journal of Infectious Disease. J Infect Dis. 2019 Nov 6; 220(12):1989-1998. doi: 10.1093/infdis/jiz405. 4. Adam D. Kenney, Temet M. McMichael, Lizhi Zhang, Lisa Dorn, Qian Wu, Foued Amara, Min Chen, Federica Accornero, Vincenzo Coppola, $Rajaram MVS, and Jacob S. Yount. IFITM3 protects the heart during influenza virus infection. Proc. Natl. Acad. Sci. U S A. 2019 Sep 10; 116(37):18607- 18612. doi: 10.1073/pnas.1900784116. 5. Headley C, Turner J, Rajaram MVS. Aging heart and Infection. Aging (Albany NY). 2019 Jul 25. doi: 10.18632/aging.102128; PMID: 31346150. 6. Moliva JI, Duncan MA, Olmo-Fontánez A, Akhter A, Arnett E, Scordo JM, Ault R, Sasindran SJ, Azad AK, Montoya MJ, Reinhold-Larsson N, Rajaram MVS, Merrit RE, Lafuse WP, Zhang L, Wang SH, Beamer G, Wang Y, Proud K, Maselli DJ, Peters J, Weintraub ST, Turner J, Schlesinger LS, Torrelles JB. The lung mucosa environment in the elderly increases host susceptibility to Mycobacterium tuberculosis infection. J Infect Dis. 2019 Mar 29. pii: jiz138. doi: 10.1093/infdis/jiz138. [Epub ahead of print]; PMID: 30923818. 7. Colwyn A. Headley, Abigail Gerberick, Sumiran Mehta, Qian Wu, Lianbo Yu, Paolo Fadda, Mahmood Khan, Latha Prabha Ganesan, Joanne Turner and Rajaram MVS. Nontuberculous mycobacterium M. avium infection predisposes aged mice to cardiac abnormalities and inflammation. Aging Cell, DOI: 10.1111/acel.12926 8. Makara MA, Curran J, Lubbers ER, Murphy NP, Little SC, Musa H, Smith SA, Unudurthi SD, Rajaram MVS, Janssen PML, Boyden PA, Bradley EA, Hund TJ, Mohler PJ. Novel Mechanistic Roles for Ankyrin-G in Cardiac Remodeling and Heart Failure. JACC Basic Transl Sci. 2018 Nov 12; 3(5):675-689. doi: 10.1016/j.jacbts.2018.07.008. 9. Guirado E, Rajaram MVS, Chawla A, Daigle J, La Perle KM, Arnett E, Turner J, Schlesinger LS. Deletion of PPARγ in lung macrophages provides an immunoprotective response against M. tuberculosis infection in mice. Tuberculosis (Edinb). 2018 Jul; 111: 170-177. doi: 10.1016/j.tube.2018.06.012. Epub 2018 Jun 21. 10. Hoang KV, Rajaram MVS, Curry HM, Gavrilin MA, Wewers MD, Schlesinger LS. Complement Receptor 3-Mediated Inhibition of Inflammasome Priming by Ras GTPase-Activating Protein during Francisella tularensis Phagocytosis by Human Mononuclear Phagocytes. Front Immunol. 2018 Mar 26; 9:561. doi: 10.3389/fimmu.2018.00561. eCollection 2018.PMID:29632532. 11. Garfoot AL, Goughenour KD, Wüthrich M, Rajaram MVS, Schlesinger LS, Klein BS, Rappleye CA. O-Mannosylation of Proteins Enables Histoplasma Yeast Survival at Mammalian Body Temperatures. MBio. 2018 Jan 2; 9(1). pii: e02121-17. doi: 10.1128/mBio.02121-17. PMID: 29295913. 12. $Rajaram MVS, Arnett E, Azad AK, Guirado E, Ni B, Gerberick AD, He LZ, Keler T, Thomas LJ, Lafuse WP, Schlesinger LS. M. tuberculosis-Initiated Human Mannose Receptor Signaling Regulates Macrophage Recognition and Vesicle Trafficking by FcRγ-Chain, Grb2, and SHP-1.Cell Rep. 2017 Oct 3; 21(1):126-140. doi: 10.1016/j.celrep.2017.09.034.PMID:28978467. 13. Michael Makara , Jerry Curran , Sean Little , Hassan Musa , Sakima Smith , Sathya Unudurthi , Rajaram MVS , Paul M.L. Janssen , Elisa Bradley , Thomas J. Hund, Peter J. Mohler. Novel mechanistic roles for ankyrin-G in cardiac remodeling, heart failure, and survival. J Am Heart Assoc. 2016 Sep 12; 5(9). pii: e003820. doi: 10.1161/JAHA.116.003820. PMID: 27620887. 14. Dodd CE, Pyle CJ, Glowinski R, Rajaram MVS, Schlesinger LS. CD36-Mediated Uptake of Surfactant Lipids by Human Macrophages Promotes Intracellular Growth of Mycobacterium tuberculosis. The Journal of Immunology 197 (12), 4727-4735. PMID: 27913648. 15. Cope FO, Abbruzzese B, Sanders J, Metz W, Sturms K, Ralph D, Blue M, Zhang J, Bracci P, Bshara W, Behr S, Maurer T, Williams K, Walker J, Beverly A, Blay B, Damughatla A, Larsen M, Mountain C, Neylon E, Parcel K, Raghuraman K, Ricks K, Rose L, Sivakumar A, Streck N, Wang B, Wasco C, Williams A, Schlesinger LS, Azad A, Rajaram MVS, Jarjour W, Young N, Rosol T, McGrath. Nucl Med Biol. 2016 Dec; 43(12):837. doi: 10.1016/j.nucmedbio.2016.10.001. PMID: 27866590 16. Makara MA, Hoang KV, Ganesan LP, Crouser ED, Gunn JS, Turner J, Schlesinger LS, Mohler PJ, Rajaram MVS. Cardiac Electrical and Structural Changes during Bacterial Infection: An Instructive Model to Study Cardiac Dysfunction in Sepsis. J Am Heart Assoc. 2016 Sep 12; 5(9). pii: e003820. doi: 10.1161/JAHA.116.003820.PMID:27620887. 17. Ganesan LP, Mates JM, Cheplowitz AM, Avila CL, Zimmerer JM, Yao Z, Maiseyeu A, Rajaram MVS, Robinson JM, Anderson CL. Scavenger receptor B1, the HDL receptor, is expressed abundantly in liver sinusoidal endothelial cells. Sci Rep. 2016 Feb 11; 6:20646. Doi 10.1038/srep20646. PMID: 26865459. 18. Yun Xiao, Juan Tang, Hui Guo, Yixia Zhao, Rong Tang, Song Ouyang, Qiuming Zeng, Chad Rappleye, Rajaram MVS, Larry S. Schlesinger, Lijian Tao, Gordon D. Brown, Wallace Y. Langdon, Belinda T. Li , and Jian Zhang. Targeting CBLB as a Potential Therapeutic Approach for Disseminated Candidiasis. Nat Med. 2016 Aug; 22(8):906-14. doi: 10.1038/nm.4141. Epub 2016 Jul 18. PMID: 27428899. 19. Yao Z, Mates JM, Cheplowitz AM, Hammer LP, Maiseyeu A, Phillips GS, Wewers MD, Rajaram MVS, Robinson JM, Anderson CL, Ganesan LP. Blood-Borne Lipopolysaccharide Is Rapidly Eliminated by Liver Sinusoidal Endothelial Cells via High-Density Lipoprotein. J Immunol. 2016 Sep 15; 197(6):2390-9. doi: 10.4049/jimmunol.1600702. Epub 2016 Aug 17.PMID:27534554. 20. Abul K. Azad, *Rajaram MVS, Wendy L. Metz, Frederick O. Cope, Michael S. Blue, David R Vera, and Larry S. Schlesinger. 99mTc-tilmanocept, a new tracer for cancer sentinel lymph nodes, binds to the macrophage mannose receptor (CD206). 2015 Sep 1; 195(5):2019-29: PMID 26202986. 21. Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Seminars in Immunology. 26:471, 2014.PMCID: 25453226. 22. Ni B, Rajaram MVS, Lafuse WO, Landes MB, Schlesinger LS. Mycobacterium tuberculosis decreases human macrophages IFN-g responsiveness through miR-132 and miR-26a. J. Immunology, 2014 Nov 1: 193 (9):4537-47: doi: 10.4049/jimmunol.1400124. Epub 2014 Sep 24. 23. Landes MB, Rajaram MVS, Nguyen H, Schlesinger LS. Role for NOD2 in Mycobacterium tuberculosis-induced iNOS expression and NO production in human macrophages. J Leukoc Biol. 2015 Mar 23. pii: jlb.3A1114-557R. [Epub ahead of print] PMID: 25801769. 24. Chesarino NM, Hach JC, Chen JL, Zaro BW, Rajaram MVS, Turner J, Schlesinger LS, Pratt MR, Hang HC, Yount JS. Chemoproteomics reveals Toll-like receptor fatty acylation. BMC Biol. 2014 Nov 5; 12(1):91. doi: 10.1186/s12915-014-0091-3. PMID: 25371237. 25. Hoang KV, Borteh HM, Rajaram MVS, Peine KJ, Curry H, Collier MA, Homsy ML, Bachelder EM, Gunn JS, Schlesinger LS, Ainslie KM. Acetalated dextran encapsulated AR-12 as a host-directed therapy to control Salmonella infection. Int J Pharm. 2014 Dec 30; 477(1-2):334-43. doi: 10.1016/j.ijpharm.2014.10.022. Epub 2014 Oct 22. PMID: 25447826. 26. Moliva JI, *Rajaram MVS, Sidiki S, Sasindran SJ, Guirado E, Pan XJ, Wang SH, Ross P Jr, Lafuse WP, Schlesinger LS, Turner J, Torrelles JB. Molecular composition of the alveolar lining fluid in the aging lung. Age (Dordr). 2014 Mar 3. [Epub ahead of print] PMID: 24584696. 27. Nicholas A. Young, Lai-Chu Wu, Craig J. Burd, Alexandra Friedman, Benjamin Kaffenberger, Rajaram MVS, Larry S. Schlesinger, Hayley James, Margaret A. Shupnik, and Wael N. Jarjour. Estrogen Modulation of Endosome-Associated Toll-Like Receptor 8: An IFNα-Independent Mechanism of Sex-Bias in Systemic Lupus Erythematosus. Accepted in Clinical Immunology. 2014 Mar; 151(1):66-77. doi: 10.1016/j.clim.2014.01.006. Epub 2014 Jan 24. PMID: 24525049. 28. Abul K. Azad, Rajaram MVS, Larry S. Schlesinger (2014) Exploitation of the macrophage mannose receptor (CD206) in infectious disease diagnostics and therapeutics. Journal of Cytology and Molecular Biology, volume 1 (1), p 1-5. PMID: 24672807. 29. Hansen JM, Golchin SA, Veyrier FJ, Domenech P, Boneca IG, Azad AK, Rajaram MVS, Schlesinger LS, Divangahi M, Reed MB, Behr MA. N-glycolylated peptidoglycan contributes to the immunogenicity but not pathogenicity of Mycobacterium tuberculosis. J Infect Dis. 2014 Apr 1; 209(7):1045-54. doi: 10.1093/infdis/jit622. Epub 2013 Nov 21. PMID: 24265438. 30. Nrusingh P. Mohapatra, Shilpa Soni, Rajaram MVS, Kristi L. Strandberg, John S. Gunn. (2013). Type-A Francisella tularensis acid phosphatases contribute to pathogenesis. PLOS ONE, Feb- 15,10.1371/journal.pone.0056834. 31. Shipan Dai, *Rajaram MVS, Heather M. Curry, Rachel Leander and Larry S. Schlesinger. Fine tuning inflammation at the front door: Macrophage Complement Receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis. PLoS Pathog. 2013 Jan; 9(1):e1003114. doi: 10.1371/journal.ppat.1003114. Epub 2013 Jan 24. PMID: 23359218. 32. Nicholas A. Young, Alexandra K. Friedman, Benjamin Kaffenberger, Rajaram MVS, Daniel J. Birmingham, Larry S. Schlesinger, Lai-Chu Wu, and Wael N. Jarjour. Novel estrogen target gene ZAS3 is overexpressed in systemic lupus erythematosus. Mol Immunol. 2013 May; 54(1):23-31. doi: 10.1016/j.molimm.2012.10.026. Epub 2012 Nov 22. PMID: 2317882. 33. Huy A. Nguyen, *Rajaram MVS, Douglas A. Meyer, and Larry S. Schlesinger. Pulmonary surfactant protein-A and surfactant lipids up-regulate IRAK-M, a negative regulator of TLR- mediated inflammation in human macrophages. Am J Physiol Lung Cell Mol Physiol. 2012 Aug 10.[Epub ahead of print]. PMID: 22886503. 34. Cremer TJ, Fatehchand K, Shah P, Gillette D, Patel H, Marsh RL, Besecker BY, Rajaram MVS, Cormet- Boyaka E, Kanneganti TD, Schlesinger LS, Butchar JP, Tridandapani S. MiR-155 Induction by Microbes/Microbial Ligands Requires NF-κB-Dependent de novo Protein Synthesis. Front Cell Infect Microbiol. 2012; 2:73. Epub 2012 Jun 1. PMID: 22919664. 35. Rajaram MVS, Jessica Morris, Michelle N. Brooks, Jordi B. Torrelles and Larry S. Schlesinger. M. tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-Activated Protein Kinase 2 (MK2) and miR125b. Proc Natl Acad Sci U S A. 2011 Oct 18; 108(42):17408-13. 36. Torrelles JB, Sieling PA, Arcos J, Knaup R, Bartling C, Rajaram MVS, Stenger S, Modlin RL, Schlesinger LS. Structural differences in lipomannans from pathogenic and nonpathogenic mycobacteria that impact CD1b-restricted T cell responses. J Biol Chem. 2011 Oct 14; 286(41):35438-46. Epub 2011 Aug 22. PMCID: PMC3195570. 37. Bret E. Betz, Abul K. Azad, Jessica D. Morris, Rajaram MVS, and Larry S. Schlesinger. B-glucans inhibit intracellular growth of Mycobacterium bovis BCG but not virulent Mycobacterium tuberculosis in human macrophages. Microbial Pathogenesis, 2011 Oct; 51(4):233-42. Epub 2011 Jul 5. 38. Michelle N. Brooks, Rajaram MVS, Abul K. Azad, Martin A. Valdivia-Arenas, and Larry S. Schlesinger. Intracellular pattern recognition receptor NOD2 plays a role in controlling the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis in human macrophages. Cellular Microbiology 2011 Mar; 13(3):402-18. 39. Rajaram MVS, Michelle N. Brooks, Jessica Morris, Jordi B. Torrelles, Abul K. Azad and Larry S. Schlesinger. Mycobacterium tuberculosis activates of human macrophage peroxisome proliferator- activator gamma linking mannose receptor recognition to regulation of immune responses. Journal of Immunology, 2010 July 15: 185(2): 929-42. 40. Nrusingh P. Mohapatra, Shilpa Soni, Rajaram MVS, My-Chan Dang, Tom J. Reilly, Jamel El benna, Larry S. Schlesinger and John S. Gunn. Francisella acid phosphatases inactivate NADPH oxidase complex components in human phagocytes by dephosphorylation. Jouranal of Immunology 2010 May 1; 184(9):5141-50. 41. Lindsay Sweet, Prachi P.Singh, Abul K.Azad, Rajaram MVS, Larry S. Schlesinger and Jeffrey Schorey. Mannose receptor- dependent delay in phagosome maturation by Mycobacterium avium glycopeptidolipids. Infection and Immunity 2010 Jan; 78(1):518-26. 42. Rajaram MVS, Jonathan P. Butchar, Kishore V Parsa, Amal Amer, Larry S. Schlesinger and Susheela Tridandapani. Akt and SHIP modulate Francisella escape from the phagosome and the induction of the Fas- mediated death pathway. PLoS ONE, 2009 Nov 20: 4(11): e 7919. 43. Kishore V. L. Parsa, Jonathan P. Butchar, Rajaram MVS, John S. Gunn, Larry S. Schlesinger, Susheela Tridandapani. Francisella gains a survival advantage within mononuclear phagocytes by suppressing host IFN gamma response. Mol Immunol. 2008 Jul; 45(12):3428-37. 44. Kishore V. L. Parsa, Jonathan P. Butchar, Rajaram MVS, Larry S. Schlesinger, Susheela Tridandapani. Syk- dependent Erk activation promotes the phagocytosis of Francisella. Mol Immunol. 2008 May; 45(10):3012-21. 45. Rajaram MVS, Latha P. Ganesan, Kishore V.Parsa, Jonathan P. Butchar, John S. Gunn and Susheela Tridandapani. Protein kinase B (Akt) modulates macrophage inflammatory response to Francisella tularensis infection, and confers a survival advantage in mice. Journal of Immunology; 2006, 177(9):6317- 24. 46. Jonathan P. Butchar, Rajaram MVS, Latha P. Ganesan, Kishore V. L,Parsa, Corey D. Clay, Larry S. Schlesinger, Susheela Tridandapani. Francisella tularensis induces Interleukin-23 production in human monocytes. Journal of Immunology. 2007, 178(7):4445-54. 47. Kishore V. L. Parsa, Latha P. Ganesan, Rajaram MVS, Mikhail A. Gavrilin, Mark D. Wewers, Larry S. Schlesinger, John S. Gunn and Susheela Tridandapani. Macrophage pro-inflammatory response to Francisella novicida infection is regulated by the SH2 domain-containing inositol 5’ phosphatase SHIP. PLoS pathogens 2006 2(7):e71.DOI:10.1371/journal.ppat.0020071. 48. Weir NM, Selvendiran,K., Kutala VK., Tong L, Vishwanath S., Rajaram MVS, Tridandapani S. Anant S. Kuppusamy P. Curcumin Induces G(2)/M Arrest and Apoptosis in Cisplatin-Resistant Human Ovarian Cancer Cells by Modulating Akt and p38 MAPK. Cancer Biol. Ther. 2007 Feb; 6(2):178-84. 49. Rajaram MVS and N.Anand 1996. Nitrogen fixation at night in nature. Current Science 70:119-120.

Book Chapter Tracy K. Carlson, Michelle N. Brooks, Lisa Henning, Douglas A. Meyer, Murugesan V.S. Rajaram, and Larry S. Schlesinger (2009). Pulmonary innate immunity: soluble and cellular host defenses of the lung. In. Regulation of Innate Immune Functions (Eds. Clay B.Marsh, Susheela Tridandapani and Melissa G. Piper) TransworldResearch Network, ISBN: 978-81-7895-492-9. pp 165-211.

Selected National and International Meeting Abstracts (from 125):

1. Matthew Pestrak, Nathan Murphy, Qian Wu, Ellen Lubbers, Dellos-Nolan, Lianbo Yu, William P. Lafuse, Latha P. Ganesan, Peter J. Mohler, Daniel Wozniak, Murugesan VS. Rajaram, 2019. MicroRNA-155 regulates the phenotype of cardiac macrophages and heart function during Pseudomonas aeruginosa infection. 2. Noushin Saljoughian, Qian Wu, Cameron Kashef, Sonali Patel, Latha P. Ganesan William P Lafuse and Murugesan V.S Rajaram, 2019. Characterization of cardiac immune cells in aged mice and their role in cardiac dysfunction during bacterial infection. Basic Cardio Vascular Sciences, AHA Meeting, August 29th-Sep2nd, Boston, MA, USA. 3. Qian Wu, Austin Hossfeld, Abigail Gerberick, Noushin Saljoughian, Charu Tiwari, Latha Prabha Ganesan, Daniel Wozniak, and Murugesan VS Rajaram, 2019. Mycobacterium tuberculosis enhances macrophage P-glycoprotein (MDR-1) expression and activity to promote intracellular survival. American Association of Immunologist Annual Meeting-2019, San Diego, CA. 4. William P Lafuse, Murugesan VS Rajaram, Qian Wu, Juan I Moliva, Jordi B Torrelles, Joanne Turner, Larry S Schlesinger. Phenotypic analysis of alveolar macrophage populations in old mice and their response to Mycobacterium tuberculosis infection. American Association of Immunologist Annual Meeting-2019, San Diego, CA. 5. Zhili Yao, Mustafa Al-Rahem, Thushara Thomas, Mark D. Wewers, Clark L. Anderson, and Murugesan V.S. Rajaram and Latha P. Ganesan (2018). Lipopolysaccharide clearance function of liver sinusoidal endothelial cells controls inflammation: Relevance to Sepsis. Shock Society meeting, Phoenix, AZ, USA. 6. Murugesan VS Rajaram, Colwyn Headley, Nathaniel Murphy, Peter J. Mohler and Joanne Turner (2017). Influence of age on mycobacterial myocarditis and heart failure. National Institute of Aging Meeting- 2017, Bethesda, Maryland, USA. 7. William P Lafuse, Murugesan VS Rajaram, Heather M Curry, Connor Nealer, Cynthia H Canan, Juan I Moliva, Jordi B Torrelles, Joanne Turner, and Larry S Schlesinger (2017). Influence of aging on the phenotype and function of alveolar macrophages. American Association of Immunologist Annual Meeting- 2017, Washington DC, USA. 8. Zhili Yao, Mustafa Al-Rahem, Thushara Thomas, Mark D. Wewers, Clark L. Anderson, and Murugesan V.S. Rajaram and Latha P. Ganesan (2018) Lipopolysaccharide clearance function of liver sinusoidal endothelial cells controls inflammation: Relevance to Sepsis. 41st Annual Conference on Shock, Scottsdale, AZ, USA. 9. Latha Prabha Ganesan, Ozan Suer, Zhili Yao, Alana Cheplowitz, Charu Tiwari, Menakshi Bhat, Sueng Chu, Erik Pong, David Szymkowski, Murugesan VS Rajaram, John Robinson, Clark Anderson. (2018). EMBO Workshop – Antibodies and Complement: Effector functions, therapies and technologies, June 28- July1st, 2018, Girona, Spain. 10. Murugesan VS Rajaram, Colwyn Headley, Nathaniel Murphy, Peter J. Mohler and Joanne Turner (2017). Influence of age on mycobacterial myocarditis and heart failure. American Association of Immunologist Annual Meeting-2017, Washington DC, USA. 11. Claire E. Dodd, Murugesan V.S. Rajaram and Larry S. Schlesinger (2015). Role of CD36 in surfactant uptake by human macrophages: Implications for tuberculosis pathogenesis. 22nd Annual Midwest Microbial Pathogenesis Conference, August 28-29, Indianapolish, IN, USA. 12. K. V. Hoang, H. Curry, M. V. S. Rajaram, M. A. Collier, H. Borteh, E. Bachelder, K. Ainslie, L. S. Schlesinger and J. S. Gunn. (2015). Needle-free Delivery of Acetalated Dextran-Encapsulated AR- 12 Protects Mice from Francisella tularensis Lethal Challenge, ICAAC &ICC 2015 meeting, San Diego, CA, September 17-21. 13. Murugesan V.S. Rajaram, Bin Ni, Eusondia Arnett, Abul K Azad and Larry S. Schlesinger (2013). FcRγ- chain, Grb2 and SHP-1 are involved in mannose receptor (CD206)-mediated signaling in relation to tuberculosis. American Association of Immunologist Annual Meeting-2015, New Orleans, LA, USA, May 8- 12, 2015. 14. Arnett E, Murugesan VS Rajaram, Schlesinger LS (2015). Identification of novel PPARγ effectors that regulate the host response to Mycobacterium tuberculosis in human macrophages. Phagocytes, Gordon Research Conference, Waterville Valley, NH, May 31-June 5. 15. Murugesan V.S. Rajaram, William Lafuse, Rachel L. Gearinger, Aleksandra I. Adamovich, Russell Ault, and Larry S. Schlesinger (2015). Molecular mechanisms of miR125b expression in human macrophages during mycobacterial infection. Keystone Symposia – Host response in tuberculosis, Jan22-27:2015, Santa Fe, New Mexico, USA. 16. Bin Ni, C. Torres, M. V. S. Rajaram and L. S. Schlesinger. Mycobacterium tuberculosis infection decreases macrophage Tpl2 protein levels over time which alters response to TLR agonists. American Association of Immunologist Annual Meeting-2015, New Orleans, LA, USA, May 8- 12, 2015. 17. Arnett E, Murugesan VS Rajaram, Joshua Keaton, and Larry S Schlesinger. Elucidate the PPAR effectors that alter macrophage responses to M. tuberculosis. PHPID Annual Meeting, May 5,2015, The Ohio State University, Columbus, OH, 43210 18. Arnett E, Murugesan VS Rajaram, Schlesinger LS. Identification of novel PPARγ effectors that regulate the host response to Mycobacterium tuberculosis in human macrophages. OSU Center for Microbial Interface Biology Symposium, Columbus, OH, Oct 2014. 19. Arnett E, Murugesan VS Rajaram, Schlesinger LS. Identification of novel PPARγ effectors that regulate the host response to Mycobacterium tuberculosis in human macrophages. OSU Davis Heart and Lung Research Institute Research Day, Columbus, OH, Nov 2014. 20. Claire E Dodd, Murugesan VS Rajaram and Larry S. Schlesinger (2014). Role of CD36 in surfactant catabolism by human macrophages: Implications for tuberculosis pathogenesis. 13th Annual OSUWMC Research Day – April 9th-10th, 2014, BRT, Columbus, OH, 43210. 21. Claire E Dodd, Murugesan VS Rajaram and Larry S Schlesinger (2014). Role of CD36 in surfactant catabolism by human macrophages: Implications for tuberculosis pathogenesis. 21st Annual MMPC - September 12th-14th, 2014, Chicago, IL. 22. Claire E Dodd, Murugesan VS Rajaram and Larry S Schlesinger (2015). Role of CD36 in surfactant catabolism by human macrophages: Implications for tuberculosis. PHPID Research Day, May 5th 2015, BRT, Columbus, OH, 43210. 23. Murugesan V.S. Rajaram, Bin Ni, Tracy Carlson and Larry S. Schlesinger (2013). Mannose receptor (CD- 206)-mediated signaling in human macrophages in the context of tuberculosis. AAI Annual Meeting, 2014, Pittsburg, PA, USA, May-2 to 6-2014. 24. Cynthia Canan, Murugesan V.S.Rajaram and Joanne Turner. Understanding altered toll –like receptor in the elderly. The American Association of Immunologist Annual meeting 2013, Honolulu, Hawaii, May 3-7. 25. Bin Ni, M. V. S. Rajaram, C. Torres, M. B. Landes, L. S. Schlesinger. MicroRNA expression profiling of M. tuberculosis-infected human macrophages: M.tb decreases host IFN-γ -responsiveness through miRNAs. 20th Annual Midwest Microbial Pathogenesis Meeting, Columbus, Ohio, August 2013 26. Ky V. Hoang, Eric Bachelder, Hassan Borteh, Murugesan V. S. Rajaram, Heather Curry, Kristy Ainslie, John S. Gunn, and Larry S. Schlesinger (2013). Use of encapsulated AR-12 as a host directed agent to control intracellular pathogens. 20th Annual Midwest Microbial Pathogenesis Meeting, Columbus, Ohio, August 2013 27. Bin Ni, Murugesan V. S. Rajaram, M. B. Landes, L. S. Schlesinger. MicroRNA expression profiling of M. tuberculosis-infected human macrophages: M.tb decreases host IFN-γ -responsiveness through miRNAs. 2013 National MD/PhD Student Conference, Keystone, Colorado, July 2013. 28. Abul K. Azad, Murugesan V.S. Rajaram, Wendy L. Metz, Frederick O. Cope, Michael S. Blue, David R Vera, and Larry S. Schlesinger (2013). γ-tilmanocept, a new tracer for cancer sentinel lymph nodes, binds to the macrophage mannose receptor (CD206). Gordon Research Conference, June 7th to 11th, 2013, USA. 29. LS Schlesinger, AK Azad, Murugesan VS Rajaram, A Agrawal, BC Abbruzzese, MS Blue, J Shuping, B Potter, WL Metz, and FO Cope (2013). Evaluation of human mannose receptor (CD206) binding of 99mTc- tilmanocept: a novel receptor-targeted lymphatic mapping agent for solid tumors. JOIC, May 27th to 30th, 2013, San Francisco, USA. 30. LS Schlesinger, AK Azad, Murugesan VS Rajaram, A Agrawal, BC Abbruzzese, MS Blue, J Shuping, B Potter, WL Metz, and FO Cope (2013). The CD206 receptor-targeted intraoperative lymphatic mapping agent 99mTc-tilmanocept has significantly higher localization in intraoral squamous cell carcinoma- associated sentinel nodes that are tumor-positive due to the recruitment of tumor proximal CD206- expressing dendritic cells in addition to macrophages. JOIC, May 27th to 30th, 2013, San Francisco, USA. 31. Murugesan V.S. Rajaram, Bin Ni, Tracy Carlson and Larry S. Schlesinger (2013). Mannose receptor (CD- Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

206) mediated signaling in human macrophages in the context of tuberculosis. Keystone Symposia on Molecular and Cellular Biology, March 13-18, 2013 Whistler, BC, Canada.

251 32. Ky V. Hoang, Eric Bachelder, Hassan Borteh, Murugesan V. S. Rajaram, Heather Curry, Kristy Ainslie, John S. Gunn, and Larry S. Schlesinger (2013). Use of encapsulated AR-12 as a host directed agent to control intracellular pathogens. PHPID meeting, June 2013, BRT, Columbus, OH, 43210 33. Danielle House, Murugesan VS Rajaram and Larry S. Schlesinger (2013). Lung surfactant contributes to the unique biologyof macrophages in their environment by up-regulation of negative regulators of immune function: Impact on tuberculosis. PHPID 34. Bin Ni, Murugesan V.S. Rajaram, Michelle B. Landes, Huy A. Nguyen, and Larry S. Schlesinger. MicroRNA expression profiling of Mycobacterium tuberculosis-infected human macrophages: M.tb decreases host IFN-γ-responsiveness through miR-132. Keystone Symposia On Molecular and Cellular Biology, March 13-18, 2013 Whistler, BC, Canada. 35. Danielle House, Murugesan VS Rajaram and Larry S. Schlesinger (2013). Lung surfactant contributes to the unique biologyof macrophages in their environment by up-regulation of negative regulators of immune function: Impact on tuberculosis. 12th Annual OSUMC Research Day- April 10th- 11th, 2013 36. Bin Ni, Murugesan V.S. Rajaram, Michelle B. Landes, Huy A. Nguyen, and Larry S. Schlesinger. MicroRNA expression profiling of Mycobacterium tuberculosis-infected human macrophages: M.tb decreases host IFN-γ-responsiveness through miR-132. 12th Annual OSUMC Research Day- April 10th- 11th, 2013. 37. Chelsea TORRES, Bin NI, Murugesan V.S. RAJARAM and Larry S. SCHLESINGER (2013). Mycobacterium tuberculosis alters macrophage signaling through p120 RAS-GAP modulation via miR132. 12th Annual OSUMC Research Day- April 10th- 11th, 2013. 38. Danielle House, Murugesan VS Rajaram and Larry S. Schlesinger (2013). Lung surfactant contributes to the unique biology of macrophages in their environment by up-regulation of negative regulators of immune function: Impact on tuberculosis. 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210. 39. Bin Ni, Murugesan V.S. Rajaram, Michelle B. Landes, Huy A. Nguyen, and Larry S. Schlesinger. MicroRNA expression profiling of Mycobacterium tuberculosis-infected human macrophages: M.tb decreases host IFN-γ-responsiveness through miR-132. 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210. 40. Abul K. Azad, Murugesan V.S. Rajaram, Wendy L. Metz, Frederick O. Cope, Michael S. Blue, David R Vera, and Larry S. Schlesinger (2013). γ-tilmanocept, a new tracer for cancer sentinel lymph nodes, binds to the macrophage mannose receptor (CD206). 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210. 41. Chelsea TORRES, Bin NI, Murugesan V.S. RAJARAM and Larry S. SCHLESINGER (2013). Mycobacterium tuberculosis alters macrophage signaling through p120 RAS-GAP modulation via miR132. 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210. 42. Ky V. Hoang, Eric Bachelder, Hassan Borteh, Murugesan V. S. Rajaram, Heather Curry, Kristy Ainslie, John S. Gunn, and Larry S. Schlesinger (2013). Use of encapsulated AR-12 as a host directed agent to control intracellular pathogens. 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210. 43. Murugesan V.S. Rajaram, Bin Ni, Tracy Carlson and Larry S. Schlesinger (2013). Mannose receptor (CD- 206)-mediated signaling in human macrophages in the context of tuberculosis. 5th Center for Microbial Interface Biology Research Retreat, April 18th- 19th 2013, The Ohio State University medical Center, Columbus, OH, 43210 44. Murugesan V.S Rajaram, Bin Ni, Tracy Carlson and Larry S. Schlesinger (2012). Unraveling the mystery of Mannose receptor- mediated signaling in human macrophages. 7th Annual Research Day, Davis Heart & Lung Research Institute, October 11, 2012, The Ohio State University medical Center, Columbus, OH, 43210. 45. Bin Ni, Murugesan V.S. Rajaram, Michelle B. Landes, Huy A. Nguyen, and Larry S. Schlesinger (2012). MicroRNA expression profiling of Mycobacterium tuberculosis-infected human Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis macrophages: M.tb decreases host IFN-γ-responsiveness through miR-132. 7th Annual Research Day, Davis Heart & Lung Research Institute, October 11, 2012, The Ohio State University medical Center, Columbus, OH, 43210.

253 46. Larry S. Schlesinger Bin Ni, Jessica Morris, Michelle N. Brooks, Tracy K. Carlson, Jordi B. Torrelles and Murugesan V.S. Rajaram, (2012). Mycobacterium tuberculosis infection alters the expression of microRNAs which regulate human macrophage immune responses. Tuberculosis-2012, Biology, Pathogenesis and Intervention Strategies, September 11-15, 2012, Institute Pasteur, Paris, France. 47. Murugesan V.S. Rajaram, Jessica Morris, Michelle N. Brooks, Jordi B. Torrelles and Larry S. Schlesinger (2011). M. tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-Activated Protein Kinase 2 (MK2) and miR125b. 2011 Keystone Symposia, Tuberculosis: Immunology, Cell Biology and Novel Vaccination Strategies, January 15-20, Vancouver, British Columbia, Canada 48. Michelle N. Brooks, Murugesan V.S Rajaram, Abul K. Azad, Amal O. Amer, Martin A. Valdivia- Arenas, Jopng-Hwang Park, Gabriel Nunez and Larry S. Schlesinger (2011). NOD2 controls the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis and M. bovis BCG in human macrophages. 2011 Keystone Symposia, Tuberculosis: Immunology, Cell Biology and Novel Vaccination Strategies, January 15-20, Vancouver, British Columbia, Canada. 49. Michelle N. Brooks, Murugesan V.S. Rajaram, Bin Ni, Huy A. Nguyen, and Larry S. Schlesinger 2011. The role of NOD2 in innate immune functions in response to Mycobacterium tuberculosis in human macrophages. The 10th Annual OSUMC Trainee Research Day - April 7, 2011. 50. Bin Ni, Murugesan V.S. Rajaram, Michelle N. Brooks, Huy A. Nguyen, Stefano Volinia, and Larry S. Schlesinger 2011. MicroRNA expression profiling of M. tuberculosis-infected human macrophages: miR- 370 target Tpl2 (MAP3K8) plays a role in mycobacterial innate immunity. The 10th Annual OSUMC Trainee Research Day - April 7, 2011.

References:

1. Jeff Schorey, Ph.D. George B. Craig Jr. Professor Department of Biological Sciences Eck Institute for Global Health Center for Rare and Neglected Diseases University of Notre Dame Notre Dame, IN 46556 Phone: 574-631-3734 Email: [email protected]

2. Sakthivel Sadayappan, PhD, MBA Professor of Internal Medicine Associate Chairman for Basic Research Department of Internal Medicine Director of Heart Branch of the Heart, Lung and Vascular Institute Division of Cardiovascular Health and Disease University of Cincinnati, College of Medicine Cardiovascular Center, Rm 4935 231 Albert Sabin Way Cincinnati, OH 45267-0542, USA Phone : 513-558-7498 Email : [email protected]

3. Jian Zhang, MD Endowed Chair in Immunology Research Professor of Pathology Department of Pathology University of Iowa 1169 Medical Laboratories 25 S. Grand Avenue Iowa City, IA 52242 Phone: 319-335-1121 Email: [email protected] NAMAL P. M. LIYANAGE

ACADEMIC APPOINTMENTS

2017- present Assistant professor of Microbial Infection and Immunity, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH. 2017- present Assistant professor of Veterinary Bioscience (secondary appointment), College of Veterinary Medicine, The Ohio State University, Columbus, OH.

EDUCATION 2010 Ph.D. Integrative Biomedical Science, University of Nebraska-Lincoln, NE. 2005 MS. Biomedical Science, University of Nebraska-Lincoln, NE.

POSTGRADUATE TRAINING AND FELLOWSHIP APPOINTMENTS:

2016 - Research Fellow, National Institutes of Health (NIH), Bethesda, MD. 2017 2011 - Postdoctoral Research Fellow / CRTA Fellow, National Institutes of Health (NIH). 2016 2010 - Postdoctoral Research Fellow / ORISE Fellow Center for Biological Evaluation and 2011 Research, The Food and Drug Administration (FDA), Bethesda, MD.

OTHER PROFESSIONAL ROLES 2017- present Member Infectious Diseases Institute, The Ohio State University 2017- present Special volunteer, National Institutes of Health (NIH), Bethesda, MD

AWARDS AND HONORS

2 Early Career Faculty Travel Grant, AAI 2020 meeting, Honolulu, Hawai 0 2 0 2 Young Investigator Award, HIV Vaccine Trial Network, Washington DC 0 1 9 2 Young Investigator Award. HIV R4P 2014 meeting Cape Town, South Africa. 0 1 4 2 Young Investigator Award. The Annual Conference on Retroviruses and Opportunistic 0 1 4 Infections. 2 Fellows Award for Research Excellence (FARE). NIH Scientific Directors. 0 1 4 2 Young Investigator Award. AIDS Vaccine meeting Barcelona- Spain. 0 1 3 2 Cancer Research Training Award. National Cancer Institute / National Institutes of Health. 0 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

1 1 2 William G. Whitmore Memorial Award. The University of Nebraska-Lincoln. 0 0 9 2 Milton E. Mohr Biotechnology Award. The University of Nebraska-Lincoln. 0 0 6 2 Scholastic Graduate Research Fellowship. Dean of Graduate Studies, University of 0 Nebraska- 0 5 Lincoln. 2 The Best poster presentation. College of Microbiologists-Sri Lanka. 0 0 2 2 Best Graduate research award. Sri Lanka Association for the Advancement of Science. 0 0 2

INVITED SPEAKER PRESENTATIONS AT CONFERENCE PROCEEDINGS

2 International Emerging Infectious Diseases Conference, Nanavati Hospital Auditorium 0 Mumbai, 1 8 India 2 Invited talk: Sri Lanka Institute of Biotechnology and research program Harmonizing 0 meeting. 1 8 National Science Foundation, Sri Lanka. 2 Keystone Symposia on HIV vaccines, Olympic Valley, California. 0 1 6 2 HIV R4P 2014 meeting, Cape Town, South Africa. 0 1 4 NAMAL P. M. LIYANAGE 1

257 2 AIDS Vaccine Meeting, Boston, MA. 0 1 2 2 HIV/AIDS & Cancer Think Tank Meeting, Bethesda, 0 MD. 1 1 2 Midwest Biomedical Research Forum, Omaha, NE. 0 0 9

POSTER PRESENTATIONS AT CONFERENCE PROCEEDINGS

2 Keystone Symposia on HIV vaccines, Steamboat Springs, Colorado. 0 1 7 2 HIV/AIDS & Cancer Think Tank Meeting, Bethesda, MD 0 1 5 2 The Annual Conference on Retroviruses and Opportunistic Infections, Boston MA. 0 1 4 2 3rd Conference of Translational Medicine on the Pathogenesis and Therapy of immune- 0 Mediated 1 4 diseases. Milan, Italy. 2 HIV/AIDS & Cancer Think Tank Meeting, Bethesda, MD. 0 1 3 2 AIDS Vaccine Meeting, Barcelona, Spain. 0 1 3 2 HIV/AIDS & Cancer Think Tank Meeting, Bethesda, MD. 0 1 2 2 Annual meeting for the Association for Research in Vision, Fort Lauderdale, FL. 0 1 0 2 Annual meeting for the Association for Research in Vision, Fort Lauderdale, FL. 0 0 6

JOURNAL REFEREE/ EDITORIAL BOARDS Reviewer: AIDS Research and Human Retroviruses Review Editor: Frontiers in Immunology Review Editor: Frontiers in Public Health Reviewer: PLOS ONE Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Reviewer: Nature Immunology

MEMBERSHIPS American Society for Microbiology (ASM) Member 2003-present American Association of Immunologists (AAI) Member 2005-present Sri Lanka Association for the Advancement of Science (SLAS) Life Member

ACADEMIC SERVICE

2017- Co Director : Special Topics in Biomedical Science I: Immunology and Infectious Disease (BIOMSCI 4810H) 2017- 2018 : Member of the Awards Committee MI &I 2019 - 2020: Member of the Space and Resource Committee 2019 – 2020: Member of the Immunology faculty search Committee VBS

PH.D Thesis Advisory Committees Aaren Kettelhut (MD. Ph.D. candidate) (Advisor : Nicholas T. Funderburg) Tiffany Claeys (Ph.D. candidate) (Advisor : Richard Robinson) Rosas Mejia (Ph.D. candidate) (Advisor : Richard Robinson) Taiwei Li (Ph.D. candidate) (Advisor : Jian Zhu)

Student and Postdoctoral Trainees:

Post-doctoral Trainees : 2018- Rajni Kant Shukla Ph.D 2017- 2018 Amila Hatharasinghe MB.BS (Currently surgery resident)

Pre-doctoral graduate trainees: 2019 - Manuja Gunasena BS. NAMAL P. M. LIYANAGE 2

259 Medical Student trainees: 2019 - 2020 Nancy Tian BS. 2020 - 2021 Jeongwon Choi BS.

Undergraduate student trainees: 2017 - Nicole Reinhold-Larsson (Currently a PhD student at Harvard Medical 2019 School) 2017- Sam Cray 2020 2017- Miguel de Real 2020 2019 Jerra Furay 2019 Tatum Skladany 2019 Will Mulhern

High School Student Summer trainees: 2018 Anusha Srivastava (Currently un undergraduate student at Vanderbilt University)

MEMBERSHIPS American Society for Microbiology (ASM) Member 2003-present American Association of Immunologists (AAI) Member 2005-present Sri Lanka Association for the Advancement of Science (SLAS) Life Member

GRANTS

K22 A1127072 Role: PI 04/01/2018 – 03/31/2021 Retooling NK cells and mucosal ILCs for an effective HIV vaccine. NIH/NIAID The goal of this project to investigate the effect of HIV vaccines on mice NK cells

WNPRC Pilot Project Grant Role: Co-PI 08/01/18 - 07/31/21 Neisseria colonization of the female reproductive tract. Wisconsin National Primate Research Center The goal of this project to investigate the immune response to Neisseria colonization in rhesus female reproductive system.

CF Pilot Project Grant Role: Co-PI 01/15/19 – 01/14/21 T cell dysfunction in CF patients with nontuberculous mycobacteria infection. NCH Investigate the Immune responses in CF patients’ lungs

PATENT International Patent WO 2015/106003A1 . RAS Pathways as Markers of Protection against HIV and Methods to Improve Vaccine efficacy.

PUBLICATIONS

PEER-REVIEWED SCIENTIFIC JOURNALS

1. Gorini G, Fourati S, Vaccari M, Rahman MA, Gordon SN, Brown DR, Law L, Chang J, Green R, Barrenäs F, Liyanage NPM, Doster MN, Schifanella L, Bissa M, Silva de Castro I, Washington- Parks R, Galli V, Fuller DH, Santra S, Agy M, Pal R, Palermo RE, Tomaras GD, Shen X, LaBranche CC, Montefiori DC, Venzon DJ, Trinh HV, Rao M, Gale M Jr, Sekaly RP, Franchini G. Engagement of monocytes, NK cells, and CD4+ Th1 cells by ALVAC-SIV Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

vaccination results in a decreased risk of SIVmac251 vaginal acquisition. PLoS Pathog. 2020 .Mar;16(3):e1008377.

NAMAL P. M. LIYANAGE 3

261 2. Silva de Castro I, Gordon SN, Liu J, Bissa M, McKinnon K, Trinh HV, Doster MN, Schifanella L, Liyanage NP, Cao J, Cheng O, Foulds K, Roederer M, Koup RA, Shen X, Tomaras GD, Venzon DJ, Forthal DN, Fouts T, Montefiori DC, Tartaglia J, Rao M, Ostrowski M, Franchini G, Vaccari M. Expression of CD40L by the ALVAC-SIV vector abrogates T cell responses in macaques. J Virol. 2020 Jan 2;. doi: 10.1128/JVI.01933-19.

3. Hayes D Jr, Harhay MO, Nicol KK, Liyanage NPM, Keller BC, Robinson RT. Lung T-Cell Profile Alterations are Associated with Bronchiolitis Obliterans Syndrome in Cystic Fibrosis Lung Transplant Recipients. Lung. 2019 Dec 5.

4. Schifanella L, Barnett SW, Bissa M, Galli V, Doster MN, Vaccari M, Tomaras GD, Shen X, Phogat S, Pal R, Montefiori DC, LaBranche CC, Rao M, Trinh HV, Washington-Parks R, Liyanage NPM, Brown DR, Liang F, Loré K, Venzon DJ, Magnanelli W, Metrinko M, Kramer J, Breed M, Alter G, Ruprecht RM, Franchini G. ALVAC-HIV B/C candidate HIV vaccine efficacy dependent on neutralization profile of challenge virus and adjuvant dose and type. PLoS Pathog. 2019 Dec;15(12).

5. Claeys TA, Rosas Mejia O, Marshall S, Jarzembowski JA, Hayes D, Hull NM, Liyanage NPM, Chun RH, Sulman CG, Huppler AR, Robinson RT. Attenuation of Helper T Cell Capacity for TH1 and TH17 Differentiation in Children With Nontuberculous Mycobacterial Infection. J Infect Dis. 2019 Oct 22;220(11):1843-1847.

6. Vaccari M, Fourati S, Gordon SN, Brown DR, Bissa M, Schifanella L, Silva de Castro I, Doster MN, Galli V, Omsland M, Fujikawa D, Gorini G, Liyanage NP, Trinh HV, McKinnon KM, Foulds KE, Keele BF, Roederer M, Koup RA, Shen X, Tomaras GD, Wong MP, Munoz KJ, Gach JS, Forthal DN, Montefiori DC, Venzon DJ, Felber BK, Rosati M, Pavlakis GN, Rao M, Sekaly RP and Franchini G. HIV vaccine candidate activation of hypoxia and inflammasome in CD14+ monocytes is associated with a decreased risk of SIVmac251. Nat.Med.2018 Jun;24(6):847-856.

7. Nooshadokht M, Kalantari-Khandani B, Sharifi I, Kamyabi H, Liyanage NP, Lagenaur LA, Kagnoff MF, Singer SM, Babaei Z, Solaymani-Mohammadi S. Stool antigen immunodetection for diagnosis of Giardia duodenalis infection in human subjects with HIV and cancer. J Microbiol Methods. 2017 Oct;141:35-41.

8. Gordon SN, Liyanage NP, Doster MN, Vaccari M, Vargas-Inchaustegui D, Pegu P, Fourati S*, Schifanella L, Shen X, Tomaras GD, Rao M, Billings EA, Schwartz J, Prado l, Bobb K, Zhang W, Montefiori DC, Foulds KE, Ferrari G, Robert-Guroff M, Roederer M, Phan TB, Forthal DN, Stablen DM, Phogat S, Venzon D, Fouts T, Franchini G. Boosting of ALVAC- SIV Vaccine-Primed Macaues with CD4-SIVgp120 Fusion Protein Elicits Antibodies to V2 Associated with Decreased Risk of SIVmac251 Acquisition. J Immunol. 2016 OCT 1;197(7):2726-37

9. Vaccari M*,Gordon S*, Fourati S*, Schifanella L*, Liyanage NP*, CameronM, Keele BF, Shen X, Tomaras G, Billings E, Rao M, Chung A, Dowell K, Bailey-Kellogg C, Brown E, Ackerman M, Vargas-Inchaustegui D, Whitney S, Doster M, Binello N, Pegu P, Montefiori D, Foulds K, Quinn D, Donaldson M, Liang F, Loré K, Roederer M, Koup R, McDermott A, Ma Z, Miller C, Phan T, Forthal D, Blackburn M, Caccuri F, Bissa M, Ferrari G, Kalyanaraman V, Ferrari M., Thompson D, Robert- Guroff M, Kim S, Kim J, Michael N, Phogat S, Barnett S, Tartaglia J, Venzon D, Stablein D, Alter G, Sekaly R* & Franchini G*. Adjuvant dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition. Nat.Med.2016 Jul;22(7):762-70. (*Equally contributed)

10. Moniuszko M*, Liyanage NP*, Doster MN, Parks RW, Grubczak K, Lipinska D, McKinnon K, Brown C, Hirsch V, Vaccari M, Gordon S, Pegu P, Fenizia C, Flisiak R, Grzeszczuk A, Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Dabrowska M, Robert-Guroff M, Silvestri G, Stevenson M, McCune J, Franchini G. Glucocorticoid treatment at moderate doses of SIVmac251-infected rhesus macaques decreases the frequency of circulating CD14+CD16++ monocytes but does not alter the tissue virus reservoir. AIDS Res Hum Retroviruses. 2015 Jan; 31(1):115-26. (*Equally contributed) NAMAL P. M. LIYANAGE 4

263 11. Liyanage NP, Gordon SN,Doster MN, Pegu P, Vaccari M, Shukur N, Schifanella L,Pise- Masison CA, Lipinska D, Grubczak K, Montiuszko M, Franchini G. Antiretroviral therapy partly reverse the systemic and mucosal distribution of NK cell subsets that is altered by SIVmac251 infection of macaques. Virology. 2014 Feb; 450-451:359-68.

12. Gordon SN, Doster MN, Kines RC, Keele BF, Brocca-Cofano E, Guan Y, Pegu P, Liyanage NP, Vaccari M, Cuburu N, Buck CB, Ferrari G, Montefiori D, Piatak M Jr, Lifson JD, Xenophontos AM, Venzon D, Robert-Guroff M, Graham BS, Lowy DR, Schiller JT, Franchini G. Antibody to the gp120 V1/V2 loops and CD4+ and CD8+ T cell responses in protection from SIVmac251 vaginal acquisition and persistent viremia. J Immunol. 2014 Dec 15;193(12):6172-83.

13. Valentin A, McKinnon K, Li J, Rosati M, Kulkarni V, Pilkington GR, Bear J, Alicea C, Vargas- Inchaustegui DA, Jean Patterson L, Pegu P, Liyanage NP, Gordon SN, Vaccari M, Wang Y, Hogg AE, Frey B, Sui Y, Reed SG, Sardesai NY, Berzofsky JA, Franchini G, Robert- Guroff M, Felber BK, Pavlakis GN. Comparative analysis of SIV-specific cellular immune responses induced by different vaccine platforms in rhesus macaques. Clin. Immunol. 2014 Nov;155(1):91-107.

14. Vargas-Inchaustegui DA, Tuero I, Mohanram V, Musich T, Pegu P, Valentin A, Sui Y, Rosati M, Bear J, Venzon DJ, Kulkarni V, Alicea C, Pilkington GR, Liyanage NP, Demberg T, Gordon SN, Wang Y, Hogg AE, Frey B, Patterson LJ, DiPasquale J, Montefiori DC, Sardesai NY, Reed SG, Berzofsky JA, Franchini G, Felber BK, Pavlakis GN, Robert-Guroff M. Humoral immunity induced by mucosal and/or systemic SIV-specific vaccine platforms suggests novel combinatorial approaches for enhancing responses. Clin Immunol. 2014 Aug; 153(2): 308-22.

15. Yang Y, Gordon SN, Doster MN, Liyanage NP, Chang J, Williams C, Agy M, Mclain R, Law L, Franchini G, Palermo R, Katze M. A systems biology approach revealed distinct host responses between ALVAC-SIV and NYVAC-SIV vaccines during an evaluation of immunogenicity in rhesus macaques (VAC7P.992). J. Immunol. 2014 May: 192 (141) 37.

16. Liyanage NP, Dassanayake RP, Kuszynski CA, Duhamel GE. Contribution of Helicobacter hepaticus cytolethal distending toxin subunits to human epithelial cell cycle arrest and apoptotic death in vitro. Helicobacter. 2013 Dec; 18(6):433-43.

17. Moniuszko M, Lipinska D, Jeznach M, Kowal K, Grubczak K, Rusak M, McKinnon K, Vaccari M, Liyanage NP, Fenizia C, Wawrusiewicz-Kurylonek N, Dabrowska M, Jablonska E, Kretowski A, Gorska M, Bodzenta-Lukaszyk A. Glucocorticoids upregulate decreased IL-7 receptor expression in asthmatic patients and simian immunodeficiency virus-infected non- human primates. J. Biol. Regul. Homeost. Agents. 2013 Apr-Jun; 27(2): 427-42.

18. Liyanage NP, Pegu P, Gordon SN, Cameron M, Foulds K, Doster MN, Vaccari M, Koup R, Roederer M, Sékaly RP, Franchini G. RV144-like trial in macaques using ALVAC-SIV & gp120 induces innate immunity and increases the frequency of NK22 & NKG2A+ cells in mucosal tissues. Retrovirology. 2012, 9(2).

19. Liyanage NP, Manthey KC, Dassanayake RP, Kuszynski CA, Oakley GG, Duhamel GE. Helicobacter hepaticus cytolethal distending toxin causes cell death in intestinal epithelial cells via mitochondrial apoptotic pathway. Helicobacter. 2010 Apr; 15(2): 98-107.

20. Liyanage NP, Fernando MR, Lou MF. Regulation of the bioavailability of thioredoxin in the lens by a specific thioredoxin-binding protein (TBP-2). Exp. Eye Res. 2007 Aug; 85(2): 270- 9.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

21. Liyanage NP, Fernando S, Malavige GN, Mallikahewa R, Sivayogan S, Jiffry MT, Vitarana T. Seroprevalence of varicella zoster virus infections in Colombo district, Sri Lanka. Indian J Med Sci. 2007 Mar; 61(3): 128-34. NAMAL P. M. LIYANAGE 5

265 22. Liyanage NP, Fernando MR, Lou MF. Cloning and Sequencing of Thioredoxin Binding Protein–2 (TBP–2) From Human Lens Epithelial Cells. Investigative & visual science 01/2005; 46(E): 3848. 23. Fernando N, Holten J, Fernando S, Liyanage NP. Helicobacter serology in the Western province of Sri Lanka. The Ceylon Journal of Medical Science. 2003, 46: 35- 43.

NAMAL P. M. LIYANAGE 6 CURRICULUM VITAE

PATRICK L. COLLINS, Ph.D.

DATE: May 2, 2020 PERSONAL INFORMATION: Sex: Male Date of Birth: April 27, 1984 Place of Birth: Silver City, NM CITIZENSHIP: U.S.A. ADDRESS AND TELEPHONE NUMBERS: a. Lab: Washington University School of Medicine Department of Pathology & Immunology 660 South Euclid Avenue, Campus Box 8118 Room 7730 CSRB St. Louis, MO 63110 Tel: (314) 362-5515 Fax: (314) 362-8888 Email: [email protected] b. Home: 1217 Oakleaf Dr Saint Louis, MO, 63119 (615) 668-3538 (Cell) PRESENT POSITION: 2019-Current Research Assistant Professor at The Ohio State University, Department of Microbial Infection and Immunity, Columbus, OH

EDUCATION: 2002-2006 Pre-doctoral: New Mexico Institute of Mining and Technology; Socorro, NM, Biology 2006-2011 Graduate: Vanderbilt University; Nashville, TN, Ph.D., Immunology. Thesis: “Distal Regulation of the Interferon Gamma Locus” (Mentor: Tom Aune)

HONORS AND AWARDS:

2006 David K. Shortess Award for Outstanding Undergraduate Student 2007-2009 Pre-doctoral training grant, Immunobiology of Blood and Vascular Systems 2008-2010 Member: Microbes and Defense Academy Honor Society 2012-2014 Post-doctoral fellowship – NIH Training Program in Immunology and Immunogenetics

PROFESSIONAL SOCIETIES AND ORGANIZATIONS: 2014-present: American Association of Immunologists

CURRENT RESEARCH: The Protective Epigenome – studying how cells establish protective chromatin at sites of DNA damage and at endogenous retroviruses In preparation Topological Interactions Establish Phosphorylated H2Ax Chromatin Domains Surrounding Double Strand Breaks 2015 Proceedings of the National Academy of the United States of America: SETDB1 Restricts Expression of Endogenous and Exogenous Retrovirues in B Lymphocytes

Regulome of Human Hematopoietic Cells in Health and Disease – collaborations that utilize epigenomics to define the regulatory logic of human hematopoietic cells In preparation Age Associated Changes in the Epigenome of Human Hematopoietic Cells (With E, Oltz, M. Artyomov) In revision Cell: Epigenetic Programming of CD56bright and CD56dim Natural Killer Subesets (With E. Oltz, M. Colonna) 2016 Cell: Distinct Gene Regulatory Pathways for Human Innate Versus Adaptive Lymphoid cells. (With E. Oltz, M. Colonna)

RESEARCH SUPPORT: CURRENT SUPPORT (CONTRIBUTED KEY PRELIMINARY DATA):

1 R01 AI118852 (Oltz, PI) NIH/NIAID 3/1/15-09/31/20

Topological Mechanisms of DNA Break Repair in Lymphocytes 5 R01 AI134035-02 (Oltz/Colonna; Co-PI) 08/13/18- 07/31/23 NIH/NIAID Cis-Regulatory Circuits For Ilc Function And Plasticity 5 R01 AI130231-03 (Oltz/Bassing; Co-PI) 09/01/19- 09/31/20 NIH/NIAID Regulation of Genome Architecture in Lymphocytes: Opposing Tether and Boundary Functions TEACHING TITLE AND RESPONSIBILITIES: 2009-2010 Teaching Assistant, Interdisciplinary Graduate Program “Foundations in Immunology” Section, VUMS: Organized and ran journal review sessions for graduate students 2009-2012 Trained Graduate Students and Summer Students in the Aune Lab, VUMS 2010-2011 Teaching Assistant, Medical Microbiology Laboratory, VUMS: Taught medical students clinically applied microbiology laboratory tests. 2012-2015 Mentor, Young Scientist Training Program, WUSTL: Mentored high school students in a summer research project completed alongside me in our laboratory 2014-2020 Trained Graduate Students, Summer Students and new Post-Docs in the Oltz Lab, WUSTL & OSU

MAJOR INVITED SEMINARS: 2016 International Congress of Immunology, Melbourne Australia

UNIVERSITY SERVICE (WASHINGTON UNIVERSITY): 2016 Founding Seminar Coordinator: DNA Damage and Metabolism Student Work in Progress

UNIVERSITY SERVICE (VANDERBILT): 2007-2009 Graduate Student Council Representative 2008-2009 Magazine Contributor: Vanderbilt A.B.S.T.R.A.C.T. Magazine 2008-2010 Seminar Coordinator: Microbes and Defense Academy Honor Society 2009-2010 President: Microbiology and Immunology Graduate Student Association

BIBLIOGRAPHY: a. Peer Reviewed Manuscripts 1. Chang, Shaojing, Patrick L. Collins, and Thomas M. Aune. "T-bet dependent removal of Sin3A-histone deacetylase complexes at the Ifng locus drives Th1 differentiation." The Journal of Immunology 181.12 (2008): 8372-8381.

2. Aune, Thomas M., Patrick L. Collins, and Shaojing Chang. "Epigenetics and T helper 1 differentiation." Immunology 126.3 (2009): 299-305.

3. Collins, Patrick L., and Thomas M. Aune. "Keeping One's Option Open." Immunity 32.5 (2010): 581-583.

4. Collins, Patrick L., Shaojing Chang, Melodie Henderson, Mohammed Soutto, Georgia M. Davis, Allyson G. McLoed, Michael J. Townsend, Laurie H. Glimcher, Douglas P. Mortlock, and Thomas M. Aune. "Distal regions of the human IFNG locus direct cell type- specific expression." The Journal of Immunology 185, no. 3 (2010): 1492-1501.

5. Hoek, Kristen L., Laura E. Gordy, Patrick L. Collins, Vrajesh V. Parekh, Thomas M. Aune, Sebastian Joyce, James W. Thomas, Luc Van Kaer, and Eric Sebzda. "Follicular B cell trafficking within the spleen actively restricts humoral immune responses." Immunity 33, no. 2 (2010): 254-265.

6. Spurlock, Charles F., Zachary T. Aune, John T. Tossberg, Patrick L. Collins, Jessica P. Aune, Joseph W. Huston, Philip S. Crooke, Nancy J. Olsen, and Thomas M. Aune. "Increased sensitivity to apoptosis induced by methotrexate is mediated by JNK." Arthritis & Rheumatism 63, no. 9 (2011): 2606-2616.

7. Collins, Patrick L., Melodie A. Henderson, and Thomas M. Aune. "Diverse functions of distal regulatory elements at the IFNG locus." The Journal of Immunology 188, no. 4 (2012): 1726-1733.

8. Collier, Sarah P., Patrick L. Collins, Christopher L. Williams, Mark R. Boothby, and Thomas M. Aune. "Cutting edge: influence of Tmevpg1, a long intergenic noncoding RNA, on the expression of Ifng by Th1 cells." The Journal of Immunology 189, no. 5 (2012): 2084-2088. 9. Collins, Patrick L., Melodie A. Henderson, and Thomas M. Aune. "Lineage-specific adjacent IFNG and IL26 genes share a common distal enhancer element." Genes and immunity 13, no. 6 (2012): 481-488. 10. Aune, Thomas Martin, Patrick L. Collins, Sarah P. Collier, Melodie A. Henderson, and Shaojing Chang. "Epigenetic Activation and Silencing of the Gene that Encodes IFN-γ." Frontiers in immunology 4 (2013): 112.

11. Gopalakrishnan, Suhasni, Patrick L. Collins, and Eugene M. Oltz. "Control of Ig gene assembly: lessons from premature activation." The EMBO journal 32, no. 10 (2013): 1350- 1351. 12. Collins, Patrick L., and Eugene M. Oltz. "Histone methylation keeps the brakes on autophagy." Molecular and cellular biology 33, no. 20 (2013): 3974-3975.

13. Pabbisetty, Sudheer K., Whitney Rabacal, Damian Maseda, Delphine Cendron, Patrick L. Collins, Kristen L. Hoek, Vrajesh V. Parekh, Thomas M. Aune, and Eric Sebzda. "KLF2 is a rate-limiting transcription factor that can be targeted to enhance regulatory T-cell production." Proceedings of the National Academy of Sciences 111, no. 26 (2014): 9579- 9584.

14. Collins, P.L., Kyle, K.E., Egawa, T., Shinkai, Y., Oltz, E.M., Moran, J.V. The histone methyltransferase SETDB1 represses endogenous and exogenous retroviruses in B lymphocytes (2015) Proceedings of the National Academy of Sciences of the United States of America, 112 (27), pp. 8367-8372.

15. Kaiko, G.E., Ryu, S.H., Koues, O.I., Collins, P.L., Solnica-Krezel, L., Pearce, E.J., Pearce, E.L., Oltz, E.M., Stappenbeck, T.S. The Colonic Crypt Protects Stem Cells from Microbiota- Derived Metabolites (2016) Cell, 165 (7), pp. 1708-1720.

16. Koues, O.I., Collins, P.L., Cella, M., Robinette, M.L., Porter, S.I., Pyfrom, S.C., Payton, J.E., Colonna, M., Oltz, E.M. Distinct Gene Regulatory Pathways for Human Innate versus Adaptive Lymphoid Cells (2016) Cell, 165 (5), pp. 1134-1146. Rohatgi, N., Zou, W., Collins, P.L., Brestoff, J.R., Chen, T.H., Abu-Amer, Y., Teitelbaum, S.L. ASXL1 impairs osteoclast formation by epigenetic regulation of NFATc1 (2018) Blood Advances, 2 (19), pp. 2467-2477. 17. Collins, P.L., Cella, M., Porter, S.I., Li, S., Gurewitz, G.L., Hong, H.S., Johnson, R.P., Oltz, E.M., Colonna, M. Gene Regulatory Programs Conferring Phenotypic Identities to Human NK Cells (2019) Cell, 176 (1-2), pp. 348-360.e12. 18. Cella, M., Gamini, R., Sécca, C., Collins, P.L., Zhao, S., Peng, V., Robinette, M.L., Schettini, J., Zaitsev, K., Gordon, W., Bando, J.K., Yomogida, K., Cortez, V., Fronick, C., Fulton, R., Lin, L.-L., Gilfillan, S., Flavell, R.A., Shan, L., Artyomov, M.N., Bowman, M., Oltz, E.M., Jelinsky, S.A., Colonna, M. Subsets of ILC3−ILC1-like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues (2019) Nature Immunology, 20 (8), pp. 980-991. 19. Park, E., Patel, S., Wang, Q., Andhey, P., Zaitsev, K., Porter, S., Hershey, M., Bern, M., Plougastel-Douglas, B., Collins, P., Colonna, M., Murphy, K.M., Oltz, E., Artyomov, M., Sibley, L.D., Yokoyama, W.M. Toxoplasma gondii infection drives conversion of NK cells into ILC1-like cells (2019) eLife, 8, art. no. e47605. 20. Cella, M., Gamini, R., Sécca, C., Collins, P.L., Zhao, S., Peng, V., Robinette, M.L., Schettini, J., Zaitsev, K., Gordon, W., Bando, J.K., Yomogida, K., Cortez, V., Fronick, C., Fulton, R., Lin, L.-L., Gilfillan, S., Flavell, R.A., Shan, L., Artyomov, M.N., Bowman, M., Oltz, E.M., Jelinsky, S.A., Colonna, M. Publisher Correction: Subsets of ILC3−ILC1-like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues (Nature Immunology, (2019), 20, 8, (980-991), 10.1038/s41590-019-0425-y) (2019) Nature Immunology, 20 (10), p. 1405. 21. Wang, Q., Robinette, M.L., Billon, C., Collins, P.L., Bando, J.K., Fachi, J.L., Sécca, C., Porter, S.I., Saini, A., Gilfillan, S., Solt, L.A., Musiek, E.S., Oltz, E.M., Burris, T.P., Colonna, M. Circadian rhythm-dependent and circadian rhythm-independent impacts of the molecular clock on type 3 innate lymphoid cells (2019) Science Immunology, 4 (40), art. no. eaay7501. 22. Purman, C.E., Collins, P.L., Porter, S.I., Saini, A., Gupta, H., Sleckman, B.P., Oltz, E.M. Regional gene repression by DNA double-strand breaks in G1 phase cells (2019) Molecular and Cellular Biology, 39 (24), art. no. e00181-19. 23. Mack, M.R., Brestoff, J.R., Berrien-Elliott, M.M., Trier, A.M., Yang, T.-L.B., McCullen, M., Collins, P.L., Niu, H., Bodet, N.D., Wagner, J.A., Park, E., Xu, A.Z., Wang, F., Chibnall, R., Council, M.L., Heffington, C., Kreisel, F., Margolis, D.J., Sheinbein, D., Lovato, P., Vivier, E., Cella, M., Colonna, M., Yokoyama, W.M., Oltz, E.M., Fehniger, T.A., Kim, B.S. Blood natural killer cell deficiency reveals an immunotherapy strategy for atopic dermatitis (2020) Science Translational Medicine, 12 (532), art. no. aay1005, . 24. Ma, H., Qian, W., Bambouskova, M., Collins, P.L., Porter, S.I., Byrum, A.K., Zhang, R., Artyomov, M., Oltz, E.M., Mosammaparast, N., Miner, J.J., Diamond, M.S. Barrier-to- autointegration factor Microbial Infection and Immunity

793 Biomedical Research Tower 460 W. 12th Avenue Columbus, OH 43210-2210 Phone: 614-292-0918 Fax: 614-292-9616

Curriculum Vitae April 2020

PAUL STOODLEY, BSc(Hons), Ph.D., FHEA, FAAM. Director Campus Microscopy and Imaging Facility Professor Dept. Microbial Infection and Immunity Dept. of Orthopaedics Dept. Microbiology The Ohio State University, Columbus OH.

Professor Microbial Tribology National Centre for Advanced Tribology (nCATS) National Biofilm innovation Centre Dept. Mechanical Eng., University of Southampton, UK

Expertise Key Words Bacterial biofilms, diagnostics, infection, orthopaedics, medical devices, mechanical properties

Education Postgraduate Certificate in Academic Practice (PCAP - MSc equivalent). Fellow of the Higher Education Academy. University of Southampton, UK. 2012. Ph.D. Biological Sciences, University of Exeter, UK. 1999. (Advisors: H. Lappin-Scott, Biology and J. Boyle, Chem. Eng.) B.Sc. (Hons) Environmental Sciences, Lancaster University, Lancaster, UK. 1982.

Research and professional experience 8/17/17- Director, Center for Campus Microscopy and Imaging Facility (CMIF), The Ohio State University. 9/1/13- Professor Dept. Microbial Infection and Immunity and Dept. of Orthopaedics, The Ohio State University, Columbus OH. 2/11/13- Adjunct Professor Microbial Tribology, National Centre for Advanced Tribology (nCATS), University of Southampton. 3/1/12-2/10/13 Reader (Associate Professor) Microbial Tribology, National Centre for Advanced Tribology (nCATS), University of Southampton. 7/1/09-2/29/12 Lecturer Microbial Tribology, National Centre for Advanced Tribology (nCATS), University of Southampton. 2005- 2009 Associate Professor Drexel University Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA. USA. 2004 – 2009 Vice Director of Imaging, Center for Genomic Sciences, Allegheny General Hospital, Pittsburgh, PA. 2003 -2004 Affiliate Associate Professor Dept Microbiology, Montana State University 2003-2004 Affiliate Associate Professor Dept. Mechanical Engineering, Montana State University 1999 – 2004 Assistant Research Professor, Center for Biofilm Engineering, Montana State University, Bozeman, MT. 1999 – 2001 Postdoctoral Associate, Center for Biofilm Engineering, Montana State University, Bozeman, MT. (Advisor: J.W. Costerton, PhD) 1994 - 2009 Scientific Consultant, Bio-Surface Technologies Inc. Bozeman, MT. 1996-1999 Research Fellow/PhD. candidate, School of Biological Sciences, Exeter University, UK.

Paul Stoodley: 716 Biomedical Research Tower, The Ohio State University, 460 West 12th Avenue, Columbus OH 43210.Tel: 1 614 6292 7826, email: [email protected], [email protected] Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

National Centre for Advanced Tribology (nCATS) Mechanical Engineering, University of Southampton, SO17 5BJ email: [email protected]

273 1987- Research Specialist/Lab Manager, Center for Biofilm Engineering, Montana State 1995 University 1982- Site Geophysicist, Cumbria and Lancashire Archaeological Unit, Lancaster 1984 University, UK. 1977- Laboratory Technician, Wessex Water Authority, Somerset, UK. 1979

Consultant Philips Oral Healthcare (active), Akzo Noble International Paint (active), Colgate (active), Semprus BioSciences, Baxter Healthcare, Biocomposites Inc. (active), Valam Inc., AMOCO Alaska, Characklis Consulting Engineering, DuPont, Northern States Nuclear Power, Novaflux (active) SC. Johnson, Trojan Nuclear, Unilever, Johnson & Johnson.

Advisory Boards MicroGen Dx Microbiology advisory board. Orlando FL. 26-27 April 2019. Member Biofilm Workgroup and consensus on Biofilm related Musculoskeletal Infections. The 2018 International Consensus Meeting on Musculoskeletal Infection. July 27-29. 3M Global Orthopaedic Summit. Philadelphia, PA. July 29-30, 2018. Infection Control Panel - Zimmer Biomet, Chicago June 10th, 2016 – 2017. Wound Biofilm Consensus Board as a “renowned expert in this field”. Smith & Nephew. 2015 – 2016. Mentor Corp. Identified as a Key Opinion Leader in Biofilms 2010. Scientific Advisory Board – Philips Oral Healthcare – 2005 – 2010.

Professional affiliations American Society for Microbiology (ASM) Society for General Microbiology (SGM) International Association for Dental Research (IADR) European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Musculoskeletal Infection Society (MSIS).

Honors 2019 Elected Fellow of the American Academy of Microbiology. 2018 Invited to serve as a reviewer on the Singapore President's Science Award (PSA), Agency for Science Technology and Research. 2014 Served on the NOW Spinoza Prize committee “of esteemed international scientific researchers” which is the highest Dutch award in science. 2012 Australian Research Council (ARC) - nominated as an 'expert of international standing' by the College of Experts. 2003 Outstanding Research Award, College of Engineering, Montana State University-Bozeman. 2002 Hanse-Wissenschaftskolleg (HWK) fellowship for 3 months study (1 month summer 2002, 1 month summer in Delmonhorst, Germany for collaborative research with Dr. deBeer at the Max Planck Institute for Marine Microbiology, Bremen.

Outreach

Invited lecture Bacterial Biofilms from your teeth to surgical site infection. “Mechanisms of Human Health and Disease”, an educational research program for advanced high school students, exposing them to a variety of scientific disciplines, such as pathophysiology, immunology, and microbiology. Nationwide Children’s Hospital, Columbus, OH. 07/11/19.

Smallpeice Trust Biomedical Engineering Course Co-Coordinator, for 24 Year 11 (US Grade 10) students. University of Southampton, 30 July - 1 August 2013.

Smallpeice Trust Biomedical Engineering Course Coordinator, for 24 Year 11 (US Grade 10) students. University of Southampton, 10 - 13 July 2012.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Smallpeice Trust Biomedical Engineering Course Coordinator, for 24 Year 11 (US Grade 10) students. University of Southampton, 12 - 15 July 2011. Overall satisfaction rating: 89%.

Smallpeice Trust Biomedical Engineering Course Co-Coordinator for 24 Year 11 (US Grade 10) students. University of

2

275 Southampton, 6 - 8 July 2010. Overall satisfaction rating: 94% which was the highest in the UK.

Biofilm based exhibit co-ordinator with Dr. L. Hall-Stoodley at the SciTech 2007, 2008, and 2009 science fairs at the Carnegie Science Center, Pittsburgh, PA USA. The exhibits were targeted to tours of school children in middle school (ages 8–12) and high school (ages 13 – 18).

Service

2019 / Feb OSU facilities and infrastructure working group 2019 / Mar External Reviewer Binghamton University, Binghamton Biofilm Research Center (BBRC). 2019 / Feb Proposal reviewer (3) OSU COM Medical Student Research Scholarship. 2019 / SP Grad student interviews – BSPG, Microbiology, Biophysics.

Invited promotion evaluation 2018 Binghamton University, Microbiology Assoc. prof. w/tenure. Nov 2018 Carnegie Mellon, Biological Sci. Assoc. prof w/o August tenure. 2017 University of Technology, Sydney, Full prof. Sept. Australia 2017 University of Texas at Austin, Physics. Assoc. prof. w/tenure. August 2017 Texas Tech University Health Full prof. July Sciences Center 2014 University of New South Wales, Senior lecturer May Australia (Assoc. prof.)

Boards and Committees Dept. MI&I Space and Resources Committee Meeting Apr 2019 - Dept. MI&I Seminars Committee Feb 2019 - OR Infrastructure and Facilities Working Group Feb 2019 - Dept. MI&I Procedures Oversight Designee (POD) Feb 2016 - Dept. MI&I Awards Committee Feb 2016 - Dept. MI&I Promotions and Tenure Committee Feb 2016 – 2018 Dept. Orthopaedics Promotions and Tenure Committee Dec 2013 - ongoing OSU Agendas Committee July 14 2014 – July 13 2016. Genetic modification and biosafety committee. University of Southampton. Oct 5th 2010 – Aug 31st 2013. Faculty of Engineering and the Environment Health and Safety Committee. Oct 31st 2011 – Aug 31st 2013. Biosafety officer for the Engineering Science Biolab 1001. Nov 15th 2010 – Aug 31st 2013.

Editorial boards (last 3 years): Editorial review board “Biofilms”, Elsevier. 1/15/19 – present. Editorial review board “Journal of Orthopaedic Research”. IF = 3.4. Sept 11 2018 – present. Editorial board “Nature Scientific Reports". IF = 4.219. Oct 1 2017 – present. Series Editor of the Springer Series on Biofilms. May 2013 – present. Assistant editor “Advances in Microbiology, Infectious Disease and Public Health”. IF = 2.012. April 2015 – present. Assistant editor of “Biofouling: Journal of Bioadhesion and Biofilm Research”. 2006 – May 2013. IF 4.41 Assistant editor of “Biofilms.” 2004 – 2005.

Journal reviewer (39): Nature, PNAS, Nature Protocols, Nature Reviews Microbiology, PLOS Pathogens, Infection and immunity, Acta Biomaterilia, African Journal Of Microbiology Research, Appl. Environ. Microbiol., Archives of Oral Biology, Biophysical Journal, Biotech. Bioeng., Biotech. Progress, BMC Medical Genomics, Environ. Microbiol., FEMS Microbiology Ecology, FEMS Medical Microbiology and Immunology, Folia Microbiologica, Interface, International Journal of Medical Microbiology, International Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Journal of Pediatric Otorhinolaryngology, ISME Journal, J. Bacteriol., J. Biomedical Materials Research: Part B - Applied Biomaterials, Chemotherapy, Interface, J. Clinical Periodontology, J. Dental Res., J. Infect. Dis., J. Materials Res., J. Microbiol. Methods, J. Molecular Biol., Journal of Theoretical Biology, J. Physical Chem, Langmuir, The Laryngoscope, Materials, Microbiology, Molecular Microbiol., Physics Reviews Letters, Proceedings of the Royal Society B: Biological Sciences, Trends Microbiol. Vaccine, Wat. Res., Wear. Jove, Scientific Reports.

Grants / proposal reviewer

3

277 39. NIH Special Emphasis Panel/Scientific Review Group “Exploration of Antimicrobial Therapeutics and Resistance”. July 8-9 2020. 38. NIH SBIR/STTR Biomedical Sensing, Measurement and Instrument. Tele meeting 37. NIH SBIR/STTR Biomedical Sensing, Measurement and Instrument. Bethesda 11/14-15 2019. 36. NIH Special Emphasis Panel/Scientific Review Group 2020/01 ZRG1 IDM-Y (82). “Exploration of Antimicrobial Therapeutics and Resistance. San Diego, 11/07/2019-11/08/2019 35. External reviewer “Center for Computational Microbiology and Medicine (C2M2)” Michigan University program grant. Sept. 2019 34. NIH SBIR/STTR SBIB-G11 Study section. Small Business: Biomedical Sensing, Measurement and Instrument. San Diego, CA. March 21-22, 2019 33. NIH SBIR/STTR grant proposal review meeting, ZRG1 SBIB-G (11). Bethesda 11/13-14 2018. 32. ZRG1 SBIB-G (83) A. Center for Scientific Review Special Emphasis R15. Panel. Bethesda 5/25/2018. 31. NIH Study section. Small Business: Biomedical Sensing, Measurement and Instrument. San Diego. 3/15/- 3/16/18. 30. Academy of Medical Sciences (UK) Starter Grant Clinical Lecturer Programme. Nov 8th 2017. 29. Department of Defense (DOD) US Army Medical Research and Materiel Command (USAMRMC) Congressionally Directed Medical Research Programs (CDMRP). 2017 Peer Reviewed Orthopaedic Research Program Nov. 15-17, 2107. 28. NIH Study section. Small Business: Biomedical Sensing, Measurement and Instrument. Bethesda. 11/02-03/17. 27. Congressionally Directed Medical Research Programs. FY17 Peer Reviewed Medical Research Program (PRMRP) Pre-Antimicrobial Resistance (Pre-AMR) peer review panel. 26. NASA MaterialsLab Biophysics Grant Review Panel. Arlington VA. May 6th 2016. 25. Peer-Reviewed Medical Research Program (PRMRP) Focused Program Award-Healthcare-Acquired Infection Reduction (FP-HCAIR) panel. Sept 2015. 24. ESCMID Trusted Grant Reviewer 2015 23. Deutsche Forschungsgemeinschaft (DFG), 2014. 22. Danish Council for Independent Research, 2014. 21. Orthopaedic Research UK. 2014. 20. Danish Council for Independent Research | Natural Science: DFF-Advanced Grant. 2012. 19. BBSRC (2012) 18. Belgian Science Policy Office (BELSPO) nominated “international experts for the “ex-ante” evaluation of the research proposals for the seventh phase of the “Interuniversity Attraction. Poles” programme (IAP, 2012- 2017). 17. Cystic Fibrosis Foundation 16. Advanced Food and Materials Network, Canada 15. Science Foundation Ireland RFP2009 BIOCHEMISTRY (BIC) Panel 14. Children’s Research Center of Michigan, Grant Program. 13. Portuguese Foundation for Science and Technology. 12. Technology Foundation STW, Netherlands. 11. National Science Foundation NSF's Merit Review (NSF). 10. Australian Research Council (ARC) - nominated as an 'expert of international standing' by the College of Experts. 9. Army Research Office (ARO) 8. Natural Sciences and Engineering Research Council of Canada (NSERC) 7. National Medical Research Council (Singapore) 6. Ad hoc reviewer, National Institutes of Health (NHLBI special emphasis panel 3/03). 5. Ad hoc reviewer, National Institutes of Health (NIH study section “Microbial Pathogenesis” 7/7/05) 4. NSF 06-607, Study Section Joint DMS/NIGMS Initiative for Mathematical Biology. 12/09-12/10/07. 3. National Institute of Biomedical Imaging and Bioengineering, NIH. NIBIB Training SEP ZEB1 OSR-C (M1) S. 5/2010 2. Health Protection Agency PhD Studentship Fund Review Group (2010) 1. Natural Sciences and Engineering Research Council of Canada 6/2010

Organizing committees

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

14. “9th ASM Biofilms Conference”. 2021. 13. “5th Annual Michael J. Patzakis, M.D. Endowed Lectureship in Orthopaedics”. Ohio State University. Sept. 5th 2018. Co-organizer with Drs. Granger and Sullivan. 12. “The mechanical properties of biofilms: state-of-the-art and research needs.”Co-organizer with with Rob

4

279 Nerenberg (PI). NSF funded workshop. Notre Dame University. April 23 – 24, 2018. 11. “8th ASM Biofilms Conference”. October 7 – 11, 2018, in Washington, DC. 10. “Eurobiofilms 2015” Workshop organized, Brno Czech republic. June 2015. 9. “Biofilms 2012”, 6th ASM Conference, Miami, Sept - Oct. 2012. Attendance 450. 8. “Eurobiofilms 2011”. Second European congress on microbial biofilms. Scientific advisory board member. Copenhagen, Denmark, July 6-8, 2011 7. “International Conference Biofilms 4: "Communities Bridging Disciplines." Sept. 1st to 3rd September, 2010. Winchester, UK. 6. “Biofilms 2009”, 5th ASM Conference, Cancun, Mexico. Co-PI. Nov. 2009. Attendance 410. 5. “First European Congress on Microbial Biofilms”, Rome, September 2 – 5, 2009. International Advisory Board, and Session Chair. 4. “Biofilm Mechanics”, MSU, Bozeman, MT June 28-29, 2007. 3. “Biofilms 2007”, 4th ASM Conference, Quebec City, March 24-27 2007. Attendance 700. 2. “Bugs and Biomaterials: Bacteria and the Biointerface.” University of Washington Engineered Biomaterials (UWEB) 9th Summer Symposium. Seattle. August 24-26, 2005. 3rd ASM Conference 1. “Biofilms 2003”, 3rd ASM Conference, Victoria, BC. Nov 1-6, 2003.

Advancement Implicit bias training, OSU. 1 hour training. 7 Aug 2017. Good Clinical Practice (GCP) Training. Southampton General Hospital. 6.5 hr course. Dec 16 2010.

Teaching

Ohio State University DENT 6400. Microbiology and Immunology. Basics of Biofilm 1 hr 04/16/20. SP2020 MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Infections – Clinical Perspective”. SP 2020. 1 hour lecture 4/13/20. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Basics of Biofilm Formation”. SP 2020. 1 hour lecture 4/10/20. BSGP 7070 Fundamentals of Grant Writing AU 2019 MICRO7724/BSGP7240 AU 2019 - Biofilms and Infection II: Case Studies MICRO7724/BSGP7240 AU 2019 - Biofilms and Infection I: Introduction to Biofilms SURGERY 8501 Career Development for Surgeons. SU2019. July 8. 0.5 hr lecture. 8833.07 Advanced Veterinary Surgical Sciences Graduate Course - Musculoskeletal. SP2019. “Bacterial Biofilms and Chronic Infections” 1 hour lecture. SP 03/07/19. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation II”. SP 2019. 1 hour lecture. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation I”. SP 2019. 1 hour lecture. BSGP 7070 Fundamentals of Grant Writing AU 2018 MICRO 7724/IBGP 7240 AU 2018. Mechanisms of tolerance in biofilms. MICRO 7724/IBGP 7240 AU 2018. Introduction to biofilms. MICRO 4998 AU18 Microbiology Undergraduate Research Projects. Anthony Li. SURGERY 8501 Career Development for Surgeons. SU2018. July 2 2018 0.5 hr lecture. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation II”. SP 2018. 1 hour lecture. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation I”. SP 2018. 1 hour lecture. MICRO 7724/IBGP 7240 AU 2017. Mechanisms of tolerance in biofilms. MICRO 7724/IBGP 7240 AU 2017. Introduction to biofilms. BSGP 7070 Fundamentals of Grant Writing AU 2017 MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation II”. SP 2017. 1 hour lecture. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation I”. SP 2017. 1 hour lecture. BSGP 7070 Fundamentals of Grant Writing Autumn 2016. 9/19 2 hr., 9/26 2hr, 10/3 2hr, 10/10 2 hr IBGP 8800.02 Therapeutics Infection & Host Pathogen Science Spring 2/28/16 “Emerging strategies for coping with bacterial multi-drug resistance and biofilm infections”. 2 hr lecture and led discussion. MICRO 4998H Microbiology Undergraduate Honors Thesis Research (Lydia Mack) 2015/16 MICRO 4998 Microbiology Undergraduate Research Projects. (Anthony Falzarano 2014/15) Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

BIOMEDE 4999H (IND) Biomedical Engineering Undergraduate Honors Thesis Research (Nick Farrar 2015/16), Michael Young (2014/2015))

5

281 BSGP 7950: Host Pathogen Research Seminar 10/19/15 MICRO 7724/IBGP 7240 Microbial Pathogenesis. Fall 2015. 9/25. IBGP 7400, Selected Topics in Microbial Pathogenesis (2 credit) Fall 2015. Two 2 hour sessions. Sept. 4 and 11. MICRBIO 4110 Microbial Pathogenesis and Immunobiology. “Biofilm Formation”. March 25th 2015. 1 hour lecture. Bench to Outcomes Seminar Series (BOSS). The role of bacterial biofilms in intravenous catheter associated infections Research Institute at Nationwide Children’s Hospital, Columbus OH. With Guliz Erdem, MD. 4th Nov 2014. IBGP 8800.02 Therapeutics Infection & Host Pathogen Science Spring 2014, 2/28/16 “Emerging strategies for coping with bacterial multi-drug resistance and biofilm infections”. 2 hr lecture and led discussion. MICRO 7724/IBGP 7240 Mol Bio Bac Pathog (2741) Spring 2014. Invited 1h guest lecture “Anti-Biofilm Surface Modification, Design and Control Strategies” OSU

University of Southampton SESM6029 -29730-12-13: 12-13-Nano & Biotribology-29730. 5 lectures. Rating 4.5/5. BIOL3057 -29860- 12-13: 12-13-Biofilms and Microbial Communities-29860. 2 lectures.

Montana State University MB500. 2003 Spring Semester. Environmental Microbiology Journal Club. Module leader. ENVE566 2001-2003 Fundamentals of Biofilm Eng. 1 lecture.

Guest lecture EPID507: Microbial Control: Sterilization, Disinfection and Manipulation. “Anti-Biofilm Surface Modification, Design and Control Strategies”. Guest Lecture. U. Michigan. Feb. 2014.

University of Copenhagen Bacterial Biofilms and Their Role in Chronic Infections. Online Course. “Basic Understanding of Biofilm”. 5 online contact hours. Fall 2018.

Bacterial Biofilms and Their Role in Chronic Infections. Online Course. “Basic Understanding of Biofilm”. 5 online contact hours. Fall 2016.

Multimedia

Approaches to Biofilm-associated Infections: Evaluating Gaps in Standardized Methods for Clinical Applications. https://www.biofilmeducation.com/. Hamood, A., James, G., Stoodley, P. North American Center for Continuing Medical Education, LLC. Approved course credit: 1.00 CME. September 2019.

Biocomposites Biofilm Series. Biofilm. https://www.biocomposites.com/patient-zone/biofilm/ 2018. Answers to some of the common questions surrounding the causes, impact and treatment of Biofilm.

Microbial Monsters - Microbial gangs are organised killers. 2015. Cosmos Magazine, Apr-May, 62:61- 73. Australia’s National Popular Science Magazine. I was interviewed on the multiucellular nature of biofilms and quoted in the article.

Über das Leben im Biofilm (Life in the Biofilm). Radio interview with Wolfgang Däuble for Austrian Public Radio Station ORF1 Originally aired 8/18/14. http://oe1.orf.at/programm/379822.

Emerging Trends in Oral Healthcare 2003. Lecture included in web cast course for continuing education program as approved by the American Academy of Dental Hygiene. Available at http://www.sonicare.com/ in the “Dental Professionals” section.

US Public Broadcasting Service TV Documentary. Fighting for Life: Emelia's Story. Emelia, a young girl with cystic fibrosis, endures many treatments to control her symptoms. This story also takes us to the Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

MSU Center for Biofilm Engineering, where scientists work to understand bacteria in a new way, as multi- cellular organisms, in order to better treat some of the complications of Emelia's disease. Produced by Sara Shier for TERRA. Distributed by KUSM/Montana PBS, MSU Bozeman.

6

283 Stoodley, P. 2003. Biofilms: An Introduction to Microbial Biofilm Science and Engineering Concepts. Science e- learning DVD short course. Cytergy LLC. www.cytergy.com (out of production).

Sonic brushing and the delivery of fluoride through Streptococcus mutans biofilms. Aspiras, M., Stoodley, P. & de Jager, M. 2008. Trends & Applications DENTALTRIBUNE Asia Pacific. 12;14-15.

Publications and Presentations (full list may be included at the end of the CV or as a separate document):

Publication Metrics (04/28/2020) 236 peer reviewed research articles, reviews, conference proceedings and book chapters.

Google Scholar: Citations: 29,807, H-Index: 73. Scopus Author ID: 7003908836, Cited docs: 191, H-index: 61. PubMed Bibliography http://www.ncbi.nlm.nih.gov/sites/myncbi/1JQQ5idesq9kl/bibliography/40262443/public/?sort=date&dire ction=asc ending

Invited to speak at over 175 conferences and workshops, including 50 International Conferences in the US, Asia and Europe.

Graduate student supervision

PhD/MS (bold indicates graduated / completed), yellow highlight indicates still active

Role Details Main supervisor Kat Sicirik, MS Bioeng. OSU. Anti-biofilm wound dressings 12/30/19 - Committee member Samantha Locke (OSU College of Veterinary Medicine) Co-supervisor Alex Jackson (Mech. Eng., Uni Southampton). “Viscoelasticity and Julian Wharton main Associated- drag of Artificial and Natural Marine Fouling Biofilms”. supervisor. Funded by University of Southampton EPSRC DTP award EP/K503130/1award and Akzo-Nobel. Sept. 2019 – Sept. 2022. Co-supervisor Katie Roe (Biological sciences, Uni. Southampton). “Oral biofilms: A Jeremy Webb main fundamental understanding to target improved oral health and supervisor. breath freshness” University of Southampton. Funded by Mondelēz International. Oct 18 – Oct ’22. Committee member Varun Lochab (Mech. And Aero Eng., Prakash PI) AU 2019 - Committee member Yiwei Liu (Wozniak PI) SU 2019 - Committee member Pranav S J B Rana (Wozniak PI) AU 2019 - Committee member Audra Fullen (Raj Deora PI) BSGP SU 2019 - Committee member Jenna Sandala (John Gunn PI) BSGP Feb 2019 - Main supervisor Amelia Staats BSGP 7930. Feb 28 2019 - Lab rotation supervisor Erin Connors BSGP 7930. SU/2018 Committee member Mengfei Li. Notre Dame. Main Supervisor Prof. Robert Nerenberg. 4/24/18 -. Main Supervisor Devine Sindeldecker. May 2017 - BSGP 8999 - 219 (1620). Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Committee member and Heather Eggleston. Mar 2016 – Jun 2017. Supervisor - Dan Chair Wozniak. Committee member Matt Pestrak. Jan 2015 – 2019. The structure and regulation of Psl production during biofilm formation. Supervisor – Dan Wozniak.

7

285 Main Supervisor Jennifer Longyear. Nov 2012 – April 2020 “Developing remote sensing tools for quantifying ship-borne marine fouling biofilms”. Sponsored by 1851 Committee award and International Paint, Akzo- Nobel. Co-supervisor –Julian Wharton, Simon Dennington and Mark Moore (NOCS). Part Time PhD, with 2 years mat leave.

Main Supervisor Stefania Fabbri. Nov 2012 – May 2016. “Interfacial instability generation in dental biofilms by high velocity fluid flow for biofilm removal and antimicrobial delivery.” Philips Oral Healthcare and Faculty funding. Stefania completed a 1 year industry post doc with International paint as is currently doing a post-doc at the University of Liverpool. Main Supervisor Lianne Niehaus. Fall Semester 2010 – 04/13/2015. “Micro computed tomography to diagnose and quantify biofilms in central venous catheters”. Joint PhD studentship between School of Medicine and School of Engineering Sciences. Co-supervisor Saul Faust (Medicine), Luanne Hall-Stoodley (Medicine), Jeremy Webb (Biology), Stuart Clarke (Medicine). Lianne is currently in medical school. Main Supervisor Natalya Doroshenko. May 2009 – April 2014. “The effect of sub- MIC concentrations of antibiotics on the EPS chemistry of Staphylococcus epidermidis biofilms as related to antibiotic resistance”. SES PhD Studentship. Co-supervisors – Andrea Russel (Chemistry), Julian Wharton (Engineering), Phil Thurner (Engineering). Natalya was awarded a World University Network (WUN) award to research in Dr. Parsek’s lab at the University of Washington in Seattle. 2015 -3 yr post doc at the University of Sheffield, School of Clinical Dentistry. Main Supervisor Amir Rmaile. July 2009 – October 2013. “Mechanical Properties and Disruption of Dental Biofilms”. Philips Oral Healthcare PhD Studentship. Co-supervisors – Julian Wharton (Engineering), Phil Thurner (Engineering). Amir has been hired by Philips Research as a Research Dentist at their High Technology Research Park in Eindhoven. Amir was the recipient of the 2014 UAE International Dental Conference & Arab Dental Exhibition (AEEDC) Young Researcher Award. Co-chair, main research B. (Laura) Purevdorj-Gage. “Pseudomonas aeruginosa biofilm supervisor structure, behavior and hydrodynamics”. Microbiology, Montana State University. 2000- 2004. Graduated 2004. (Chair: J.W. Costerton). Committee member and Brett W. Towler. ”Modeling the non-linear response of mixed culture main research supervisor biofilm structures to and research supervisor turbulent flow”. Civil Engineering, Montana State University. 2002-2003. Graduated 2004. (Chair: L. McKittrick). Committee member B. Klayman. “Tracking the movement of biofilm cells.” Civil Engineering, Montana State University. 2003. (Chair A. Camper). Committee member and Todd Shaw. “Measuring and modeling the viscoelastic properties of main research supervisor biofilms.” Mathematics, MSU. 2002-2003. (Chair I. Klapper).

Industry Doctoral Training Centre (IDTC) EngD Environmental Advisor

Joanne Davies “Composite materials/structures – Damage tolerance criteria for aged ships“. March 2012 - Lauren Wallis. “Novel battery technologies for electric and hybrid electric vehicle applications” March 2012 - Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Matthew Streeter. “Integrated design for production to ensure sustainability in marine transportation” March 2012 - Michal Lewandowski. “Structural integrity of hydrogen pressure vessels for the transport industry” March 2012 – Sarah Neenan -

MS Supervisor Kat Sikiric (Biomedical Engineering. Fall 2019 - Supervisor Xiaoyu Li. Advanced Mechanical Engineering. The use of micro CT to detect bacterial biofilms 8

287 on medical devices. Co-supervisor – Phil Thurner (Engineering). Feb -Sept 2012. Supervisor Ponkrit Mekkhunthod. MSc Maritime Engineering. Antibiofouling properties of electrospray ionization surface coatings. UG Electrical engineering. Royal Thai Navy Academy. Co- supervisor – Simon Dennington (Engineering). Feb – Sept. 2012. Co-chair: Patrick M. Norris. “Ultrasonically controlled antibiotic release from hydrogel coatings for biofilm prevention”. Mechanical Engineering, Montana State University. 2002-2004. Graduated 2004. (Chair: A. Vinogradov). Research advisor: Michelle Wolf. ” Statistical analysis of biofilm structure”. Statistics, MSU. 2002. (Chair: M. Hamilton).

Graduate defense examiner / Graduate Student Faculty Representative

PhD External examiner Pavithra Raghuraman. “Anti-biofilm function-spacer-lipid coatings on medical surfaces”. Faculty of Design and Creative Technologies, Auckland University of Technology, New Zealand. Prof Stephen Henry (Primary Supervisor). Michael Lockwood. Modelling Biofilm Formation in a Dairy Wastewater Irrigation System. Process Eng. Massey University, New Zealand. Process Engineering. Main Supervisor Prof Steve Flint. Mar. 2018. Maijken Sonderholm. University of Copenhagen, Denmark. Main Supervisor: Prof. Thomas Bjarnsholt. 5/2317. Sudhir Kumar Shukla. Bhabha Atomic Research Centre, Trombay, Mumbai– 400085 INDIA 29th of February 2016. Steffen Eickhardt-Dalbøge. University of Copenhagen, Denmark. Main Supervisor: Prof. Thomas Bjarnsholt. 1/8/16. Ashkan Safari. Engineering and Materials Science Centre University College Dublin. “A Combined Experimental and Numerical Study of Biofilm Detachment.” Supervisors. Prof. Alojz Ivankovi, Prof. Eoin Casey. 3/10/15. Mahmoud Y. Alkawareek, School of Pharmacy, Queens University Belfast. “Antimicrobial applications of atmospheric pressure non-thermal plasma”. Main supervisor Brendan Gilmore. 927/13. Lindsey Glew, Plymouth University, School of Biomedical and Biological Sciences. “The effects of oxygen and reactive oxygen species on antibiotic resistance and microbial communities in chronic wounds. Supervisor Dr. Karen Tait. Summer 2013. Martin Weiss Nielsen. “Next generation in vitro systems for biofilm studies - a cystic fibrosis patient’s airway perspective”. Department of Systems Biology, DTU, Copenhagen (Chair Claus Sternberg). Fall 2012. Kim Everuss. “Marine microbial biofilms degrading squid pen, a natural chitinous substratum”. Professor Amanda Goodman, Flinders University Australia. Dec 2011. Joe Harrison. “Multimetal resistance and tolerance in microbial biofilms”, Calgary University. (Chairs Dr. Ray Turner and Howard Ceri) June 2008. N. Barraud. “Nitric oxide-mediated differentiation and dispersal in bacterial biofilms.” Biotechnology and Biomolecular Sciences, U. New South Wales, Australia. 2007. (Chair Prof. S. Kjelleberg). A.W. Cense. “A spray based method for biofilm removal”. Biomedical Engineering, Eindhoven Technical University, Eindhoven, The Netherlands. Graduated 2005. (Chair: Prof. M.E.H. van Dongen).

Internal Examiner Nicola Gibbins. Investigating a role for nitric oxide in the control of biofilm and zoonotic pathogen colonization of the spinach phylloplane. Supervisor: Prof. Bill Keevil (Biology). 7/2/13. David Walker. “Biofouling and its control for in situ lab-on-a-chip marine environmental sensors”. Supervisor: Prof. Bill Keevil (Biology). 2013. Nicholas Churton. Supervisor Dr. Jeremy Webb (Biol) and Stuart Clarke (Med) Maria Salta. “Natural products as marine antifoulants”. Engineering Sciences. University of Southampton. Supervisor Dr. Julian Wharton. 2011. Enyioha Kennedy. “Mutability and survival of Pseudomonas aeruginosa in multi-species drinking water biofilm communities. Biological Sciences, University of Southampton. Supervisor: Dr. Jeremy Webb. 2011. (Biology). Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Stephane Werwinski. “Sensor actuated smart interfaces incorporating bio-hybrid materials (biofilm sensing)”. Engineering Sciences. University of Southampton. Supervisor: Dr. Julian Wharton. 2010

Graduate Faculty Representative A. Yousefi Koupaei ”Interaction between fluid flow and biomaterials: Applications in cardiovascular and ocular biomechanics”. BIOMEDE-PH Prof. Cynthia Roberts 4/9/2020.

9

289 Junfeng Shi. “Development of Nanoelectroporation-based Biochips for Living Cell Interrogation and Extracellular Vesicle Engineering”. MECHENG-PH, OSU. 08/14/17. Jibin Zhang. “Identification of the important genes in specific tissues using microarray analysis, bioinformatical and molecular tools”. ANIMSC-PH, OSU. 04/08/15.

Mentoring Name Program Project Title Dates

Medical School Students (highlight and italics indicates still active) Justine 1st year Med Student OSU Development of a Tendon May Schneider12 Soft Tissue Biofilm Model 2020- for ACL Failure: A Cadaveric Laboratory Study Lisa 4th year Med Student and MPH Sept. Coleman 19 - Nick 1st year Med Student OSU Persistent Cultures After Sept Farrar12,18 Long- Term Antibiotic 16 - Release in Total Joint Arthroplasty James 1st year Med Student OSU Mapping biofilms on the Sept Moley4 failed 16 – total hips and knees Jun 18. Daniel 1st year Med Student. U. Toledo Antibiotic elution from bone Jun – O’Reilly cement Aug 2015 Iris H.D. 1st year Med Student. Indiana PNA FISH staining on Jun – Petersen failed ACLs Aug 2015 Alex 3rd year Med Student OSU Mapping biofilms on the Jun15 Dibartola5 failed - total knees and ACLs Nov 16 Douglas 1st year Med Student OSU S. aureus invasion of Apr Kyrouac osteoblasts 15- Mar 16 Cory 1st, 4th year Med Student OSU Developing Diagnostics for Jan 14 Knecht4 Advanced Competency Credit Detecting Bacterial Biofilms – in Jun 16 Failed Orthopaedic Implants Scott 1st year Med Student OSU Developing Antimicrobial Jan Diamond4,5 Materials and Strategies for 14- Reducing the Incidence of May Bacterial Biofilm Infection in 16 Orthopaedic Implants

Undergraduate Students Mentored

Peter Biomedical Fall Burback Science: 2019 - [email protected] Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

David [email protected]. Antibiotic tolerant Fall Cavanagh edu populations in 2019- biofilms Nick Biology, OSU Determining the impact of Sept Chandler [email protected] host- synovial fluid factors 18 - .edu on Staphylococcus aureus aggregation

Doug Microbiology, OSU Determining the impact of Sept1 Guzion8 host- synovial fluid factors 8- Aug on Staphylococcus aureus 19. aggregation

Anthony Li8 Pharmaceutical sci., OSU. Antibiotic Efficacy in Jun18 Treating Variant - Pseudomonas aeruginosa Jun19 and Staphylococcus aureus Biofilms

10

291 Jack Biochemistry, OSU. Analysis of antibiotic loaded Jun Brooks8,14, materials on Pseudomonas 17- 15,16,17 aeruginosa and sStaphylococcus aureus biofilm eradication. Honors Thesis Nov 2019. Fatima Microbiology, OSU. Antibiotic tolerant populations Jun 17 Warda in - Jama biofilms Feb 18 Katherine Biochemistry, OSU. Biofilms in orthopaedic Jan- Currie11 infections May ‘17 McGrath Health Sciences, OSU. Biofilms in orthopaedic Jan 16 Mary11,13 infections – Jun 18 Kelly Biology, OSU. Determination of the Elution Jan 16 Moore19,8 Kinetics of Antibiotic-Loaded - Calcium Sulfate Beads and Powdered Antibiotic Bolus for Surgical Site Infections". Luke Health Science, OSU. S. aureus invasion of Oct 15 Bushrow osteoblasts – Jul 16 Michael Biomed Engineering OSU. Assessment of Mechanical and Aug Young8 Antibiotic Elution Properties of 15- Novel Hollow Tube Surgical May Mesh 16. Lydia Mack7,8 Microbiology OSU. Pseudomonas aeruginosa Jan 15 Short- Range Signaling Protein – Influence May 16 On Biofilm Phenotypic Expression Nick Biomed Engineering OSU. Mechanical properties and Sep 14 Farrar6,7,8 antibiotic release from bone – cement. May ’15 - Casey Aerospace Eng. OSU. Testing of Various Antibacterial Sept Peters5,7 Materials Against Biofilm ‘14 – Formation via Surgical Beads. Sept 19. Anthony Microbiology OSU. Persister cell phenotype Sep 13 Falzarano9 – Jun 16 Aneka Jhass3 4th year Med Student UoS Chronic infection of mandibular Dec osteosynthesis plates has a 2012- biofilm July etiology*** 2013. Methini Medical Student, UoS The effect of sub-inhibitory Dec. Himayakanth concentrations of vancomycin 2012 an on biofilm matrix composition. Richard Mech Eng. UoS Biofilm growth on novel Feb Fraser engineered surfaces. 2012 Vidullan Aeronautics and Astronautics, Shear stress caused by Oct Surendran UoS powered 2010 - Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

brushing in a model of human teeth and gums

Krista Microbiology, MSU Proteomics of detached biofilm. 2002- Cooperstein1 2004 Paul Gentile Mech. Eng., MSU Mobile biofilm unit 2003- 2004 Zack Bell Microbiology, MSU. Proteomics of detached biofilm. 2002- 2003. Matt Winston Mechanical Eng., MSU Biofilm viscoelasticity and 2002- detachment 2003 Cory Rupp2 Mechanical Eng., MSU Biofilm viscoelasticity (Keck 2001- Fellow) 2003 Krista Ind. Eng., MSU Ultrasonic delivery of antibiotics 2002- vanBuren 2003 Margo Statistics, MSU Detached biofilm particles 2002 Schurman Scott Electrical Eng., MSU Rheometry of biofilms (Keck 2002 Warnick Fellow) Ryan Cargo Mechanical Eng., MSU Biofilm viscoelasticity (Keck 2001- 2002

11

293 Fellow) Chayne Chemistry, MSU Cell signaling proteins in biofilms 2000- Piscitelli1 2001 Jennie Microbiology, MSU Detached biofilm particulates 2000 Graham Susan Microbiology, MSU Detached biofilm particulates 1999 Ruone

OSU – Ohio State University, UoS - University of Southampton, MSU - Montana State University

Undergraduate and Medical Student Awards and Scholarships 1Recipient USP Scholarship, Honors Program. 22003 Goldwater Scholarship Nominee, 2003 William Parkins Engineering-Physics Award, 2003 Boeing Scholarship, 2002 Summer internship at the Princeton Plasma Physics Laboratory, 2003 Summer internship at the Los Alamos National Laboratory - Dynamics Summer School Program, National Science Foundation Graduate Fellowship, worth $121,500 over three years. 3Best BMedSci project. 4Recipient Bennett Medical Student Research Scholarship. 5Exceptional poster $1000 travel award at the OSU College of Medicine Trainee Research Day. 6Superior Poster award for his poster: N. Farrar, S. Diamond, C. Knecht, C. Peters, M. Swearingen, T. Nocera, P. Stoodley. “Short Term Release from Calcium Sulfate and Polymethyl Methacrylate Beads” at the Sigma Xi, Scientific Research Society conference November 7-9 at the Renaissance Glendale Hotel in Glendale, Arizona. Sigma Xi is an honor society for scientists and engineers, recognizing high school, undergraduate, and graduate student research. 7Recipient Research Scholar Award of $1000 from OSU Undergraduate Office. 8Research distinction. 9Poster prize UG research Denman Forum 10Advanced Competency credit 11 Recipient $ 3,500 OSU UGRO scholarship for 2017 summer research. 12 COM Medical Student Research Scholarship (MDSRS) funded by the Roessler Medical Student Research Scholarship. 13 2018 Denman Forum award for her poster "Mapping of Bacterial Biofilms on Orthopaedic Implants: A Case Study Approach Testing the Novel Candle Dip Method". Mary S. McGrath, James P. Moley BS, Alex DiBartola MD, Jeffrey F. Granger MD, Anne C. Sullivan MD, Devendra H. Dusane PhD, Paul Stoodley PhD. 14 MacNevin Memorial Award from the Department of Chemistry & Biochemistry scholarship of $2,000. 15 Enrichment grant from the Undergraduate Student Government of $700. 16 Second prize in the 24th Annual Richard J. and Martha D. Denman Undergraduate Research Forum in Emerging Human Infections category. “Analysis of Calcium Sulfate Antibiotic Bead Arrangement on Methicillin Resistant Staphylococcus aureus and Pseudomonas aeruginosa Biofilm Eradication”. Jacob Brooks, Casey Peters, Devendra Dusane, Jeff Granger, Anne Sullivan, Paul Stoodley. 17 Academic enrichment grant of $ 700 for Summer 2019 research “Antibiotic loaded calcium cement beads for treating orthopaedic infections” from OSU USG and an Arts and Sciences Honors Committee Undergraduate Research Scholarship/ International Research Grant award of $ 5,000 for his research proposal "Analysis of Antibiotic Bead Arrangement and Combination on Biofilm Eradication". 18 $ 1000 travel award at the 2019 OSU Research Day in the "Medical Student" category for his poster "Antibiotic Resistance and Variant Colony Production of MRSA Biofilms on Periprosthetic Joints". 19 $2000 Second-Year Transformational Experience Program (STEP) to participate in undergraduate research for the summer of 2019. 20 2020 COM Medical Student Research Scholarship (MDSRS) funded by a Medical Student Research Scholarship for $ 3000.

Medical Resident Intern Mentoring Shivani Chlorhexidine transport through biofilms 2008 Anand DMD Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Laura Mejía Modeling biofilm EPS 2008 MD Bimaljeet Biofilms on infected sutures and meshes 2008 Singh MD

Undergraduate mentoring: Research Experience for Undergraduates (REU) program. Aaron Jacobsen Biological Systems in Eng., University of Nebraska 2000 “Mechanical properties of biofilms” Kimberly White Microbiology, University of Missouri 2001 12

295 “Antimicrobial resistance of detached biofilm clumps” Heather Adams Genetics/Molecular Cell Biology, Washington State University 2002 “Detachment of an oral biofilm by powered brushing” Paul Gentile Mech. Eng. MSU. 2003 “Design and Development of a Mobile biofilm unit”. Sunhoo Kim Carnegie Mellon University 2005 “Biofilm formation in S. aureus”. Thomas Huy Vu Biological Sci/ University of Pittsburgh 2005 “Biofilm formation in S. aureus”. Amar Mehta Mathematics/Pre-Med, University of Pittsburgh 2006 “Staining and quantification of biofilm EPS” Ajeet Mehta Mathematics/Pre-Med, University of Pittsburgh 2006 “Staining and quantification of biofilm EPS” Ashley Boucek Molecular biology, Westminster College 2007 “Bacterial biofilms on surgical mesh” Brian Zembruski , Community College of Allegheny County 2007 “Assessing anti-biofilm urinary catheters” Andrew West, Phillips Academy Andover 2007 “Bacterial biofilms on surgical mesh” Megan Mather Biology / Pre-Med, Virginia Tech 2007 “Bacterial biofilms on surgical mesh” Elizabeth Hill Biology, CMU 2008 “Development of FISH techniques for tissues” Zachary Brown Chemistry, Washington and Jefferson College 2008 “Usnic acid in bone cement as an antibiofilm agent” Charlie Magovern Allegheny College 2008 “Biofilm growth systems for rheometry”

Visiting scientists hosted Nan Jiang, MD. Nanfang Dept of Orthopaedics & Traumatology, Nanfang Hospital, 9/1/19-8/31/20 Southern Medical University, Guangzhou, China. Prof. Ellen Gewalt, Duquense University. Antibiofilm surface modification for metals. 6-7/2012 Baroncini, Fabrizio. (PhD Student of Iolonda Francolini) 2011/12 Sapienza University of Rome (3-months) Grande, Rossella. (Post-Doc). Department of Drug Sciences, 2011 University of "G. d'Annunzio" Chieti-Pescara (2 months) deBeer, D. PhD. Max Planck Institute for Marine Microbiology, Bremen, Germany. 2007 “Microelectrodes and FISH on clinical and dental samples.” von Ohle, C. DMD. University Hospital, Tübingen, Germany. 2005 “Dental biofilms.” Gieseke, A. PhD. Max Planck Institute for Marine Microbiology, Bremen, Germany. 2005 “Microelectrodes and FISH on clinical samples.”

Visiting scientists trained in lab Alicia Macías-Valcayo. (PhD candidate, University Hospital Jimenez Díaz. Spain. Jul-Oct ‘19 Grande, Rossella. (Post-Doc). Department of Drug Sciences, 2008 University of "G. d'Annunzio" Chieti-Pescara (3 months) Iannitelli, Antonio. (PhD Candidate). Department of Drug Sciences, 2008 University of "G. d'Annunzio" Chieti-Pescara(3 months) Thornton, Ruth. University of Western Australia, Perth, Australia, PhD Candidate. 2005 “Imaging biofilms on adenoids”. Gondim-Almeida, Lys M. A., M.D. (Otolaryngology). Hospital Nossa Senhora de Lourdes, Brasil 2005 “Imaging biofilms on adenoids”. Francolini, Iolonda. Roma University. PhD Candidate, Chem Eng. (Chair: G. Donelli). 2003 “Ultrasonic delivery of antibiotics from hydrogels to bacterial biofilms”. O’Sullivan, Catherine. University of Queensland. PhD Candidate Env. Eng. (Chair: L. Blackall). 2003 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

13

297 “Cellulose degradation by biofilms”. Dunsmore, Braden. University of Exeter. PhD Candidate Biological Sciences (Chair H. Lappin-Scott). 2000 “Formation and behavior of SRB biofilms.” Allegheny-General Hospital resident research program *#Greenleaf, R. MD (orthopedic surg.) 2007-2009 “Antibiofilm poly methyl methacrylate additives” *Tower, I. MD. () (Main advisor: S. Kathju) 2007-2009 “Biofilms on sutures and meshes” Sekhon, S. MD. (internal med.) 2007-2008 “Biofilm infections of surgical meshes” Gahlot, V. MD. (internal med) 2007-2008 “Biofilm infection-resistant implants for otologic application” Hillman, T. MD. (ENT). (Main Advisor: C.J. Post ) 2005-2007 “Biofilm resistant myringotomy tubes” Braxton, E. MD. (Neurosurgery) (Main advisor C.J. Post) 2005 “Biofilm infections in brain shunts”

* Presented at resident day research program. # Recipient AGH Residents Research Day 2009 1st Prize

Mentoring /Supervising Post-Docs Dr. Niraj Gupta (OS). Orthopaedic biofilms. March ’20 - present Dr. Erin Gloag (OSU). Rheology of dental biofilm. April 1 ‘19 - present Dr. Tripti Gupta (OSU). Attachment of S. aureus aggregates to orthopaedic materials. Feb ’19 - present Dr. Matt Pestrak (OSU). Kinetics of S. aureus aggregate formation. Jan ’19 – Oct ’19. Young Investigator award. Montana Biofilm Meeting July 16-18 2019. $750 travel stipend and waived registration. Dr. Robert Hull (Biology U. Southampton) Feb ’18 – Feb ‘20 Dr. Yalda Khosravi (OSU). Dental biofilm models. Sep ‘16 - present Dr. Devendra Dusane (Ohio State University) Jan ‘15 – Dec ‘19 Young Investigator travel award to Montana Biofilm Meeting, the Center for Biofilm Engineering, Bozeman MT to present his work “Strategies to treat intracellular and biofilm forming Staphylococcus aureus in orthopedic infections.” July 19-21, 2016. Dr. Matt Swearingen (Ohio State University) Oct ‘13 – Sept ‘15 Dr. Simon Dennington (Engineering, U. Southampton, UK) Apr ’16 – Apr ‘17 Recipient NIH/NAID T32 1 year Post-Doctoral Training Award Sept. 2014. Dr. Robert Howlin (Molecular Microbiology, U. Southampton, UK) 2012-2017 Dr. Karthik Sambamamoorthy (Molecular Microbiology, ASRI, 2007-2008 Pittsburgh) Dr. Laura Nistico (Clinical Microbiology, ASRI, Pittsburgh) 2004-2009 Dr. Duc Nguyen (Chemical Engineering, ASRI, Pittsburgh) 2004-2006

RESEARCH SUPPORT Current

Akzo-Nobel and EPSRC Doctoral Training Partnership (DTP) University of Southampton, UK. Sept. 26 19-Sept. 25 22 PI “Viscoelasticity and Associated-drag of Artificial and Natural Marine Fouling Biofilms”. £ 87,500 (£ 52,497 cash and £ 24,000 in kind A-N) 0 calendar

NIH S10 S10OD025008 PI “Structured Illumination Microscope for high resolution imaging of cells and tissues”. 09/04/2019 – 09/03/2020 $593,440 0 calendar This award is supplemented with a $ 155,000 CCC IRP award (46050-110439) and an OR award of $ 100,000 for remodeling and microscope upgrade. $155K using IRP funds. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Co-I (Prakash, Shaurya PI) “Detection, identification, and quantification of microbial contamination 0 calendar

14

299 in P&G production lines through multi-technique probing informed by machine learning”. Procter & Gamble Co $ 170,000 (DC+IDC) 07/01/2019 - 06/30/2020

PI. “Efficacy of antibiotic loaded high purity calcium sulfate beads against single and multispecies biofilms: an in vitro study.” Biocomposites Ltd. $ 403,305 (DC+IDC) (£ 322,644)06/1/19-5/31/21

PI. “The influence of solutions of an active agent on pregrown biofilms grown from human saliva / plaque using indentation rheometery and confocal microscopy”. Colgate-Palmolive. $ 65,000 May 1 ‘19 – Oct 31 ‘19.

Co-I. PhD studentship “Oral biofilms: A fundamental understanding to target improved oral health and breath freshness” University of Southampton. Mondelēz International Oct ‘18 – Sept ‘22

NIH R01 GM124436-01 “The role of Staphylococcus aureus aggregate formation in establishing biofilms in chronic orthopaedic periprosthetic joint infection.” Role PI. $ 2,041,856 (DC+IDCs) 9/1/2018 – 8/31/22 4.0 calendar

Past research support

PI. “Development of a Typodont Biofilm Model for Assessing Oral Plaque Biofilm Management Strategies”. Colgate- Palmolive. $ 360,000 (inc. of IDCs). (£ 259,750)Sept 1 2016 – Jun 30 2020 1.5 calendar

PI “Developing remote sensing tools for quantifying ship-borne marine fouling biofilms”. Royal Commission for the 1851 Exhibition and International Paint. £ 39,340 ($ 62,944). Jan 1 2013 – Jan 31 2020. University of Southampton.

PI. “Optimization of antibiotic calcium sulphate beads to combat periprosthetic biofilm infections”. University of Southampton. Biocomposites Ltd. £ 50,020. 1 Jan 19 – 30 May 19.

Co-I / Jeremy Webb PI. “A model oral system for oral healthcare risk assessment”. University of Southampton. NBIC/BBSRC and Unilever £ 79,470. 1 Dec 18 – 30 Apr 19.

PI: “Development of synthetic biofilm for calibrating the effect of coatings on reducing marine viscoelastic drag”. University of Southampton. NBIC/BBSRC and Akzo-Nobel £ 59,724 1 Dec 18 – 30 Apr 19.

PI: “Oral malodor microbiological testing” Role: PI 0.5 calendar Biolase Inc. $ 21,000 (DC+IDCs) 12/1/18 – 2/19/19

PI. “Antibiotic loaded high purity calcium sulfate beads for augmenting the antimicrobial efficacy of PMMA spacers in vitro”. Biocomposites Ltd. $285,884 (£ 206,273). 11/28/2017 - 05/31/2019. 1.8 calendar

Co-I “Benchmarking different antibacterial technologies using an oral in vitro system”. Unilever. Through University of Southampton. £ 160,336 ($ 222,216). 10/01/2017 to 09/30/2018. 1.2 calendar

PI. “Influence of Repeated Microspray Exposure on Community Development in an Oral Biofilm Model”. Philips Oral Healthcare. $ 120,000 (inc. IDCs). (£ 86,584) Oct 1 2016 – Sep 31 2018. 1.2 calendar

PI. “Biofilm infections in burn wounds”. PHPID. Co-I’s Shaurya Prakash and Vish Subramaniam (Mech and Aero Eng.) Chandan Sen and Sashwati Roy (Surgery), Dan Wozniak, (Microbial Infection and Immunity), Gayle Gordillo (). Public Health Preparedness for Infectious Disease. $ 100,000 (£ 72,153) (directs only). Jun 1 2016 – May 31 2018. 0 calendar

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

PI. “Measuring predictors of drag penalty of viscoelastic ship-fouling biofilms” Innovate UK in partnership with International Paint. £ 59,977.82 ($ 89,966.73) FEC funded at 80%. BBSRC BB/P004164/1 Feb 4 2016 – Feb 3 2017.

15

301 PI. “Influence of antibiotic-loaded Stimulan® on antibiotic efficacy and biofilm prevention and treatment – Biocomposites Phase IIV”. £104,654. Sept 1 2015 – Aug 31 2016.

Co-I “Indicators of bacterial colonization in failed ACL reconstructions” 06/01/2015 - 07/30/2016 . Arthroscopy Assn of North America. $24,945.00. PI: Flanigan, David C. OSU proposal GRT00037410. OSU.

PI. “Development of rheometer method for investigating the impact of marine biofilms on coating hydrodynamic properties”. International Paint. £ 36,144.50 ($ 55,499.70). 1/8/2015 – 6/30/2016.

Co-I “Targeting intracellular Staphylococcus aureus to lower recurrence of orthopaedic infection”. 1/1/15 – 12/31/15. Orthopedic Trauma Association. PI: Laura Phieffer. OSU. $29,849.38. OSU Project 60043856.

PI. “Delivery of fluoride and antimicrobial agents into dental plaque biofilms”. Philips Oral Healthcare PhD Studentship. £ 61,853 ($ 99,000). Nov/19/12-Nov/18/15. University of Southampton.

PI. “Development of rheometry for rapid screen of marine biofouling”. Feb 2015 – July 31 2015. £ 20,739 ($ 31,257). Akzo-Nobel.

PI. “Influence of antibiotic loaded Stimulan® on antibiotic efficacy and biofilm prevention and treatment Phase III”. Biocomposites. 18 June 2014 – 17 March 2015. £78,700 ($133,790). University of Southampton.

Reducing the burden of intravenous drug/nutrition delivery system infections using novel anti-biofilm strategies. PI Stoodley, P. and Faust, S. PhD Scholarship. £ 21,000 ($ 33,600). Oct 1 2010 – Sept 31 2014. University of Southampton.

Nano-graphene impregnated coatings for antimicrobial applications. Knowledge Transfer Partnership between the University of Southampton and Haydale Limited. Technology Strategy Board Project 508710. 2 years 2012 – 2013.

Development of ultrasonic technology for decontamination, debridement and cleaning. Co-I T. Leighton, P. Birkin, C.W. Keevil, P. Stoodley, R. Oreffo. Royal Society Mercer Award. £244,703.00. ($ 391,525) 10/1/2011- 10/31/2013. University of Southampton.

Influence of antibiotic loaded Stimulan® on antibiotic efficacy and biofilm prevention and treatment Phase II. Biocomposites. 7 Oct 2013 – 6 April 2014. £ 49,300 ($78,880). University of Southampton.

Influence of antibiotic loaded Stimulan® on antibiotic efficacy and biofilm prevention and treatment. Biocomposites. 21 Jan – 22 April 2013. £ 29,240 ($ 46,784). University of Southampton.

Mechanical Properties and Disruption of Dental Biofilms. Philips Oral Healthcare PhD Studentship. £127,892 ($ 204,000). 3 Years. 2009-2012. University of Southampton.

Antibiotic resistance of Staphylococcus epidermidis biofilms. Student exchange for my PhD student, Natalya Doroshenko, to spend 3 months in the lab of Dr. Parsek at the University of Washington. Worldwide Universities Network (WUN) Exchange Program. £ 4,954.37 ($ 9,822.92). Aug-Nov. 2011. University of Southampton.

Full biofilm study on PICC catheters. P. Stoodley and S. Faust. Semprus BioSciences. $ 13,543 (£8,473.48) 24 Jan 2011. University of Southampton.

R44 DC008907-02A1. Biofilm polymicrobial infection-resistant implants for otologic application. NIH – NIDCD. Labib, Mohamed E (Contact), Stoodley, Paul. $ 603,627. 2 years. 08/01/2009 to 07/31/2011. AGH, Pittsburgh. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Biofilm formation in IV systems. (PI) Baxter Healthcare. 4/1/09-7/1/09 $ 26,000. AGH, Pittsburgh.

Biofilm Affected by Power Toothbrushes IV (PI). Philips Oral Health Care. $117,594. 1/1/09-12/31/09. AGH, Pittsburgh.

16

303 Co-I R305M050264 (Hall-Stoodley, L. PI) Mentoring Teachers Through Pedagogical Content Knowledge Development Department of Education. 9/1/2005 – 8/31/2008. 1.2 yr. $638,554 AGH, Pittsburgh.

Whole Genome Amplification for Rapid Low Cost Genotyping (PI). J. Craig Venter Institute (NIH RO1 subcontract) $ 65,804. 7/1/08 – 6/30/10. AGH, Pittsburgh.

No Grant Number (Ehrlich, G. PI). Bayer Materials Science Anti-biofilm Coating Project Bayer 7/1/2006 – 6/30/2009 0.6 yr $58,632 AGH, Pittsburgh.

Biofilm Affected by Power Toothbrushes III (PI). Philips Oral Health Care. $106,000. 3/1/08-12/31/08. AGH, Pittsburgh.

Multi arm in vitro Chronic Rhinosinusitis (PI) Valam Corporation. $ 8,930. July – Aug., 2008. AGH, Pittsburgh.

Biofilm polymicrobial infection-resistant implants for otologic application (Labib PI). SBIR Subcontract with Novaflux. NIH SBIR -R43 DC 8907 PI Labib. 6/07-4/08. $ 71,997. AGH, Pittsburgh.

Laser disruption of biofilm (PI) Valam Corporation. $ 13,500. July 2007 - May 2008. AGH, Pittsburgh.

Optimization of Multiple Displacement Amplification (MDA) to Differentially Amplify Human and Bacterial DNA from Adenoid Biopsies. (PI). The Institute for Genomic Research (TIGR) subcontract on NIH RO1. 1/1/07 – 12/31/07. 1 yr $90,944.00 AGH, Pittsburgh.

Biofilm formation on endotracheal tubes. (co-PI with Kelly Wood M.D.). Allegheny General Auxiliary. 7/1/06-6/30/07. 1yr $ 5,000. AGH, Pittsburgh.

Otologic Implant Polymers for Biofilm Control in Chronic Otitis Media. Deafness Research Foundation (co- PI with Hillman, T.). 7/1/2006 – 6/30/2007. 0.6yr $18,182. AGH, Pittsburgh.

Conference on Biofilms (PI). NIH-R13DE018309-01ASM. 2007. $ 20,000. AGH, Pittsburgh.

Biofilm Affected by Power Toothbrushes II (PI). Philips Oral Health Care. $106,000. 12/1/06-12/31/07. MSU.

Biofilm affected by power toothbrushes. (PI) Philips Oral Health Care. $50,000. 8/1/05-12/31/05. MSU.

Development of Mobile Biofilm Unit (MBU). (PI) W.M. Keck Foundation through Portland State University subcontract with Sherry Cady, Associate Professor Geomicrobiology. $ 81,185.00. 1/1/2004-10/31/04. MSU.

Biofilm Remediation for Restoration of Contaminated Army Sites (Co-PI). DOD. STTR with MSE, Butte, MT. $ 99,966.00. 2004. MSU.

Rheology of Dental Plaque Biofilms. (PI) Philips Oral Health Care. $39,162.94. 4/1/03-12/31/03. MSU.

Development of Mobile Biofilm Unit (MBU). (PI) W.M. Keck Foundation through Portland State University subcontract with Sherry Cady, Associate Professor Geomicrobiology. $111,575. 10/1/2002-9/30/03. MSU.

2R01GM060052. Viscoelasticity and detachment in biofilms. (PI) National Institutes of Health RO1 grant, National Institute of General Medical Sciences. $ 716,000. 09/30/1999 - 09/29/2003. MSU.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Engineering Research Center for Interfacial Microbial Process Engineering (Co-PI, W.J. Costerton PI) ERC collaboration between CBE and Buddy Ratner, University of Washington Engineered Biomaterials Center. NSF $299,999 9/1/2002 – 8/31/2004. MSU.

Modelling antibiotic resistance in bacterial biofilms. (Collaborating investigator, Phil Stewart PI) NIH/NSF Joint Initiative $1,577,047. 1/1/00 - 12/31/04. MSU.

17

305 Bacterial biofilm formation as a coordinated multicellular process. (Collaborating investigator on 6 member team, Phil Stewart PI) W.M. Keck Foundation. $ 800,000, 2001-2005. MSU.

Visual evaluation of powered toothbrushing in-vitro and rheological properties of oral biofilms. (PI) Philips Oral Health Care. $ 59,586. 6/1/02-10/31/02. MSU.

Visualization of biofilm removal utilizing a powered ultrasonic toothbrush. (PI) Philips Oral Health Care. $44,030, Aug 1, 2001 to Jul 31, 2002. MSU.

Biofilm Capture of Chemical Warfare Agents in an Aqueous Stream. (PI) MSE Technology Applications, Inc. / DARPA $ 32,015. 11/1/01-11/1/02. MSU.

Assessment of attachment and shear removal of Pseudomonas aeruginosa cells from fluoropolymer surfaces. (PI) DuPont. 12/01/01 - 2/01/02. $ 14,787. MSU.

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

18

307 CURRICULUM VITAE

NAME: Purnima Dubey, Ph.D.

CURRENT ACADEMIC TITLE: Associate Professor

ADDRESS: Department of Microbial Infection & Immunity The Ohio State University 460 West 12th Avenue Columbus, OH 43210 (614) 292-9447 (office) E-mail: [email protected]

EDUCATION:

1986 The University of Chicago Chicago, IL A.B. (Biology)

1996 The University of Chicago Department of Pathology Chicago, IL Ph.D. (Immunology) Advisor: Hans Schreiber, M.D., Ph.D.

POSTDOCTORAL TRAINING:

1996-1997 Postdoctoral Fellow, University of Chicago, Dept. of Pathology Advisor: Hans Schreiber, M.D., Ph.D.

1997-2003 Postdoctoral Fellow, University of California, Dept. of Microbiology & Molecular Genetics Advisor: Owen N. Witte, M.D. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 2

EMPLOYMENT:

Academic Appointments

The Ohio State University

9/1/2017- present: Associate Professor Department of Microbial Infection & Immunity College of Medicine

4/1/2018-present: Courtesy faculty, Department of Microbiology Member, Microbiology Graduate Program College of Arts & Sciences

Wake Forest School of Medicine

7/2017-9/2017 Associate Professor with tenure Department of Pathology-Comparative Medicine

7/2015-9/2017 Associate Professor Department of Pathology-Comparative Medicine

2003-June 2015 Assistant Professor Department of Pathology-Tumor Biology

10/2003-9/2017 Cross-appointed faculty, Department of Microbiology & Immunology

7/2005-9/2017 Associate faculty, Molecular Medicine and Translational Science Graduate Program

7/2007-9/2017 Cross-appointed faculty, Department of Cancer Biology

OTHER PROFESSIONAL APPOINTMENTS AND INSTITUTIONAL SERVICE: Institutional Service: The Ohio State University Spring 2020 Poster judge COM Research Day Winter 2020- Co-leader, Immunology emphasis, BSGP program August 2019- M.D. application screener, College of Medicine Spring 2019- MII Space and Resource Committee Summer 2019 MII Retreat Committee January 2019- MII Seminar Committee 8/2018-present Member, Microbiology Graduate Studies Committee DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 3

6/2018 Graduate Faculty Representative for thesis defense (Megan Cramer) 10/2017-Dec 2019 Faculty advisor, Microbial Infection & Immunity Graduate Students (MIIGS) 10/2017-present MI&I Faculty Engagement and Networking Committee 9/2017 Judge, DHLRI Research Day

Institutional Service: Wake Forest School of Medicine

2003-9/2017 Member, Tumor Progression and Recurrence program Comprehensive Cancer Center

2004, 2005, 2007, 2014 Medical School Applicant Interview committee 2015

2004-2014 Evaluator, Standard Patient Assessment (SPA) exams for 1st and 2nd year medical students 2004-2007 Recruitment committee member Molecular & Cellular Pathobiology graduate program Department of Pathology

2005 Judge, Medical Student Research Day

2005-2007 Organizer, Bi-weekly floor meeting Department of Pathology, Section on Tumor Biology

2005-2009 Institutional Biosafety Committee

2005-2011 Recruitment committee member Molecular Medicine & Translational Science program Department of Internal Medicine

2006-2008 Member, Health & Effectiveness Council

2007-2011 Co-chair of Graduate Student Recruitment Molecular Medicine and Translational Science Ph.D Program

2008-4/17 Director, Cell and Virus Vector Lab (CVVL) Comprehensive Cancer Center 2009, 2010 Judge, Graduate Student Research Day

2010 Chair of Recruitment, Molecular and Cellular Biosciences (Track 4) Implementation committee 2011-2017 Recruitment Chair, Molecular and Cellular Biosciences DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 4

(Track 4)

2012-2014 Institutional Chemical Safety Committee 2012-2018 Institutional Animal Care and Use Committee

2016-9/2017 Member, Center for Vaccines at the Extremes of Aging WFUHS

EXTRAMURAL APPOINTMENTS AND SERVICE:

Journal Reviewer: Journal of Biomedical Optics Journal of Molecular Imaging and Biology Cancer Research Cancer Prevention Research Plos One Molecular Cancer Research Journal of Immunotherapy Journal of Pathology MBio Microbial Pathogenesis Pathogens and Disease Emerging Pathogens and Diseases Microbiology Journal of Immunology

Funding Agency Reviewer: Department of Defense Study Section Member Prostate Cancer Research Program, 2007, 2009-2012, 2015, 2016

Department of Defense Study Section Chair Prostate Cancer Research Program 2012-2014

Department of Defense Study Section Member Prostate Cancer Research Program, 2018

Ad hoc member, Vaccines Against Microbial Diseases (VMD) study section, February 2020, June 2020.

Editorial Boards: The Open & Nephrology Journal (2008-present)

Abstract Reviewer: World Molecular Imaging Congress, 2009, 2011-2015

Consultant: CellSight Technologies, San Francisco, CA.

External site visitor: George Washington University, M.S program in Bioinformatics and Molecular Biochemistry. Washington D.C. May 2016 DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 5

PROFESSIONAL MEMBERSHIPS: 2017-present American Society for Microbiology 2004-present American Association for Cancer Research 2004-present Society for Molecular Imaging

HONORS AND AWARDS:

1997-1998 Tumor Cell Biology Training Grant, UCLA 1998-2001 Cancer Research Institute Postdoctoral Fellowship 2001-2004 CaPCURE Young Investigator Award, Prostate Cancer Foundation 2006-2007 Prostate Cancer Foundation Competitive Research Award 2007 Participant, NIH/NCI Cancer Research Imaging Camp 2012-2013 Career Development for Women Leaders, Office for Women i n Medicine & Science, Wake Forest University 2017-2018 FAME Program, The Ohio State University

INVITED PRESENTATIONS AND SEMINARS:

International Research Group on Immunoscintigraphy & Therapy Congress, “Positron- emission tomography based imaging of anti-tumor T cell responses in vivo” Capri, Italy, June 2003. University of Chicago “ Non-invasive imaging of CD8+ T cell localization and function”, Chicago, IL. June 2006 Hanes Magnet School, “How does cancer develop?” Winston-Salem, NC Fall 2008 American Cancer Society, “Imaging immune responses to prostate cancer” Hillsboro, NC Fall 2009

Taiwan Society for Molecular Imaging, September 2010 “Imaging Immune Cell Trafficking in Cancer, with a Focus on Bioluminescent and PET Imaging”

Wake Forest School of Medicine, Comprehensive Cancer Center 2006, 2008, 2010. Wake Forest School of Medicine, Lipid Sciences Seminar Series 2008, 2010. American Cancer Society, “Immune responses to castration-resistant prostate cancer” Winston Salem, NC, 2011. Winston Salem State University MARC U*STAR program “Immune responses to castration-resistant prostate cancer”, Winston Salem, NC 2012. Winston Salem State University MARC U*STAR program “Immune inhibitory pathways in castration- resistant prostate cancer: Winston Salem, NC 2013. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 6

Wake Forest University, Department of Urology, Prostate Cancer Symposium “Amplification of immune inhibitory mechanisms in castration-resistant prostate cancer”, Winston-Salem, NC, March 2013. Genentech-Oncology-Biomarkers Early Stage, LLC “An immunological see-saw created by androgen ablation promotes castration-resistant prostate cancer growth”. South San Francisco, CA. July 2013. Tumor Immunology symposium, University of Chicago “Amplification of immune inhibitory mechanisms following androgen ablation therapy for prostate cancer”, Chicago, IL February 2014. Prostate Cancer Symposium “Challenges and Solutions”: “Prostate Stem Cell Antigen: Biomarker and Therapeutic Target” Winston-Salem, NC June 2014 Janssen Research and Development, LLC “An immunological see-saw created by androgen ablation promotes castration-resistant prostate cancer growth”. Spring House, PA June 2014.

Respiratory Pathogens Research Center Annual External Advisory Board Meeting “Enhancement of anti- pertussis immune responses by Bordetella Colonization Factor A (BcfA)” NIH, Rockville, MD, March 2016

3rd Annual Prostate Cancer Symposium: Back to the Future “Immunotherapy for prostate cancer” Winston Salem, NC. April 2016

Ohio State University Microbial Infection and Immunity Department “Remodeling Immune Responses to Improve Therapeutic Efficacy in Murine Models of Cancer and Infectious Disease” Columbus, Ohio, September 2016

National Institute of Immunology, New Delhi, India “Remodeling Immune Responses to Improve Therapeutic Efficacy in Murine Models of Cancer and Infectious Disease” New Delhi, India, December 2016

Respiratory Pathogens Research Center Annual External Advisory Board Meeting “Enhancing the efficacy of pertussis vaccines: Modulation of immune responses by BcfA” Rochester, NY, March 2017

Texas BioMed, Southwest National Primate Research Center “Enhancing the efficacy of pertussis vaccines by the novel adjuvant, BcfA” San Antonio, Texas, April 2017 DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 7

Vaccines R&D 2017 “Reshaping acellular pertussis vaccine-induced immune responses by the novel adjuvant, BcfA” Washington D.C., November 2017

The Ohio State University, Department of Microbial Infection & Immunity “Development of improved acellular vaccines against Bordetella species” November 2017

The Ohio State University, Department of Microbiology “Development of improved acellular pertussis vaccines that prevent B. pertussis colonization and transmission” January 2018

The Ohio State University, Infectious Disease Institute “Defining vaccine-induced T cell correlates of protection against B. pertussis” March 2018

New Albany Middle School “Careers in Biological Sciences” New Albany, OH, May 2018

Vaccines R&D 2018 “Improving acellular pertussis vaccine efficacy by intranasal delivery of BcfA-adjuvanted vaccines” Baltimore, MD, November 2018

International Emerging Infectious Diseases Conference 2018 “Development of next generation acellular pertussis vaccines to combat pertussis resurgence” Mumbai, India, December 2018

Advanced Centre for Treatment Research & Education in Cancer, India “Modulation of the tumor immune microenvironment of castration-resistant prostate cancer” Mumbai, India, December 2018

The Everett D. Reese Immunology and Rheumatology Research Seminar Series Ohio State University “Modulation of the tumor immune microenvironment to prevent castration-resistant prostate cancer growth” January 2019

University of Ferrara “Personalized neo-antigen vaccines to prevent progression of castration-resistant prostate cancer” Ferrara, Italy, March 2019 DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 8

NIAID Conference: Pertussis Waning Immunity “Durable Vaccine Immunity” September 2019, Frederick, MD

GRANTS:

Current:

NIH/NIAID 03/05/2020-03/04/2022 1R21AI151867 “Subunit TB vaccines adjuvanted with BcfA” The goal of this project is to test the protective efficacy in a mouse model of heterologous prime-pull immunization with acellular TB vaccines containing immunogenic M.tb proteins adjuvanted with BcfA Role: P.I.

NIH/NIAID 06/01/2016-05/31-2021 1R01AI125560-04 (Dubey (contact), Deora, M.P.I.) “Enhancing efficacy of pertussis vaccines” The goal of this proposal is to investigate the ability of BcfA to elicit improved immune responses to the acellular pertussis vaccine, by remodeling antibody and T cell responses to the more protective Th1 phenotype. Role: MPI (contact)

Merck contract (Bitting, P.I.) 3/2016-8/2020 “Single-arm phase II combination study of low-dose paclitaxel with pembrolizumab in platinum- refractory urothelial carcinoma” Role: Subcontract P.I

Pending grants

NIH/NIAID 07/01/2020-06/31/2025

“Identification of novel immunogenic proteins from B. pertussis” The goal of this project is to identify novel proteins that are presented on MHC Class II and elicit CD4+ T cell responses. Role: P.I. Under revision for resubmission

Selected completed grants

Ohio State University, James Comprehensive Cancer Center 7/2018-12/2018 Molecular Carcinogenesis & Chemoprevention Seed Grant Award “Neo-antigens in prostate cancer” Role: P.I. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 9

NIH/NIAID 04/20/2016-3/31-2018 1R21AI123805-01 (Deora, P.I.) “Regulation of biofilm formation and pathogenesis in Bordetella pertussis” The goal of this proposal is to understand the role of an essential regulator, BpsR, in biofilm formation. Role: Co-I

NIAID/RPRC Innovation Concept Grant (P.I. Deora) 09/15/2014-09/14/2017 “Enhancement of anti- pertussis immune responses by Bordetella Colonization Factor A (BcfA)” The goal of this project is to investigate the adjuvant function of BcfA, and its ability to improve protective immune responses induced by the acellular pertussis vaccine. Role: Co-I

Wake Forest Innovations SPARK Award (Dubey (contact), Wadas, 3/2013- MPI) 6/2014 “Non-invasive imaging of regulatory T cells in vivo”: Role: P.I. American Cancer Society Research Scholar Grant 7/2007- 6/2012 “Effector T Cell function in Androgen-Independent Prostate Cancer” Role: PI

NIH/NCI 1R21 CA 124457 4/2007- 3/2009 “Non-invasive Imaging of Tumor-reactive T Cell Subsets in vivo” Role: P.I. Prostate Cancer Foundation Competitive Research Award 2/2006- 2/2007 "In vivo evaluation of PSCA as a T cell target for immunotherapy of prostate cancer" Role: P.I.

CCCWFU Partner Grant 6/ 2006-4/2007 "Non-Invasive Imaging of Immune Modulation for Treatment of Melanoma" Role: P.I.

CaP CURE Young Investigator Award 7/2001-6/2004 "Positron emission tomography localization of cytotoxic T lymphocytes to bone metastases of prostate cancer." Role: P.I. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 10

BIBLIOGRAPHY:

Journal Articles:

1. Seung LP, Rowley DA, Dubey P and Schreiber H. 1995. Synergy between T-cell immunity and inhibition of paracrine stimulation causes tumor rejection. Proc Natl Acad Sci USA 1995;92:6254-6258.

2. Dubey P, Hendrickson RC, Meredith SC, Siegel CT, Skipper JCA, Engelhard VH, Shabanowitz, J, Hunt DF Schreiber H. The immunodominant antigen of an ultraviolet- induced regressor tumor is generated by a somatic point mutation in the DEAD box helicase p68. J Exp Med 1997;185:695-705.

3. Wick M, Dubey P, Koeppen H, Siegel CT, Fields PE, Fitch FW, Chen L, Bluestone JA and Schreiber H. Antigenic cancer cells can grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J Exp Med 1997;186:229-238.

4. Dubey P, Meredith SC, Siegel CT and Schreiber H. Tumor cells induce cytolytic T cells to a single immunodominant mutant peptide. J Immunother 1998; 21:277-282.

5. Dubey P, Wu H, Reiter RR and Witte ON. Alternative pathways to prostate carcinoma activate PSCA expression. Canc Res 2001;61: 3256-3261.

6. Ide H, Seligman DB, Memarzadeh S, Xin L, Horvath S, Dubey P, Flick MB, Kacinski BM, Palotie A and Witte ON. Expression of colony stimulating factor 1 receptor during prostate development and prostate cancer progression. Proc Natl Acad Sci USA 2002;11:14404- 14409.

7. Dubey P, Su H, Adonai N, Du S, Braun J, Rosato A, Gambhir SS, and Witte ON. Quantitative imaging of the T cell anti-tumor response by positron emission tomography. Proc Natl Acad Sci USA 2003;100:1232-1237.

8. Xin L, Ide H, Kim Y, Dubey P, Witte ON. In vivo regeneration of murine prostate from dissociated cell populations of postnatal epithelia and urogenital sinus mesenchyme. Proc Natl Acad Sci USA 2003;100:11896-11903.

9. Kim YJ, Dubey P, Ray P, Gambhir SS, Witte ON. Multimodality imaging of lymphocytic migration using lentivirus-based transduction of a fusion reporter gene. Mol Imaging Biol 2004;6:331-340.

10. Ali K, Dubey P, Roten S, Kute T. Effects of soy extracts on the growth of herceptin sensitive and resistant breast cancer cells in vitro and in vivo. JNCAS 2006;122:19-28.

11. Moore ML, Teitell MA, Kim Y, Watabe T, Reiter RE, Witte ON, Dubey P. Deletion of PSCA increases metastasis of TRAMP-induced prostate tumors without altering primary tumor formation. Prostate. 2008;68:139-151.

12. Akins J, Dubey P. Non-invasive imaging of cell-based therapies for the treatment of cancer. J. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 11

Nuc. Med, 2008, 49:180S-195S; doi: 10.2967/jnumed.107.045971.

13. Wang G, Shen H, Liu Y, Cong A, Cong W, Wang Y, Dubey P. Digital spectral separation methods and systems for bioluminescence imaging. Opt Express, 2008, 16:1719-1732.

14. Sukumar N, Love C, Conover MS, Kock ND, Dubey P. Deora R. Active and passive immunization with Bordetella Colonization Factor A (bcfA) protects mice against respiratory challenge with Bordetella bronchiseptica. Infect Immun, 2009, 77:885-95. Epub 2008 Dec 8.

15. Akins, EJ, Moore ML, Tooze JA, Willingham, MC, Dubey P. Sequestration of effector cytolytic T cells in prostate tumor stroma is alleviated by concomitant castration and regulatory T cell depletion. Cancer Research, 2010. 70:3473-82. Epub April 20th 2010.

16. Tang S, Moore ML, Grayson JM, Dubey P. Increased CD8+ T cell function following castration and immunization is countered by parallel expansion of regulatory T cells. Cancer Res 2012; 72:1975-85. Epub 2012 Feb 28

17. Tang S, Mishra M, Frazier D, Moore ML, Inoue K, Deora R, Sui G, Dubey P. Positive and negative regulation of Prostate Stem Cell Antigen expression by Yin Yang 1 in prostate epithelial cell lines. Plos One. 2012; 7(4):e35570.doi:10.1371/journal.pone.0035570

18. Dubey, P. Reporter gene imaging of immune responses to cancer. Theranostics.2012;2:355-62. Epub 2012 Apr 4. Invited review.

19. Stovall DB, Wan M, Zhang Q, Dubey P, Sui G. DNA vector-based RNA interference to study gene function in cancer. J Vis Exp. 2012 Jun 4;(64). pii: 4129. doi: 10.3791/4129

20. Krishnan B, Smith TL, Dubey P, Zapadka ME, Torti FM, Willingham MC, Tallant EA, Gallagher PE. Angiotensin-(1-7) attenuates metastatic prostate cancer and reduces osteoclastogenesis. Prostate. 2013 Jan;73(1):71-82. doi: 10.1002/pros.22542. Epub 2012 May 29.

21. Tang S, Dubey P. Opposing effects of androgen ablation on immune responses to prostate cancer. Oncoimmunology. 2012 Oct 1;1(7):1220-1221.

22. Nagalla S, Chou JW, Willingham MC, Ruiz J, Vaughn JP, Dubey, P, Lash TL, Hamilton- Dutoit SJ, Sotiriou C, Black MA, Miller LD. Interactions between immunity, proliferation and molecular subtype in breast cancer prognosis. Genome Biol. 201314(4):R34.

23. Taneja NK, Ganguly T, Bakaletz LO, Nelson KJ, Dubey P, Poole LB, Deora R. D-alanine modification of a protease-susceptible outer membrane component by the Bordetella pertussis dra locus promotes resistance to antimicrobial peptides and polymorphonuclear leukocyte- mediated killing. J Bacteriol. 2013 Nov;195(22):5102-11. doi: 10.1128/JB.00510-13. Epub 2013 Sep 6

24. Cattelan N. Dubey P, Arnal L, Yantorno OM, Deora R. Bordetella biofilms: a lifestyle leading to persistent infections. Pathog Dis. 2016 Feb;74(1). pii: ftv108. doi: 10.1093/femspd/ftv108. Epub 2015 Nov 19

25. Cattelan N, Jennings-Gee J, Dubey P, Yantorno O, Deora R. Hyperbiofilm formation by Bordetella pertussis strains correlates with enhanced virulence traits. Infect Immun. 2017 Sep 11. pii: IAI.00373-17. doi: 10.1128/IAI.00373-17. Print 2017 Dec 26. Jennings-Gee J, Quataert S, Ganguly T, D’Agostino R, Deora R, Dubey P. The adjuvant DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 12

Bordetella Colonization Factor A attenuates alum-induced Th2 responses and enhances Bordetella pertussis clearance from mouse lungs. Infect Immun, 2018. Mar 12. pii: IAI.00935-17. doi: 10.1128/IAI.00935-17. [Epub ahead of print]. *Selected for Spotlight 27. Dubey P. Visualization of Immune Cell Reconstitution by Bioluminescent Imaging. Chapter in: Reporter Gene Imaging: Methods and Protocols. Methods Mol Biol. 2018;1790:127-136. doi: 10.1007/978-1-4939-7860-1_10. 28. Caution K, Yount K, Deora R, Dubey P. Evaluation of Host-Pathogen Responses and Vaccine Efficacy in Mice. J Vis Exp. 2019 Feb 22;(144). doi: 10.3791/5893 29. Yount KS, Jennings-Gee J, Caution, K. Quataert S, Deora R, Dubey P. Bordetella Colonization Factor A (BcfA) elicits protective immunity against Bordetella bronchiseptica in the absence of an additional adjuvant. Infect Immun. 2019 Jul 15. pii: IAI.00506-19. doi: 10.1128/IAI.00506-19. [Epub ahead of print] *Selected for Spotlight 30. Wang P, Huo CX, Lang S, Caution K, Nick ST, Dubey P, Deora R, Huang X. Chemical Synthesis and Immunological Evaluation of a Pentasaccharide Bearing Multiple Rare Sugars as a Potential Anti-pertussis Vaccine. Angew Chem Int Ed Engl. 2020 Apr 16;59(16):6451-6458. doi: 10.1002/anie.201915913. Epub 2020 Feb 25.

Book chapters:

Dubey P, Seung LP and Schreiber H. The role of stroma in the growth or rejection of antigenic solid tumors. Review. In “The Immunology of Ocular Tumors” Editor Zierhut, M.; Ksander BR, Jager, M. 2002.

Dubey P. Visualization of Immune Cell Reconstitution by Bioluminescent Imaging. Chapter in: Reporter Gene Imaging: Methods and Protocols. Methods Mol Biol. 2018;1790:127-136. doi: 10.1007/978-1- 4939- 7860-1_10

Book editor:

Methods in Molecular Biology: Reporter Gene Imaging: Protocols and Methods. Editor: P. Dubey Springer. June 2018

Selected Abstracts

1. P Dubey, RC Hendrickson, SC Meredith, CT Siegel, DF Hunt, H Schreiber. The immunodominant antigen of a UV-induced regressor tumor is generated by a tumor-specific somatic point mutation in a DEAD-box protein. FASEB/American Association of Immunology, June 1996. *Invited speaker 2. P Dubey, T Watabe, KJ Singh, M Lin, A Donjacour, GR Cunha, RE Reiter, ON Witte. The basic biology of PSCA. CaP CURE, Oct 1999 3. P Dubey, H Su, N Adonai, S Du, J Braun, SS Gambhir, ON Witte. Visualization of adoptively transferred T lymphocytes homing to the tumor using positron emission tomography. Poster presentation, CaP CURE, Sep. 2001. Lake Tahoe, NV

4. P Dubey, H Su, N Adonai, S Du, J Braun, SS Gambhir, ON Witte. Visualization of adoptively transferred T lymphocytes homing to the tumor using positron emission tomography. Poster presentation, AACR Special Meeting on Prostate Cancer, Dec. 2001. Naples, FL DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 13

5. P Dubey, H Su, N Adonai, S Du, J Braun, SS Gambhir, ON Witte. Visualization of adoptively transferred T lymphocytes homing to the tumor using positron emission tomography. Poster presentation, Nanotriangle meeting, California Nanosystems Institute Marina del Rey, CA. July, 2002.

6. P Dubey, H.Su, N. Adonai, S Du, A Rosato, J Braun, SS Gambhir, ON Witte.. Quantitation of the T cell anti-tumor response by positron emission tomography. Society for Molecular Imaging. Boston, MA, August 2002.

7. P Dubey, H Su, N Adonai, S Du, A Rosato, J Braun, SS Gambhir, ON Witte. Quantitation of the T cell anti-tumor response by positron emission tomography. Poster presentation. Midwinter Conference of Immunologists. January 2003. Asilomar, CA

8. P Dubey, Y Kim, T Watabe, A Chin, R Reiter, M Teitell, ON Witte. Prostate Stem Cell Antigen (PSCA) is a modifier of epithelial cancer development and progression. Prostate Cancer Foundation, Lake Tahoe, NV. November 2003. 9. SK George, ML Moore, EJ Akins, P Ray, SS Gambhir and P Dubey. Introduction of molecular imaging reporter genes into murine lymphoid progenitors in vivo by lentivirus-mediated transduction of bone marrow. Society for Molecular Imaging, Colonne, Germany. September 2005.

10. EJ Akins, ML Moore, MC Willingham, and Dubey P. Decreased CTL Function Accompanies Increased Treg Localization to Prostate Tumors Following Androgen Ablation. AACR Special Conference on Tumor Immunology, Miami, FL, 2008.

11. E J. Akins, ML. Moore, MC. Willingham, P Dubey. Preferential localization to prostate tumor versus stroma of FoxP3+ cells over time after androgen ablation therapy. NCI Meeting 2009

12. S Tang, JA Tooze, P Dubey. Tumor-associated neutrophils have anti-tumor potential in prostate cancer. Cancer Research Institute Annual Meeting New York, NY. October 2009.

13. EJ Akins, ML. Moore, P. Dubey. Tracking CD8+ T cell localization and function using bioluminescent imaging. Society for Molecular Imaging, Honolulu, HI, September 2009.

14. ML Moore, S Tang, MC Willingham, P Dubey. PSCA haploinsufficiency enhances prostate tumor growth in prostate-specific Pten KO mice. First AACR International Conference on Frontiers in Basic Cancer Research. Boston, MA., October 2009.

15. S Tang, ML Moore, P Dubey. Tumor-specific CD8+ T cell responses are transiently magnified by concomitant castration and regulatory T cell depletion. AACR Special Conference in Tumor Immunology, Miami, FL. December 2010.

16. S Tang, JA Tooze, P Dubey. Treg infiltration in castration-resistant prostate cancer is controlled by tumor-associated neutrophils. AACR Annual Meeting, Chicago, IL. March-April 2012.

17. S. Tang, JA Tooze, JC Wood, R Deora, P Dubey. CCL20-dependent cooperation between Tregs and TANs promotes castration-resistant prostate cancer growth. AACR Special Conference in Tumor Immunology, Miami FL. December 2012. DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 14

18. P Dubey, J Jennings-Gee, Quataert S, Deora R. Remodeling of acellular pertussis vaccine immune responses by Bordetella Colonization Factor A heightens B. pertussis clearance from mouse lungs. 11th Annual Bordetella Symposium, Buenos Aires, Argentina, April 2016.

COLLABORATIONS

1. Sally Quataert, PhD University of Rochester Medical Center

2. Elaine Mardis, PhD Institute for Genomic Medicine Nationwide Childrens’ Hospital

3. Hans Schreiber, MD, PhD, University of Chicago

4. Jennifer Maynard, PhD, University of Texas at Austin

5. Ronald Hendrickson, PhD, Memorial-Sloan Kettering Cancer Center

6. Karen Scanlon, PhD and Nicholas Carbonetti, PhD, University of Maryland

7. Rhonda Bitting, MD, Wake Forest University School of Medicine

8. Jacob Yount, PhD, Ohio State University

9. Rich Robinson, PhD, Ohio State University

10. Debasish Sundi, MD, Ohio State University

11. Eugene Oltz, PhD, Ohio State University

12. Ken Oestreich PhD, Ohio State University

13. Ramesh Ganju, PhD, Ohio State University

14. Stephanie Seveau, PhD, Ohio State University DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 15

TEACHING PORTFOLIO

WAKE FOREST UNIVERSITY SCHOOL OF MEDICINE

Medical Students:

2004 Fall Semester Doctor-Patient Relationships (8/04-2/05) (Course Director: F. Celestino, M.D.) 30 contact hours

2013 Spring Semester Case Centered Learning (Course Director: David Jackson, M.D.) (3/13-5/13) 36 contact hours Graduate Students: LECTURES: 2004 Fall Semester Fundamentals of Immunology Department of Microbiology/Immunology Lectures “T cell receptor, CD4 and CD8 in T cell signaling” “Autoimmunity” 4 contact hours

2005 Spring Semester Topics in Cancer Biology Department of Cancer Biology Lecture “T cell recognized antigens as targets for immunotherapy” 3 contact hours

2005 Spring Semester Advanced Topics in Immunology Lecture “Non-invasive Molecular Imaging Techniques” 2 contact hours

2005 Fall Semester Fundamentals of Immunology Department of Microbiology/Immunology Lectures "T cell receptor, CD4 and CD8 in T cell signaling" "Autoimmunity" "Inherited immunodeficiencies, immune evasion, tumor immunity, DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 16 transplantation." 6 contact hours

2005 Fall Semester Advanced Topics in Immunology Lecture "Systems biology of tumors" 2 contact hours

2005, 2006, 2007 Molecular Basis of Human Disease Facilitator for case studies 12 contact hours

2006 Fall Semester Fundamentals of Immunology "T cell receptor, CD4 and CD8 in T cell signaling" "Autoimmunity" "Inherited immuno-deficiencies, immune evasion, tumor immunity, transplantation." Lectures 6 contact hours

2006 Fall Semester Topics in Cancer Biology Department of Cancer Biology Lecture “T cell recognized antigens as targets for immunotherapy” 3 contact hours

2007 Fall Semester Advanced Topics in Cancer Biology Facilitator 1 contact hour

2008-2016 Foundations of Translational Science Facilitator 6 contact hours

2008 Fall Semester Advanced Topics in Cancer Biology Facilitator 1 contact hour

2008 Fall Semester Fundamentals of Immunology Lecture “Tumor Immunity and Transplantation” 2 contact hours

2008 Fall Semester Cancer Biology Research Progress Report Facilitator 1.5 contact hours

2009 Spring Semester Cancer Biology Research Progress Report Facilitator 1.5 contact hours DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 1 7 2009-2012 Cancer Biology 712 Facilitator 1.5 contact hours

2009 Spring Carcinogenesis, DNA Damage and Semester Repair Lecturer 2 contact hours

2010 Fall Semester Advanced Topics in Cancer Biology Facilitator 1 contact hour

2010 Pathology 702 Lecturer “Autoimmunity” 1.5 contact hours

2011 Cancer Biology 711 Journal Club facilitator 1 contact hour

2012, 2013, 2014 MCB 702 Molecular & Cellular Biosciences 2015, 2016, 2017 Lecturer (Tumor Immunology, Autoimmunity & Transplantation) 3 contact hours each term 2012, 2013, 2016, MCB 723 Topics in Cancer Biology 2017 Lecturer/Journal Club facilitator “Tumor Immunology” 3 contact hours

2012, 2013 CABI 705 Lecturer/Facilitator “Tumor Immunology” 3 contact hours

2012, 2013, 2014 MCB 761 Lecturer “Tumor Immunology” 1.5 contact hours

The Ohio State University

2018 CBG7010/MICR7010: Cellular and Molecular Immunology 1. T cell maturation (Feb 2018) 2. T cell activation (March 2018) 3. Helper T cells (March 2018) 4. Tumor immunology (April 2018) 4 contact hours DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 18

2019 CBG7010/MICR7010: Cellular and Molecular Immunology 1. T cell maturation (Feb 2019) 2. T cell activation (Feb 2019) 3. Helper T cells (March 2019) 4. Tumor immunology (April 2019) 4 contact hours

2020 MII7010/MICR7010: Cellular and Molecular Immunology 1. Introduction to cells of the immune system (Jan 2020) 2. Dendritic cells (Jan 2020) 3. B cell maturation and activation (Feb 2020) 4. T cell maturation (March 2020) 5. T cell activation (March 2020) 6. Tumor immunology (April 2020) 6 contact hours

Oct 2018-- Co-director CBG7010/MICR7010: Cellular and Molecular Immunology (Lafuse/Dubey)

Fall 2019 IBGP7400 Selected Topics in Microbial Pathogenesis (facilitator for a 2 week block)

Fall 2019 Systems and Integrated Biology Journal Club Facilitator for 2 sessions

Fall 2019 MICR 7110 Microbial Pathogenesis and Immunobiology Lecturer, 1.5 hours

Trainees: Wake Forest School of Medicine

Graduate Students Elizabeth Joy Akins (Molecular Medicine) (2005-2010; Graduation May 2010) Shuai Tang (Molecular Pathology) (2008-2012; Graduation date August 2012)

Rotation Students: Wake Forest School of Medicine Rebecca Ivey (Molecular and Cellular Pathobiology; Fall 2004) Latoya Mitchell (Microbiology and Immunology; Spring 2005) Nicole Beauchamp (Molecular Medicine; Spring 2006) Tanya Sanders (Microbiology and Immunology; Spring 2008) Peter Alexander (Cancer Biology; Fall 2008) Guerry Jeanne Cook (Cancer Biology, Fall 2009) David Lemler (Cancer Biology, Winter 2010) Kacy Yount (MCB, Fall 2016) Aaron Deal (MCB, Winter 2017)

Medical Students Heather Clark (Summer 2006) DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 19

Interns Jacqueline Simmons (Forsyth Technical College) (Summer 2006) Bailey Fearing (Guilford College) (Spring 2007) Himani Vaidya (Salem College) (Spring 2009) Marisela Velasquez De Kamm (Fall 2016-Fall 2017)

Center for Excellence in Research Teaching and Learning (CERTL) Students Emeraghi David (Summer 2007) Anvita Bhardwaj (Summer 2008) Uma Gaddamanugu (Summer 2010) Neha Chatrath (Summer 2012)

Postdoctoral Fellows Sunil K. George, Ph.D (2004-2005) Meenu Mishra Ph.D (2007-2009) Jamie Jennings-Gee Ph.D (2015- 2017)

Technicians Miranda (2003- Moore 2010) Terry Bennett (2008- 2013) Nathan Iyer, (2008- M.S. 2013) Yelena (2013- Karpova 2017) Jane Strupe (2015- 2017)

Member of Thesis Committee: Wake Forest School of Medicine

Latoya Mitchell (Microbiology and Immunology, graduated 2009) Neelima Sukumar (Microbiology and Immunology, graduated 2009) Hetal Pandya (Molecular Genetics, graduated June 2011) Bhavani Krishnan (Molecular Genetics, graduated Summer 2011) Sarah Ryan (Cancer Biology, graduated April 2012) Stephanie Rockett (Chemistry, graduated June 2013) Guerry J. Cook (Cancer Biology, April 2012-2016) Marcela Haro (Microbiology and Immunology, December 2012-2016) Theresa Swanson Isbell (Chemistry, defended Dec 2014) Ivy Mead (Molecular Medicine and Translational Science, June, 2015) Sambad Sharma (Cancer Biology, April 2015-2016) Eric Routh (Cancer Biology, August 2015-August 2017) Allison Dyevoich (Physiology/Pharmacology, August 2016-August 2017) Anthony Dominijani (Cancer Biology, graduated M.S. July 2017)

Member of Thesis Committee: Ohio State University Megan Cramer (BSGP) Lauren Johnson (Micro) Kaitlin Reid (BSGP) Devin Jones (BSGP)

Trainees: The Ohio State University DUBEY, PURNIMA, PH.D. CURRICULUM VITAE PAGE 20

Kacy Yount (Biological Sciences Graduate Program, OSU) (2017-present) Co-mentor Kyle Caution, PhD (Postdoctoral fellow, March 2018-present) Jessica Brown (Biological Sciences Graduate Program) October 2018- Mohamed Shamseldin (Microbiology program) August 2019- Michael Haught Undergraduate student, 8/2017-12/2019 Pranav Rajan Undergraduate student, 6/2019- Rahul Rodrigues Undergraduate student, 6/2019-8/2019 Rotation students Marissa Gittrich (Micro) 10/2018-11/2018 Rajendar K Deora, PhD.

Academic Title Associate Professor

Address Department of Microbial Infection and Immunity The Ohio State University Wexner Medical Center Biomedical Research Tower 460 W. 12th Ave. 782 Columbus, OH 43210 (P) 614.688.9627; (F) 614.292.9616 [email protected]

Education

Ph.D., Microbiology University of Illinois-Chicago; 1991-1997 Dissertation: “Transcription of Pathogenesis Related genes in Staphylococcus aureus”. Advisor: Dr. Tapan K. Mishra

M.Sc., Biochemistry University of Calcutta–India; 1989-1991

B.Sc., Chemistry Honors University of Calcutta–India; 1986-1989

Postdoctoral Training

University of California at Los Angeles; 1997-2003

Academic Appointments Current Primary Department of Microbial Infection and Immunity; The Ohio State University Wexner Medical Center Secondary 1. Department of Microbiology; The Ohio State University 2. Department of Veterinary Biosciences; College of Veterinary Medicine at The Ohio State University

Page | 1 WFSM Primary 2010-2017 Associate Professor; Department of Microbiology and Immunology 2003-2010 Assistant Professor; Department of Microbiology and Immunology Adjunct 2016-2017 Center for Vaccines at the Extremes of Aging 2015-2017 Center for Molecular Communication and Signaling 2012-2017 Member, Molecular Medicine and Translational Sciences 2010-2017 Faculty Researcher, Office of Women in Medicine & Science 2009-2017 Wake Forest Institute of Regenerative Medicine 2003-2011 Program in Molecular Genetics

NIH Study Sections and other Grant Review Committees • Bacterial pathogenesis study section, BACP, Meeting 2020/10 • Small Business: Non-HIV Microbial Vaccine Development, 2020/05, ZRG1- IMM- N-07 • Small Business: Non-HIV Microbial Vaccine Development, 2020/05 ZRG1- IMM- R-12 • Member, Topics in Bacterial Pathogenesis, Meeting 2020/01. ZRG1-IDM-B- 80 • Review of Centers of Biomedical Research Excellence (COBRE) P20 Applications. 2019/10 ZGM1 RCB-4 (C1). June 2019. • CSR, NIH Review Panel, ZAI1-NVM-M, 2019/05. March 2019 • CSR, NIH review Panel, ZRG1 IDM-R (02); 2019/01. • NIH Support for Conferences and Scientific Meetings (R13); 2018/05; ZAI1 TS-M (M1) 1; 2018 • Member, Topics in Bacterial Pathogenesis, 2018/05 ZRG1 IDM-B (80) S, 2018 • Vidi programme NWO/ZonMw, The Netherlands, 2017 • Member, NIAID Adjuvant Development BAA panel, 2017 • CSR, NIH, Meeting 2017/10; ZRG1-IDM-T, 07/2017 • National Institute of General Medical Sciences, Special Emphasis Panel, 2017/10 ZGM1 RCB-4 (S1). SCORE applications, 06/2017 • National Institute of General Medical Sciences, Special Emphasis Panel, ZGM1 RCB-7 (SC), SCORE applications, 08/2016 • Special Emphasis Panel/Scientific Review Group 2016/05 ZRG1 IMM-R, 2016 • Special Emphasis Panel/Scientific Review Group 2016/05 ZRG1 IMM-R, 2016 Page | 2 • NIH Support for Conferences & Scientific Meetings (Parent R13), 2016/01 ZAI1 NVM-M (J2) 1, 2016 • Small Business: Non HIV Microbial Vaccine Research ZRG1 IMM-R(12), November 2015 • Small Business: Non HIV Microbial Vaccine Research ZRG1 IMM-R(12), July 2015 • Grant competition for Georgian microbiologists CDRF, 2014 • Special Emphasis Panel, NIH, December 2014 • Special Emphasis Panel, NIH, June 2014 • American Heart Association Microbiology BSc 1 Spring 2014 Peer Review Panel • NIH IDM-A study section, 2011 • American Heart Association Region 1&2 Microbiology and Microbial Pathogenesis Peer review panel, 2009 • External review panel Canadian Cystic Fibrosis Foundation, 2008, 2009 • American Heart Association Region II Microbiology and Immunology Peer review session, Tampa, Florida, 2008 • National Science Foundation, 2008 • Agency for Science, Technology and Research’s (A*STAR) Biomedical Research Council (BMRC), Singapore, 2008 • Wellcome Senior Research Fellowship in Basic Biomedical Science, 2004

Editorial Board Member/Scientific Reviewer

• Member, Editorial Board; Pathogens. 2019- • Editor. Pathogens and Disease. 2019- • Review Editor; Molecular Bacterial Pathogenesis for Frontiers Cellular and Infection Microbiology. 2018- • Member, Editorial Board; AIMS Microbiology. 2016-

Adhoc reviewer

Annals of Clinical Microbiology and Antimicrobials, AIMS Bioengineering, Applied and Environmental Microbiology, BMC Genomics, BMC Microbiology, BMC Veterinary Research, Biofouling; Cell, Clinical and Vaccine Immunology, Clinical and Experimental Immunology, Emerging Microbes and Infections, Infection and Immunity, Journal of Bacteriology, Journal of Biological Chemistry, Molecular Microbiology, Microbiology, Molecular and Cellular Biology, Pathogens and disease, Plos ONE, Plos Pathogens, Proceedings of National Academy of Sciences, Vaccine, Veterinary Microbiology

Institutional Service (Wake Forest School of Medicine) 2016 Medical School Curriculum Redesign Committee Page | 3 2015- Subcommittee on Pre-clinical Education 2015- Institutional Radiation Safety Committee 2014 LCME Accreditations Task Force 2014 AAALAC Accreditations Task Force 2013- Chair; Institutional Animal Care and Use Committee 14 2012- Vice-Chair, Institutional Animal Care and Use Committee 13 2012 Facilitator–Searching for Excellence and Diversity Workshop 2012 Molecular Medicine and Translational Science Graduate Program Policies & Procedures Committee 2012- Heightened Awareness Committee 13 2011- Institutional Animal Care and Use Committee 2011 MCB Recruitment Committee 2010- Diversity celebrations sub-committee 12 2010- WFUSM Dean’s Search Committee 11 2010 Member, Coordinating Committee for Strategic Plan Rollout 2010 Facilitator, Promotions Workshop Focus Group 2010 Faculty Council Working Group 2009- Council for Inclusion and Diversity 2009- Intramural Research Support Committee 12 2009 Trainee, Anti-Racism Workshop, Institute for Dismantling Racism 2008- Director, Bacteriology graduate course 13 2009- Nano-Technology Sub Committee, EH&S 11 2008- Chemical Safety Committee 11 2008- Dean’s Advisory Committee 11 2006- Institutional Animal Care and Use Committee 09 2007- Director, Microbiology and Immunology Departmental Seminar 10 Series 2006- Microbiology and Immunology Graduate Recruitment Committee 10 Institutional Service (Ohio State University) 2019- Faculty member. Committee on Academic Misconduct 2019- Education & Curriculum Committee: Charged with drafting and revising plans for Masters and PhD degree programs 2018- Member, Grant Mentoring Committee for Dr. Luanne Hall-Stoodley 2018 Member, IDI Interdisciplinary Seed Grant Review Team 2018 Judge, OSUWMC Research Day 2017- Departmental representative FAME

Teaching (Wake Forest School of Medicine) Medical School 2014-17 Cellular and Subcellular Processes, First year Medical Students Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

2014 Member, LCME Accreditations Task Force. Page | 4

338 2004-13 Medical Microbiology Infectious Disease 2004-15 Standardized patient assessment student faculty interaction and examination. 2013-17 Case centered learning

Graduate 2016-17 Director of MCB732; Fundamentals of Bacteriology 2008-14 Director of the Bacteriology Graduate Course 2011-17 MCB701, Molecular and Cellular Biosciences 2011-14 Director of MCB732; Fundamentals of Bacteriology 2004-14 MICR 750, Advanced Topics in Microbiology and Immunology 2008-11 MOGN731, Molecular Genetics 2003-10 MICR 703, Fundamentals of Bacteriology 2003-05 MICR 707, Introduction to the Scientific Method and Evaluating Scientific Literature

Honors and Awards 2017 Invited Editor, mBIO 2016 Member, Scientific Program Committee, Session co-chair, 11th International Bordetella Conference, Buenos Aires, Argentina 2014 Organizer and Convener, FASEB Meeting on Microbial Glycobiology 2012 Convener and Speaker, “Microbial Glycobiology and Glycobiotechnology”, American Society for Microbiology, 112th General Meeting 2009 Moderator, Biothreat Agent Workshop, Chapel Hill, NC 2006 FASEB Young Investigator Travel grant 2004- Thomas H. Davis Research Award, American Lung Association 05 North Carolina 1998- Microbial Pathogenesis Training Grant (NIH T32A107323) 00 1997- NIH Tumor Cell Biology Training Grant 98 1996 Wattamul Academic Excellence Award, University of Illinois, Chicago 1996 Travel award, Graduate College and Graduate Student Council, University of Illinois at Chicago 1994 Student Travel Grant, American Society for Microbiolog

Ongoing Research Support NIH/NIAID 1R21AI151867 03/05/2020-03/04/2022 “Subunit TB vaccines adjuvanted with BcfA” The goal of this project is to test the protective efficacy in a mouse model of heterologous prime-pull immunization with acellular TB vaccines containing immunogenic M.tb proteins adjuvanted with BcfA Role: Co-I, (3% effort).

NIH/NIAID 1R01AI25560-01 6/01/2016-5/31/2021 Page | 5 Enhancing efficacy of pertussis vaccines The goals of this project are to understand the mechanism of action of the adjuvant, BcfA, and to determine its ability to improve acellular pertussis vaccine-induced protective immunity. Role: PI Total Direct costs: $1,270,190

NIH/NHLBI 1R01 HL143076-01A1 9/1/19-6/30/22 Bioengineered Multi-Cell Type Organoids for Airways Disease Modeling The goals of this project are to generate bioengineered multi-cell type 3D airway organoids, supported by a lung extracellular matrix (ECM) biogel, to preserve airway epithelial phenotype and function. These organoids will be used to model CF disease pathology and the pharmacodynamic/pharmacokinetic efficacy of CFTR- modulators. Role: CoI, Murphy (PI)

Past Grant History NIH/NIAID 1R21AI123805-01 4/20/2016-3/31/2020 Regulation of biofilm formation and pathogenesis in Bordetella pertussis This proposal seeks to understand the regulation of biofilm development and pathogenesis in Bordetella pertussis. Total direct costs: $275,000; Role: PI

NIAID Contract 09/09/2014-08/30/2017 Enhancement of anti-pertussis immune responses by Bordetella Colonization Factor A (BcfA). This proposal focuses on examining the efficacy of the BcfA protein in enhancing B. pertussis immunity. Role: PI Total direct cost: $249,000. Wake Forest Innovations 2016-2017 Proof- of-concept studies for BcfA-mediated enhancement of vaccine-induced responses to B. bronchiseptica; Role: PI

Wake Forest Innovations and Commercialization 12/1/14-11/31/15 Fast-tracking the commercial potential of BcfA for animal and human therapy, $70,000

FASEB SRC R13 on Microbial Glycobiology. NICHD 6/01/14-4/01/15 $3,000

NIH RO1DK097697 2/1/11-1/31/16 Oxalate handling and Oxalobacter formigenes colonization in a mouse model Role: Co-I (PI: Dr. John Knight) Page | 6 NIH/NIAID 1R01AI075081 09/15/08-08/31/14 Bordetella Biofilms and Pathogenesis The goal of this project is to examine the function of the Bps polysaccharide in Bordetella biofilm development and virulence; Direct costs: $250,000/yr

Mid-Atlantic Affiliate Research Committee of the American Heart Association; 7/01/07-6/30/10; Role of a polysaccharide locus in the in vivo biofilm formation and virulence of Bordetella. Direct costs $60,000/yr

2R01HL058334-09A1 11/30/07-11/30/12 Control of P. aeruginosa algD transcription. The goal of this project is to examine the regulation of gene expression by the regulatory protein AmrZ, Role: Co-I (PI: Dr. Daniel Wozniak)

NIH/NIAID 1R21AI071054–01 9/01/06-8/30/10 Total direct costs $275,000 Role: PI

USDA 6/01/06-5/31/10 Identification of Novel Virulence Factors of Bordetella bronchiseptica by Functional Genomics The goal of this project was to study novel Bvg-regulated virulence factors of Bordetella bronchiseptica. Direct costs: $200,000/yr

WFUHS venture grant 11/01/06-10/31/07 Role of a Putative Polysaccharide Locus in Bordetella Biofilm Development Total direct costs $40,000. Role: PI American Lung Association of North Carolina 2004-2005 Thomas H. Davis Research Award Total direct costs $12,000 Role: PI Industry support

2010-2012 Boehringer Ingelheim ‘Bordetella vaccines’ 2008-2009 Merial Inc. ‘Characterization of B. bronchiseptica strains’

Patents and Licensed Technology

Bordetella Outer-membrane protein antigens and methods of making the same Deora, R, Mishra M, Sukumar N. Issued November 2010

Page | 7 Bibliography

• Wang P, Huo CX, Lang S, Caution K, Nick ST, Dubey P, Deora R* and Huang X*. Chemical Synthesis and Immunological Evaluation of a Pentasaccharide Bearing Multiple Rare Sugars as a Potential Anti-pertussis Vaccine. 2020. Angew Chem Int Ed Engl. 59(16):6451-6458. *Corresponding author.

Booth WT, Davis RR, Deora R, Hollis T. 2019. Structural mechanism for regulation of DNA binding of BpsR, a Bordetella regulator of biofilm formation, by 6-hydroxynicotinic acid. PLoS One. 14(11):e0223387.

Yount KS, Jennings-Gee J, Caution K, Fullen AR, Corps KN, Quataert S, Deora R*, Dubey P*. Bordetella Colonization Factor A (BcfA) Elicits Protective Immunity against Bordetella bronchiseptica in the Absence of an Additional Adjuvant. Infect Immun. 2019. 87(10). Print 2019 Oct. Selected for Spotlight *co-corresponding authors

Bracey DN, Seyler TM, Jinnah AH, Smith TL, Ornelles, DA, Deora R, Parks GD, Van Dyke ME and Whitlock PW. A porcine xenograft-derived bone scaffold is a biocompatible bone graft substitute: an assessment of cytocompatibility and the alpha-Gal epitope. 2019. Xenotransplantation. 2019. 26(5):e12534.

Caution K, Yount K, Deora R and Dubey P. Evaluation of Host-Pathogen Responses and Vaccine Efficacy in Mice. 2019. J Vis Exp. 22:(144). PMID:30855568 Cattelan N, Yantorno OM and Deora R. Structural Analysis of Bordetella pertussis Biofilms by Confocal Laser Scanning Microscopy. 2018. Bio Protoc. 2018, 8(15). pii: e2953. PMID:30160901

Little DJ, Pfoh R, Le Mauff F, Bamford NC, Notte C, Baker P, Guragain M, Robinson H, Pier GB, Nitz M, Deora, R, Sheppard DC, Howell PL. PgaB orthologues contain a glycoside hydrolase domain that cleaves deacetylated poly-β(1,6)-N-acetylglucosamine and can disrupt bacterial biofilms. 2018. Plos Pathogens. 14(4):e1006998. PMID:29684093.

Guragain M, Jennings-Gee J, Cattelan N, Finger M, Conover MS, Hollis T, Deora R. The transcriptional regulator BpsR controls the growth of Bordetella bronchiseptica by repressing genes involved in nicotinic acid degradation. 2018. J Bacteriol. Mar 26. pii: JB.00712-17. PMID:29581411.

Jennings-Gee J, Quataert S, Ganguly T, D'Agostino R Jr, Deora R∗, Dubey P∗. The adjuvant Bordetella Colonization Factor A attenuates alum-induced Th2 responses and enhances Bordetella pertussis clearance from mouse lungs. Page | 8 Infect Immun. 2018 Mar 12. pii: IAI.00935-17. doi: 10.1128/IAI.00935-17. [Epub ahead of print]. PMID:29531137. Featured in the spotlight section of the Journal. ∗ Corresponding author. Dorji D, Mooi F, Yantorno O, Deora R, Graham RM, Mukkur TK. Bordetella Pertussis virulence factors in the continuing evolution of whooping cough vaccines for improved performance. 2018. Med. Microbiol Immunol. 207(1):3-26. PMID:29164393

Cattelan N, Jennings-Gee J, Dubey P, Yantorno OM and Deora R. Hyperbiofilm Formation by Bordetella pertussis Strains Correlates with Enhanced Virulence Traits. 2017. Infect. Immun. 2017. 85(12). pii: e00373-17. PMID:28893915

Nicholson T, Brockmeier S, Sukumar N, Paharik A, Lister J, Horswill, A, Kehrli, M, Loving C, Shore S. and Deora R. 2017. The Bordetella Bps Polysaccharide is required for Biofilm Formation and Enhances Survival in the Lower Respiratory Tract of Swine. 2017. Infect. Immun. 19:85(8); pii: e00261-17. PMID: 28559403.

Carbonetti NH, Wirsing von König CH, Lan R, Jacob-Dubuisson F, Cotter PA, Deora R. Merkel TJ, van Els CA, Locht C, Hozbor D and Rodriguez ME. Highlights of the 11th International Bordetella Symposium: from Basic Biology to Vaccine Development. 2016. Clin Vaccine Immunol. 23(11):842-850. PMID:27655886.

Cattelan N, Dubey P, Arnal L, Yantorno OM and Deora R. Bordetella biofilms: a lifestyle leading to persistent infections. 2016 Pathog Dis. 74(1). PMID:26586694.

Little DJ, Milek S, Bamford NC, Ganguly T, DiFrancesco, BR, Nitz, M, Deora R∗ and Howell PL∗. BpsB is a Poly-β-1,6-N-acetyl-D-glucosamine Deacetylase Required for Biofilm Formation in Bordetella bronchiseptica. 2015. J. Biol. Chem. 290:22827-40. PMID:26203190. ∗ Joint corresponding author

Sukumar N, Nicholson TL, Conover MS, Ganguly T and Deora R. Comparative Analyses of a cystic fibrosis isolate of Bordetella bronchiseptica reveals differences in important pathogenic phenotypes. 2014. Infect. Immun. 82:1627- 37. PMID:24470470

Ganguly T, Johnson JB, Kock ND, Parks GD and Deora R. The Bordetella pertussis Bps Polysaccharide Enhances Lung Colonization by Conferring Protection from Complement-mediated Killing. 2014. Cellular Microbiology. 16(7):1105-18. PMID:24438122

Kumra-Taneja N, Ganguly T, Bakaletz LO, Nelson JK, Dubey P, Poole LB and Page | 9 Deora R. D-alanine Modification of a Protease-Susceptible Outer Membrane Component by the Bordetella pertussis dra Locus Promotes Resistance to Antimicrobial Peptides and PMN-mediated Killing. 2013. J. Bacteriol. 195:5102- 11. PMID:24013634 Knight J, Deora R, Assimos DG and Holmes RP. The genetic composition of Oxalobacter formigenes and its relationship to colonization and calcium oxalate stone disease. 2013. Urolithiasis. 2013. 41:187-96. PMID:23632911

Nicholson TL, Conover MS and Deora R. Transcriptome profiling reveals stage- specific production and requirement of flagella during biofilm development in Bordetella bronchiseptica. 2012. Plos One. 7(11):e49166. PMID:23152870

Tang S, Mishra M, Frazier DP, Moore ML, Inoue K, Deora R, Sui G, and Dubey P. Positive and negative regulation of Prostate Stem Cell Antigen expression by Yin Yang 1 in prostate epithelial cell lines. 2012. Plos One. 2012. Plos One. 7(4):e35570. PMID:22536409

Serra DO, Conover MS, Arnal L, Sloan GP, Rodriguez ME, Yantorno OM and Deora R. FHA mediated cell-substrate and cell-cell adhesions are critical for Bordetella pertussis biofilm formation in vitro and in the respiratory tract. 2011. PLoS ONE. 6(12): e28811. PMID:22216115

Register KB, Sukumar N, Palavecino EL, Rubin BK and Deora R. 2011. Bordetella bronchiseptica in a Pediatric Cystic Fibrosis Patient: Possible Transmission from a Household Cat. 2012. Zoonoses and Publ. Health 59(4):246-50. PMID:22212633.

Conover MS, Redfern CJ, Ganguly T, Sukumar N, Sloan G, Mishra M and Deora R. BpsR Modulates Bordetella Biofilm Formation by Negatively Regulating the Expression of the Bps Polysaccharide. 2012. J. Bacteriol. 194(2):233-42. PMID:22056934 Conover MS, Mishra M and Deora R. Extracellular DNA Is Essential for Maintaining Bordetella Biofilm Integrity on Abiotic Surfaces and in the Upper Respiratory Tract of Mice. 2011. Plos One. 6(2): e16861. PMID:21347299

Waligora EA, Ramsey DM, Pryor EE Jr, Lu H, Hollis T, Sloan GP, Deora R and Wozniak DJ. AmrZ beta- sheet residues are essential for DNA binding and transcriptional control of Pseudomonas aeruginosa virulence genes. 2010. J. Bacteriol. 192:5390-401. PMID:20709902.

Conover MS, Sloan GP, Love CF, Sukumar N and Deora R. The Bps polysaccharide of Bordetella pertussis promotes colonization and biofilm formation in the nose by functioning as an adhesin. 2010. Mol Microbiol. 77(6):1439-55. PMID:20633227

Page | 10 Sukumar N, Sloan GP, Conover M, Love C, Mattoo S, Kock, ND and Deora, R. Cross-species protection mediated by a Bordetella bronchiseptica strain lacking antigenic homologs present in the acellular pertussis vaccines. 2010. Infect. Immun. 78(5):2008-16. PMID:20176797

Sukumar N, Love C, Conover M, Kock ND, Dubey P and Deora R. Active and Passive Immunization with Bordetella Colonization Factor A (BcfA) Protects Mice against Respiratory Challenge with Bordetella bronchiseptica. 2009. Infect. Immun. 77:885-95. PMID:19064638. Featured in the spotlight section of this issue

Matthysse AG, Deora R, Mishra M and Torres AG. 2008. The polysaccharides cellulose, poly-ss-1,6-N- acetyl-D-glucosamine, and colanic acid are required for optimal binding of E. coli O157:H7 strains to alfalfa sprouts and K12 strains to plastic but not for binding to epithelial cells. 2008. Appl. Environ. Microbiol. 74:2384-90. PMID:18310435

Sloan-Parise G, Love CF, Sukumar N, Mishra M and Deora R. The Bordetella Bps Polysaccharide is Critical for Biofilm Development in the Mouse Respiratory Tract. 2007. J. Bacteriol. 189:8270-6. PMID:17586629

Inhibition of Pseudomonas aeruginosa Biofilm Formation with Bromoageliferin Analogs. Huigens R W 3rd, Richards JJ, Parise G, Ballard TE, Zeng W, Deora R and Melander C. 2007. Journal of the American Chemical Society. 129:6966-7. PMID:17500516

Sukumar N, Mishra M, Sloan GP, Ogi T and Deora R. Differential Bvg-Phase Dependent Regulation and Combinatorial Role in Pathogenesis of Two Bordetella Paralogs, BipA and BcfA. 2007. J. Bacteriol. 189:750-60. PMID:17351043

Parise G, Mishra M, Itoh Y, Romeo T and Deora R. Role of a Putative Polysaccharide Locus in Bordetella Biofilm Development. 2007. J. Bacteriol. 189:750-60. PMID:17114249

Lopez-Boado YS, Cobb L and Deora R. Bordetella Flagellin is a Pro- inflammatory Determinant for Airway Epithelial Cells. 2005. Infect. Immun. 73:7525-34. PMID:16239555

Mishra M and Deora R. Mode of Action of the Bordetella BvgA Protein: Transcriptional Activation and Repression of the B. bronchiseptica bipA Promoter. 2005. J. Bacteriol. 187:6290-6299. PMID:16159761

Page | 11 Mishra M, Parise G, Jackson KD, Wozniak, DJ and Deora R. The BvgAS signal transduction system regulates biofilm development in Bordetella. 2005. J. Bacteriol. 187:1474-1484. PMID:15687212

Dulatov S, Hodes A, Dai L, Mandhana N, Liu M,Deora R, Simons RW, Zimmerly S and Miller JF. Tropism Switching in Bordetella phage defines a family of diversity-generating retroelements. 2004. Nature. 431:476-81. PMID:15386016

Guo S, Gregory Z. Ferl Deora R, Reidinger M, Sheng Y, Kervin J, Loo JA and Witte O. A tyrosine phosphorylation site in Btk selectively regulates the efficiency of B cell calcium signaling by altering PLCgamma activation. 2004. Proc. Natl Acad. Sci. 101:14180-5. PMID:15375214.

Deora R. Multiple mechanisms of bipA gene regulation by the Bordetella BvgAS phosphorelay system. 2004. Trends in Microbiology. 12:2 63-65. PMID:15036321

Deora R. Differential regulation of Bordetella bipA gene: Distinct roles for different BvgA binding sites. 2002. J. Bacteriol. 184: 6942-6951. PMID:12446644.

M Liu, Deora R, Doulatov SR, M Gingery, Eiserling FA, Preston A, Maskell DJ, Simons RW, Cotter PA, Parkhill J, and Miller JF. Reverse Transcriptase- Mediated Tropism Switching in Bordetella Bacteriophage. 2002. Science. 295:2091-2094. PMID:11896279.

Deora R, Bootsma HJ, Miller JF and Cotter PA. Diversity in the Bordetella virulence regulon: transcriptional control of a Bvg-intermediate phase gene. 2001 Mol. Microbiol. 40:669-683. PMID:11359572.

Singh VK, Xiong A, Usgaard TR, Chakrabarti S, Deora R, Misra TK and Jayaswal RK. ZntR is an autoregulatory protein and negatively regulates the chromosomal zinc resistance operon znt of Staphylococcus aureus. 1999. Mol. Microbiol. 33:2000-2007. PMID:10411736

Deora R, Tseng T and Misra TK. Alternative transcription factor sigmaSB of Staphylococcus aureus: characterization and role in transcription of the global regulatory locus sar. 1997. J. Bacteriol. 179:6355- 59. PMID:9335283

Deora R and Misra TK. Characterization of the primary sigma factor of Staphylococcus aureus. 1996. J. Biol. Chem. 271:21828-34. PMID:8702982

Deora R and Misra TK. Purification and characterization of DNA dependent RNA polymerase from Staphylococcus aureus. 1995. Biochem. Biophys. Res. Commun. 208:610-616. PMID:7695614

Page | 12 Formal Presentations

2020 IDI annual microbial communities symposium. Invited speaker. Canceled due to COVID-19. 2020 Faculty seminars in Immunopathology. Invited speaker. 2020 MI&I Faculty work in Progress. 2020 Invited speaker, Indian Institute of Science Education and Research, Kolkata, India. Canceled due to COVID-19 2019 Invited speaker, ‘Changing Paradigms in Infectious diseases.’ Nanavati Super Speciality Hospital, Mumbai, India. 2019 Purdue University, Department of Biological Sciences 2019 Ohio University, Athens 2018 Ohio State University Gateway in India, Nanavati Hospital Mumbai, India 2018 University of Georgia, Athens 2018 Midwest Microbial Pathogenesis conference. 2018 “The Discover Seminar Series”. Nationwide Children’s Hospital 2017 Featured faculty speaker, Annual research Symposium Department of Microbiology; Ohio State University 2017 Veterinary Biosciences Graduate Seminar Series; Ohio State University 2017 Vaccines Research & Development; Baltimore, Maryland 2016 The South Texas Center for Emerging Infectious Diseases; University of Texas at San Antonio 2016 Translational Health Science and Technology Institute, Faridabad, India 2016 Department of Microbial Infection and Immunity, Ohio State University 2016 The University of Texas Rio Grande Valley Health and Biomedical Sciences 2016 International Bordetella Symposium at Buenos Aires, Argentina. 2015 RPRC Annual External Advisory Review Meeting; NIAID Respiratory Pathogens Research Center at the University of Rochester 2015 Molecular Communications and Signaling; Wake Forest University 2014 FASEB SRC Conference, Microbial Glycobiology; Eaglewood Resort, Itasca, IL 2014 Department of Basic Sciences, Touro University Nevada Page | 13 2014 Department of Biology; Appalachian State University 2013 Wake Forest Innovations Lunch & Learn seminar 2013 Program in Molecular Structure & Function; University of Toronto 2012 Convener and Speaker, “Microbial Glycobiology and Glycobiotechnology”. American Society for Microbiology, 112th General Meeting 2012 Indian Science Congress, Bhubaneswar, India 2011 Microbial polysaccharides of medical, agricultural, and industrial importance Carefree, Arizona 2011 University of Nebraska Medical Center Omaha 2011 Infectious Disease Research Conference, WFUHS 2010 Microcon 10, IMAM, India 2010 ‘Michelob Seminar’; Cancer Center and Cancer Biology; WFUHS 2010 Pennsylvania State University 2009 University of Minnesota, Minneapolis 2008 Microbial Functional Genomics Awardee Workshop, CSREES, USDA Washington DC 2008 Veterinary and Microbiological Sciences, North Dakota State University 2007 Indian Institute of Microbial Technology, Chandigarh, India 2006 FASEB Summer Research Conferences; Microbial Polysaccharides of Medical, Agricultural, and Industrial Importance, Tucson, AZ 2006 Department of Biophysics and Molecular Biology, University of Calcutta, Calcutta, India 2006 Department of Biochemistry, University of Calcutta, Calcutta, India 2005 Department of Microbiology and Immunology, University of Iowa 2005 Mid-Atlantic Microbial Pathogenesis Meeting, Wintergreen, Virginia 2004 College of Veterinary Medicine, North Carolina State University 2004 “Research in Progress” Department of Microbiology and Immunology, University of North Carolina at Chapel Hill 2002 Department of Microbiology and Immunology, Wake Forest University Health Sciences 2002 Department of Microbiology, University of Alabama at Birmingham 2002 Department of Microbiology and Immunology, West Virginia University Page | 14 2002 Montana State University 2001 Department of Microbiology and Immunology, Oregon Health Sciences University, Portland, Oregon 2000 Department of Microbiology and Immunology, University of California at Los Angeles 1999 Micorscience-STM Study day; Imperial College, London, UK

Podcast ASM MicroTalk: Infectious conversations about microbes: WHOOP WHOOP! THE PERTUSSIS VACCINE: https://www.asm.org/index.php/podcasts/microtalk/item/6899- whoop-whoop-the-pertussis- vaccine-rajendar-deora-ph-d

Convener/Chair/Organizer

2016 Member, Scientific organizing Committee, International Bordetella Symposium at Buenos Aires, Argentina 2014 Organizer chair and Speaker, FASEB conference on Microbial Glycobiology 2012 Convener and Speaker. “Microbial Glycobiology and Glycobiotechnology”, American Society for Microbiology, 112th General Meeting 2009 Co-Chair, The Biofilm Matrix, The 5th ASM Conference on Biofilms, Cancun, Mexico 2009 Biothreat Agent Workshop, Chapel Hill, NC

International Thesis Adjudicator

2018 Amrita Vishwa Vidyapeetham; Amrita Center for Nanosciences and Molecular Medicine; India 2018 National Institute of Technology Rourkela; Rourkela; India.

Graduate Students–Thesis Advisor WFSM

2003-2007 Gina Parise 2004-2009 Neelima Sukumar 2005-2010 Cheraton Love 2006-2011 Matt S Conover 2008-2011 Sonja Milek Page | 15 2011-2015 Crystal Redfern

OSU

2018- Audra Fullen 2017- Kacy Yount

Postdoctoral Fellows WFSM 2003-2008 Dr. Meenu Mishra 2007-2008 Dr. Banabihari Giri 2007-2009 Dr. Manish Bhardwaj 2009-2011 Dr. Neelima Sukumar 2009-2011 Dr. Gina Parise Sloan 2009-2010 Dr. Diego Serra 2010-2011 Dr. Neetu-Kumra Taneja 2011-2012 Dr. Matt S. Conover 2011-2012 Dr. Olcay Boyacioglu 2011-2014 Dr. Tridib Ganguly 2015-2017 Dr. Jamie Jennings-Gee 2016-2019 Dr. Manita Guragain 2016 Dr. Natalia Cattelan 2019-2020 Dr. Sharbani Kaushik

OSU 2018- Dr. Kyle Caution; Joint mentor with Dr. Purnima Dubey 2017-2018 Dr. Manita Guragain 2017-18 Dr. Natalia Cattelan 2019-2020 Dr. Sharbani Kaushik

Medical Student/Scientist/Visiting Fellows

2009 Pavani Thotakura 2013-2015 Dr. Dan Bracey 2009 Diego Serra 2015-2018 Dr. Natalia Cattelan 2018-Present Jessica Lizzeth Gutiérrez Ferman

Page | 16 Technicians

Anju Karki Katie Rice Haiping Liu Srija Ghosh Lubaina Puri

Undergraduate Research Sponsorships

LaTonya Mills Post-baccalaureate student, WFUHS Daya Stridiron Post-baccalaureate student, WFUHS Anju Karki Salem College Rouhani Pouya East Carolina University Surabhi Mathur United Kingdom Van Nguyen WFU undergraduate Ying Vang WFU undergraduate Srija Ghosh WFU undergraduate Lavern Eugene Keitt WFU undergraduate Caleb Murray WFU undergraduate Spencer Carter WFU undergraduate Anna-Grace Tribble WFU undergraduate Ayana Stukes Post-baccalaureate student, WFUHS Amy Medford WFU undergraduate Mary Finger WFU undergraduate Betlihem Ayalew Senior thesis student, WFU Carter Max Senior thesis student, WFU

OSU Bhavneep Kaur Kiersten Marion Meghan Deutsch Galen Silver

Service on Graduate Student Thesis Committees

WFSM Debbie M. Ramsey (PI Dan Wozniak) Kara Caitlin Mattos (PI Griffith Parks) Amity Roberts (PI D. Jackson (PI Dan Wozniak) Shayla Sean Reid) Stacy Reeves (PI Leslie Poole) Christie West (PI William Swords) Belen Belete (PI Young (PI Sean Reid) Lily Gordon (PI Sean Reid) Dan Wozniak) Anne Hasse Tart (PI Dan Chris Holder (PI Sean Reid) Wozniak) Chris Doern (PI Sean Reid) Ryan Davis (PI Thomas Hollis) Elizabeth Waligora Kyle Murrah (PI Ed Swords) (PI Dan Wozniak) Brittney Dodge (PI Tom Hollis) Amy Braden (PI Sean Reid) Cynthia Ryder (PI Dan Wozniak)

Page | 17 Amanda Hyre (Subhaschandrabose)

OSU

Yiwei Liu (PI Dan Wozniak) Medhat Shamseldin (PI Dubey Purnima)

Rotation Students WFS M 2004 Curtis Henry 2006 Matthew Byrd 2007 Caitlin Mattos 2009 Marlena Pichon 2011 Crystal Redfern 2011 Christie Young 2012 Jerome Mckay 2016 Shaina Aleese Yates

OSU 2017 Jenna Sandala 2017-18 Audra Fullen

Page | 18 CURRICULUM VITAE

Richard T. Robinson PhD Associate Professor

OFFICE ADDRESS: The Ohio State University HOME ADDRESS: 403 West North St College of Medicine Ostrander, OH 43061 Biomedical Research Tower Phone: 414-405-3528 460 West 12th Avenue Email: [email protected] Columbus, OH 43210 Phone: 614-292-0918 E-mail: [email protected]

PLACE OF BIRTH: Columbia, South Carolina

CITIZENSHIP: United States of America

EDUCATION: 9/1997 - 5/2001 BS, Anderson College, Anderson, SC 9/2002 - 5/2007 PhD, Dartmouth Medical School, Hanover, NH

POSTGRADUATE TRAINING AND FELLOWSHIP APPOINTMENTS: 5/2007 - 8/2009 Postdoctoral Fellow, Mentor: Dr. Andrea Cooper, The Trudeau Institute, Saranac Lake, NY 9/2009 - 9/2010 Ruth L. Kirschstein NRSA Postdoctoral Fellow, The Trudeau Institute, Saranac Lake, NY

FACULTY APPOINTMENTS: 1/2011 - 6/2017 Assistant Professor, MCW Dept of Microbiology & Immunology, Milwaukee, WI 7/2017 - 4/2018 Associate Professor, MCW Dept of Microbiology & Immunology, Milwaukee, WI 5/2018 - present Associate Professor, OSU Dept of Microbial Infection & Immunity, Columbus, OH

AWARDS AND HONORS: 2004 - 2006 Dartmouth, Immunobiology of Myeloid and Lymphoid Cells Training Grant (T32AI007363) 2006 – 2007 Dartmouth, Autoimmunity and Connective Tissue Biology Training Grant (T32AR007576) 2007 – 2009 Trudeau Institute, Research Training in Immunology and Infectious Diseases Training Grant (T32AI049823) 2008 Best Oral Poster Presentation Award – Riken RIKEN Research Center for Allergy and Immunology (RCAI) International Summer Program; Yokohama, Japan 2013, 2014 Medical College of Wisconsin Excellence in Medical Education Award - Immunology 2014 Anderson College Distinguished Alumni Award 2014 Richard C. Knudsen Memorial Publication Award, American Biological Safety Association 2015 Panel Discussion Leader, “Endpoints and readouts in vaccine efficacy studies in different hosts”, Many Hosts of Mycobacteria: A Comparative Symposium (Covington, LA) 2015 Co-Chair, “Molecular Regulation of Cytokine/Chemokine and Receptor Function” Block Symposium, AAI Annual Meeting (New Orleans, LA). 2015 AAI Early Career Travel Award and Invited Speaker, AAI Annual Meeting (New Orleans, LA). 2017, 2019 Panel Discussion Leader, “Nontuberculous Mycobacteria Infection.” Many Hosts of Mycobacteria 7th Annual Meeting (2017, Fort Collins, CO), as well as Many Hosts of Mycobacteria 8th Annual Meeting (2019, Bronx, NY) 2019 Member, Scientific Advisory Board, New York Immunology Conference (Albany Medical Center) 2019 Member, TB Advisory Board, AbbVie Pharmaceuticals (Clinical Translational Research Division) 2021 Lead Organizer, Many Hosts of Mycobacteria 9th Annual Meeting (Columbus, OH)

MEMBERSHIPS IN HONORARY AND PROFESSIONAL SOCIETIES: 2007 – Present American Association of Immunologists (Member) 2008 – Present American Society for Microbiology (Member) 2014 – Present International Cytokine and Interferon Society (Member) Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

NATIONAL ELECTED/APPOINTED LEADERSHIP AND COMMITTEE POSITIONS: 03/2 Reviewer, UK Medical Research Council Infections and Immunity Board 011 10/2 Reviewer, NIH Topics in Bacterial Pathogenesis Special Emphasis Panel 016 (IDM-B 80) 05/2 Reviewer, French National Research Agency (ANR), Immunology Panel 017 (CE15) 07/2 Reviewer, NIH Immunity and Host Defense Study Section (IHD) 018 10/2 Reviewer, NIH AIDS and Related Research Special Emphasis Panel (ZRG 018 AARR K)

356 06/2 Reviewer, University of Washington/Fred Hutch Center for AIDS Research 019 07/2 Reviewer, NIH U.S.-Brazil Collaborative Biomedical Research Program (ZRG1 019 IMM-N (50) R) 10/2 Reviewer, NIH Immunity and Host Defense Study Section (IHD) 019 12/2 Reviewer, NIH TB Meningitis Special Emphasis Panel (ZRG1 IDM-T (50) R) 019 01/2 Reviewer, Foundation Bettencourt Schueller (France) 020 03/2 Reviewer, Topics in Bacterial Pathogenesis Special Emphasis Panel (IDM-B 81) 020

EDITORIAL SERVICE

Ad Hoc reviewer: Infection & Immunity, Human Genetics, International Immunopharmacology, Clinical Microbiology Reviews, Clinical and Vaccine Immunology, Elsevier S&T Books, Cytokine, European Journal of Immunology, Journal of Immunological Methods, PLoS One, PLoS Neglected Tropical Diseases, PLoS Pathogens, Trends in & Metabolism, Journal of Immunology, mBio

Editorial Board: Clinical and Vaccine Immunology, ASM Press (term: 2016-2019); International Immunopharmacology, Elsevier (term: 2016-2019); Cytokine, Elsevier (term: 2017-2020); Lung, Springer Nature Press (term: 2020-2023)

RESEARCH GRANTS/AWARDS/CONTRACTS/PROJECTS:

ACTIVE Title: Department Startup Support Source: OSU Dept Microbial Infection & Immunity Role: Principal Investigator Dates: 05/1/2018 – 4/30/2022

Title: Regulation of human IL12/IL23 responsiveness by IL12RB1 Source: NIH – NIAID (R01AI121212) Role: Principal Investigator Dates: 05/1/2016 – 4/30/2021 (Y1-Y2: MCW; Y3-Y5: OSU) Direct Funds: $1,250,000 (total all years)

Title: T cell dysfunction in CF patients with nontuberculous mycobacteria infection Source: Cystic Fibrosis Foundation (Pilot & Feasibility Award Program) Role: Principal Investigator Dates: 02/1/2019 – 1/31/2020 Direct Funds: $140,000 (total all years)

Title: Single-cell RNA sequencing (scRNA-seq) of TB mouse lung Source: OSU Department of Microbial Infection & Immunity Pilot Award Role: MPI (Namal Liyanage, Haitao Wen, Rich Robinson) Dates: 04/19/19-12/31/20 Amount: $26,600

PENDING Title: Acellular TB vaccines adjuvanted with BcfA Source: NIH – NIAID (R21 AI151867) Role: MPI (Purnima Dubey, Kara Corps, Rajendar Deora, Richard Robinson) Dates: 04/01/2020- 03/31/2022 Amount: $275,000 (total over Y1-2) Note: Received fundable score; ZRG1 IMM-R (90) review on 11/21/19; award notice pending.

Title: Developing Cry3Aa-TB subunit antigen crystals as an oral TB vaccine Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Source: Hong Kong Food and Health Bureau, Health and Medical Research Fund (HMRF grant # 19180082) Role: Co-PI, (PI: Michael Kenneth Chan, The Chinese University of Hong Kong) Dates: 04/01/20-03/31/23 Direct Funds: HK$1,499,917 (subcontract to OSU: HK$516,039 = US$66,066) Note: This would fund BSL3 costs for our testing a novel oral vaccine formulations in our TB mouse model.

Title: Evaluation of immunogenicity of mycobacterial strains in C3HeB/FeJ mouse model of TB Source: National Council of Science and Technology (CONACYT) Role: Co-I. (PI: Mario Alberto Flores-Valdez, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco | CIATEJ · Guadalajara, Mexico) Dates: 06/01/20-05/31/22

358 Direct Funds: MXN$800,000 (subcontract to OSU: MXN$657,530 = US$35,000) Note: This would fund stipend/fringe support for Mexico®US Graduate Student exchange from CIATEJ to test various BCG mutants in the TB mouse model.

PREVIOUS Title: Zoonotic Tuberculosis (zTB) Workshop Source: OSU Infectious Disease Institute, Internal Event Mechanism Role: PI (Co-PI Shu-Hua Wang) Dates: 07/15/19 Direct Funds: $1,500

Title: Host Susceptibility to Pediatric Nontuberculous Mycobacterial Infection Source: MCW Research Affairs Committee Role: Co-Investigator (PI: Anna Huppler, MD; Pediatric Infectious Disease) Dates: 1/1/17-6/1/18 Direct Funds: $25,000

Title: M.tuberculosis-induced Alternative Processing of IL12RB1 mRNA Source: NIH - NIAID (R21 AI099661) Role: Principal Investigator (Co-PI, Mark McNally) Dates: 2/1/2013 - 2/1/2015 Direct Funds: $275,000 (total for all years)

Title: Influence of IL12Rβ1 on TH1/TH17 differentiation Source: BD Biosciences Role: Principal Investigator Dates: 6/1/2011 - 6/1/2014 Direct Funds: $10,000

Title: Characterization of aerosols generated during animal lung homogenization Source: American Biological Safety Association (ABSA) Role: Principal Investigator Dates: 2/1/2013 - 2/1/2015 Direct Funds: $20,000

Title: IL12Rβ1 Alternative Splice Variant in the Regulation of IL12 Family Signaling Source: NIH – NIAID (F32 AI084397) Role: Principal Investigator Dates: 9/2009 - 8/2010 Direct Funds: $50,000

PEER REVIEWED and/or INVITED PRESENTATIONS:

Regional 1. Robinson RT, Gorham JD, "Analysis of TCR Vβ-usage by intrahepatic T helper cells in a mouse model of autoimmune hepatitis." 2004. Poster, 30th Annual New England Immunology Conference., Woods Hole, MA, 2004 2. Robinson RT, Kitzmiller TJ, Gorham JD, "Experimental restriction of the CD4+ T cell receptor repertoire prevents immune mediated necroinflammatory liver disease in BALB/c-TGFβ1-/- mice.", 31st Annual New England Immunology Conference, Woods Hole, MA, 2005 3. Robinson, RT, SA Khader, ST Smiley and AM Cooper, (Oral presentation) "Yersinia pestis evades TLR4-dependent IL12p40-to-IL12(p40)2 conversion by dendritic cells in a temperature-restricted manner" 2008. Upstate Immunology Conference, 11th Annual Upstate New York Immunology Conference, Bolton Landing, NY, 2008 4. Claeys, TA* and RT Robinson. (*Oral Presenter) “Human T cell differentiation in pediatric nontuberculous mycobacterial disease.” Midwest Microbial Pathogenesis Conference, University of Notre Dave, South Bend, IN, 2017 5. Robinson, RT*. (*Oral Presenter) “Potential impact of environmental mycobacteria on TB diagnostics.” Zoonotic TB Workshop, Beaumont Healthcare Center, Westland, MI (07/15/19)

National 1. Poster: Acevedo, R, RT Robinson, S Cox and WS Baldwin, "4-Nonylphenol alters steroid hormone metabolism in liver of FVB mice." , Society of Toxicology 41st Annual Meeting, Nashville, TN, 2002 2. Poster: Robinson RT, Gorham JD, "Analysis of TCR Vβ-usage by intrahepatic T helper cells in a mouse model of autoimmune hepatitis.", American Association for the Study of Liver Diseases 55th Annual Meeting, Boston, MA, 2004 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

3. Poster: Robinson RT, Kitzmiller TJ and Gorham JD, "Experimental restriction of the CD4+ T cell receptor repertoire prevents immune mediated necroinflammatory liver disease in BALB/c-TGF- 1-/- mice.", 56th Annual Meeting American Association for the Study of Liver Diseases, Boston, MA, 2005

360 4. Poster: Robinson, RT, MA French, TJ Kitzmiller and JD Gorham, "Restriction of the CD4+ TCR repertoire prevents liver disease in TGF-β knockout mice: evidence for antigen specificity in a model of autoimmune hepatitis." Keystone Symposia: Autoimmunity and Immune Regulation. Breckenridge, CO, 2006 5. Poster: Robinson, RT, SA Khader, ST Smiley and AM Cooper, "Yersinia pestis evades TLR4-dependent IL12p40-to- IL12(p40)2 conversion by dendritic cells in a temperature-restricted manner", 95th Annual Meeting of the American Association of Immunologists, San Diego, CA, 2008 6. Invited Talk: “Regulation of Mycobacterium tuberculosis infection by IL12RB1”, Midwest Microbial Pathogenesis Conference, Milwaukee, WI, September 2012. 7. Invited Talk: “Regulation of Mycobacterium tuberculosis infection by IL12RB1”, Jeffrey Modell Foundation Human Immunology Symposium, Milwaukee, WI, October 2012. 9. Poster: Reeme, AE and RT Robinson, "Vitamin D plays a dynamic role in the Kramnik model of Mycobacterium tuberculosis infection", Novel Therapeutic Approaches to Tuberculosis (Keystone Symposia), Keystone, CO, 2014 10. Miller, HE and RT Robinson, "Human IL12RB1 mRNA transcripts are extensively hypermutated in individuals with otherwise normal IL12RB1 genomic alleles." Novel Therapeutic Approaches to Tuberculosis (Keystone Symposia), Keystone, CO, 2014 8. Robinson, RT*. (*Oral Presenter) “Endpoints and readouts in vaccine efficacy studies in different hosts.” Many Hosts of Mycobacteria VI: A Comparative Symposium, Covington, LA, 2015 9. Reeme, AE* and RT Robinson. (*Oral Presenter) “Dynamic effects of vitamin D on the innate and adaptive immune response during experimental Mycobacterium tuberculosis infection.” Annual Meeting of the American Association of Immunologists, New Orleans, LA, 2015 10. Robinson, RT*. (*Oral Presenter) “The introduction of RNA-DNA differences underlies inter-individual variation in the human IL12RB1 mRNA repertoire.” Annual Meeting of the American Association of Immunologists, New Orleans, LA, 2015 11. Invited Talk: “Animal models of tuberculosis”, Argonne National Laboratory, Division of Environment Safety and Health, Lemont, IL January 2015. 12. Reeme, AE and RT Robinson. “Dietary Vitamin D3 suppresses pulmonary immunopathology associated with late state tuberculosis in C3HeB/FeJ mice.” Colorado Mycobacteria Conference, Colorado Springs, CO, 2016 Invited Talk: “Regulation of mycobacterial disease resistance by IL12RB1”, University of Virginia, Department of Pathology, Charlottesville, VA, April 2017 13. Invited Talk: “Regulation of mycobacterial disease resistance by IL12RB1”, Ohio State University, Department of Microbial Infection & Immunity, Columbus, OH, November 2017 14. Robinson, RT*. (*Oral Presenter) “Human nontuberculous mycobacterial diseases.” Albert Einstein College of Medicine, Many Hosts of Mycobacteria VII Conference, Bronx, NY (03/04/19) 15. Robinson, RT*. (*Oral Presenter) “Immunological, microbiological and lung structure determinants of mycobacterial infection susceptibility in adults with Cystic Fibrosis (CF).” Upstate New York Immunology Conference, Cooperstown, NY (10/31/19)

International 1. Robinson, RT*, SA Khader, RM Locksley, E Lien, ST Smiley and AM Cooper . (*Oral Presenter) "Yersinia pestis elicits growth-condition dependent patterns of dendritic cell IL12p40 production and CCL19-dependent chemotaxis", RIKEN Research Center for Allergy and Immunology, Yokohama, Japan, 2008. 2. Robinson, RT, JJ Fountain, CA Martino, E Torrado, JE Pearl and AM Cooper, IL12p40-responsiveness is required for tubercular granuloma formation, Immunochemistry and Immunobiology Gordon Research Conference, Les Diablerets, Switzerland, 2010. 13. Robinson, RT*. (*Oral Presenter) “Progress in the HIV/TB Research Space.” The Foundation for Neglected Disease Research, Bangalore, India (April 2020)

Internal 1. MCW Invited research talks given to MCW Departments/Divisions: Biomedical Resource Center (10/09/13), Children’s Research Institute (10/18/13), Blood Research Institute (11/14/2013), Infectious Disease (12/17/2015), Pathology (10/20/2016), Biochemistry (11/02/2016) Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

2. OSU Invited research talks given to OSU Departments/Divisions: Cure CF Columbus (C3) research program at OSU and NCH (01/09/2019); Cystic Fibrosis Foundation (CFF) external advisory board for the OSU and NCH C3 research program (04/17/2019)

COMMITTEE SERVICE:

Medical College of Wisconsin 2011-2013 Microbiology and Molecular Genetics Department Representative, Graduate School Council Recruitment Committee, Graduate School 2012 Member, Immunology Faculty Search Committee, Immunobiology, Blood Center of Wisconsin (BCW) 2013 Member, Immunology & Inflammation White Paper Work Group (Dean Appointed), 2014 Advisor, Institutional Animal Care and Use Committee (IACUC) Semi-Annual Program Review 2014-2017 Voting Member, MCW Graduate School Council (GSC) Curriculum and Evaluation 2016-2017 Virology Faculty Search Committee, Department of Microbiology & Immunology

362 2015-2018 Voting Member, Institutional Animal Care and Use Committee (IACUC), Office of Research, 2017-2018 Voting Member, Institutional Biosafety Committee (IBC), Office of Research

The Ohio State University 2018-2019 Member, ad hoc Advisory Committee for targeted faculty recruitment (contributed to the successful recruitment of Drs. Ken Oestreich and Hazem Ghoneim) 2018- Member, BSL3 Users Group 2019 Member, OSU Infectious Disease Institute (IDI) Interdisciplinary Seed Grant Review Panel (05/07/19) 2020 Member, OSU Institutional Animal Care and Use Committee (IACUC)

TEACHING ACTIVITIES (with Instructor Contact Hours) Medical College of Wisconsin (MCW) MCW Medical School Education

2012 – 2014 Advisor and Discussion Leader, Global Health Pathway (MCW Medical Course # PWY- P02GH). Cumulative Instructor Contact Hours per Academic Year: 5 hr 2012 – 2014 Advisor and Discussion Leader, Physician Scientist Pathway (MCW Medical Course # PWY- P04PS). Cumulative Instructor Contact Hours per Academic Year: 5 hr 2012 – 2017 Lecturer, Infection and Host Immunity (MCW Medical Course #280-M1D102). Lecture topics: “Cells & Tissues of the Immune System”, “Innate Immunity”, “Cytokines & Soluble Mediators”, “Immune Tolerance”, “Hypersensitivity”, “Whooping Cough: Small Group Discussion”, “Integrating the Foundations of Immunity: Small Group Discussion” Cumulative Instructor Contact Hours per Academic Year: 8 hr 2015 – 2016 Lecturer, Endocrinology & Reproduction (MCW Medical Course #999-M2D208). Lecture topics: “Male and Female Lower Urinary Tract Dysfunction”, “Immune Response to Urinary Tract Infection.” Cumulative Instructor Contact Hours per Academic Year: 3 hr

MCW Graduate School Education

2011 – 2015 Lecturer, Mucosal Pathogenesis (MCW Graduate Course # 25260). Lecture topics: “Urogenital Host Defenses”, “Immune Defenses Against UPEC”, “Primary Literature Review”, “Teaching Mucosal Pathogenesis to an Undergraduate Student Audience “, “Teaching Mucosal Pathogenesis to a Medical Student Audience” Cumulative Instructor Contact Hours per Academic Year: 4.5 hr 2015 – 2017 Lecturer, Cell Signaling (MCW Graduate Course #16250). Lecture topic: “Toll-like receptor (TLR) and T-cell receptor (TCR) signaling.” Cumulative Instructor Contact Hours per Academic Year: 2 hr 2013 – 2017 Lecturer, Cellular and Molecular Immunology ( MCW Graduate Course # 25234). Lecture topics: “Cytokines and other Soluble Immune Mediators”, “Innate Immunity I”, “Innate Immunity II”, “Approaches to Studying Immunology”, “Integrated Paper Discussion”, “Immune Responses to Bacteria”, Cumulative Instructor Contact Hours per Academic Year: 7.5 hr

The Ohio State University (OSU)

OSU Undergraduate Education

2018 – Present Course Director and Lecturer, Special Topics in Biomedical Science I: Immunology and Infectious Disease (BIOMSCI 4810H). Co-course director: Namal Liyanage, PhD. Lecture Topics: “Mycobacterial pathogens and their associated diseases”, “Tuberculosis: history and current problem”, “Roundtable Discussion: TB Silent Killer (PBS Documentary)”, “Immune responses to M. tuberculosis infection”, “In- class debate: Will new drug development solve the world TB problem?”, “In-class debate: Should the world malaria problem be managed by gene-edited mosquitos?”, “Roundtable Discussion: AIDS in Black America (PBS Documentary)”, “Roundtable Discussion: LGBTQ+ Youth Experiences, Risks and Resilience (w/ guest speaker from Kaleidoscope Youth Center)”, “Roundtable Discussion: What is needed to improve HIV care in Ohio? (w/ guest speakers from Equitas Health)” Cumulative Instructor Contact Hours per Academic Year: 17 hr

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

OSU Graduate School Education

2018 – Present Course Director and Lecturer, Concepts in Biomedical Science: Host Defense block (BSGP 7000). Co- course directors: Aaron Goldman, PhD; Noah Weisleder, PhD. Lecture Topics: “Adaptive Immunity: Paper presentation and discussion”, “Immune tolerance: Paper presentation and discussion”, “T cell trafficking: Paper presentation and discussion”, “Innate and adaptive immunity to bacterial pathogens”, “Clinical relevance of immunity to bacterial pathogens”, “Autophagy and Bacteria: Paper presentation

364 and Discussion”, “Antibodies and Virology: Paper presentation and discussion.” Cumulative Instructor Contact Hours per Academic Year: 7 hr

STUDENTS, FACULTY, RESIDENTS, AND CLINICAL/RESEARCH FELLOWS MENTORED:

Undergraduate Students

• Alexis Rwatambuga, Wisconsin Lutheran College, 5/2011 - 8/2011 Mentor, MCW Summer Program in Undergraduate Research (SPUR). Current Position: Research Associate, Oregon Health and Science University (Portland, OR) • Jessica Jimenez-Rico, Alverno College, 2/2012 - 5/2012 Mentor, MCW Program for Undergraduate Research Experience (PURE). Current Position: Graduate Student • Jacob L. Fohtung, Marquette University, 5/2013 - 12/2013 Mentor, MCW Program for Undergraduate Research Experience (PURE). Current Position: Entrepeneur and MBA student, Babson College (Boston, MA) • Shane Patzlsberger, University of Wisconsin - Madison, 5/2013 - 7/2013 Mentor, MCW Summer Program in Undergraduate Research (SPUR). Current Position: Software Developer, Epic Systems (Versona, WI) • Katrina Monson, University of Wisconsin - Whitewater, 2014 Mentor, MCW Summer Program in Undergraduate Research (SPUR). Current Position: Clinical Trial Coordinator, Froedtert Hospital (Milwaukee, WI) • Brady Brooks, Saint Leo University, 2015 Mentor, MCW Summer Program in Undergraduate Research (SPUR). Current Position: DO Student, University of Pikeville-Kentucky College of Osteopathic Medicine (Pikeville, KY) • Oscar Rosas Mejia, Cardinal Stritch University, 2016 Mentor, MCW Summer Program in Undergraduate Research (SPUR). Current Position: OSU BSGP Graduate Student.

Medical Students

• Matthew Rodgers, MCW Medical Scientist Training Program, 2011 Lab Rotation Mentor • Joan Abi Ashcraft, MCW Medical School, 2012 - 2014 Global Health Pathways Mentor. o Current Position: Emergency Room Physician, Monroe Clinic Hospital (Monroe, WI) • Jason Sieber, MCW Medical Scientist Training Program, 2013 Lab Rotation Mentor Graduate Students

PhD Students Advised • Maciej Czarneki, MCW Graduate School, 2011 (Rotation) • Matthew Forsberg, MCW Graduate School, 2011 (Rotation) • Rajrupa Chakraborty, MCW Graduate School, 2011 (Rotation) • Allison Reeme, MCW Graduate School, 2012 – 2016 (PhD Student) o Current Position: Infection Prevention & Control Specialist, Froedtert Hospital • Ahmed Al-Muhairi, MCW Graduate School, 2015-2017 (MS Student) o Current Position: Engineering Technician, Harley Davidson (Milwaukee, WI) • Tiffany Claeys, OSU Graduate School, 2016 - Present (PhD Student) • Oscar Rosas Mejia, OSU Graduate School, 2018 – Present (PhD Student) • Marisa Ruane-Foster, OSU Graduate School, 2020 (Rotation) PhD Committees • Allison Reeme, MCW Graduate School, 2012 – 2016 • Katherine Zappia, MCW Medical Scientist Training Program, 2014 – 2016 • Matthew Forsberg, MCW Graduate School, 2012 – 2017 • Michael Curtis, MCW Graduate School, 2014 – 2018 • Vanessa Yuan, MCW Graduate School, 2015 – 2018 • Clinton Piper, MCW Graduate School, 2016 – 2018 • Kaitlin Schmitz, MCW Graduate School, 2016 – Present • Tiffany Claeys, OSU Graduate School, 2016 – Present • Oscar Rosas Mejia, OSU Graduate School, 2016 – Present Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

• Jenny Resiliac, OSU Graduate School, 2016 – Present

Postdoctoral Fellows/Visiting Scholars

• Nicole Ford (PhD, Princeton University, Princeton, NJ), 2011 – 2013 o Current Position: Assistant Professor, Kansas City University (Kansas City, MO). • Agumas Shibabaw (PhD Candidate, University of Gondar, Gondar, Ethiopia), 2018-2019 • Naiquan Yao (PhD, Jilin Agricultural University, Changchun, China), 2019 – Present • Ayesha Zafar (PhD Candidate, University of the Punjab, Lahore, Pakistan), 2019 – Present

366 COMMUNITY SERVICE ACTIVITIES: 2005 Participant, American Cancer Society US Senate/House Lobbying Day 2008 – 2010 Board Member, Northern Lights Waldorf School (Saranac Lake, NY) 2012 – 2013 Milwaukee School of Engineering SMART team faculty mentor (Grafton High School, Wisconsin) 2012 – 2016 Organizer, Microbiology Department Summer Picnic 2015 American Association of Immunology Focus Group (5/9/15) 2014 – 2018 Roosevelt Elementary School Parent Teacher Association 2015 – 2018 Wauwatosa School District Safe Routes to School Committee 2015 – 2018 Guest Speaker on the topic of “DNA”, Girl Scouts of American Troop #3430 2018 – Present Kaleidoscope LGBTQ+ Youth Center

BIBLIOGRAPHY: Refereed Journal Publications/Original Papers

1. Shibabaw, A., B. Gelaw, W. Gebreyes, R.T. Robinson, S. Wang, B. Tessema “The burden of pre- extensively and extensively drug-resistant tuberculosis among MDR-TB patients in Amhara regional state, Ethiopia” Accepted, PLoS One. 2. Larsen, MH*, K Lacourciere, TM Parker, A Kraigsley, JM Achar, LB Adams, KM Dupnik, LH Stoodley, T Hartman, C Kanipe, SL Kurtz, MA Miller, LCM Salvador, RT Robinson*. "The Many Hosts of Mycobacteria 8 (MHM8): A Conference Report." In revision, Tuberculosis. *Co-corresponding authors. 3. Hayes D Jr, Harhay MO, Nicol KK, Liyanage NPM, Keller BC, Robinson RT. (2020) “ Lung T-Cell Profile Alterations are Associated with Bronchiolitis Obliterans Syndrome in Cystic Fibrosis Lung Transplant Recipients.” In press, Lung. doi: 10.1007/s00408-019-00298-1 4. Claeys, T.A., Rosas-Mejia, O., S. Marshall, J. Jarzembowski, D. Hayes Jr., N. Hull, N. Liyanage, R. Chun, C.G. Sulman, A.R. Huppler, and R.T. Robinson. (2019) " Claeys, T.A., Rosas-Mejia, O., S. Marshall, J. Jarzembowski, D. Hayes Jr., N. Hull, N. Liyanage, R. Chun, C.G. Sulman, A.R. Huppler, and R.T. Robinson. (2018) "Attenuated TH1 and TH17 differentiation underlies susceptibility to nontuberculous mycobacteria infection in otherwise healthy children." Journal of Infectious Diseases 220:1843-1847. 5. Miller, H.E., K.E. Johnson, V.L. Tarakanova, and R.T. Robinson. (2019) “ g-herpesvirus latency attenuates Mycobacterium tuberculosis infection in mice.” Tuberculosis 116:56-60. 6. Reeme, A.E., T.A. Claeys, P. Aggarwal, A.J. Turner, J.M. Routes, U. Broeckel, and R.T. Robinson. (2019) “Human IL12RB1 expression is allele-biased and produces a novel IL12 response regulator.” Genes & Immunity 20:181-197. 7. Claeys, T.A. and R.T. Robinson. (2018) “The many lives of nontuberculous mycobacteria.” Journal of Bacteriology pii: JB.00739-17 (Invited Minireview) 8. R.T. Robinson. (2017) “T cell production of GM-CSF protects the host during experimental tuberculosis.” mBio 8:e02087-17. (Invited Commentary) 9. Curtis, M., B. Hahn, K. Zhang, C. Li, R.T. Robinson, and J. Coburn. (2017) "Characterization of Stress and Innate Immunity Resistance of Wild-type and Δp66 Borrelia burgdorferi"” Infection & Immunity 86: pii e00186-17 10. R.T. Robinson* and A.R. Huppler*. (2017) “The Goldilocks model of immune symbiosis with Mycobacteria and Candida colonizers” Cytokine 97:49-65. (*Co-Corresponding Authors) 11. Reeme, A.E. and R.T. Robinson. (2016) “Dietary Vitamin D3 suppresses pulmonary immunopathology associated with late stage tuberculosis in C3HeB/FeJ mice.” Journal of Immunology 196:1293-304. 12. Turner, A.J., P. Aggarwal, H.E. Miller, J. Waukau, J.M. Routes, U. Broeckel, and R.T. Robinson. (2015) “The introduction of RNA-DNA differences underlies inter-individual variation in the human IL12RB1 mRNA repertoire.” Proceedings of the National Academy of Sciences 112:15414-15419. 13. Robinson, R.T., I.M. Orme, A.M. Cooper. (2015) “The onset of adaptive immunity in the mouse model of tuberculosis and the factors that compromise its expression.” Immunology Reviews 264:46- 59. Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

14. Orme, I.M., R.T. Robinson, A.M. Cooper. (2015) “The balance between protective and pathogenic immune responses in the TB-infected lung.” Nature Immunology 16:57-63. 15. R.T. Robinson. (2015) "IL12Rβ1: The cytokine receptor that we used to know." Cytokine 71:57-63. 16. Ray, A.A., J.J. Fountain, H.E. Miller, A.M. Cooper, and R.T. Robinson. (2015) "IL12Rβ1ΔTM is a secreted product of il12rb1 that promotes control of extrapulmonary tuberculosis." Infection and Immunity 83:560-71. (Chosen for cover photograph) 17. Miller, H.E., S. Patzlsberger, D.J. Rodi and R.T. Robinson. (2014) "Characterization of respirable aerosols generated during routine laboratory procedures." Applied Biosafety 19:19-27. 18. Reeme, A.E., H.E. Miller, and R.T. Robinson. (2013) "IL12B expression is sustained by a heterogeneous population of myeloid lineages during tuberculosis" Tuberculosis. 93:343-56. 19. Miller, H.E., and R.T. Robinson. (2012) "Early control of Mycobacterium tuberculosis infection requires il12rb1 expression by rag1-dependent lineages" Infection and Immunity. 80:3828-41. (Chosen for cover photograph) 20. Ford, N.R., H.E. Miller, A.E. Reeme, and R.T. Robinson. (2012) "Inflammatory signals direct expression of human IL12RB1 into multiple distinct isoforms." Journal of Immunology. 189:4684-94. 21. Torrado, E., J.J. Fountain, R.T. Robinson, C.A. Martino, J.E. Pearl, J. Rangel-Moreno, M. Tighe, R. Dunn, A.M. Cooper. (2013) "Differential and site specific impact of B cells in the protective immune response to Mycobacterium tuberculosis in the mouse." PLoS One. 8:e61681.

368 22. Torrado E., Robinson R.T., Cooper A.M. (2011) "Cellular response to mycobacteria: balancing protection and pathology." Trends in Immunology 32:66-72. 23. Robinson, RT., S.A. Khader, C.A. Martino, J.J. Fountain, M. Teixeira-Coelho, J.E. Pearl, Stephen T. Smiley, G.M. Winslow, D.L. Woodland, M.J. Walter, J.R. Conejo-Garcia, U. Gubler, A.M. Cooper. (2010) "Mycobacterium tuberculosis infection induces il12rb1 splicing to generate a novel IL12Rβ1 isoform that enhances DC migration." Journal of Experimental Medicine. 207:591-605. 24. Robinson, R.T., J. Wang, J.G. Cripps, M.W. Milks, K.A. English, T.A. Pearson, and J.D. Gorham (2009) "End- organ damage in a mouse model of fulminant liver inflammation requires CD4+ T cell production of IFN-γ but is independent of Fas." Journal of Immunology 182:3278-3284. 25. Milks, M.W., J.T. Cripps, H. Lin, J. Wang, R.T. Robinson, J.L. Sargent, M.L. Whitefield, R.M. Strieter, and J.D. Gorham. (2009) "The role of IFNγ in alterations in liver gene expression in a mouse model of fulminant autoimmune hepatitis." Liver International 29:1307-1315. 26. Robinson, R.T., S.A. Khader, R.M. Locksley, E. Lien, S.T. Smiley, and A.M. Cooper. (2008) "Yersinia pestis evades TLR4-dependent induction of IL12(p40)2 by dendritic cells and subsequent migration." Journal of Immunology 181:5560-5567. 27. Robinson, R.T. and J.D. Gorham. (2007) "TGF-β1 regulates antigen-specific CD4+ T cell responses in the periphery" Journal of Immunology, 179:71-79. 28. Robinson, R.T., M.A. French, T.J. Kitzmiller, and J.D. Gorham (2006) "Restriction of the CD4+ T- cell receptor repertoire prevents immune pathology in TGF-β1 knockout mice." Laboratory Investigation, 86:815-828. 29. Robinson, R.T., B.C. Drafts, and J.L. Fisher (2003) "Fluoxetine increases GABAA receptor activity through a novel modulatory site." Journal of Pharmacology and Experimental Therapeutics, 304:978- 984.

Books, Chapters, and Reviews

1. A.M. Cooper and R.T. Robinson. "Acquired Immunity: Chronic bacterial infections" In: Immunology of Infectious Diseases. S.H.E. Kaufmann, B. Rouse and D. Sacks (Eds.), ASM Press. Herndon, VA. 2010.

Patents

1. Robinson, R.T. and A.M. Cooper. "Novel use of an IL-12 receptor splice variant and molecular assay to quantify expression thereof." US Patent 8,394,593 B2; filed 02/07/2011. 2. Robinson, R.T. and A.M. Cooper "Use of an IL12 receptor-beta 1 splice variant to diagnose active tuberculosis." US Patent 8,877,212 B2; filed 02/07/2013.

Invention Disclosures

1. Robinson, R.T. Invention No. MCW 1855: “Suppression of IL12Rß1 Activity as a Means to Treat Psoriasis and Other Chronic Inflammatory Diseases”; NIH Grant Support: AI099661; IR No. 0046001- 14-0006. 2. Robinson, R.T. Invention No. MCW 1881: “IL12Rß1 RNA-DNA Difference Measurement as a Companion Diagnostic to Predict the Efficacy of IL12Rß1-Targetted Therapies”; NIH Grant Support: AI099661; IR No. 0046001-15-0008. CURRICULUM VITAE Stephanie Marthe Marie Seveau

PERSONAL INFORMATION Place of birth: France Citizenship: USA Address: 6033 Glenbarr Place, Dublin, 43017 Ohio.

CURRENT APPOINTMENT Associate Professor Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio. Phone: (614) 247-7671 Email: [email protected]

EDUCATION 1997 Doctoral Degree (Ph.D.), Université Pierre & Marie Curie, Paris, France. Institut National de la Sante et de la Recherche Medicale (INSERM), Hôpital Necker, Paris, France. Title: Mechanisms of neutrophil polarization and migration. Supervisor: Dr. L. Halbwachs-Mecarelli. 1993 Diplôme d’Etudes Approfondies (M.S.) “Cellular and Molecular Biology Program” Université Pierre & Marie Curie, Paris, France. 1992 Maîtrise (B.S.) “Biochemistry and Immunology” Université Pierre & Marie Curie, Paris, France. POSTDOCTORAL TRAINING

2001- Postdoctoral Fellow, Department of Cell Biology and Infection, Pasteur Institute, 2005 Paris, France. Adviser: Dr. Pascale Cossart. 2002 Live-cell microscopy course Fellow, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany. 1998- Postdoctoral Fellow, Department of Biochemistry, Cornell Medical College of 2001 Cornell University, New York, USA. Adviser: Dr. Frederick R. Maxfield. POSITIONS HELD

2014- Associate Professor, Department of Microbiology (70% appointment), 2015 Department of Microbial Infection and Immunity (30% appointment), The Ohio State University. 2006- Assistant Professor, Department of Microbiology (70% appointment), 2014 Department of Microbial Infection and Immunity (30% appointment), The Ohio State University. 2005- Research Investigator (Faculty), Department of Microbiology and Immunology, 2006 University of Michigan Medical School, Ann Arbor, MI. Adviser: Dr. Joel A Swanson.

MEMBERSHIP IN PROFESSIONAL SOCIETIES 2013-present The American Association of Immunologists 2011- The American Society for Microbiology 1998- The American Society for Cell Biology 2013- Faculty member of the Food Innovation Center, The Ohio State University. 2006- Faculty member of the Center for Microbial Interface Biology, The Ohio State University. 2007- Faculty member of the Public Health Preparedness for Infectious Diseases, The Ohio State University.

1 2011- Faculty member of the Center for Clinical and translational Sciences, The Ohio State University. HONORS 2013 Travel Award. PHPID, The Ohio State University. 2011 Travel Award. PHPID, Ohio State University. 2007 Keynote Speaker. The 16th International Symposium on Problems of listeriosis (Isopol XVI), Savannah, USA. 2002- Fellow, Association pour la Recherche contre le Cancer, France. 2004 2002 Fellow, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany. 2001 Fellow, Fondation Roux, Institut Pasteur, France. 1998- Fellow, Association pour la Recherche contre le Cancer, France. 1999 1997 Distinguished Ph.D. Dissertation Award (awarded to top 10% graduate students) 1995- Fellow, European Molecular Biology Organization (EMBO), Germany 1996 (ASTF8598 and ASTF8713, two awards were obtained). 1994- Fellow, Association Nationale de la Recherche Technique, Paris, France. 1997 Program CIFRE co-sponsored by the pharmaceutical company Guerbet, Villepinte, France.

EDITORIAL ACTIVITIES 2016-present Associate Editor, Critical Reviews in Microbiology 2016-present Associate Editor, Frontiers in Cellular and Infection Microbiology 2014-2018 Associate Editor, Microbial Pathogenesis 2013- Editorial board member, Microbial Pathogenesis 2013- Editor for the Special Research Topic “Advances in Listeria monocytogenes Infection Biology”, Frontiers in Cellular and Infection Microbiology. 11 manuscripts published in 2014. 2010-present Editorial board member, Frontiers in Cellular and Infection Microbiology

2006-present Ad hoc Reviewer for the following Journals Archives of Medical Research Canadian Journal of Microbiology BMC Microbiology Philosophical Transactions (Royal Society, PloS ONE UK) Cellular Microbiology PNAS Science Elife EMBO Journal PLoS Pathogens Expert Opinion on Drug Discovery FEBS Letters Future Microbiology Frontiers in Cellular Infection Microbiology Host Cell and Microbe Innate Immunity Journal of cell Biology Journal of Leukocyte Biology Mediators of Inflammation Journal of Bacteriology mBio Virulence Microbes and Infections Microbial Pathogenesis Molecular Biology of the Cell Molecular Microbiology Placenta Infection and Immunity Frontiers in Microbiology FEMS Microbiology Letter RNA biology Precision Clinical Medicine Biophysical Reviews mSphere Microbiology Open Scientific reports Journal of the Royal Society Interface

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

2

372 GRANT REVIEW PANELS 2019 (November 7) NIH (NIAID) F13 Infectious Diseases and Microbiology Fellowship Review Group Meeting 2019/11 ZRG1-F13-Z-20 2019 (March 21) NIH (NIAID) F13 Infectious Diseases and Microbiology Fellowship Review Group Meeting 2019/05 ZRG1-F13-Z-20 2018 (November 14) NIH (NIAID) R01 Special Emphasis panel Meeting 2019/01 ZRG1-IDM-R-02 2018 (November 6) NIH (NIAID) Topics in Bacterial Pathogenesis IDM-B Meeting 2019/01 ZRG1- IDM-B-81 2018 (July 31) NIH (NIAID) R01 Special Emphasis panel Meeting 2018/10 ZRG1-IDM-T-02 2018 (July 12) NIH (NIAID) R01 Special Emphasis panel Meeting 2018/10 ZRG1-F13-Z-20

2018 (March 22) NIH (NIAID) F13 Infectious Diseases and Microbiology Fellowship Review Group 2017 (November) NIH (NIAID) Topics in Mechanisms of Bacterial Virulence and Pathogenesis (ZRG1 IDM-V-02) 2017 (October) NIH (NIAID) Special Emphasis Panel Member conflicts Topics in Infectious Diseases (ZRG1 IDM-X (02) M) 2017 (July) NIH (NIAID) Topics in Bacterial Pathogenesis IDM-B 2017 (March) NIH (NIAID) Infectious Diseases and Microbiology Fellowship Review Panel 2016 (Oct.) NIH (NIAID) BACP, Bacterial pathogenesis 2016 (March & July) NIH (NIAID) Topics in Bacterial Pathogenesis IDM-B 2015 (July & Oct.) NIH (NIAID) Topics in Bacterial Pathogenesis IDM-B 2014 (July) NIH (NIGMS) SCORE review panel 2012 Postdoctoral fellowships, Wellcome Trust, London, UK 2010 Grants, Agence Nationale de la Recherche, Paris, France

ADMINISTRATIVE SERVICE Departmental Committees 2019-present Microbial Infection and Immunity, Speaker selection Committee 2019-present Microbial Infection and Immunity, Equipment and Resources Committee 2017-2019 Microbial Infection and Immunity, Education & Curriculum Committee 2016-2018 Microbial Infection and Immunity, DTI faculty search committee. OSU 2011-2016 Microbial Infection and Immunity, Professional development and mentoring Committee. OSU 2017 Microbial Infection and Immunity, Application review committee for the Hodges Family Legacy Fund Trainee Travel Award for Infectious Diseases 2016-2016 Microbial Infection and Immunity, Departmental survey and retreat organization committee. 2009-2016 Microbiology Award Committee, Member. OSU 2007-2010 Center for Microbial Interface Biology Retreat Organizing Committee, Member. 2006-2010 Microbiology Graduate Program, Admission Committee. OSU

College and University Committees and Service 2017- present University Senate Faculty Compensation and Benefits Committee 2017- present College of Medicine Faculty Council and College Assembly Representative 2017- present College of Medicine Medical students Admission committee and MSTP sub-committee 2018-2019 Pelotonia Fellowship Awards Committee 2016-2019 Faculty liaison for the Immunology Research Emphasis. Biomedical Sciences Graduate Program (BSGP), OSU. 2016-2019 Biomedical Sciences Graduate Program T32 BSGP fellowship selection committee. 2016 Graduate Faculty Representative thesis Defense (12/9/2016 Prof. H. Lisle Gibbs) 2016 Graduate Faculty Representative. Graduate program: EEOB-PH. (12/9/2016), OSU

3 2014 Graduate Faculty Representative. Graduate program: Mechanical Engineering, candidate John Larson (05/14/2014), OSU 2014 College Committee: Harlan Hatcher award review committee (02/28/2014) 2014 Biological Sciences Scholars Program, College of Arts and Sciences. Lab visit and presentation to undergraduate students (01/24/2014) 2013 OSUMC Research day, poster judge, OSU 2013 GHI Undergrad Mix & Mingle Research Mix & Mingle, co-hosted by Global Health Initiative, the Undergraduate Research Office, and PHPID, OSU 2012 The Landacre Medical Student Research Fair. Department of Microbial Infection and Immunity, OSU 2012 PHPID annual meeting, posters judge, OSU 2010 Fundraising, OSU Campaign kick-off. Center for Microbial Interface Biology, OSU 2009 - Chair of the Center for Department of Microbial Infection and Immunity faculty 2016 grant chalk talks (previously CMIB), OSU 2009 Maximus competition, student orientation, OSU 2009 Maximus competition, student orientation, Ohio State University 2009 - Chair of the Center for Department of Microbial Infection and Immunity faculty 2016 grant chalk talks (previously CMIB), OSU 2008 - Denman Undergraduate Research Forum, Poster judge, Ohio State University 2016 2008 Faculty representative for student orientation IBGP Graduate Program. OSU 2007 - Center for Microbial Interface Biology retreat, posters and short talks judge, 2018 OSU 2007 - Undergraduate Research Colloquium, College of Arts and Sciences, Poster 2016 judge, OSU

TEACHING

2018-present BSGP 8010 “Selected Topics in Advanced Immunology” Course Director and new Curriculum Design. Teaching 4 h about Immunity during Pregnancy 2017-present BSGP 7240 “Listeria infection Biology” (1 h) 2017-present Micro 7010 “Anatomy of Lymphoid Organs and Innate Immunity” (1h 2017; 3 h 2018) 2016- present BSGP 7000/Concepts in Biomedical Science, (1 h/year) 2014-present Lecturer (1 h/year), Fungi and Parasite Immunology/Bacterial Toxins. Graduate course IBGP7000, The Ohio State University 2014-2016 Organizer, Department of Microbiology, Micro 7899 2013-2016 Lecturer (43 h/year + 25 h/year office hours). Undergraduate course: Immunology and Microbial Diseases, Micro 4110, Spring semester, enrolment: 120-150). 100% responsible for course curriculum, teaching, and grading. 2013-2016 Journal Club (1 h/year). Graduate course: Principles of Microbiology, Micro 6010, The Ohio State University 2008-2012 Lecturer (42 h/year + 11 h/year office hours). Undergraduate course: Mechanisms of Microbial Diseases, Micro 524, spring quarter, enrolment 50-60). 100% responsible for course curriculum, teaching, and grading, The Ohio State University 2007-present Lecturer (1 lecture/year), Listeria monocytogenes Infection Biology. Graduate course, Molecular Biology of Bacterial Pathogens, BSGP/Micro 7240, The Ohio State University 2005 Lecturer (2 h), Quantitative fluorescence microscopy. Graduate course: Molecular biology of the cell, Pasteur Institute, Paris, France Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

2004-2005 Lecturer (2 h/year), Quantitative fluorescence microscopy. Graduate course: General Microbiology, Pasteur Institute, Paris, France 2004 Lab course (postdoctoral level, “High-speed FRET imaging using conventional microscopy”). Pasteur Institute, Paris, France.

STUDENT ADVISOR & POSTDOCTORAL TRAINING Undergraduate Research Advisor (The Ohio State University, Columbus, USA) 2019 Lizzie Puelo/ Sophia Kozlowski/ Bryce Piper/ Bella Cho

4

375 2018 Lizzie Puelo/ Madison McQuate/ Bella Cho 2017 Lizzie Puelo/Shirui Yan/ Madison McQuate 2016 Jasneet Singh/Shirui Yan 2015 Jasneet Singh/ Shirui Yan 2014 Eric Weber/ Jasneet Singh 2013 Eric Weber/ Jasneet Singh/ Adam Smith 2012 Colleen Nackerman /Victoria Liang 2011 Victoria Masson/Danielle Chapell/Colleen Nackerman 2010 Victoria Masson/Alex Coe 2007 Peter El-Dahda/Bachir Paraag/Andrew Schneider/Andrew Bruggeman 2009 Siwen Dong 2008 Andrew Bruggeman /Katerine Daprile/Timothy Baker/Siwen Dong

Fellowships and Awards won by Undergraduates in my lab (Ohio State University, Columbus, USA) Dean’s Undergraduate Research Fund: Timothy Baker (2008), Andrew Bruggeman (2008), Siwen Dong (2009), Victoria Masson (2010), Colleen Nackerman (2012) Mayers summer fellowship: Siwen Dong (2009), Colleen Nackerman (2011) Choose Ohio First for Bioinformatics Scholarship: Danielle Chapell (2011) Arts and Sciences Honors Undergraduate Research Scholarship: Alex Coe (2010), Colleen Nackerman (2012) The Ohio State University Denman Undergraduate Research Forum: Colleen Nackerman awarded first place in Biological Sciences, 2012.

Undergraduate Senior Honors Thesis Adviser (Ohio State University, Columbus, USA)

2017 Shirui Yan; Graduated with Honors in Microbiology. Thesis defense in April 2017 2012 Colleen Nackerman. Graduation with Research Distinction 2008 Andrew Bruggeman. Graduation with Research Distinction

Graduate Students (Ohio State University, Columbus, USA) Thesis Adviser Past: 2008-2013 Eusondia Arnett, Microbiology Graduate Program Recipient of the Ohio State University Presidential fellowship 2011-2012 Recipient of poster travel awards (Public Health Preparedness for Infectious Diseases Annual meeting, Ohio State University, 2012; Ohio State University Medical College Research Day, 2013) 2008-2013 Stephen Vadia, Microbiology Graduate Program Recipient of the Ohio State University Presidential fellowship 2011-2012 Recipient of poster and travel awards (Ohio State University Medical College Research Day, 2013; The Food Innovation Center, Ohio State University, 2013) 2014-2018 Jonathan Lam, Microbiology Graduate Program Recipient of poster and travel awards (Microbial Infection and Immunity Travel Award for ASCB meeting Dec. 2017)

2014-2019 Christopher Phelps, Microbiology Graduate Program Recipient of poster and travel awards (). Oral presentation 2017 Ohio Branch ASM/ASM annual meeting 2017.

Current:

2017- Lauren Johnson, Microbiology Graduate Program

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

5

377 NIH Award (05/23/2018 $ 1,000.00) to present her research at the IFPA meeting in September 2018 (Tokyo). Oral presentation Ohio Branch ASM 2018. 2017- Siavash Azari, Microbiology Graduate Program.

Thesis Committee Member

Past: 2007-2010 Dennis Horvath, OSBP Graduate Program 2007-2011 Hannah Cummings, Microbiology Graduate Program 2007-2012 Eric Holbrook, Microbiology Graduate Program 2007-2012 Kiley Dare, Microbiology Graduate Program 2009-2013 Geoffrey Gonzales, OSBP Graduate Program 2008-2014 Tracy Keiser, Microbiology Graduate Program 2010-2014 Bin Li, MD/PhD Program BSGP 2011-2016 Gayathri Natarajan, Microbiology Graduate Program 2014-2017 Michelle Gerst, Microbiology 2016-2018 Rachael Hardisson, BSGP Graduate Committee, Chair 2016-2019 Hannah S. Bekebrede, MCDB 2016-2020 Colwyn Headley, BSGP 2016-2020 Goughenour Kristie D, Microbiology

Current: 2017- Mark Hahn, BSGP 2018- Qi Yan, MCDB 2018- Asmaa, Badr, BSGP Program 2019- Daily, Kylene, MD/PhD BSGP 2020- Lidan Wang, Comparative and Veterinary Medicine Graduate Program

Postdoc Adviser Past: 2014-2017 Dr. Sarika Pathak-Sharma 2015-2016 Dr. Joanna Marshall

INVITED SEMINARS

Intramural

2017 Center for Microbial Interface Biology (CMIB-WIP), Ohio State University 2016 Center for Microbial Interface Biology (CMIB-WIP), Ohio State University 2015 Center for Microbial Interface Biology (CMIB-WIP), Ohio State University 2013 Microbiology Departmental Seminar, The Ohio State University 2012 Microbiology Symposium 2012, The Ohio State University 2012 Center for Microbial Interface Biology, The Ohio State University 2011 Center for Microbial Interface Biology, The Ohio State University 2010 Division of Infectious Diseases, The Ohio State University 2010 Advanced Microscopy Meeting, Ohio State University 2010 Center for Microbial Interface Biology, Ohio State University 2009 Inaugural Imaging and Flow Cytometry Research Day, Mid-Ohio Cytometry Associates, Ohio State University 2007 Division of Infectious Diseases, The Ohio State University 2006 Microbiology Symposium 2006, The Ohio State University 2006 Center for Microbial Interface Biology, The Ohio State University 2003 Department of Cell Biology and Infection, Pasteur Institute, Paris

6 Extramural

2015 Department of Microbiology and Immunology, University of Michigan 2013 Magee-Womens Research Institute, University of Pittsburgh, Pennsylvania, USA 2012 Rocky Mountain Research Laboratories, National Institute of Allergy and Infectious diseases (NIAID, NIH), Hamilton, Montana, USA 2011 Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA 2008 Department of Microbiology, Miami University, Oxford, Ohio, USA 2007 Department of Biological Sciences, University of Toledo, Ohio, USA 2006 Mycobacteria Research Laboratory, Colorado State University, USA 2006 Center for Microbial Interface Biology, Ohio State University 2004 Centro Nacional de Biotechnologia, Madrid, Spain 2004 Department of Immunology and Microbiology, University of Michigan, Ann Arbor, USA 2004 Institut des Cordeliers, INSERM U505, Paris, France 2000 CNRS-Université Bordeaux 2, France 2000 Department of cell Biology and Infection, Institut Pasteur, France 2000 Institut fur Pathologie der Universitat Bern, Switzerland

Seminars at Meetings

2016 Invited speaker. Isopol-XIXth International Symposium on Problems o f Listeriosis- EMBO Conference, Pasteur Institute, Paris, France. 2015 The 3rd Prato Conference on Pore Forming Proteins, Italy. 2013 18th International Symposium on Problems of Listeriosis, Goa, India 2012 International Toxicology Summit 2012, San Antonio, Texas, USA 2008 VIème Colloque "Smal G proteins", CNRS, France 2007 16th International Symposium on problems of listeriosis, Savannah, GA, USA 2004 44th meeting of The American Society for Cell Biology, Washington DC, USA 2003 Meeting GREMI (Group Study on Inflammatory Mediators), Paris, France 2000 15th European Cytoskeletal Forum, Blankenberge, Belgium 1999 4th World Congress on Inflammation, Paris, France 1997 12th Meeting of the European Cytoskeletal Forum, Sienna, Italy

CURRENT FUNDING R03AI149371 (S. Seveau, PI) 12/01/2019-11/30/2021 NIH/NIAID $156,000 Title: Establishing the roles of lncRNAs in placental infection by Listeria monocytogenes.

ProteoSense (Seveau, PI) 02/12/2019-present Title: generation of recombinant listeriophage $141,224

MI&I (S. Seveau, PI; Kara Rood, PI) 09/01/2019-08/31/2021 MI&I Pilot Funding Program application $30,000 Title: Mechanistic study of the infection of the placenta by Listeria monocytogenes

PAST FUNDING

1R21AI105588-01A1 (Seveau, PI) 09/01/2014-08/31/2018 NIH/NIAID $ 423,000.00 Title: Mechanistic study of human placental infection by Listeria monocytogenes

Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

7

380 1R01AI107250-01A1 (Seveau, PI) 02/01/2014-01/31/2019 NIH/NIAID $ 1,896,605 Title: Multifaceted activity of listeriolysin O during host cell invasion by Listeria monocytogenes

PUBLICATIONS

38- High-throughput Measurement of Plasma Membrane Resealing Efficiency in Mammalian Cells. Lam JGT, Song C, Seveau S. 2019. J Vis Exp. Jan 7;(143). doi: 10.3791/58351.

37- Relative Roles of Listeriolysin O, InlA, and InlB in Listeria monocytogenes Uptake by Host Cells. Phelps CC, Vadia S, Arnett E, Tan Y, Zhang X, Pathak-Sharma S, Gavrilin MA, Seveau S. 2018. Infect Immun. Sep 21;86(10). pii: e00555-18. doi: 10.1128/IAI.00555-18. Print 2018 Oct.

36- Checks and Balances between Autophagy and Inflammasomes during Infection. Seveau S, Turner J, Gavrilin MA, Torrelles JB, Hall-Stoodley L, Yount JS, Amer AO. 2018. J Mol Biol. 19;430(2):174-192. doi: 10.1016/j.jmb.2017.11.006. Epub 2017 Nov 21.

35- Caspase-4 promotes autophagosome formation in response to bacterial infection. Kathrin Krause, Kyle Caution, Asmaa Badr1, Kaitlin Hamilton1, Abdulmuti Saleh1, Khushbu Pate, Stephanie Seveau, Luanne Hall- Stoodley, Rana Hegazi1, Xiaoli Zhang, Mikhail A. Gavrilin and Amal O. Amer. 2018. Autophagy 2018;14(11):1928-1942. doi: 10.1080/15548627.2018.1491494. Epub 2018 Aug 31

34- Host cell perforation by listeriolysin O (LLO) activates a Ca2+-dependent cPKC/Rac1/Arp2/3 signaling pathway that promotes Listeria monocytogenes internalization independently of membrane resealing. Lam JGT, Vadia S, Pathak-Sharma S, McLaughlin E, Zhang X, Swanson J, Seveau S. 2018. Mol Biol Cell. Feb 1;29(3):270-284. doi: 10.1091/mbc.E17-09-0561. Epub 2017 Nov 29.PMID: 29187576

33- High-Throughput Microplate-Based Assay to Monitor Plasma Membrane Wounding and Repair. Pathak- Sharma S, Zhang X, Lam JGT, Weisleder N, Seveau SM. 2017. Front Cell Infect Microbiol. Jul 14;7:305. 32- Targeting and inactivation of bacterial toxins by human defensins. Kudryashova E, Seveau SM, Kudryashov DS. 2017. Biol Chem. Sep 26;398(10):1069-1085 31- Distinct surveillance pathway for immunopathology during acute infection via autophagy and SR-BI. Pfeiler S, Khandagale AB, Magenau A, Nichols M, Heijnen HF, Rinninger F, Ziegler T, Seveau S, Schubert S, Zahler S, Verschoor A, Latz E, Massberg S, Gaus K, Engelmann B. 2016. Sci Rep. Oct 3;6:34440. doi: 10.1038/srep34440. 30- Retrocyclins neutralize bacterial toxins by potentiating their unfolding. Kudryashova E, Seveau S, Lu W, and Kudryashov DS. 2015. Biochem J. Feb 29- CXCR3 expression defines a novel subset of innate CD8+ T cells that enhance immunity against bacterial infection and cancer upon stimulation with IL-15. Oghumu S, Terrazas CA, Varikuti S, Kimble J, Vadia S, Yu L, Seveau S, Satoskar AR. 2015. FASEB J. Dec 2. 28- Human trophoblasts confer resistance to viruses implicated in perinatal infection. Bayer A, Delorme- Axford E, Sleigher C, Frey TK, Trobaugh DW, Klimstra WB, Emert-Sedlak LA, Smithgall TE, Kinchington PR, Vadia S, Seveau S, Boyle JP, Coyne CB, Sadovsky Y. 2015. Am J Obstet Gynecol. August 26.

8 27- Human defensins facilitate local unfolding of thermodynamically unstable regions of bacterial protein toxins. Kudryashova E, Quintyn R, Seveau S, Lu W, Wysocki VH, Kudryashov DS. 2014. Immunity. Nov 20;41(5):709-21. 26- LysPGS formation in Listeria monocytogenes has broad roles in maintaining membrane integrity beyond antimicrobial peptide resistance. Dare K, Shepherd J, Roy H, Seveau S, Ibba M. 2014. Virulence. 6; 5(4). 25- Multifaceted activity of Listeriolysin O, the key virulence factor of Listeria monocytogenes Seveau S. 2014 Subcellular Biochemistry, Book Chapter, Springer 24- Fluxes of Ca2+ and K+ are required for the LLO-dependent internalization pathway of Listeria monocytogenes. Vadia S and S Seveau. 2014. Infection and Immunity.82 (3): 1084-91 23- Listeriolysin O is degraded by the neutrophil metalloprotease MMP-8 and fails to protect Listeria monocytogenes from intracellular killing in neutrophils. Arnett E, Vadia S, Nackerman CC, Oghumu S, Satoskar AR, McLeish KR, Silvia M. Uriarte ¶, and S Seveau. 2014. Journal of Immunology. 192 (1): 234-44 22- Yersinia enterocolitica inhibits Salmonella enterica and Listeria monocytogenes cellular uptake. Habyarimana F, Swearingen MC, Young GM, Seveau S, and BMM Ahmer. 2014. Infection and Immunity. 82 (1): 174-83 21- A pseudo-tRNA modulates antibiotic resistance in Bacillus cereus. Rogers TE, Sandro FA, Dare K, Katz A, Seveau S, Roy H, and M Ibba. 2012. Plos One. 7(7):e41248. 20- Critical role for PI3Kγ in parasite invasion and disease progression of cutaneous leishmaniasis. Cummings H, Barbi J, Reville P, Zorko N, Oghumu S, Sarkar A, Keiser T, Lu B, Rückle T, Lezama-Davila C, Wewers M, Whitacre C, Radzioch D, Rommel C, Seveau S, and AR Satoskar. 2012. Proc Natl Acad Sci U S A. 109(4):1251-6. Cited in Faculty of 1,000 (1) 19- The pore-forming toxin listeriolysin O mediates a novel entry pathway of L. monocytogenes into human hepatocytes. Vadia S, Arnett E, Haghighat AC, Wilson-Kubalek EM, Tweten RK, and S Seveau*. 2011. Plos Pathog. 7(11):e1002356. cited in Faculty of 1,000 (3) 18- The Multifaceted Activities of Mammalian Defensins. Arnett E and S Seveau. 2011. Current Pharmaceutical Design. 17(38):4254-69. 17- Defensins enable macrophages to inhibit the intracellular proliferation of Listeria monocytogenes. Arnett E, Lehrer RI, Pratikhya P, Lu W, and S Seveau*. 2011. Cell Microbiol. (4):635-51. 16- Quantification of host-microbe interactions by automated fluorescence microscopy. Haghighat AC and S Seveau. 2010. J Immunol Methods. 352(1-2):186-91. 15- Live Cell Fluorescence Microscopy to study microbial pathogenesis. Hoppe A, Seveau S and JA Swanson. 2009. Cell Microbiol. 11(4):540-50. 14- Molecular mechanisms exploited by Listeria monocytogenes during host cell invasion. Seveau S, Pizarro-Cerda J, and P Cossart. 2007. Microbes Infect. 10:1167-75. 13- Histone modifications induced by a family of bacterial toxins. Hamon MA, Batsché E, Régnault B, Tham TN, Seveau S, Muchardt C, and P Cossart. 2007. Proc Natl Acad Sci USA. 104 (44):17555. Cited in Faculty of 1,000 (1) 12- Adapter Protein SH2-B{beta} Stimulates Actin-based Motility of Listeria monocytogenes in a VASP- dependent Fashion. Diakonova M, Helfer E, Seveau S, Swanson JA, Kocks C, Rui L, Carlier MF, and C Carter- Su.

9 2007. Infect Immun. 75:3581-3593. 11- A FRET analysis to unravel the role of cholesterol in Rac1 and PI 3-kinase activation in the InlB/Met signalling pathway. Seveau S, Tham TN, Payrastre B, Hoppe AD, Swanson J, and P Cossart. 2007. Cell Microbiol. 9:790-803. 10- Ku70, a component of DNA-dependent protein kinase, is a receptor involved in Rickettsia conorii invasion of mammalian cells. Martinez JJ, Seveau S, Veiga-Chacon E, Matsuyama S and P Cossart. 2005. Cell. 123(6):1013-23. Cited in Faculty of 1,000 (8) 9- Distinct signaling pathways are involved in leukosialin (CD43) downregulation, membrane blebbing and phospholipids scrambling during neutrophil apoptosis. Nusbaum P, Lainé C, Bouaouina M, Seveau S, Cramer EM, Masse JM, Lesavre P, and L Halbwachs-Mecarelli. 2005. J Biol Chem. 280(7):5843-53. 8- Role of lipid rafts in E-cadherin- and HGF-R/Met-mediated entry of Listeria monocytogenes into host cells. Seveau S, Bierne H, Giroux S, Prevost MC, and P Cossart. 2004. J Cell Biol. 166(5):743-53. 7- Early membrane events in polymorphonuclear cell (PMN) apoptosis: membrane blebbing and vesicle release, CD43 and CD16 down-regulation and phosphatidylserine externalization. Nusbaum P, Laine C, Seveau S, Lesavre P, and L Halbwachs-Mecarelli. 2004. Biochem Soc Trans. 32:477-9. 6- Chemotactically-induced redistribution of CD43 as related to polarity and locomotion of human polymorphonuclear leucocytes. Dehghani ZA, Seveau S, Halbwachs-Mecarelli L, and HU Keller. 2003. Biol Cell. 95(5):265-73. 5- Membrane lipid organization is critical for human neutrophil polarization. Pierini LM, Eddy RJ, Fuortes M, Seveau S, Casulo C, and FR Maxfield. 2003. J Biol Chem. 278(12):10831-41. 4- Cytoskeleton-dependent membrane domain segregation during neutrophil polarization. Seveau S, Eddy RJ, Maxfield FR, and LM Pierini. 2001. Mol Biol Cell. 12:3550-62. 3- Neutrophil polarity and locomotion are associated with surface redistribution of leukosialin (CD43), an anti- adhesive membrane molecule. Seveau S, Keller H, Maxfield FR, Piller F, and L. Halbwachs- Mecarelli. 2000. Blood. 95:2462-70. 2- CD43 (sialophorin, leukosialin) shedding is an initial event during neutrophil migration, which could be closely related to the spreading of adherent cells. Lopez S, Seveau S, Lesavre P, Robinson MK, and L. Halbwachs-Mecarelli. 1998. Cell Adhes Commun. 5(2):151-60. 1- Leukosialin (CD43, sialophorin) redistribution in uropods of polarized neutrophils is induced by CD43 cross- linking by antibodies, by colchicine or by chemotactic peptides. Seveau S, Lopez S, Lesavre P, Guichard J, Cramer EM, and L. Halbwachs-Mecarelli. 1997. J Cell Sci. 110:1465-75.

10 CURRICULUM VITAE

EUGENE M. OLTZ, Ph.D.

DATE: May 19, 2020 PERSONAL INFORMATION: Sex: Male Date of Birth: November 9, 1960 Place of Birth: Cortland, NY CITIZENSHIP: U.S.A. ADDRESS AND TELEPHONE NUMBERS:

The Ohio State University, College of Medicine, Department of Microbial Infection & Immunity 460 W. 12th Ave, Columbus, OH 43210 Biomedical Research Tower, Room 776A Tel: (614) 293-7570; email: [email protected]

PRESENT POSITION: 2019 Professor & Chair, Department of Microbial Infection & Immunity, Ohio State University Wexner School of Medicine, Columbus, OH EDUCATION: 1978-1982 Pre-doctoral: Cornell University; Ithaca, NY, AB (with distinction), Chemistry 1982-1987 Graduate: Columbia University; New York, NY, Ph.D., Chemistry. Thesis: “Bioorganic Studies of the Tunichromes: A Class of Reducing Blood Pigments from Sea Squirts” (Mentor: Koji Nakanishi) ACADEMIC POSITIONS / EMPLOYMENT: 1987-1991 Postdoctoral Fellow, Dept. of Biochemistry, Columbia University College of Physicians & Surgeons (Mentor: Fred Alt) 1991-1993 Postdoctoral Fellow, Children’s Hospital & Dept. of Genetics, Harvard University, (Mentor: Fred Alt) 1993-2000 Assistant Professor, Dept. of Microbiology & Immunology, Vanderbilt University Medical School 2000-2005 Associate Professor, Dept. of Microbiology & Immunology, Vanderbilt University Medical School 2000-2006 Director of Graduate Studies, Dept. of Microbiology & Immunology, Vanderbilt University Medical School 2005-2009 Professor, Dept. of Microbiology & Immunology, Vanderbilt University Medical School 2009-2018 Professor, Dept. of Pathology & Immunology, Washington University School of Medicine 2015-2017 Director, Immunology Graduate Program, WUSM 2015-2017 Co-Leader, DNA Metabolism and Repair Group, Siteman Cancer Center, WUSM 2017-2018 Vice-Chair for Faculty Development, Dept. of Pathology & Immunology, WUSM 2018-2023 Editor-In-Chief, The Journal of Immunology 2019- Professor, Chair, and Samuel Saslaw Professor in Infectious Disease, Department of Microbial Infection & Immunity, Ohio State University Wexner School of Medicine Eugene M. Oltz Curriculum Vitae Page 2

UNIVERSITY APPOINTMENTS AND COMMITTEES (VANDERBILT):

1994-2009 Applicant Reviewer, Interdisciplinary Graduate Program (IGP) 1994 Consultant, Faculty Search Committee, Dept. of Biochemistry 1994-1996 Consultant, Faculty Search Committee, Vanderbilt-Ingram Cancer Center 1994-1996 Faculty Committee, Immunology Workshop for Middle Tennessee High School Science Teachers, VUMS 1994-2003 Member, Part I Qualifying Exam Committee (Departmental) 1994-2009 Member, Departmental Graduate Education Committee 1995-1998 Consultant, HIV Search Committee, VUMS 1995-2009 New Student Orientation, MSTP Program, Vanderbilt Medical School 1996 Reviewer, Vanderbilt-Ingram Cancer Center Pilot Grants 1996 Reviewer, Introduction to Biomedical Research, Student Presentations, Vanderbilt Medical School 1996 Departmental Representative, University Strategic Initiative Task Force, Scientific Faculty Committee 1996 Faculty Participant, University Strategic Initiative Task Force, Diversity Workshop 1996 Lecturer, Community Outreach, Immunology for High School Students (Martin Luther King High School), Vanderbilt Medical School 1996-1997 Planning Committee, Departmental Scientific Retreat 1997-1999 Member, Immunogenetics Faculty Search Committee, VUMC 1997 IGP Curriculum Review Committee 1997-2009 Member, Sydney P. Colowick Award Committee 1997-2000 Coordinator, Departmental Seminar Committee 1998 Departmental Representative, LCME Accreditation Committee, Graduate Education in Basic Sciences Subcommittee 1998 Consultant, Dept. of Infectious Diseases, HIV Faculty Search Committee 1998 Chair, Departmental Scientific Retreat Planning Committee 1999-2000 Ad Hoc Member, Part I Qualifying Exam Committee, Dept. of Cell Biology 2000-2006 Director of Graduate Studies, Dept. of Microbiology & Immunology 2000-2006 Chair, Departmental Graduate Education Committee 2001 Member, Part I Qualifying Exam Committee, Dept. of Molecular Biophysics & Physiology 2001-2009 Member, Division of Animal Care, Small Animal Review Committee 2005 Coordinator and Chair, Dept. of Microbiology & Immunology Graduate and Post- Graduate Education Retreat 2001-2006 Member, Graduate Faculty Delegate Assembly 2002-2007 Member, Steering Committee, Program for Functional Genomics of Inflammation, VUMS 2003-2009 Chairman’s Advisory Committee, Dept. of Microbiology & Immunology, Ph.D. Thesis Committee, Katrina Moreshead, Harvard University 2004 Faculty Advisor, HHMI Summer Undergraduate Training Program 2004-2009 Member, Postgraduate Education Committee 2004-2005 Consultant, Bacterial Pathogenesis Faculty Search Committee, VUMS 2004-2007 Consultant, Cellular Immunology Faculty Search Committee, VUMS 2005-2009 Faculty Promotions Committee (Departmental) 2007-2009 Dean’s Faculty Advisory Council, VUMS Eugene M. Oltz Curriculum Vitae Page 3

UNIVERSITY APPOINTMENTS AND COMMITTEES (WUSM): 2010-present Co-Director, Work-In-Progress Seminar Series 2010-present Member, Graduate Education Steering Committee (Chair 2015-present) 2010-present Member, Graduate Admissions Committee (Chair 2015-present) 2010-2015 Member, Physician Scientist Training Program, Selection Committee 2015-2017 Director, Immunology Graduate Program 2015-2017 Co-Leader, DNA Metabolism and Repair Group, Siteman Cancer Center 2015-2017 Co- Director, Immunology Seminar Series 2016-present Tenure and Promotions Committee (Immunobiology & LGM) 2016-present Scientific Review Committee, Siteman Investment Program (Cancer Center) 2017-present Vice-Chair for Faculty Development, Dept. of Pathology & Immunology

UNIVERSITY APPOINTMENTS AND COMMITTEES (OSU): 2019-present Co-Chair, Transplant Immunology Search Committee 2019-present Co-Chair, Chair of Cancer Biology & Genetics Search Committee 2019-present Pelotonia Institute for Immuno-Oncology Strategic Planning Committee 2019-present Scientific Advisory Board for Central Ohio Spinal Cord Injury Model System 2019-present College Council Agenda Committee 2020-present Co-Chair, Convalescent Plasma Working Groups 2020-present Chair, Virus Neutralization Working Group 2020-present Chair, MI&I COVID-19 Working Groups 2020-present Member, University Research Recovery Committee 2020-present Member, BSL3 Operations Group

MEDICAL LICENSURE AND BOARD CERTIFICATION: None HONORS AND AWARDS: 1984 Distinguished Teaching Award, Columbia University 1985-1986 American Chemical Society Graduate Fellowship 1987 Hammett Award for Outstanding Graduate Student, Columbia University Chemistry Department 1987-1990 Postdoctoral Fellowship, Irvington Institute 1990-1993 Postdoctoral Fellowship, Cancer Research Institute 1996-1998 Vanderbilt University Vice Chancellor’s Research Scholar 1996-2001 NIH Research Career Development Award 2007 Fellow, Japan Society for Promotion of Science 2008 Impact Award, Vanderbilt Ingram Cancer Center 2010 Transformative Research Grant, NIH Director’s Fund 2012-2016 Member, Publications Committee, American Association of Immunologists 2013-2016 Member, CMI-B Study Section 2013-2016 Chair, Publications Committee, American Association of Immunologists 2013-2020 Ex officio member, Council, American Association of Immunologists 2016-2018 Chair, CMI-B Study Section 2016- Board Member, Aging Biology Foundation 2017 Distinguished Service Award, American Association of Immunologists 2017-2018 Chair, Program Committee, American Association of Immunologists Eugene M. Oltz Curriculum Vitae Page 4

EDITORIAL & PEER REVIEW RESPONSIBILITES:

1994-present Ad hoc manuscript reviews for: Immunity, Science, Nature, Nature Immunology, Molecular Cell, PNAS, Molecular and Cellular Biology, Journal of Biological Chemistry, Journal of Experimental Medicine, Blood, and Journal of Immunology 1999-2004 Journal of Immunology; Associate Editor 1999 NIH Site Visit, Program on Molecular Biology of Viral Tumorigenesis, Johns Hopkins School of Medicine 2001 Site reviewer for U.S. Department of Defense Chemical-Biological Agent Point Detection Program 2001 Immunologic Research; Guest Editor 2003 NIH Mammalian Genetics Study Section, ad hoc 2003-2010 NSF, grant reviewer 2003-2012 NIH Cellular and Molecular Immunology (B) Study Section, ad hoc 2005 NCI Cluster Review for Program Project Grants NIH Genes, Genomics and Genetics NRSA Study Section 2007-2011 Journal of Immunology; Section Editor 2008 NIH IMM-H Study Section, ad hoc 2009 NIH Molecular Genetics (C), ad hoc 2010 NIH CSRS & NIH IMM-D Study Sections, ad hoc 2010 External Reviewer, NCI Experimental Immunology Division 2010 Co-Editor, Current Opinions in Immunology (“Lymphocyte Development”) 2010 NIEHS Superfund Program Study Section 2014- Siteman Cancer Center grant review panel 2013-2018 CMI-B Study Section (Chair, 2016-2018) 2016-2019 Scientific Review Committee, Siteman Investment Program (Cancer Center) 2018-2023 Editor-In-Chief, The Journal of Immunology PROFESSIONAL SOCIETIES AND ORGANIZATIONS: 1995-present: American Association of Immunologists 2019-present American Association for the Advancement of Science MAJOR INVITED PROFESSORSHIPS AND LECTURESHIPS:

1994 University of Texas Health Sciences Center, San Antonio, Dept. of Microbiology and Immunology 1996 FASEB Meeting, B Cell Development Section, New Orleans, LA University of Minnesota, Dept. of Genetics University of Wisconsin, Madison, Dept. of Genetics 1997 Keystone Conference, B Cells in Health and Disease, Steamboat Springs, CO 1997 University of Texas, Southwestern (Dallas), Dept. of Immunology 1999 Keystone Conference, B Cells in Health and Disease, Keystone, CO Signaling & Gene Expression in the Immune System, Symposium, Cold Harbor Laboratories 2000 University of Massachusetts Medical Center, Worcester, Dept. of Microbiology and Immunology 2001 International Immunology Congress, Stockholm, Sweden University of Wisconsin, Dept. of Genetics 2003 University of Alabama (Birmingham), Dept. of Microbiology & Immunology Eugene M. Oltz Curriculum Vitae Page 5

MAJOR INVITED PROFESSORSHIPS AND LECTURESHIPS: (continued) 2004 Harvard University, Dept. of Genetics. Vriej University Medical Center Symposium, Amsterdam, The Netherlands International Immunology Congress, VDJ Recombination Workshop 2005 Chair, VDJ recombination, Somatic Hypermutation and Ig Isotype Switching Workshop; Keystone Meeting: B Cells in Health and Disease FASEB Meeting: Lymphocytes and the Immune System: Molecular, Cellular and Integrative Mechanisms SUNY Downstate; Dept. of Microbiology & Immunology University of Pennsylvania, Dept. of Immunology Albert Einstein School of Medicine, Dept. of Cell Biology 2006 NIH, Laboratory of Cellular and Molecular Biology Sloan Kettering Memorial Cancer Institute 2007 Tokyo University of Science Research Institute Riken Institute, Yokohama Tokyo, Nagasaki, and Kumamoto Universities 2007 Kyoto University 2008 University of Virginia, Dept. of Microbiology & Immunology Emory University, Atlanta, Dept. of Microbiology & Immunology 2008 Signaling & Gene Expression in the Immune System, Symposium, Cold Spring Harbor Duke University, Dept. of Immunology Mayo Clinic, Dept. of Microbiology & Immunology Washington University, Dept. of Pathology & Immunology Scripps Florida, Dept. of Cancer Biology Medical College of Georgia, Cancer Center 2011 Wells Research Center, Indiana University Medical School 2012 University of Pennsylvania, Immunology Program 2012-present Lecturer, AAI Advanced Course in Immunology 2013 Riken Institute, Yokohama and Wako, Japan 2014 AAI Annual Meeting; Pittsburgh, PA Intl. Conf. on Genome Architecture & Cell Fate Regulation; Hyderabad, India 2015 Michigan State University, Dept. of Microbiology & Molecular Genetics AAI Annual Meeting; New Orleans, LA International Conference on Malignant Lymphoma; Lugano, Switzerland Wake Forest Cancer Center Midwestern University National Jewish Center for Immunology; Denver, CO University of North Dakota School of Medicine 2016 University of Illinois Chicago, Dept. of Microbiology & Immunology AAI Annual Meeting, Seattle, WA ICI, Melbourne, Australia University of Florida, Center for Epigenetics Eugene M. Oltz Curriculum Vitae Page 6 MAJOR INVITED PROFESSORSHIPS AND LECTURESHIPS: (continued) 2017 Emory University, Distinguished Scientist Seminar Series Columbia University, Department of Microbiology & Immunology University of Missouri School of Medicine Rockefeller, Sloan-Kettering, Cornell: Immunology & Microbial Pathogenesis University of Pennsylvania, Immunology Program Max Planck Institute of Immunobiology & Epigenetics, Freiburg, Germany University of Pittsburgh, Dept. of Immunology Vanderbilt University Medical School, Center for Immunology 2018 Colorado University School of Medicine; Microbiology & Immunology George Washington University, Cancer Center University of Texas Health Sciences Center; Microbiology & Immunology Medimmune, Astra-Zeneca University of Massachusetts, Worcester; Microbiology & Immunology 2019 University of Miami; Microbiology & Immunology University of Michigan; Microbiology & Immunology FASEB; Molecular mechanisms of Immune Cell Function & Development National Institutes of Health (NIH) 2020 Western Michigan University, Center for Immunobiology Cleveland Clinic, Lerner Research Institute, Inflammation & Immunity Nationwide Children’s Hospital

CONSULTING RELATIONSHIPS AND BOARD MEMBERSHIPS:

2013- Ex officio member, Council, American Association of Immunologists 2016- Board member, Aging Biology Foundation 2000- Board member & President (2000-05), Scottie’s Place Camp for at-risk children 2017-2018 Chair, Program Committee, American Association of Immunologists RESEARCH SUPPORT: ACTIVE 1 R01 AI118852 (Oltz, PI) 3/1/15-2/29/20 NIH/NIAID Topological Mechanisms of DNA Break Repair in Lymphocytes The major goal of this project is to understand the crosstalk between DNA damage responses and 3D chromosome architecture, which may have a profound effect on the resolution of potentially oncogenic lesions. Our overarching hypothesis is: the spread of altered chromatin in response to a DNA break is limited by inherent topological features around the lesion, forming a spatially compact platform to stabilize association of chromosome ends and focus repair. Several aspects of this hypothesis will be examined, including: (i) How physiologic DNA breaks impact chromosome conformation and DNA end stability, (ii) Do focal DNA breaks alter structural and regulatory loops?, and (iii) Are g-H2AX domain structures and chromosome end stability determined by the location of a DNA break? Collectively, this project will explore entirely new concepts of how spatial mechanisms generate repair platforms to restrict DSB interactions with the rest of the genome, thereby preventing oncogenic translocations. Eugene M. Oltz Curriculum Vitae Page 7

RESEARCH SUPPORT: (continued) 1R01AI130231 Oltz/Bassing (Co-PIs) 09/25/17-08/31/22 NIH/NIAID Topological Control of Antigen Receptor Loci During Lymphocyte Development The goal of this project is to determine the roles of transcription, chromatin states, and architectural proteins in developmental changes in Tcrb topology that help sculpt the primary repertoire of T cell antigen receptors. Insights from this project will be broadly applicable to transcriptional regulatory mechanisms at many multi-gene loci.

1 R01 CA188286-01A1 (Payton/Oltz; Co-PI) 5/1/15-4/30/20 NIH/NCI Sequence –Specific Chromatin Modifiers; Novel Protein Therapeutics for B Cell lymphoma The major goals of this proposal are to select a prioritized set of lymphoma-specific enhancers based on several metrics, including potency in a massively parallel reporter assay. The pathogenic chromatin state of this prioritized set will be targeted for reversal with sequence-specific chromatin modifiers (SSCMs) in vitro. Finally, the inherent cell-penetrant properties of zinc finger SSCMs will be exploited to develop these molecules as protein therapeutic molecules for direct delivery in a mouse xenograft model of lymphoma.

1R01AI134035-01 (Oltz/Colonna; Co-PI) 9/13/18-09/12/23 NIH/NIAID Cis-Regulatory Circuits for ILC Function and Plasticity The major goals of this project are to decipher the regulatory logic governing expression programs in innate lymphoid cells (ILCs). We will focus on the dissection of a super-enhancer that controls expression of signature ILC3 cytokines, IL-22 and IL-26, as well as enhancers that regulate multigenic loci involved in autoimmune disorders. The final Aim will identify key regulatory elements and transcription factors that allow ILC3 to transform into ILC1 under inflammatory conditions in vivo.

COMPLETED GRANTS

1994- Molecular Control of Antigen Receptor Gene Assembly. NIAID (R01; Oltz, PI) 2003 1995- Molecular Genetics of Antigen Receptor Gene Assembly and Lymphocyte 1997 Development. Joe C. Davis Scholarship. VUMS, Vice Chancellor's Scholar Award 1994- Molecular Control of Antigen Receptor Gene Assembly. NIAID (R01; Oltz, PI) 2003 1995- Molecular Genetics of Antigen Receptor Gene Assembly and Lymphocyte 1997 Development. Joe C. Davis Scholarship. Vanderbilt University Medical School, Vice Chancellor's Scholar Award 1995- DNA Damage Response and Lymphopoiesis in RSB-1 Deficient Mice. 1996 Center for Molecular Toxicology, VUMC, Pilot Project 1996- Molecular Control of Antigen Receptor Gene Assembly. NIAID (K02; Oltz, PI) 2001 1998- Identification and Characterization of NF-κB Regulated Genes in Pre-B 1999 Cells, University Research Council, Vanderbilt University 1998- Identification and Characterization of NF-κB Regulated Genes in Pre-B 1999 Cells, Vanderbilt-Ingram Cancer Center, Pilot Project 2001- Program in Functional Genomics of Inflammation. NHBLI (PI of the B Lymphocyte 2006 Component) 2003- Signal-Dependent Phosphorylation and Function of IKKgamma. NIAID (R01, Co- 2009 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Investigator) 2005- Deregulation of Cellular lκB Kinases by HTLV1 Tax. NCI (R01, Co-Investigator) 2009 2008- Chemical Library Screen for Histone Demethylase Inhibitors. NCI (Vanderbilt- 2009 Ingram Cancer Center, Pilot Project)

391 Eugene M. Oltz Curriculum Vitae Page 8

COMPLETED GRANTS: (continued) 2009 In Vivo Function of NEMO as a Sensor of K63-linked Polyubiquitination. NIAID (R21, Co-Investigator) 2009- Long-range Genetic and Epigenetic Control of Tcrb Gene Assembly. NIAID (R21; PI) 2011 2009- Long-range Genetic and Epigenetic Control of Igh Gene Assembly. NIAID (R21; PI) 2012 2009- Control of thymocyte development and Rag gene expression by the zinc finger 2012 protein ZFP608. NIAID (R01, Co-Investigator) 2008-2014 Accessibility Control of Antigen Receptor Gene Assembly (R01; PI) 2010-2012 Localized Revision of Epigenetic Landscapes Induced by DNA Double-Strand Breaks (R21; Co-PI) 2015- Regulation of Genome Architecture in Lymphocytes: Opposing Tether and Boundary 2017 Functions (R21; PI) 2010- Targeting Epigenomic Signatures in Non-Hodgkin Lymphoma for Novel 2015 Therapeutics (Transformative R01; Co-PI) 2015- Chromatin-Based Discovery of ILC Gene Regulatory Circuits (R21; Co-PI) 2017 2015- Chromatin-Based Discovery and Function of Novel TCR Regulatory Elements 2017 (R21; Co-PI) 2017-2019 Deconvolution of Epigenomic Data to Characterize Cellular Subpopulations (R21; Co-I)

PATENTS: 1989 Nakanishi, K.; Oltz, E. & Grunberger, D. "Caffeic Acid Esters and Methods for Preparing and Using Same." U.S. Serial No. 369,060.

CLINICAL TITLE AND RESPONSIBILITIES: None

TEACHING TITLE AND RESPONSIBILITIES:

1994- Lecturer, Interdisciplinary Graduate Program “Defense Mechanisms” Section, VUMS 2009 1994- Lecturer/Preceptor, Immunology Workshop for High School Science Teachers, 1995 VUMS 1994- Lecturer, “Molecular and Cellular Immunology”, Dept. of Microbiology and 2009 Immunology, VUMS 1994- Coordinator, Research-In-Progress Seminar Series, Dept. of Microbiology and 1997 Immunology, VUMS 1995 Preceptor, “Microbial Topics”, Vanderbilt Medical School 1995- Director, “Molecular and Cellular Immunology”, Dept. of Microbiology and 2000 Immunology, VUMS 1996 Lecturer, “Receptors”, Dept. of Pharmacology, VUMS 1996- Preceptor, Graduate Education Course, “Foundations in Microbiology and 2009 Ohio State University Master’s of Science in Immunology and Microbial Pathogenesis

Immunology”, Dept. of Microbiology and Immunology. 1997- Lecturer, Microbiology and Immunology, Vanderbilt Medical School 2009 2006 Preceptor, Functional Genomics and Proteomics, Dept. of Microbiology & Immunology 2006- Director, “Molecular and Cellular Immunology”, Dept. of Microbiology and 2009 Immunology, VUMC 2008 Lecturer, Epigenetic Control of Gene Expression in Health and Disease, Vanderbilt Medical School

393 Eugene M. Oltz Curriculum Vitae Page 9

TEACHING TITLE AND RESPONSIBILITIES (continued): 2009-present Lecturer, Immunobiology I, Dept. of Pathology & Immunology, WUSTL 2010-2013 Lecturer, Contemporary Molecular Immunology, WUSTL Medical School 2010-present Lecturer & Co-Director, Advanced Topics in Immunology, Dept. of Pathology & Immunology, WUSTL 2010-2012 Lecturer, Pillars in Immunology, Dept. of Pathology & Immunology, WUSTL 2010-present Discussion Leader, Ethics, WUSTL Medical School 2012-present Lecturer, Bio 548, Gene expression component 2013-present Lecturer, AAI Course in Advanced Immunology, Boston, MA

2013-2015 Lecturer, Medical School Immunology 2015-2016 Co-Director, Immunobiology I, Dept. of Pathology & Immunology, WUSTL 2015-present Co-Director, Advanced Topics in Immunology, Dept. of Pathology & Immunology, WUSTL 2016-2017 Co-Director, Immunobiology II, Dept. of Pathology & Immunology, WUSTL 2016-present Lecturer, DNA Repair

BIBLIOGRAPHY: a. Peer Reviewed Manuscripts 1. Nakanishi K, Kuryanagi M, Nambu H, Oltz E, Verdine G, Zask A: Recent applications of circular dichroism to structural problems, especially oligosaccharide structures. Pure Appl Chem 1984;56:1031.

2. Bruening R, Oltz E, Furakawa J, Nakanishi K, Kustin K: Isolation and structure of tunichrome B- 1, a reducing blood pigment from the tunicate Ascidia nigra (Linnaeus). J Am Chem Soc 1985;107:5298-5300.

3. Bruening R, Oltz, E, Furakawa J, Nakanishi K, Kustin K: Isolation of tunichrome B-1, a reducing blood pigment from the sea squirt Ascidia nigra. J Nat Prod 1986;49(2):193-204. PMID: 3755466

4. Oltz E, Nakanishi K, Yagen B, Corley G, Rottinghaus G, Tempesta M: CD additivity in trichothecene benzoates: Application as a microanalytical method for trichothecene characterization. Tetrahedron 1986;42:2615.

5. Oltz E, Bruening R, Smith M, Kustin K, Nakanishi K: The Tunichromes: A class of reducing blood pigments from sea squirts: isolation, structures, and vanadium chemistry. J Am Chem Soc 1988;110(18):6162-6172.

6. Grunberger D, Banerjee R, Eisinger K, Oltz E, Efros L, Caldwell M, Estevez V, Nakanishi K: Preferential cytotoxicity on tumor cells by caffeic acid phenethyl ester isolated from propolis. Experentia 1988;44(3):230-232. PMID: 2450776 Eugene M. Oltz Curriculum Vitae Page 10

Peer Reviewed Manuscripts (continued) 7. Eldefrawi E, Eldefrawi M, Konno K, Mansour N, Nakanishi K, Oltz E, Usherwood P: Structure and synthesis of a potent glutamate receptor antagonist in wasp venom. Proc Natl Acad Sci 1988;85:4910-4943. PMID: 2838850

8. Oltz E, Pollack S, Delohery T, Smith M, Ojika M, Lee S, Kustin K, Nakanishi K: Distribution of tunichrome and vanadium in sea squirt blood cells sorted by flow cytometry. Experentia 1989;45(2):186-190. PMID: 2920805

9. Yancopoulos G, Oltz E, Rathbun G, Berman J, Smith R, Lansford R, Rothman P, Okada A, Lee G, Morrow M, Kaplan K, Prockop S, Alt F: Isolation of coordinately regulated genes that are expressed in discrete stages of B-cell development. Proc Natl Acad Sci USA 1990;87(15):5759-5763. PMCID: PMC54407

10. Oltz EM, Yancopoulos GD, Morrow MA, Rolink A, Lee G, Wong F, Kaplan K, Gillis S, Melchers F, Alt FW: A novel regulatory myosin light chain gene distinguishes pre-B cell subsets and is IL- 7 inducible. EMBO J 1992;11(7):2759-2767. PMCID: PMC556751

11. Ma A, Fisher P, Dildrop R, Oltz E, Rathbun G, Achacoso P, Stall A, Alt FW: Surface IgM mediated regulation of RAG gene expression in Eµ-N-myc B cell lines. EMBO J 1992;11:2727- 2734. PMID: 1628630

12. Shinkai Y, Rathbun G, Lam K-P, Oltz EM, Stewart V, Mendelsohn M, Charon J, Datta M, Young F, Stall AM, Alt FW: RAG-2 deficient mice lack mature lymphocytes due to inability to initiate VDJ rearrangements. Cell 1992;68(5):855-867. PMID: 1547487

13. Xu Y, Baldessare M, Fisher P, Rathbun G, Oltz E, Yancopoulos GD, Jessell TM, Alt F: LH-2: a LIM/homeodomain gene expressed in developing lymphocytes and neural cells. Proc Natl Acad Sci USA 1993;90(1):227-231. PMCID: PMC45633

14. Taccioli GE, Rathbun G, Oltz E, Stamato T Jeggo P, Alt FW: Impairment of V(D)J recombination in double-strand break repair mutants. Science 1993;260:207-210. PMID: 8469973

15. Rathbun G, Oltz EM, Alt FW: Comparison of RAG gene expression in normal and transformed precursor lymphocytes. Int Immunol 1993;5(8):997-1000. PMID: 8398992

16. Oltz EM, Alt FW, Lin W-C, Chen J, Desiderio S, Rathbun G: A V(D)J recombinase inducible B cell line: Role of transcriptional enhancer elements in directing V(D)J recombination. Mol Cell Biol 1993;13(10):6223-6230. PMCID: PMC364681

17. Demengeot J, Oltz E, Alt F: Promotion of V(D)J recombinational accessibility by the intronic Eκ element: Role of the κB motif. Int Immunol 1995;7(12):1995-2003. PMID: 8746569

18. Scherer D, Brockman J, Bendall H, Zhang G, Ballard D, Oltz E: Corepression of RelA and c-Rel inhibits immunoglobulin kappa gene transcription and rearrangement in pre-B lymphocytes. Immunity 1996;5(6):563-574. PMID: 8986716 Eugene M. Oltz Curriculum Vitae Page 11

Peer Reviewed Manuscripts (continued) 19. O’Brien DP, Oltz EM, Van Ness BG: Coordinate transcription and V(D)J recombination of the kappa immunoglobulin light-chain locus: NF-kappaB-dependent and -independent pathways of activation. Mol Cell Biol 1997;17(7):3477-3487. PMCID: PMC232201

20. Bendall HH, Scherer DC, Edson CR, Ballard DW, Oltz EM: Transcription factor NF-kappaB regulates inducible Oct-2 gene expression in precursor B lymphocytes. J Biol Chem 1997;272(46):28826-28828. PMID: 9360945

21. Sikes ML, Gomez RJ, Song J, Oltz EM: A developmental stage-specific promoter directs germline transcription of DβJβ gene segments in precursor T lymphocytes. J Immunol 1998;161(3):1399- 1405. PMID: 9686603

22. Sikes ML, Suarez C, Oltz EM: Regulation of V(D)J recombination by transcriptional promoters. Mol Cell Biol 1999;19(4):2773-2781. PMCID: PMC84070

23. Sikes ML, Oltz EM: An inducible cell model system for studies of V(D)J recombinational control. J Immunol Methods 1999;224(1-2):25-29. PMID: 10357203

24. Bendall HH, Sikes ML, Ballard DW, Oltz EM: An intact NF-κB signaling pathway is required for maintenance of mature B cell subsets. Mol Immunol 1999;36(3):187-195. PMID: 10403484

25. Hicks GG, Singh N, Nashabi A, Mai S, Boek G, Klewes L, Oltz EM, Van Kaer L, Ruley HE: Fus deficiency in mice results in defective B-lymphocyte development and activation, high levels of chromosomal instability and perinatal death. Nature Genet 2000;24(2):175-179. PMID: 10655065

26. Fields EM, Seufzer BJ, Oltz EM, Miyamoto S: A switch in distinct I kappa B alpha degradation mechanisms mediates constitutive NF-kappa B activation in mature B cells. J Immunol 2000;164(9):4762-4767. PMID: 10779782

27. Sleckman BP, Khan WN, Xu W, Malynn BA, Copeland NG, Bardon CG, Davidson L, Oltz EM, Jenkins NA, Berman JE, Alt FW: Cloning and functional characterization of the early-lymphoid- specific Pb99 gene. Mol Cell Biol 2000;20(12):4405-4410. PMCID: PMC85807

28. Bendall HH, Sikes ML, Oltz EM: Transcription factor NF-kappa B regulates Ig lambda light chain gene rearrangement. J Immunol 2001;167(1):264-269. PMID: 11418658

29. Bender CF, Sikes ML, Sullivan R, Huye LE, Le Beau MM, Roth DB, Oltz EM, Petrini JHJ: Cancer prediposition and hematopoietic failure in Rad50S/S mice. Genes Dev 2002;16(17):2237-2251. PMCID: PMC186667

30. Sikes ML, Meade A, Tripathi R, Krangel MS, Oltz EM: Regulation of V(D)J recombination: a dominant role for promoter positioning in gene segment accessibility. Proc Natl Acad Sci USA 2002;99(19):12309-12314. PMCID: PMC129441 Eugene M. Oltz Curriculum Vitae Page 12

Peer Reviewed Manuscripts (continued) 31. Cao S, Bendall H, Hicks GG, Nashabi A, Sakano H, Shinkai Y, Gariglio M, Oltz EM, Ruley HE: The HMG box protein Ssrp1/T160 is essential for cell viability in E3.5 mouse embryos. Mol Cell Biol 2003;23(15):5301-5307. PMCID: PMC165710

32. Osipovich O, Milley R, Meade A, Tachibana M, Shinkai Y, Krangel MS, Oltz EM: Targeted inhibition of V(D)J recombination by a histone methyltransferase. Nature Immunol 2004;5(3):309-316. PMID: 14985714

33. Carter RS, Pennington KN, Arrate P, Oltz EM, Ballard DW: Site-specific monoubiquitination of IκB kinase IKKβ regulates its phosphorylation and persistent activation. J Biol Chem 2005;280(52):43272-43279. PMID: 16267042

34. Afshar R, Bolland D, Pierce S, Corcoran A, Oltz EM: Regulation of IgH gene assembly: role of the intronic enhancer and 5'DQ52 region in targeting DHJH recombination. J Immunol 2006;176(4):2439-2447. PMID: 16456003

35. Oestreich K, Cobb R, Pierce S, Chen J, Ferrier P, Oltz EM: Regulation of TCRβ gene assembly by a promoter/enhancer holocomplex. Immunity 2006;24(4):381-391. PMID: 16618597

36. Zhang F, Thomas LR, Oltz EM, Aune TM: Control of thymocyte development and recombination- activating gene expression by the zinc finger protein Zfp608. Nature Immunol 2006;7(12):1309- 1316. PMID: 17057722

37. Bolland DJ, Wood AL, Afshar R, Featherstone K, Oltz EM, Corcoran AE: Antisense intergenic transcription precedes IgH D-to-J recombination and is controlled by the intronic enhancer, Emu. Mol Cell Biol 2007;27(15):5523-5533. PMCID: PMC1952079

38. Osipovich O, Cobb RM, Oestreich KJ, Pierce S, Ferrier P, Oltz EM: Essential role for SWI/SNF chromatin remodeling complexes in promoter-directed assembly of TCRβ genes. Nature Immunol 2007;8(8):809-816. PMID: 17589511

39. Ni C, Wu Z, Florence WC, Parekh V, Arrate MP, Pierce S, Schweitzer B, Van Kaer L, Joyce S, Miyamoto S, Ballard DW, Oltz EM: K63-linked polyubiquitination of NEMO modulates TLR signaling and inflammation in vivo. J Immunol 2008;180(11):7107-7111. PMCID: PMC2601684

40. Thomas LR, Miyashita H, Cobb RM, Pierce S, Tachibana M, Hobeika E, Reth M, Shinkai Y, Oltz EM: Functional analysis of histone methyltransferase G9a in B and T lymphocytes. J Immunol 2008;181(1):485-493. PMCID: PMC2497432

41. Osipovich OA, Subrahmanyam R, Pierce S, Sen R, Oltz EM: Cutting Edge: SWI/SNF mediates anti-sense Igh transcription and locus-wide accessibility in B cell precursors. J Immunol 2009;183(3):1509-1513. PMCID: PMC2753546

42. Kondillis-Mangum HD, Cobb RM, Osipovich O, Srivatsan S, Oltz EM, Krangel MS: Transcription-dependent mobilization of nucleosomes at accessible TCR gene segments in vivo. J Immunol 2010;184(12):6970-6977. PMCID: PMC2909652 Eugene M. Oltz Curriculum Vitae Page 13

Peer Reviewed Manuscripts (continued) 43. Ji Y, Little AJ, Banerjee JK, Hao B, Oltz EM, Krangel MS, Schatz DG: Promoters, enhancers, and transcription target RAG1 binding during V(D)J recombination. J Exp Med 2010;207(13):2809- 2816. PMCID: PMC3005232

44. Ise W, Kohyama M, Schraml BU, Zhang T, Schwer B, Basu U, Alt FW, Tang J, Oltz EM, Murphy TL, Murphy KM: The transcription factor BATF controls the global regulators of class-switch recombination in both B and T cells. Nature Immunol 2011;12(6):536-545. PMCID: PMC3117275

45. Guo C, Gerasimova T, Hao H, Ivanova I, Chakraborty T, Selimyan R, Oltz EM, Sen R: Two forms of loops generate the chromatin conformation of the immunoglobulin heavy-chain gene locus. Cell 2011;147(2):332-343. PMCID: PMC3685183

46. Alhawagri M, Yamanaka Y, Ballard D, Oltz E, Abu-Amer Y: Lysine 392, a K63-linked ubiquitination site in NEMO mediates inflammatory osteoclastogenesis and osteolysis. J Orthop Res 2012;30(4):554-560. PMCID: PMC3272311

47. Reed NP, Henderson MA, Oltz EM, Aune TM: Reciprocal regulation of Rag expression in thymocytes by the zinc-finger proteins, Zfp608 and Zfp609. Genes Immun 2013;14(1):7-12. PMCID: PMC4028662

48. Lee BS, Gapud EJ, Zhang S, Dorsett Y, Bredemeyer A, George R, Callen E, Daniel JA, Osipovich O, Oltz EM, Bassing CH, Nussenzweig A, Lees-Miller S, Hammel M, Chen BP, Sleckman BP: Functional intersection of ATM and DNA-PKcs in coding end joining during V(D)J recombination. Mol Cell Biol 2013;33(18):3568-3579. PMCID: PMC3753869

49. Gopalakrishnan S, Majumder K, Predeus A, Huang Y, Koues O, Verma-Gaur J, Loguerico S, Su AI, Feeney AJ, Artyomov M, Oltz EM: A unifying model for molecular determinants of the pre- selection Vβ repertoire. Proc Natl Acad Sci, USA 2012;110(34):E3206-3215. PMCID: PMC3752219

50. Choi NM, Loguercio S, Verma-Gaur J, Degner SC, Torkamani A, Su AI, Oltz EM, Artyomov M, Feeney AJ: Deep sequencing of the murine igh repertoire reveals complex regulation of nonrandom v gene rearrangement frequencies. J Immunol 2013;191(5):2393-2402. PMCID: PMC3778908

51. Predeus AV, Gopalakrishnan S, Huang Y, Tang J, Feeney AJ, Oltz EM*, Artyomov MN*: Targeted chromatin profiling reveals novel enhancers in immunoglobulin heavy and light chain loci. J Immunol 2014;192(3):1064-1070. PMCID: PMC4096788

52. Dorsett Y, Zhou Y, Tubbs AT, Chen B, Purman C, Lee B, George R, Bredemeyer AL, Zhao J, Sodergen E, Weinstock GM, Reyes A, Oltz EM, Dorsett D, Misulovin Z, Payton JE, Sleckman BP: HCoDES reveals chromosomal DNA end structures with single nucleotide resolution. Molec Cell 2014;56(6):808-818. PMCID: 4272619 Eugene M. Oltz Curriculum Vitae Page 14

Peer Reviewed Manuscripts (continued) 53. Majumder K, Koues OI, Chan EAW, Kyle KE, Horowitz JE, Yang-Iott K, Bassing CH, Taniuchi I, Krangel MS, Oltz EM: Lineage-specific compaction of Tcrb requires a chromatin barrier to protect the function of a long-range tethering element. J Exp Med, 2015;212(1):107-120. PMCID: 4291525

54. Koues OI, Kowalewski R, Chang L-W, Pyfrom S, Schmidt JA, Luos H, Sandoval LE, Hughes TB, Bednarski JJ, Cashen A, Payton JE*, Oltz EM*: Enhancer sequence variants and transcription factor deregulation synergize to construct pathogenic regulatory circuits in B cell lymphoma. Immunity, 2015;42(1):186-198. PMCID: 4302272 *Co-corresponding authors

55. Collins PL, Kyle KE, Egawa T, Shinkai Y, and Oltz EM: The histone methyltransferase SETDB1 represses endogenous and exogenous retroviruses in B lymphocytes. Proc. Natl. Acad. Sci., USA, 2015;112(27):8367-8372. PMCID: 26100872

56. Majumder K, Rupp LJ, Yang-Iott KS, Koues OI, Kyle KE, Bassing CH*, Oltz EM*: Domain- specific and stage-intrinsic changes in Tcrb conformation during thymocyte development. J. Immunol., 2015;195(3):1262-1272. PMCID: 26101321

57. Zhao Y, Ma CA, Wu L, Iwai K, Ashwell JD, Oltz EM, Ballard DW, and Jain A: CYLD and the NEMO zinc finger regulate tumor necrosis factor signaling and early embryogenesis. J. Biol. Chem., 2015; 290(36):22076-22084. PMCID: 26224629

58. Gu J, Stevens M, Xing X, Li D, Zhang B, Payton JE, Oltz EM, Jarvis JN, Jiang K, Cicero T, Costello JF, Wang T: Mapping variable DNA methylation across multiple cell types defines a dynamic regulatory landscape of the human genome. G3: Genes, Genomes, Genetics, 2016; g3.115.025437. PMCID: 26888867.

59. Koues OI, Collins PL, Cella M, Robinette ML, Porter SI, Pyfrom SC, Payton JE, Colonna M*, Oltz EM*: Distinct Gene Regulatory Pathways for Human Innate Versus Adaptive Lymphoid Cells. Cell, 2016. 165(5):1134-1146. PMCID:27156452.

60. Raju S, Kretzmer LZ, Koues OI, Payton JE, Oltz EM, Cashen A, Polic B, Schreiber RD, Shaw AS, Markiewicz MA. NKG2D-NKG2D ligand interaction inhibits the outgrowth of naturally arising low- grade lymphoma in vivo. J. Immunol., 2016. 196(11):4805-4813. PMID:27183590.

61. Kaiko GE, Ryu SH, Koues OI, Collins PL, Solnica-Krezel L, Pearce EJ, Pearce EL, Oltz EM, & Stappenbeck TS: The colonic crypt protects stem cells from microbiota-derived metabolites. Cell, 2016, 165(7):1708-1720. PMID: 27264604.

62. Luo H, Schmidt JA, Lee, Y, Oltz EM, Payton JE: Targeted repression of a lymphoma oncogene by sequence-specific histone modifiers induces apoptosis in DLBCL. Leukemia and Lymphoma, 2016. June 7:1-12. PMID: 27268204.

63. Huang Y, Koues OI, Zhao JY, Liu R, Pyfrom SC, Payton JE*, & Oltz EM*: Pathogenic Cis- Regulatory Circuitry for the NEK6 Mitosis-Associated Kinase Is Super-Enhancer-Independent in Transformed B Cells. Cell Reports, 2017, 18(12):2918-2931. PMID: 28329684. Eugene M. Oltz Curriculum Vitae Page 15

Peer Reviewed Manuscripts (continued) 64. Zhao J-y, Osipovich O, Koues OI, Majumder K, & Oltz, EM: Activation of Mouse Tcrb: Uncoupling RUNX1 Function from its Cooperative Binding with ETS1. J. Immunol., 2017, 199(3):1131-1141. PMID: 28637900.

65. Barajas-Mora EM, Kleiman E, Xu J, Carrico NC, Lu H, Oltz EM, Murre C, & Feeney AJ: A B-Cell- Specific Enhancer Orchestrates Nuclear Architecture to Generate a Diverse Antigen Receptor Repertoire. Mol. Cell, 2019, 73(1): 48-60. PMID: 30449725.

66. Collins PL, Cella M, Porter SI, Li S, Gurewitz GL, Hong HS, Johnson RP, Oltz EM*, & Colonna M*: Gene Regulatory Programs Conferring Phenotypic Identities to Human NK Cells. Cell, 2019, 176(1-2):348-360. PMID:30595449.

67. Cella M Gamini R, Sécca C, Collins PL, Zhao S, Peng V, Robinette ML, Schettini J, Zaitsev K, Gordon W, Bando JK, Yomogida K, Cortez V, Fronick C, Fulton R, Lin LL, Gilfillan S, Flavell RA, Shan L, Artyomov MN, Bowman M, Oltz EM, Jelinsky SA* and Colonna M*: Subsets of ILC3−ILC1- like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues. Nature Immunol, 2019, 20(8):980- 991. PMID 31209406

68. Wang Q, Robinette ML, Billon C, Collins PL, Bando JK, Fachi JL, Sécca C, Porter SI, Saini A, Gilfillan S, Solt LA, Musiek ES, Oltz EM, Burris TP, and Colonna M: Circadian rhythm-dependent and circadian rhythm-independent impact of the molecular clock on type 3 innate lymphoid cells. Science Immunol., 2019, 4(40), doi:10.1126/sciimmunol.aay7501. PMID 31586012.

69. Park E, Patel S, Wang Q, Andhey P, Zaitsev K, Porter S, Hershey M, Bern M, Plougastel-Douglas B, Collins P, Colonna M, Murphy KM, Oltz E, Artyomov M, Sibley LD, and Yokoyama WM: Toxoplasma gondii infection drives conversion of NK cells into ILC1-like cells. eLife, 2019, doi: 10.7554/eLife.47605. PMID 31393266.

70. Purman C, Collins PL, Porter SI, Saini A, Gupta H, Sleckman BP, and Oltz EM: Regional gene repression by DNA Double-Strand Breaks in G1 Phase Cells. Mol. Cell. Biol., 2019, 39(24), doi:10.1128/MCB.00181-19. PMID 3151143.

71. Mack MR, Brestoff JR, Berrien-Elliott MM, Trier AM, Yang TB, McCullen M, Collins PL, Niu H, Bodet ND, Wagner JA, Park E, Xu AZ, Wang F, Chibnall R, Council ML, Heffington C, Kreisel F, Margolis DJ, Sheinbein D, Lovato P, Vivier E, Cella M, Colonna M, Yokoyama WM, Oltz EM, Fehniger TA, and Kim BS: Blood natural killer cell deficiency reveals an immunotherapy strategy for atopic dermatitis. Sci. Transl. Med., 2020, 12 (532), doi: 10.1126/scitranslmed.aay1005. PMID:32102931.

72. Ma H, Qian W, Bambouskova M, Collins PL, Porter SI, Byrum AK, Zhang R, Artyomov M, Oltz EM, Mosammaparast N, Miner JJ, and Diamond MS. Barrier-to-Autointegration Factor 1 protects against basal cGas-STING response. mBio, 2020, 11(2), doi: 10.1128/mBio.00136-2. PMID:32156810.

73. Collins PL, Purman C, Porter SI, Nganga V, Saini A, Hayer KE, Gurewitz GL, Sleckman BP, Bednarski JJ, Bassing CH, and Oltz EM: DNA Double-Strand Breaks Induce H2Ax Phosphorylation Domains in a Contact-Dependent Manner. Nature Comm., in press.

*Co-corresponding authors Eugene M. Oltz Curriculum Vitae Page 16

Invited Publications 1. Bruening R, Oltz E, Nakanishi K: The tunichromes, in chemistry and biotechnology of biologically active natural products. R. Vlahov ed. (1985).

2. Alt FW, Rathbun G, Oltz EM, Taccioli G, Shinkai Y: Function and control of recombination activating gene activity. CD5 B Cells in Development and Disease: Ann New York Acad Sci 1992;651:277.

3. Alt FW, Oltz EM, Young F, Gorman J, Taccioli G, Chen J: VDJ recombination. Immunol Today 1992;13(8):306-314. PMID: 1510813

4. Taccioli GE, Rathbun G, Shinkai Y, Oltz EM, Cheng H, Whitmore G, Stamato T, Jeggo P, Alt FW: Activities involved in V(D)J recombination. Curr Topics in Microbiol Immunol 1992;182:107-114. PMID: 1490344

5. Lansford R, Okada A, Chen J, Oltz E, Blackwell TK, Alt F, Rathbun G: Mechanism and Control of Immunoglobulin Gene Rearrangement, in Molecular Immunology: Frontiers in Molecular Biology, 2nd ed. Hames, B.D. and Glover, D.M., eds. Oxford Univ. Press, Oxford, 1996;1-100.

6. Oltz EM: Regulation of antigen receptor gene assembly in lymphocytes. Immunol Res 2001;23(2- 3):121-133. PMID: 11444378

7. Oltz EM, Hawiger J: Immunology at Vanderbilt. Immunol Res 2001;23:97-98. PMID: 11444395

8. Cobb R, Oestreich, K, Osipovich O, Oltz EM: Accessibility control of V(D)J recombination. Adv Immunol 2006;91:45-109. PMID: 16938538

9. Sen R, Oltz E: Genetic and epigenetic regulation of IgH gene assembly. Curr Opin Immunol 2006;18(3):237-242. PMID: 16616470

10. Oltz EM, Osipovich O: Targeting V(D)J recombinase: Putting a PhD to work. Immunity 2007;27(4):539-541. PMID: 17967406

11. Thomas LR, Cobb RM, Oltz EM: Dynamic regulation of antigen receptor gene assembly. Adv Exp Med Biol 2009;650:103-115. PMID: 19731805

12. Osipovich O, Oltz EM: Regulation of antigen receptor gene assembly by genetic-epigenetic cross- talk. Seminars in Immunology 2010;22(6):313-322. PMCID: PMC2981692

13. Oltz E, Hogquist K: Editorial Overview, Lymphocyte Development Issue. Current Opinion in Immunology, 2011;23:153-155. PMID: 21300532 PMC N/A

14. Sikes ML, Oltz EM: Genetic and epigenetic regulation of TCRβ gene assembly. Current Topics in Molecular Immunology 2012;356:91-116. PMID: 21688096 PMC N/A Eugene M. Oltz Curriculum Vitae Page 17

Invited Publications (continued) 15. Sleckman BP, Oltz EM: Preparing targets for V(D)J recombinase: transcription paves the way. J Immunol 2012;188(1):7-9. PMID: 22187481

16. Gopalakrishnan S, Collins PL, Oltz EM: Control of Ig gene assembly: lessons from premature activation. EMBO J 2013;32(10):1350-1351. PMCID: PMC3655476

17. Collins PL, Oltz, EM: Histone methylation keeps the brakes on autophagy. Mol Cell Biol 2013;33(20):3974-3975. PMCID: PMC3811685

18. Majumder K, Bassing CH, Oltz EM: Regulation of Tcrb Gene Assembly by Genetic, Epigenetic, and Topological Mechanisms. Adv Immunol 2015; 128:273-306. PMCID: 26477369

19. Koues OI, Oltz EM*, Payton JE*: Short-circuiting gene regulatory networks: Origins of B cell lymphoma. Trends in Genetics 2015; 31(12):720-731. PCMID: 26604030

20. Oltz, EM: A New JI: The Home for All Immunology. J. Immunol., 2018; 201(1):1-2. PMCID: 29914934

21. Oltz, EM: Immunity to Influenza: Closing in on a Moving Target. J. Immunol., 2019; 202:325-6. PCMID: 30617112

22. Oltz, EM: Neuroimmunology: To Sense and Protect. J. Immunol., 2020; 204(2):239-240. PMID 31907263.

*Co-corresponding authors

MS in Immunology and Microbial Pathogenesis Full Proposal

Appendix F: Letters of Concurrence and Support

To be requested following faculty approval of this document.

403