SCRIPPS FLORIDA ANNUAL REPORT FOR THE YEAR ENDING JUNE 30, 2017

PART 1: SCIENTIFIC ACHIEVEMENTS AND GRANT AWARDS

A: SCIENTIFIC ACHIEVMENTS

Scripps Florida 2017 Scientific Report July 1, 2016 – October 4, 2017

Part 1: New Faculty and Scientific Administration

TSRI Completes Executive Team Build Out

LA JOLLA, CA – August 1, 2016 – The Institute (TSRI) today announced the completion of its California executive team build out, which began with the promotion of James R. Williamson to Vice President of Academic Affairs, followed this summer by the addition of four new executive team leaders—Richard King, Jennifer Crosby-Meurisse, Cara Miller and Julia Ronlov–the promotion of Jared Machado to Chief Financial Officer and the appointment of Douglas A. Bingham to Executive Vice President and General Counsel. With the new executive leadership team now in place, Steve Kay has announced that he has made the personal decision to return to the University of Southern California (USC) as a faculty member in the Keck School of Medicine and Director of the USC Michelson Center. “The new leadership team brings together a diverse and highly experienced executive team whose expertise will be critical in driving forward TSRI’s strategic vision, and I am thrilled to have them on board,” said Peter G. Schultz, Chief Executive Officer. “I’d like to personally thank Steve for his numerous contributions and leadership over the last year, as well as his dedication to assembling such a strong team that will carry forward our focus on innovative science and translational research. We wish him all the best in his future endeavors.”

Alongside these appointments, TSRI has initiated a long-term strategic planning process that will build on the history of TSRI’s academic pursuits, while also further differentiating and solidifying TSRI’s position at the forefront of biomedical research.

“I am incredibly pleased with the continued momentum and progress toward the future envisioned for TSRI—which will ensure the Institute’s position as one of the leading academic institutions in the world,” said Schultz. “We look forward to continuing to build upon its scientific heritage with transformational alliances and partnerships, and ongoing scientific discovery.”

Executive Appointments and Promotions  Dr. James R. Williamson was promoted in September 2015 to Vice President, Academic Affairs to oversee academic planning, faculty affairs and training program partnerships. He remains Professor and Dean of Graduate and Postdoctoral Studies at TSRI.

August 30, 2017 1 Summer 2016

 Richard King was named Chief Operating Officer. King was formerly president and CEO of AcelRx Pharmaceuticals, Inc.  Douglas A. Bingham was named Executive Vice President and General Counsel, and Board Secretary.  Jared Machado was promoted to Chief Financial Officer and Board Treasurer following his tenure as Controller; Vice President, Finance; and a brief interim period as Acting CFO at TSRI.  Jennifer Crosby-Meurisse, Vice President, Human Resources joined TSRI from Oregon Health & Science University, where she served as Director, Research and Academic HR Team and the Office of International Affairs.  Cara Miller assumed the role of Vice President, Communications. Miller joined TSRI from Gilead Sciences, Inc., where she was Senior Director, Public Affairs.  Julia Ronlov has been appointed Executive Director, New Business Development, and joins TSRI on August 1. Ronlov previously served as Director, Strategic Partnerships, for Oregon Health & Science University.

The Scripps Research Institute Announces New Board Members and Chairman of the Board

LA JOLLA, CA & JUPITER, FL – February 21, 2017 – The Scripps Research Institute (TSRI), one of the world's largest, private, non-profit research organizations, today announced the appointment of nine new members to its Board of Directors, including John D. Diekman, who will serve as chairman of the board. Diekman replaces Richard A. Gephardt, who is retiring from the board after seven years of service.

“I am pleased to welcome such an esteemed, world-renowned group of education, business and scientific leaders to TSRI's Board of Directors,” said Peter G. Schultz, Ph.D., President of TSRI and the California Institute for Biomedical Research (Calibr). “The unique and extensive expertise of our existing and newly appointed board members will be critical to the advancement of our vision of a self-sustaining, 'bench-to-bedside,' non-profit model that not only advances science through basic research but also rapidly translates new discoveries into life-saving medicines for the public benefit.

“On behalf of the entire Institute, I would also like to thank Richard Gephardt for his insightful leadership and steadfast support over the past seven years. His wide-ranging experience and commitment to advancing our mission is greatly appreciated, and we wish Richard all the best.”

TSRI Board of Director Appointees Chairman John D. Diekman, Ph.D. Diekman is a Founding Partner of 5AM Ventures, a life science venture capital firm in San Francisco. Prior to founding 5AM in 2002, Diekman was a Founder and Managing Director of Bay City Capital. Previously, he was Chairman and Chief Executive Officer of Affymetrix, and Chairman and Managing Director of Affymax.

August 30, 2017 2

Diekman currently serves as chairman of the board of IDEAYA Biosciences and as a board member of Wildcat Discovery Technologies. He is a Charter Trustee of Princeton University and a former Trustee of The California Institute of Technology and of The Scripps Research Institute. Diekman serves on the Schaeffer Center for Health Policy and Economics Advisory Board at the University of Southern California and is an Honorary Officer of the Order of Australia.

He received his doctoral degree from and holds an honorary doctoral degree from Australia’s Monash University.

Herb Boyer, Ph.D.

Boyer is internationally recognized for his pioneering discovery of recombinant DNA, which has led to numerous life-saving medicines. He co-founded Genentech in 1976 with the late venture capitalist Robert Swanson, breaking new ground for both life science technology and new business models. He is now retired and is Professor Emeritus of Biochemistry and Biophysics at the University of California, San Francisco.

In 2004, Business Week magazine named Boyer one of the "Greatest Innovators of the Past 75 Years," and he was featured on the cover of TIME magazine in 1981 with a story titled, "Shaping Life in the Lab: The Boom in Genetic Engineering, Genentech's Herb Boyer." Boyer has received numerous prestigious awards for his work including the National Medal of Technology and Innovation, the National Medal of Science, the Lemelson-MIT Prize and the Albany Medical Prize. He is an elected member of the National Academy of Sciences, the American Academy of Arts and Sciences and the National Inventors Hall of Fame. He earned his doctoral degree from the University of Pittsburgh.

Gerald Chan, Ph.D.

Chan co-founded Morningside, a private equity and venture capital firm, in 1986. In life science, Morningside has been the founding or major investor in many biotechnology companies. In oncology, the investments include oncolytic viruses (Biovex, DNATrix), immuno-oncology (Aduro), modified cytotoxic agents (Nucana) and novel therapeutic targets (CellCentric, K-Gen, Vigeo). Investments in infectious diseases include antibiotics (MicuRx, Artugen), prophylactic (Matrivax) and antivirals (Atea). Investments in other therapeutic areas include metabolic diseases (CVI), autoimmune diseases (Kezar), CNS disorders (Orthogonal, Pinteon, Cognoa, Cognito) and rare orphan diseases (Stealth, Apellis). Chan received his doctoral degree from Harvard University and conducted his post-doctoral fellowship at Dana-Farber Cancer Institute.

Mark Edwards

As Founder and Managing Director of Recombinant Capital (Recap) from 1988 until its sale to Deloitte in 2008, Edwards supervised the creation and maintenance of several databases relating to the development and commercialization of pharmaceutical products, including the Recap Corporate Alliances Database. Over this period, Edwards and Recap were retained by

August 30, 2017 3 more than 50 companies to assist in the negotiation of biopharma alliances. In 2011, Edwards founded Bioscience Advisors, Inc. (Biosci), a consulting and database firm focused on biopharma alliances.

Edwards serves on the boards of directors of AcelRx and Calibr. In 2008, Edwards was awarded a Lifetime Achievement Award by the American Liver Foundation for "two decades of leadership, thoughtful insights and detailed analysis of the biotechnology industry." He holds a Master of Business Administration from Stanford University.

Isy Goldwasser

Goldwasser is Co-Founder and Chief Executive Officer of Thync, Inc. Prior to founding Thync, Goldwasser was Chief Executive Officer of Symyx Technologies, where he was the company's first employee. In addition to his 16 years at Symyx, Goldwasser was Khosla Ventures' Entrepreneur-in-Residence in 2010. He is a named inventor on more than 30 U.S., European and Canadian patents.

Goldwasser has served on Calibr’s board since its inception and brings two decades of experience in managing corporate operations and business development. He holds a Master of Science degree from Stanford University.

William R. Hearst III

Hearst is Chairman of the Board of Hearst Corporation, one of the nation's largest diversified media and information companies, and he has been a Director of Hearst Corporation for more than 30 years. He is also President of the charitable William Randolph Hearst Foundation, and he has been actively engaged in the charitable activities and programs of the Hearst Foundation for the last 20 years.

Hearst joined the venture capital firm of Kleiner Perkins Caufield & Byers (KPCB) in 1995 as a Managing Partner and has continued his work with KPCB as an Affiliated Partner since 2006. He currently serves on the boards of numerous organizations, including the Carnegie Institution for Science, FORA.tv, the San Francisco Film Society and The Center for Investigative Reporting. Hearst is a fellow of the American Association for the Advancement of Science and was recently appointed an associate in mathematics by the Department of Mathematics at Harvard University. He earned his bachelor’s degree from Harvard University.

Ge Li, Ph.D.

Li is the Founder, Chairman and Chief Executive Officer of WuXi AppTec, a leading research and development capability and technology platform company serving the global pharmaceutical, biotechnology and medical device industries. Prior to founding WuXi in 2000, Li was a founding scientist at Pharmacopeia Inc., a leading combinatorial chemistry platform company.

August 30, 2017 4

Li has received numerous prestigious awards and honors and was named one of Forbes 25 Notable Chinese-Americans and one of the 25 Most Influential People in Biopharma by FierceBiotech. He received his doctoral degree from Columbia University.

Christopher T. Walsh, Ph.D.

Walsh is a Consulting Professor to the Stanford University Department of Chemistry and was previously the Hamilton Kuhn Professor of Biological Chemistry and Molecular Pharmacology at Harvard Medical School for 26 years when he took emeritus status. He has had extensive academic leadership experience, including Chairmanship of the Massachusetts Institute of Technology Chemistry Department and of the Harvard Medical School Department of Biological Chemistry & Molecular Pharmacology, as well as serving as President and Chief Executive Officer of the Dana-Farber Cancer Institute.

Walsh has been involved in a variety of venture-based biotechnology companies since 1981 and currently serves on the boards of Ironwood Pharmaceuticals, Proteostasis Therapeutics and Calibr, and on the scientific advisory boards of Hua Medicine, Abide Therapeutics, Cidara Therapeutics and Flex Pharma, Inc. He serves as an advisor to HealthCare Ventures and is a limited investor in HealthCare Ventures, MPM Capital, Clarus Financial Technology and the Longwood Fund.

Walsh is a member of the U.S. National Academy of Sciences, the U.S. National Academy of Medicine, the American Academy of Arts and Sciences, the American Philosophical Society and is a co-recipient of the 2010 Welch Award in Chemistry. He earned a doctoral degree from .

Faculty Board Appointee Paul Schimmel, Ph.D.

Schimmel is Hahn Professor in the Department of Molecular Medicine and Chemistry at TSRI. He was formerly MacArthur Professor of Biochemistry and Biophysics at the Massachusetts Institute of Technology.

Author or co-author of several hundred scientific publications, he is an elected member of the American Academy of Arts and Sciences, the U.S. National Academy of Sciences, the National Academy of Medicine (Institute of Medicine), the National Academy of Inventors and the American Philosophical Society. He is the co-founder/founding director of seven biopharmaceutical corporations that went on to be NASDAQ-listed and publicly traded, and of a number of private corporations. Schimmel earned his doctoral degree from the Massachusetts Institute of Technology.

Current TSRI Board Members Peter G. Schultz, Ph.D., Vice Chair of the Board and President, The Scripps Research Institute; President, Calibr Peter C. Farrell, Ph.D., D.Sc., Founder and Chairman, ResMed Claudia S. Luttrell, President, The Skaggs Institute for Chemical Biology Mark Pearson, Co-Founder, Vice Chairman of the Board, Drawbridge Realty Trust Bernard Saint-Donat, President, Saint-Donat & Co.

August 30, 2017 5

Philip Dawson Named Dean of The Scripps Research Institute’s Graduate Program

Dawson brings to his new role more than 25 years’ experience at TSRI as a student, alumnus, faculty member and associate dean.

LA JOLLA, CA – March 27, 2017 – The Scripps Research Institute (TSRI) today announced the promotion of Philip Dawson, Ph.D., from Associate Dean to Dean of the TSRI Graduate Program.

Dawson, who earned his Ph.D. from TSRI in 1996, replaces Jamie Williamson, Ph.D., who has taken on a new role as Vice President of Academic Affairs.

“As one of TSRI’s earliest alumni, Phil possesses a unique perspective on the program, as well as a proven track record of leadership success as both a researcher and an administrator,” said Williamson. “In his role as Associate Dean, Phil has been instrumental in the management and direction of the program and he has a clear vision for continuing to build the program’s reach and reputation.”

As the graduate program prepares to celebrate its 25th graduating class this May, Dawson sees his number one responsibility as continuing the great tradition of academic and scientific excellence that has earned the program international prestige, including annual top-10 rankings in U.S. News & World Report’s Best Grad Schools.

“I am both humbled and excited by the opportunity to build on the momentum established under Jamie’s leadership and lead this incredible program into the future,” Dawson said. “As someone who has been involved in all facets of the TSRI graduate program, I know exactly how transformational this experience can be for students.”

Dawson’s own research revolves around the development and utilization of methods to incorporate unnatural chemical groups into proteins. He earned his bachelor’s degree in chemistry from Washington University in St. Louis, Missouri, graduating magna cum laude. After earning his doctoral degree at TSRI, Dawson conducted postdoctoral work at the California Institute of Technology before returning to TSRI in 1997 as Assistant Professor in the department of Cell Biology. He was recently promoted to Professor of Chemistry and has continued to be active in the graduate program through teaching, as well as directing the graduate admissions committee under the role of Associate Dean.

Dawson has authored more than 160 publications, was an Alfred P. Sloan Research Fellow and has earned such honors as the Max-Bergmann Gold Medal, the Vincent du Vigneaud Award and Leonidas Zervas Award. He explained that what makes TSRI’s graduate program unique is the opportunity, from the very beginning of their education, for students to work in a variety of disciplines, with a diversity of world-renowned researchers.

“What our program does best is teach students how to ask important questions and be experimentally fearless in solving them,” Dawson said. “Our graduate program is about more than just learning how to conduct experiments. We nurture independent thought and

August 30, 2017 6 interdisciplinary work. As a result of this training, many of our alumni are performing groundbreaking research in diverse fields and have carved career paths through all areas of industry and academics.”

The Scripps Research Institute (TSRI) Expands New Board of Directors with the Appointment of Yucaipa Companies Founder and Philanthropist Ron Burkle

LA JOLLA, CA – April 3, 2017 –The Scripps Research Institute (TSRI), one of the world's largest, private, non-profit research organizations, today announced the appointment of Ronald W. Burkle to its Board of Directors—adding another world-renowned business leader and philanthropist to its newly restructured board.

Burkle is Founder and Chairman of the Ronald W. Burkle Foundation, whose mission is to positively influence people around the world and their communities. He also founded The Yucaipa Companies, a premier investment firm that has established a record of fostering economic value through the growth, repositioning and responsible development of individual companies. He is widely recognized as one of America’s preeminent investors in the retail, distribution, technology, entertainment, sports and hospitality industries.

“Ron’s business acumen and philanthropic mindset will be critical to realizing the new self- sustaining ‘bench-to-bedside’ model we are building,” said Peter G. Schultz, Ph.D., President of TSRI and the California Institute for Biomedical Research (Calibr). “I am thrilled to welcome yet another incredibly credentialed individual to the TSRI board who can help guide our efforts to take on new challenges in advancing human health through basic science and drug discovery.”

In addition to his role as co-chairman of the Burkle Center for International Relations at the University of California, Los Angeles, Burkle is trustee of the Carter Center, the National Urban League and AIDS Project Los Angeles. He also has been an active member of the Frank Lloyd Wright Conservancy. Burkle has served as Chairman of the Board and controlling shareholder of numerous companies, including Soho House, Golden State Foods, Dominick's, Fred Meyer, Ralphs and Food4Less.

The Scripps Research Institute Appoints Alexandria Real Estate Equities, Inc. CEO Joel Marcus to Board of Directors

LA JOLLA, Calif. – May 30, 2017 – The Scripps Research Institute (TSRI), one of the world's largest, private, non-profit research organizations, today announced the appointment of Joel S. Marcus to its Board of Directors.

Marcus is Chairman, Chief Executive Officer and Founder of Alexandria Real Estate Equities, Inc. (NYSE: ARE), an urban office real estate investment trust (REIT) uniquely focused on collaborative life science and technology campuses in the top innovation cluster locations in the United States.

August 30, 2017 7

Marcus co-founded Alexandria in 1994 as a startup with a business plan and $19 million in seed capital and has since led its growth into an investment-grade rated S&P 500 company. Alexandria has a total market capitalization of approximately $15 billion and a significant market presence in key locations, including Greater Boston, San Francisco, New York City, San Diego, Seattle, Maryland and Research Triangle Park.

“Joel’s professional experience and business leadership add to the expertise of our current board members,” said Peter G. Schultz, Ph.D., President of TSRI and the California Institute for Biomedical Research (Calibr). “His creativity, insight and commitment to research and public health will be invaluable in our efforts to create a new model for funding basic and translational research in the nonprofit sector. We are thrilled to welcome him to the board.”

“TSRI is a world-class biomedical science research and discovery Institute,” said Marcus. “I am honored to join the TSRI board and look forward to helping the Institute achieve its goals of translating new scientific breakthroughs into life-saving medicines under the visionary leadership of Pete Schultz.”

In addition to his work in life science real estate, Marcus is the founder of Alexandria Venture Investments, which focuses on investing in the biopharmaceutical, diagnostic, research tools, agtech, digital health and technology sectors. He also co-founded the Alexandria Summit, an annual invitation-only meeting that convenes the world’s foremost thought leaders from the pharmaceutical, biotechnology, agribusiness, technology, medical, academic, venture capital, philanthropic, patient advocacy and government communities to address the most critical challenges in global healthcare, agriculture and the environment.

Marcus serves on the boards of directors for several biotechnology companies, including the Accelerator Corporation (which he co-founded), AgTech Accelerator Corporation, for which he serves as chairman, Atara Biotherapeutics and Intra-Cellular Therapies. He also serves on the boards of the Biotechnology Innovation Organization (BIO) and the Foundation for the National Institutes of Health (FNIH).

Marcus earned his undergraduate and Juris Doctor degrees from the University of California, Los Angeles and was a recipient of the Ernst & Young Entrepreneur Of The Year Award (Los Angeles – Real Estate).

Part 2: Grant Awards and Licensing Agreements

Scripps Research Institute Chemist Jin-Quan Yu Wins MacArthur Fellowship

LA JOLLA, CA—September 22, 2016—Chemist Jin-Quan Yu of The Scripps Research Institute (TSRI) has won a 2016 MacArthur Fellowship, sometimes called a “genius grant.”

August 30, 2017 8

Yu, who is the Frank and Bertha Hupp Professor of Chemistry at TSRI, will receive a $625,000 fellowship over five years from the John D. and Catherine T. MacArthur Foundation. The grant comes with no specific obligations or reporting requirements.

“Jin is an extraordinarily creative chemist,” said TSRI CEO Peter Schultz. "This is a well- deserved honor and, following the MacArthur Fellowship awarded to TSRI’s Phil Baran in 2013, is a wonderful recognition of the remarkable science being conducted here at The Scripps Research Institute. I offer my heartiest congratulations to Jin—I always knew he was a genius, but this confirms it!”

MacArthur Fellowships are awarded to individuals who have shown extraordinary originality and dedication in their creative pursuits and a marked capacity for self-direction. Individuals cannot apply for the award; they must be nominated. Typically, 20 to 30 fellows from a wide variety of fields are selected each year.

Yu’s work in the field of organic synthesis focuses on the development of new strategies and tools to accelerate catalytic C-H activation reactions.

“At a time when the science and concepts were on the wish list of dream reactions not yet feasible, Jin-Quan Yu systematically and single-handedly transformed the field, developing powerful new synthetic methods for selective C–H activation,” said Dale Boger, who is the Richard and Alice Cramer Professor and chair of the Department of Chemistry at TSRI. “He pioneered many of the first practical and robust carbon-hydrogen (C–H) bond-activation reactions now used in nearly every sector of chemical science.

“His methods have transformed how we think about making molecules, how we conduct medicinal chemistry and how tools are created for chemical biology,” Boger continued. “He is, simply, a brilliant scientist.”

Yu, a graduate of East China Normal University (1987) and University of Cambridge (1999), joined the TSRI faculty in 2007 after an appointment at Brandeis University and a Royal Society fellowship at the University of Cambridge (United Kingdom). A fellow of the American Association for the Advancement of Science and the Royal Society of Chemistry, Yu is the recipient of many awards and honors. For more information, see his faculty web page.

One of the nation’s largest independent foundations, the MacArthur Foundation supports creative people and effective institutions committed to building a more just, verdant, and peaceful world. In addition to selecting the MacArthur Fellows, the Foundation works to defend human rights, advance global conservation and security, make cities better places and understand how technology is affecting children and society. For more information, see the websites of the MacArthur Foundation and its fellows program.

August 30, 2017 9

Scripps Florida Scientists Awarded Special Grant to Develop Memory-Altering Medication for Addiction

JUPITER, FL – October 13, 2016 – In a development that brings the world one step closer to a time when destructive addiction-fueling memories can be erased with a single treatment, scientists from the Florida campus of The Scripps Research Institute (TSRI) have been given a grant by the National Institutes of Health through the Blueprint Neurotherapeutics Network and the National Institute of Drug Abuse.

TSRI Associate Professor Courtney Miller, an early pioneer in this emerging research, will be a principal investigator of the new five-year grant, along with TSRI investigators Professor Patrick Griffin and Associate Professor Ted Kamenecka. The first year’s funding is for $640,000, followed by the possibility of additional support in the millions, including outsourced studies. Each year’s funding is based on reaching certain milestones, with the goal of a Phase 1a clinical trial in the fifth year.

“We’re excited that the NIH recognizes the outstanding potential of our research,” Miller said. “We have a solid target, a viable drug candidate and a clear development path. Now our research is on an accelerated footing toward clinical trials.”

Miller’s approach has been compared to the cult film classic “Eternal Sunshine of the Spotless Mind,” in which Jim Carrey’s character attempts to get over a romantic breakup by erasing his memories. But for methamphetamine abusers, memories are not only painful, they can also trigger relapse. As a 2015 article highlighting Miller’s research in The Washington Post concluded: “A spotless mind…[is] a chance for an addict to start over again.”

In their research, Miller and colleagues are examining a drug candidate called blebbistatin. The compound enables scientists to take advantage of the fact that meth-related memories are more unstable than other memories. In animal models, the team was able to show that a single treatment with blebbistatin was sufficient to produce a seemingly permanent loss of meth- associated memories without affecting other types of recall. The potential of such an effect with a single treatment significantly reduces the typical safety and toxicity concerns of making a new drug.

The grant, number UH2NS096833, was made as part of the NIH’s Blueprint Neurotherapeutics Network, which aims to generate novel compounds and data on their activity, toward the goal of discovering and developing new drugs that will ultimately be commercialized and benefit the public.

The Scripps Research Institute and California Institute for Biomedical Research Sign Strategic Affiliation to Accelerate the Development of New Medicines

Lays the Foundation for a New Evergreen Biomedical Research Model

LA JOLLA, CA – October 20, 2016 – The Scripps Research Institute (TSRI) and the California Institute for Biomedical Research (Calibr) – two leading non-profit research organizations –

August 30, 2017 10 today announced the signing of a strategic affiliation that combines the two organizations into a new biomedical research entity with the tools and know-how to rapidly translate its scientific discoveries into life-saving medicines for the public benefit.

This integration of basic scientific and translational research will reduce the costs and timelines associated with the crucial early stages of drug development, and lead to the creation of a self- sustaining model for non-profit research in which drug development successes drive the funding of new scientific discoveries and medicines many years into the future.

The union of these two non-profit organizations – both led by Peter Schultz, Ph.D. – provides the ability to rapidly move the basic scientific research carried out at TSRI into Calibr, whose expertise and infrastructure can accelerate drug development from discovery through early clinical trials, complementing the pharmaceutical industry's increasing focus on the later stages of clinical development.

“The affiliation between TSRI and Calibr is the first of its kind. Unlike other research institutes and universities that have sought to establish translational capabilities within academic centers, we aren’t building from scratch – rather, we are integrating the strengths of two proven non- profit organizations, enabling TSRI to remain committed to basic biological and chemical discovery and the understanding of disease processes, while turning these insights into innovative new medicines for unmet needs ranging from cancer and degenerative disease to childhood and neglected diseases through the affiliation with Calibr,” said Schultz, President of both organizations. “Ultimately, I believe this ‘bench-to-bedside’ model has the potential to become self-sustaining, with the value we create being reinvested back into research, education and additional clinical studies.”

As part of the formal affiliation, the two organizations will now share a combined Board of Directors, as well as scientific and administrative resources that will be consolidated over time. Schultz, who has an outstanding track record in both basic research and drug discovery, is overseeing the integration of the two institutes’ distinct cultures and operations. Both institutes will continue to collaborate with their other partners in the academic and commercial sectors.

“I want my work to have an impact on the lives of people suffering from life-threatening diseases, and the TSRI integration with Calibr provides an important mechanism for moving molecules faster and further along the research to development continuum, while also creating additional value for TSRI and Calibr,” said Jeff Kelly, Ph.D., Lita Annenberg Hazen Professor of Chemistry, Chairman, Department of Molecular Medicine at TSRI.

TSRI has a strong history of success in basic science – the research that lays the groundwork for new medical advances. The Institute is internationally recognized for its scientific impact in the fields of chemistry, structural and molecular biology, immunology and neuroscience, and for its many scientific breakthroughs that have led to therapies that address infant respiratory distress, rheumatoid arthritis, lupus, hemophilia and blood cancers.

Each of the 10 regulatory agency-approved medicines emerging from TSRI took an average of more than 20 years to move from drug discovery to market availability. The new affiliation

August 30, 2017 11 between TSRI and Calibr aspires to accelerate that timeline, as well as to increase the number of basic science discoveries that are translated into first-in-class drugs.

Founded in 2012, Calibr has leveraged its strengths in small molecule discovery, protein engineering, and translational research through collaborations with academic, foundation and industry partners. To date, Calibr has generated a pipeline of candidate medicines for a number of disease areas – including new approaches to osteoarthritis, tuberculosis, childhood diarrhea, heart disease, fibrosis, and cancer – creating opportunities to move five to 10 new molecules into the clinic within the next two years.

“The partnership with TSRI is critical to our long-term vision for Calibr,” said Matt Tremblay, Ph.D., Calibr’s Chief Operating Officer. “We’re thrilled to work alongside TSRI’s world-class faculty and bring Calibr’s capabilities and focus to bear in creating a self-renewing source of new drug discovery opportunities well into the future.”

The two organizations have already successfully collaborated on several research programs in recent years, including the development of an antibody engineering platform that could improve treatments for chronic diseases such as diabetes and COPD, and innovative immune therapies for the treatment of cancer. These collaborations have led to several candidate therapies, two of which Calibr has partnered with major pharmaceutical companies. Moreover, these collaborations are providing a unique training environment for students and fellows at the interface of basic and translational research, resulting in high-impact publications, and preparing them to tackle some of the most important biomedical challenges in academia and industry.

Scripps Florida Scientist Awarded $5 Million Outstanding Investigator Grant to Study the Biology of Memory

JUPITER, FL – December 20, 2016 – Ron Davis, chair of the Department of Neuroscience on the Florida campus of The Scripps Research Institute (TSRI) has been awarded a $5 million Outstanding Investigator Grant, one of the first of its kind, by the National Institute of Neurological Disorders and Stroke (NINDS) of the National Institutes of Health (NIH).

The new eight-year grant will focus on the biological processes that underlie memory formation, targeting the brain mechanisms that mediate forgetting, how the brain organizes memories, and the role for genes that suppress memory formation.

The grant is fully funded for the first five years. At the end of the fifth year, an administrative review of the program’s progress will decide on an additional three-year renewal.

Davis, who has pioneered the study of memory formation, particularly how the brain actively forgets certain memories, will be the principal investigator for the new grant, which was launched by NINDS earlier this year.

“This new type of grant will fund our research program, not just one single project,” Davis said. “It’s a long-term endorsement of our work. I’m pleased to have the NINDS endorsement

August 30, 2017 12

and extremely grateful for their support. It will help us build on what we’ve accomplished over the last ten years.”

The new funding will cover virtually all the Drosophilia research done in the lab, Davis noted. “We use Drosophilia—the common fruit fly—to better understand the basic principles of active memory formation and as a guide in our drug discovery efforts,” he said. “For our drug discovery efforts, our studies with the fly help us filter through genes and brain proteins that might be important targets for developing cognitive enhancers as therapeutics.”

Davis’s research into how the brain actively forgets certain memories represents a breakthrough in what has been a largely unstudied area of memory formation and offers tremendous opportunities for making new discoveries in the molecular biology of the process.

“The overall results of our research going forward will offer an unprecedented view of the constraints the brain uses to limit memory formation,” Davis said. “There’s a rich medical importance to this research, given the well-documented problems of cognition associated with numerous neurological and psychiatric disorders. In fact, the majority of human neurological and psychiatric disorders involve some impairment in learning and memory.”

According to NINDS, the Outstanding Investigator Grant is designed specifically to give scientists the freedom to “pursue longer range, innovative, high-risk research without feeling pressured to generate results quickly to renew short-term grants.”

The new grant also had a shorter submission process to reduce the amount of time scientists spend writing and administrating multiple grant awards.

The number of the grant is 1R35NS097224-01.

Intarcia and Calibr Announce Collaboration and In-Licensing Deal Aimed At Delivering a Next Generation Combination Product For Diabetes & Obesity

 Collaboration aims to develop a novel peptide therapeutic derived from Calibr’s Stapled-Peptide Technology Platform that can be combined with ITCA 650, Intarcia’s late stage investigational GLP-1 therapy for Type 2 Diabetes.  The Calibr deal expands Intarcia’s existing combination product strategy and pipeline that is targeting diabetes, obesity and other potential indications.  Intarcia’s Medici Drug Delivery SystemTM will be leveraged to optimize the combination therapy and then deliver it in a once- or twice-yearly mini-pump.

Boston, MA, and San Diego, CA, January 9, 2017 – Intarcia Therapeutics, Inc. and the California Institute for Biomedical Research (Calibr) today announced a strategic research collaboration focused on the development of a novel peptide therapeutic leveraging Calibr’s Stapled-Peptide Platform Technology. Intarcia plans to develop and administer this novel therapy in combination with Exenatide (a GLP-1 receptor agonist and active agent in Intarcia’s ITCA 650 product), by leveraging its proprietary Medici Drug Delivery SystemTM.

August 30, 2017 13

Under the terms of the agreement, Calibr will receive an upfront grant of Intarcia equity, with additional shares vesting over key development milestones, as well as undisclosed cash payments on achievement of predetermined regulatory and sales milestones. In addition to the milestone payments, Calibr will be eligible to receive tiered royalties on product sales.

Kurt Graves, Chairman, President and CEO of Intarcia Therapeutics, commented: “We are committed to disruptive innovation leveraging our proprietary Medici Drug Delivery SystemTM and we now have several combination products in our pipeline that we believe we can uniquely optimize and deliver in a once- or twice-yearly mini-pump. We’re excited to advance a combination program with Calibr that is targeting significant advances for patients suffering from type 2 diabetes and obesity.”

“This collaboration is another example of Calibr’s drive to work with innovative partners who are committed to making a difference in human health”, said Matt Tremblay, Ph.D., Calibr’s Chief Operating Officer. “We look forward to working with Intarcia to leverage our peptide technology platform to create transformative new medicines.”

About the Medici Drug Delivery System™

Intarcia’s novel technology platform, the Medici Drug Delivery System, is a proprietary subcutaneous delivery system comprised of three unique technologies:

 A stabilization technology that allows for proteins, peptides, antibody fragments, and other highly potent small molecules to be stabilized at or above human body temperatures for up to three years or more  A matchstick-sized osmotic mini-pump that is placed just under the dermal layer of skin to deliver a continuous and consistent flow of medication  A placement technology including proprietary tools designed to provide an optimal user experience

Once a drug is approved, a trained healthcare professional can place the small device in an in- office procedure. Once in place under the skin, water from the extracellular fluid enters the pump device at one end – by diffusing through a semi-permeable membrane directly into an osmotic engine – that expands to drive a piston at a controlled rate. This allows the drug within the pump to be released in a steady, consistent fashion at the other end of the device. Each osmotic mini-pump is designed to hold an appropriate volume of drug to treat a patient for up to a full year.

The Scripps Research Institute Signs Collaboration Agreement with to Advance DNA-Encoded Library Technology

LA JOLLA, CA – January 10, 2017 – The Scripps Research Institute (TSRI), a leading non- profit biomedical research institute, today announced a research collaboration and license agreement with Pfizer Inc. (NYSE: PFE) to pioneer new DNA-encoded library (DEL) technology, including new synthetic chemistry for the creation of next-generation DELs, a potentially transformative technology for early stage drug discovery research.

August 30, 2017 14

Under the terms of the collaboration, Pfizer will pay a technology access fee and thereby gain access to innovative chemical synthesis technology developed at TSRI. Members of the TSRI chemistry department—Professors Phil Baran, Ph.D., Dale Boger, Ph.D., Jin-Quan Yu, Ph.D., K. Barry Sharpless, Ph.D., and others—will work alongside Pfizer scientists to adapt these chemical methods for use in creating DELs, which require stringent processes that are tolerant of the delicate DNA backbone. TSRI and Pfizer may choose to expand the scope of the joint research to include other technologies relevant for enabling DEL-based drug discovery. Financial terms of the agreement are not disclosed.

“TSRI’s chemistry department is known for its rich history of innovation, and that has never been more true than today," said TSRI President Peter Schultz, Ph.D. “We are delighted to partner with Pfizer in an area where pioneering chemical methodologies can have a significant impact on the discovery of new medicines for unmet medical needs.”

In contrast to conventional drug screening where a few million small molecules are evaluated in biological systems, DEL screening uses DNA-based “barcodes” to survey billions of small molecules, potentially increasing the ability of researchers to identify promising chemical leads. While this technology was originally conceived at TSRI by Richard Lerner, M.D., and Sydney Brenner, Ph.D., in the early 1990s, the reduction to practice has taken decades and required technological advances in DNA sequencing and informatics in order to be more fully realized.

“We look forward to continuing our longtime relationship with TSRI,” said Tony Wood, Senior Vice President and Head of Medicinal Sciences, Pfizer. “We hope that this new collaboration will contribute to the creation of novel chemistries that can be used by Pfizer to prepare unique libraries for screening which, together with our parallel medicinal chemistry expertise, will have the potential to help us accelerate the discovery of new medicines for patients.”

Scripps Florida Team Awarded $1.8 Million Grant to Develop Drugs for Heart Disease and Rheumatoid Arthritis

JUPITER, FL – January 24, 2017 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have been awarded approximately $1.8 million from the National Institute of General Medical Sciences of the National Institutes of Health to develop a series of drug candidates for a number of diseases, including heart disease, rheumatoid arthritis and several neurodegenerative disorders.

The co-principal investigators of the three-year project are TSRI Professor William R. Roush and Associate Professor Derek Duckett.

“This is an exciting project, and I think our combined efforts are well-positioned to make significant progress over the next three years,” said Roush.

The new project will focus on an enzyme known as ASK1, which is involved in mediating cell survival and programmed cell death, or apoptosis. ASK1 is part of a larger family of mitogen- activated protein kinases (MAP kinases), enzymes that help control a cell’s response to stress.

August 30, 2017 15

A number of studies have shown that animal models lacking ASK1 have decreases in the size of myocardial infarctions (heart attacks) and a marked resistance to heart cell death.

“ASK1 has been highlighted as a therapeutic target in several stress-related diseases and we expect that inhibitors of its activity will provide significant benefit in multiple disease states,” Duckett said.

Under a previous grant, Duckett and his colleagues completed a high throughput screening campaign aimed at identifying the best lead molecules to target ASK1, ultimately producing a series of small molecule inhibitor candidates from six different structural classes.

“This new grant is the result of another fruitful collaboration between Derek’s lab and mine,” Roush noted. “The starting point for our effort to develop a selective and brain penetrant ASK1 inhibitor originated from high throughput screening, molecular modelling for compound design, and initial rounds of synthesis and testing.”

The optimization strategy going forward, Duckett explained, will focus on developing inhibitor properties sufficient for use in pre-clinical testing, and ultimately, in safety assessment and clinical trials.

The number of the grant is 1R01GM122109-01.

Scripps Florida Collaboration Awarded $3.3 Million to Develop Next-Generation Breast Cancer Therapies

JUPITER, FL, February 14, 2017 – A pair of scientists from the Florida campus of The Scripps Research Institute (TSRI) have been awarded up to $3.3 million from the National Cancer Institute of the National Institutes of Health (NIH) to create the next generation of breast cancer treatments for the thousands of patients whose current treatment options are limited.

Ben Shen, TSRI professor and co-chair of the Department of Chemistry, and Christoph Rader, TSRI associate professor in the Department of Immunology and Microbiology, will co-lead the new five-year study.

The researchers aim to develop a potent type of therapy known as an antibody-drug conjugate (ADC). This new class of anti-cancer drugs combines the specificity of antibodies, which attack only cells they recognize, with a highly toxic payload designed to kill specific cancer cells with far greater efficiency than most currently available treatments. So far, only three of these combination therapies have been approved by the U.S. Food and Drug Administration (FDA).

The new ADC approach targets HER2-postive and ROR1-positive breast cancers, which are often aggressive and harder to treat with conventional chemotherapy and hormone drugs.

The new grant builds on the work done in both the Shen and Rader labs.

August 30, 2017 16

Shen and his colleagues recently uncovered a new class of natural products called tiancimycins, (TNMs) which kill selected cancer cells more rapidly and more completely compared with the toxic molecules already used in FDA-approved ADCs.

Rader, who has spent most of his scientific career at TSRI and the NIH, has been studying and developing site-specific ADCs to treat cancer.

“This grant matches my lab’s work on advancing antibody engineering and conjugation technologies with the world-class natural product-based drug discovery in Ben Shen’s lab,” Rader said. “It’s precisely what I came to Scripps Florida for: to build new molecules at the interface of chemistry and biology that can advance medicine. I’m very pleased that the NIH continues to invest in our ideas.”

Since HER2 and ROR1 expression is highly complementary, the new collaboration could provide new treatment options for at least 50 percent of breast cancer patients, Shen noted.

“At Scripps Florida we not only do great science, but we have even greater opportunities to collaborate on projects like this,” Shen added. “The combination of Christoph Rader’s antibody technology and the tiancimycins, which have been proven to be exquisitely potent, should produce an antibody drug conjugate that we hope to move very quickly into the clinic.”

The grant number is 1R01CA204484-01A1.

Scripps Florida Scientist Awarded $4.8 Million to Bring HIV Closer to Human Trials

JUPITER, FL – April 11, 2017 – Professor Michael Farzan, co-chair of the Department of Immunology and Microbiology on the Florida campus of The Scripps Research Institute (TSRI) has received $4.8 million in funding through a 2017 Avant-Garde Award for HIV/AIDS research from the National Institutes of Health’s National Institute on Drug Abuse (NIDA). The new funding will support a five-year project, led by Farzan, to bring a potential HIV vaccine closer to human clinical trials.

This work will build on a 2015 study, published by Farzan and colleagues in the journal Nature, showing that researchers can use a gene-therapy approach to prompt muscle tissue to produce HIV-fighting antibodies or antibody-like molecules. Further studies from the Farzan lab have shown that this method works as a vaccine to protect nonhuman primates from HIV.

With the new funding, Farzan and his lab will explore the development of an “off switch” that halts production of these antibodies and antibody-like molecules. Their goal is to design a way to counteract any bad reaction to the vaccine and make the vaccine safe for long-term exposure.

“To really make this vaccine concept work, we have to have a way to turn it off,” Farzan said.

August 30, 2017 17

Farzan is one of three researchers to win a 2017 NIDA Avant-Garde Award, which aims to stimulate high-impact research for the prevention and treatment of HIV/AIDS in drug users.

“With nearly 37 million people living with HIV worldwide, it is essential that researchers continue to develop effective prevention and treatment strategies for those suffering from this devastating disease, including people with substance use disorders,” said NIDA Director Nora D. Volkow, M.D., in a statement. “These scientists are pioneering exciting new approaches aimed at preventing and treating new cases of HIV and helping people at risk live longer, healthier lives.”

The grant number is DA043912-01.

HitGen Announces Drug Discovery Collaboration with the Scripps Research Institute and the California Institute for Biomedical Research (Calibr)

CHENGDU, China--HitGen Ltd., a privately held biotech company focused on technology for effective and efficient small molecule drug discovery and development, announced today a collaboration with The Scripps Research Institute (TSRI) and its affiliate, the California Institute for Biomedical Research (Calibr). This collaboration is to discover and develop potential new therapies in areas of unmet clinical need, with an initial focus in oncology, regenerative medicine and virology.

HitGen’s technology platform, centered around the design, synthesis, and interrogation of multi‐billion component libraries of DNA encoded small molecules, will be deployed to identify new small molecule leads for therapeutic targets identified by the collaboration. Leaders in basic and translational research at TSRI and Calibr will provide target materials and relevant know‐how to prosecute drug targets of interest. The parties will contribute in‐kind to the discovery efforts and anticipate progressing any drug candidates that emerge to clinical proof‐of‐concept studies.

“We are delighted to partner with HitGen to further the combined TSRI‐Calibr mission to create new medicines for unmet needs. HitGen’s commitment to developing and deploying DEL technology is a great fit for our new model for translational research in the non‐profit sector,” commented Peter Schultz, President and Professor of Chemistry at TSRI, and CEO of Calibr.

“We are excited to collaborate with Prof. Schultz and his colleagues at TSRI‐Calibr as a strategic partner for these discovery efforts, and we acknowledge the fundamental contributions of TSRI Professors Sydney Brenner and Richard Lerner in initiating the field of DNA‐encoded chemistry technology,” said Dr. Jin Li, Chairman & CEO of HitGen Ltd. He added that “HitGen continues to grow as a world‐class drug discovery organization. We are keenly focused on engaging with world‐leading academic institutions to provide new solutions for unmet medical need.”

August 30, 2017 18

Scripps Florida Scientist Wins $2 Million Grant to Study Childhood Disorder Linked to Behavioral Problems

JUPITER, FL – May 31, 2017 – Assistant Professor Seth Tomchik of the Florida campus of The Scripps Research Institute (TSRI) has received $2 million in funding from the National Institutes of Health’s National Institute of Neurological Disorders and Stroke (NINDS). The new five-year grant funding will support the study of neurofibromatosis type I, an inherited disorder that results from genetic mutations affecting a protein called neurofibromin (Nf1).

There is currently no FDA-approved treatment for neurofibromatosis 1. The disease begins in childhood with symptoms ranging from harmless spots to nerve tumors. It also predisposes individuals to a suite of behavioral symptoms, including attention-deficit/hyperactivity disorder (ADHD), autism-like symptoms, learning disabilities and chronic pain. The prevalence of behavioral symptoms suggests that Nf1 can affect the development or function of neurons in the brain that regulate activity levels.

Because neurofibromin is a large protein with multiple potential signaling roles in cells, Tomchik’s research will focus on the functions of Nf1 at several levels, examining its molecular biology, genetic interactions and effects on signaling and development of neuronal circuits.

“Aside from surgery to remove tumors, there are really no treatment options for those who suffer from neurofibromatosis,” Tomchik said. “We intend to look at how the underlying protein regulates behavioral hyperactivity, determining how specific neuronal circuit alterations underlie its behavioral effects.”

The grant number is 1R01NS097237.

The Scripps Research Institute and ShangPharma Innovation Announce Translational Research Collaboration

World-class research institute and new innovation center partner to accelerate early translational research

La Jolla, California (June 8, 2017) – The Scripps Research Institute (TSRI) and ShangPharma Innovation, Inc. (SPII) today announced a strategic collaboration to accelerate the development of innovative drug candidates through scientific collaboration, a services relationship with Shanghai ChemPartner Co. Ltd. (ChemPartner), and sponsored research at TSRI and its drug discovery affiliate, California Institute for Biomedical Research (Calibr). The parties aim to leverage the drug discovery pipeline, expertise, and momentum emerging from the TSRI and Calibr affiliation announced late last year.

SPII facilitates and accelerates drug discovery by offering funding, incubator space, and other essential support to its collaborators. The partnership with TSRI marks a major new initiative for SPII to enable its mission to support therapeutic research at the earliest stages with the ultimate goal of getting innovative new treatments to patients. Under the terms of the

August 30, 2017 19 collaboration, institute scientists will put forth translational research projects to a joint committee comprising drug discovery experts across TSRI, Calibr, and SPII. In turn, SPII will contribute cash and in-kind services totaling up to $15 million over an initial term of three years. TSRI and Calibr will retain control over assets resulting from the collaboration, with SPII receiving a significant share of future value.

“We are delighted to partner with an experienced group that has complementary infrastructure and resources,” said Peter G. Schultz, Ph.D., President of TSRI and Calibr. “This new initiative allows us to further accelerate our mission of creating new medicines for unmet needs in a nimble partnership structure designed to mature the programs before out-licensing, creating significant value for patients as well as for the institute.”

“Biomedical R&D is one of the most challenging areas of science across all disciplines and industries. Strategic partnerships often drive success in making the impossible possible. That is exactly what we plan to do in this alliance with TSRI and Calibr,” stated Walter H. Moos, Ph.D., CEO of SPII. “Unlocking the potential of top investigators at TSRI and Calibr to advance novel therapies and ultimately to save and improve lives is our joint goal.”

Scripps Florida Scientist and Collaborators Win $7 Million Grant to Develop New ALS Treatments

JUPITER, FL – June 15, 2017 – Professor Matthew Disney of the Department of Chemistry on the Florida campus of The Scripps Research Institute (TSRI), together with scientists from Mayo Clinic’s Florida campus and Johns Hopkins School of Medicine, has been awarded $7.2 million from the National Institute of Neurological Disorders and Stroke of The National Institutes of Health to create new RNA-based treatments for the most common form of amyotrophic lateral sclerosis (ALS), as well as a type of frontotemporal dementia (FTD).

Disney is the principal investigator on the five-year project, along with Leonard Petrucelli, chair of neuroscience at Mayo Clinic’s Florida campus, and Jeffrey D. Rothstein of Johns Hopkins School of Medicine.

While the scientists will initially focus on small-molecule drug candidates for ALS and FTD, the grant establishes the RNA Therapeutics Center at TSRI for the continued study of RNA- based therapies for a number of untreatable diseases.

“Our work has changed the view of what is considered a druggable target, and this grant is a clear recognition of that accomplishment," Disney said. "The funding will allow us to more rigorously assess if a small molecule drug can be developed to target the RNA that causes the most common inherited form of ALS and FTD. The question we can now try and answer is if these studies can help to advance the science to bring a drug to ALS patients.”

ALS is usually fatal two to five years after diagnosis. There is no effective treatment for FTD, a neurodegenerative disease that destroys neurons in the frontal lobes of the brain. This study will focus on a lead compound that interferes with the synthesis of an abnormal protein— known as a repeat RNA expansion—that plays a key role in the development of both diseases.

August 30, 2017 20

Disney has been a pioneer in the development of potential treatments for a number of diseases caused by defects in RNA, which affect millions worldwide. His success in identifying drug- like small molecules that bind to RNA is the result of his lab’s broad, bottom-up, computational approach known as Inforna, which can deep mine information against such genome sequences and cellular RNAs.

“RNA plays critical roles and causes many incurable diseases, and even more common diseases that have limited treatment options, such as cancers. The work of the new center will be to study and develop insights into developing treatments that target RNAs broadly, which were not thought to be targetable before,” Disney said. “Our work will apply to any disease.”

The initial research is a particularly good test case for RNA-based treatments, said Disney.

“The new grant supports the idea that RNA is a legitimate target to treat disease,” Rothstein added. “We believe that this approach—to truly treat the starting point of ALS—can lead to effective and impactful therapies.”

“We want to further evaluate bioactive small molecules targeting the repeat expansion in preclinical models of ALS,” Petrucelli noted.

The number of the grant is 1P01NS099114-01.

New Equipment at Scripps Florida Will Advance Drug Discovery

JUPITER, FL – May 22, 2017 – The Nuclear Magnetic Resonance (NMR) facility at the Florida campus of The Scripps Research Institute (TSRI) is becoming one of the most advanced in South Florida, thanks to a new $600,000 grant from the National Institutes of Health (NIH). The grant will fund the purchase of a 600 Mhz NMR machine, which will help researchers develop drugs and understand how those drugs act in the body.

The TSRI’s Florida campus currently houses three NMR machines, but as the scientists have continued to focus on translational research—studies that aim to “translate” laboratory findings into potential treatments for disease—those machines are constantly in use. “We were basically maxed out on instrument time,” said TSRI Associate Professor Douglas Kojetin, project leader on the new grant. “All of our instruments were all near 100 percent usage.”

The researchers use these machines throughout the drug development process. NMR applies a strong magnetic field to sample, which interacts with the individual atoms in a molecule, allowing scientists to discern that molecule’s structure. Chemists at TSRI regularly use the two lower-field NMRs the institute currently owns, which apply a magnetic field around 400MHz, to confirm the structures of molecules they have either synthesized in the lab or isolated from natural sources. TSRI also has one 700mhz NMR, which Kojetin and his colleagues use to understand how small molecules interact with other molecules in the body, including proteins, receptors and RNA.

August 30, 2017 21

The new 600mhz NMR will be able to support all types of research, with many of the capabilities of both the higher and lower field machines.

For example, Kojetin’s lab studies how drugs for type II diabetes interact with their receptors. While effective at treating diabetes, these drugs also have a wide range of undesirable side effects. Using NMR, Kojetin’s group is working to gain a better understanding of how these drug molecules cause both their positive and negative effects, with the intent of designing a molecule that could treat the disease while minimizing a patient’s side effects.

Kojetin added that the new equipment will not only expedite more than 20 NIH-funded projects currently in progress at TSRI, but it will help generate new data that will lead to more grant funding and more discoveries. “The addition of this instrument will place us within the top NMR facilities in South Florida,” said Kojetin.

The grant number is 1S10OD021550-01A1.

Part 3: Scientific Accomplishments

Scripps Florida Scientists Link Bipolar Disorder to Unexpected Brain Region

JUPITER, FL – July 8, 2016 – While bipolar disorder is one of the most-studied neurological disorders—the Greeks noticed symptoms of the disease as early as the first century—it’s possible that scientists have overlooked an important part of the brain for its source.

Scientists from the Florida campus of The Scripps Research Institute (TSRI) have shown for the first time that ensembles of genes within the striatum—a part of the brain that coordinates many primary aspects of our behavior, such as motor and action planning, motivation and reward perception—could be deeply involved in the disorder. Most modern studies of bipolar disorder have concentrated on the brain’s cortex, the largest part of the brain in humans, associated with higher-level thought and action.

“This is the first real study of gene expression in the striatum for bipolar disorder,” said Ron Davis, chair of the Department of Neuroscience at TSRI, who directed the study. “We now have a snapshot of the genes and proteins expressed in that region.”

The study, published recently online ahead of print in the journal Molecular Psychiatry, also points to several pathways as potential targets for treatment.

Bipolar disorder is a mental illness that affects about 2.6 percent of the U.S. adult population— some 5.7 million Americans—with a sizable majority of these cases classified as severe. The disease runs in families, and more than two-thirds of people with bipolar disorder have at least one close relative with the illness or with unipolar major depression, according to the National Institute of Mental Health.

August 30, 2017 22

In the new research, tissue samples from 35 bipolar and non-bipolar control subjects were analyzed. The number of genes differentially expressed in tissue samples from the two groups turned out to be surprisingly small—just 14 in all. However, co-expression network analysis also revealed two modules of interconnected genes that were particularly rich in genetic variations associated with bipolar disorder, suggestive of a causal role in the disorder. One of these two modules was particularly striking, as it seemed to be highly specific to the striatum.

“Our finding of a link between bipolar disorder and the striatum at the molecular level complements studies that implicate the same brain region in bipolar disorder at the anatomical level, including functional imaging studies that show altered activity in the striatum of bipolar subjects during tasks that involve balancing reward and risk,” said TSRI Research Associate Rodrigo Pacifico, who was first author of the new study. Analyzing reactions to risk was important because bipolar patients may act impulsively and engage in high-risk activities during periods of mania.

Pathway analysis also found changes in genes linked to the immune system, the body’s inflammatory response, and cells’ energy metabolism. Davis noted, “We don’t know if these changes are a cause of the disease or the result of it. But they provide additional gene markers in bipolar disorder that could potentially lead to the future development of diagnostics or treatments.”

The study, “Transcriptome Sequencing Implicates Dorsal Striatum-Specific Gene Network, Immune Response and Energy Metabolism Pathways in Bipolar Disorder,” was supported by funding from the State of Florida.

Study Suggests New Drug Candidate Could Treat Both Type 2 Diabetes and Bone Loss

JUPITER, FL – July 27, 2016 – In addition to its more obvious ills, type 2 diabetes is a condition closely associated with bone fractures, increasing the risk of fractures twofold. To make matters worse, certain anti-diabetic drugs further increase this risk, particularly in postmenopausal women, severely limiting their treatment options.

A new study, co-led by Patrick R. Griffin, a professor on the Florida campus of The Scripps Research Institute (TSRI), and B. Lecka-Czernik, a professor at the University of Toledo, has shown that a new class of drug candidates developed at TSRI increases bone mass by expanding bone formation (deposition of new bone) and bone turnover (a normal process of replacement of old bone). A proper balance of these two processes is critical to healthy bone maintenance, and this balance is frequently negatively affected in diabetic patients.

The result is a new dual-targeting drug candidate—or, as Griffin describes, “one drug addressing multiple therapeutic indications”—that could treat both diabetes and bone disease. The compound has been referenced as “SR10171.”

The study was published recently online ahead of print by the journal EBioMedicine.

August 30, 2017 23

Diabetes affects more than 29 million people in the United States, according to a 2012 report from the American Diabetes Association. In 2013, estimated direct medical costs of the disease were $176 billion.

Over the past decade, Griffin and his colleague, TSRI Associate Professor Theodore Kamenecka, have focused on the details of molecules that increase sensitivity to insulin (a hormone that regulates blood sugar). Using newly discovered information, the researchers made significant advances in developing a family of drug candidates that target a receptor known as peroxisome proliferator-activated receptors gamma (PPARγ), a key regulator of stem cells controlling bone formation and bone resorption and a master regulator of fat.

Anti-diabetic drugs known as glitazones (TZDs) target the PPARγ protein, but that interaction leads to severe bone loss and increased fractures. Stem cells in the bone marrow can differentiate either into bone cells or fat cells, and the glitazones drive them to fat at the expense of bone.

But SR10171 is designed to avoid this troubling outcome. In animal models treated with the compound, fat formation in the bone marrow was successfully blocked independent of their metabolic state (healthy or diabetic).

“Using structural biology technigues and rational design synthetic chemistry, SR10171 was constructed to engage the PPARγ protein in a unique way possessing an optimal balance with the receptor’s other family member, PPARa, to treat diabetes and, at the same time, improve bone health,” Griffin said. “This targeted polypharmacological approach demonstrates that the target isn’t the problem if you target it correctly.”

The compound increases bone mass by protecting and increasing the activity of bone cells in various stages of normal bone maintenance, utilizing mechanisms that overlap those that regulate whole-body energy metabolism.

“SR10171 improves bone mass regardless of body mass index, normal to obese,” Griffin added. “So you could use such a drug to treat osteoporosis whether patients are diabetic or not.”

The first author of the study, “PPARG Post-Translational Modifications Regulate Bone Formation and Bone Resorption,” is L.A. Stechschulte of the University of Toledo, Ohio. Other authors include P.J. Czernik, Z.C. Rotter and F.N. Tausif of the University of Toledo; C.A. Corzo, D.P. Marciano, A. Asteian, J. Zheng and T.M. Kamenecka of TSRI; J. B. Bruning of The University of Adelaide, Australia; and C.J. Rosen of the Maine Medical Center Research Institute.

The study was supported by the National Institutes of Health (grant numbers DK080261 and DK105825); the American Diabetes Association (award 7-13-BS-089), the Abrams Charitable Trust and the Klorfine Family Fellowship.

August 30, 2017 24

New Study Shines Light on Mutations Responsible for Debilitating Heart Conditions

JUPITER, FL – August 11, 2016 – The leading cause of death in the world remains cardiovascular diseases, which are responsible for more than one third of overall mortality, according to the World Health Organization. Obesity and diet are obvious culprits behind heart disease but, over the past decade, research has also pointed to genetic factors, specifically mutations in cell adhesion components—the forces that bind cells together.

In a new study, scientists from the Florida campus of The Scripps Research Institute offer new molecular insights into how the interaction between specific genetic mutations and a cytoskeletal protein critical for the proper development and maintenance of heart tissue can lead to conditions such as dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM)—and ultimately heart failure.

The new study, which was led by Associate Professor T. Izard of the Florida campus of TSRI, is published this week in an early online edition of the journal Proceedings of the National Academy of Sciences. The new insights could aid in the development of drug therapies to strengthen the hearts of patients suffering from age-related heart failure.

The study focuses on the protein vinculin and a variant form known as metavinculin, which is found only in muscle tissue. Vinculin has been shown to reinforce the myocardial cell cytoskeleton, improving heart muscle contractility and prolonging life, while metavinculin plays an essential role in the development and function of the heart.

Both vinculin and metavinculin regulate cell adhesion and migration by linking the cell’s cytoskeleton to adhesion receptor complexes via a process known as dimerization—the joining of two similar subunits. Control of the dimerization process is crucial for normal protein function in cell adhesion sites.

But mutations in the variant metavinculin, either inherited or spontaneous, corrupt this process, altering dimerization and, the study suggests, producing a decreased ability to stabilize critical cell adhesions, weakening the heart muscle over time.

The researchers found that these mutations—specifically, a mutation known as R975W in metavinculin—dictate the type of interaction during dimerization and can actually block the process. That, in turn, results in heart muscles that are far more susceptible to stress-induced heart disease.

The first author of the study, “Differential Lipid Binding of Vinculin Isoforms Promotes Quasiequivalent Dimerization,” is Krishna Chinthalapudi of TSRI. Other authors include Erumbi S. Rangarajan of TSRI and David T. Brown of the University of Mississippi Medical Center.

The laboratory is supported by the National Institutes of Health, the Department of Defense, the American Heart Association and the State of Florida.

August 30, 2017 25

Scripps Florida Study Finds 'Missing Evolutionary Link' of a Widely Used Natural Drug Source

JUPITER, FL – August 19, 2016 – A well-known family of natural compounds, called “terpenoids,” have a curious evolutionary origin. In particular, one question relevant to future drug discovery has puzzled scientists: exactly how does Nature make these molecules?

A study from scientists on the Florida campus of The Scripps Research Institute (TSRI) has now filled in a missing piece of the evolutionary puzzle, determining a previously unknown structure of a family of proteins that are key to making these compounds.

Terpenoids, mostly produced by plants, are a family of molecules that encompass some of the most well-known and successful drugs derived from natural sources, for example the cancer treatment Taxol®. In total, there are more than 65,000 known terpenoids.

Natural compounds, such as terpenoids, are typically made by numerous enzymes that stitch the parts together, like building a model from Lego®. In the new study, the team focused on enzymes known as terpene synthases, which are found in both plants and bacteria.

The study, led by TSRI Professor Ben Shen, was recently published online ahead of print by the Journal of the American Chemical Society. In addition to the advance in the structural biology of this class of proteins, the new findings could also affect drug discovery, making engineering of these proteins, and thus the compounds they make, easier in the future.

Using x-ray crystallography—freezing the atomic structure of a protein, then bombarding it with x-rays to create a snapshot of it—Shen and his colleagues developed a detailed structural model that provides a far better understanding of bacterial diterpene synthases and how this class of enzymes work in a natural setting.

“What we found out is that the bacterial version is structurally very similar to the plant version and may support the idea of a gene fusion event that created the bifunctional plant enzymes,” said TSRI Research Associate Jeffrey Rudolf, a co-first author of the paper with TSRI Research Associate Liao-Bin Dong. “We were also able to map which parts of the enzyme are important, giving us an idea of how to engineer the protein for structural diversity.”

Dong added, “This new information not only allows us to engineer structural diversity into both bacterial and plant terpenoids, it also helps us identify new diterpenoids of bacterial origin, which are rare and could lead to exciting new natural compounds with interesting biological activities.”

In addition to Shen, Rudolf and Dong, other authors of the study include Hongnan Cao and George N. Phillips of Rice University; Catherine Hatzos-Skintges, Jerzy Osipiuk, Michael Endres, Gyorgy Babnigg and Andrzej Joachimiak of Argonne National Laboratory; Chin-Yuan Chang and Ming Ma of TSRI.

August 30, 2017 26

The study, “Structure of the ent-Copalyl Diphosphate Synthase PtmT2 from Streptomyces platensis CB00739, a Bacterial Type II Diterpene Synthase,” was supported by the National Institute of General Medical Sciences Protein Structure Initiative (grants GM094585 and GM098248) and the National Institute of General Medical Sciences (grants GM109456 and GM114353).

Scripps Florida Scientists Shed New Light on the Role of Calcium in Learning and Memory

JUPITER, FL – August 26, 2016 – While calcium’s importance for our bones and teeth is well known, its role in neurons—in particular, its effects on processes such as learning and memory—has been less well defined.

In a new study published in the journal Cell Reports, scientists from the Florida campus of The Scripps Research Institute (TSRI) offer new insights how calcium in mitochondria—the powerhouse of all cells—can impact the development of the brain and adult cognition.

In particular, the team showed in fruit flies, a widely used model system, that blocking a channel that brings calcium to the mitochondria, called “mitochondrial calcium uniporter,” causes memory impairment but does not alter learning capacity.

“When we knocked down the activity of the uniporter, we found that flies have a deficit memory,” said Ron Davis, chair of the TSRI’s Department of Neuroscience. “Intact uniporter function is necessary for full and complete memory in the adult fly. What surprised us is that they were still able to learn—albeit with a fleeting memory. But we thought they wouldn’t be able to learn at all.”

The mitochondrial calcium uniporter protein, first identified in 2011, allows calcium ions to move from the cell's interior into mitochondria—like coal moving through a shoot into a furnace room. It is regulated by other proteins known as MICU1, MICU2 and EMRE. Davis noted that human patients with mutations in MICU1 can exhibit learning disabilities.

“The new study’s conclusion is that mitochondrial calcium entry during development is necessary to establish the neuronal competency for supporting adult memory,” said TSRI Research Associate Ilaria Drago, the first author of the study.

Drago noted the team found evidence that inhibiting mitochondrial calcium uniporter function led to a decrease in the content of synaptic vesicles (miniscule sacs within the cell where various neurotransmitters are stored) and an increase in the length of axons (the slender filaments of neurons).

While these structural problems were clearly observed, she added, what they mean in terms of neuronal development remains tantalizingly unclear.

August 30, 2017 27

“The discovery of a developmental role for the mitochondrial calcium uniporter complex in regulating memory in adult flies is especially intriguing and deserves more exploration,” said Davis.

The study, “Inhibiting the Mitochondrial Calcium Uniporter during Development Impairs Memory in Adult Drosophila,” was supported by the National Institutes of Health (grants R37NS19904 and R01NS052351) and the Iris and Junming Le Foundation.

TSRI's Peter Schultz Wins Prestigious Heinrich Wieland Prize

LA JOLLA, CA—September 29, 2016—Peter Schultz, president of The Scripps Research Institute (TSRI), has been awarded the international Heinrich Wieland Prize, one of Germany’s most prestigious scientific prizes.

The award, presented by the Boehringer Ingelheim Foundation, honors “distinguished scientists for their outstanding research on biologically active molecules and systems in the fields of chemistry, biochemistry and physiology as well as their clinical importance.” Previous Heinrich Wieland Prize winners have included subsequent Nobel Laureates Michael Stuart Brown, Joseph L. Goldstein, Bengt Samuelsson and James Rothman.

Schultz, who also holds the Scripps Family Chair Professor of Chemistry at TSRI, was selected for “his fundamental contributions to the biologically inspired synthesis of new molecules including his groundbreaking work on expanding the genetic code. Using his toolbox to extend the genetic code, over 100 amino acids with novel functions have been introduced site- specifically into proteins allowing to probe cellular processes in vivo in unprecedented detail. Schultz has applied this method to generate new medicines and also to create organisms with enhanced properties. In addition, he has pioneered the development of large combinatorial chemical libraries together with cell-based screens leading to drugs for degenerative diseases, cancer, autoimmune and neglected diseases.”

“Peter Schultz has given us a wide array of invaluable tools which enable us to rationally design molecules and organisms to understand the processes of life and to treat medical conditions,” said Professor Wolfgang Baumeister, who chairs the Selection Committee of the Heinrich Wieland Prize.

Awarded annually since 1964, the prize, which is named after 1927 Nobel Laureate in Chemistry Heinrich Wieland, is endowed with 100,000 euros. Schultz will receive the prize and present the laureate’s lecture at an award symposium on October 13 in Munich, Germany.

For further information on Schultz’s research, visit his faculty webpage and lab website. For more information on the Heinrich Wieland Prize, see the award’s website.

August 30, 2017 28

Scripps Florida Scientists Identify Potent New Anti-Obesity, Anti-Diabetes Target

JUPITER, FL – October 12, 2016 – It is no secret that losing weight is difficult. Each year brings a new popular diet, “guaranteed” to succeed, yet the explosion of obesity in the United States continues.

Current anti-obesity medications are only partly effective, due to substantial side effects, the temporary nature of the weight loss and the non-responsiveness of a considerable number of patients.

In a recently published series of studies led by TSRI Assistant Professor Anutosh Chakraborty of the Florida campus of The Scripps Research Institute (TSRI), scientists have identified a new therapeutic target—a key protein that promotes fat accumulation in animal models by slowing the breakdown and expenditure of fat and encouraging weight gain. The studies were published in The International Journal of Biochemistry & Cell Biology, Molecular Metabolism and, most recently, The Journal of Clinical Investigation.

“We found that the protein IP6K1 is a viable target in obesity and type 2 diabetes,” Chakraborty said. “We also discovered that an inhibitor of the protein known as TNP decelerates what is known as diet-induced obesity and insulin resistance.”

In the last few years, enhancing energy expenditure has emerged as an attractive strategy to combat obesity and diabetes, although how this might be accomplished remains something of a mystery simply because the mechanisms by which the body maintains its energy balance, is complex.

The new studies point to IP6k1 (inositol hexakisphosphate kinase-1), specifically its inhibition, as a potentially rich target. Chakraborty and his colleagues found that deleting IP6K1 in fat cells enhanced energy expenditure and protected animal models from diet-induced obesity and insulin resistance.

The expenditure of fat energy is preceded by a process called lipolysis that breaks down stored fat or triglycerides (specifically into free fatty acids and glycerol) to be used as energy in cells. The team discovered that deletion of IP6K1 affects interaction with another regulating protein and enhances the breakdown of fats.

To determine the therapeutic possibilities of the IP6K1 pathway, the team looked at the impact of an IP6K inhibitor known as TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl) purine] on diet-induced obesity in animal models. As it turns out, TNP significantly slows the initiation of diet-induced obesity and insulin resistance.

“In addition, the compound facilitates weight loss and improves metabolic parameters when used in animals that are already obese,” said Chakraborty.

The first author of The Journal of Clinical Investigation study, “Adipocyte-Specific IP6K1 Deletion Reduces Diet-Induced Obesity by Enhancing AMPK Mediated Thermogenesis,” is

August 30, 2017 29

Qingzhang Zhu of TSRI. Other authors include Sarbani Ghoshal, Ana Rodrigues, Su Gao, Alice Asterian and Theodore M Kamenecka all of TSRI at the time of the study and James C. Barrow of the Lieber Institute (Baltimore, MD). The first author of The International Journal of Biochemistry & Cell Biology study, “Inositol hexakisphosphate kinase-1 Interacts with perilipin1 to Modulate Lipolysis,” is Sarbani Ghoshal of TSRI at the time of the study. Other authors include Qingzhang Zhu of TSRI and Richa Tyagi of the Johns Hopkins School of Medicine. The first author of the Molecular Metabolism study, “TNP [N2-(M-Trifluorobenzyl), N6-(P- Nitrobenzyl) Purine] Ameliorates Diet Induced Obesity and Insulin Resistance via Inhibition of the IP6K1 Pathway,” is Sarbani Ghoshal of TSRI at the time of the study. Other authors include Qingzhang Zhu, Alice Asteian, Hua Lin, Haifei Xu, Baoji Xu, Michael D. Cameron and Theodore M. Kamenecka of TSRI; and Glen Ernst and James C. Barrow of the Lieber Institute (Baltimore, Maryland).

The work was supported by the National Institutes of Health (grant R01DK103746) and the State of Florida.

Scripps Florida Scientists Uncover New Facets of Zika-Related Birth Defects to Help Develop Treatment

JUPITER, FL – October 17, 2016 – In a study that could one day help eliminate the tragic birth defects caused by Zika virus, scientists from the Florida campus of The Scripps Research Institute (TSRI) have elucidated how the virus attacks the brains of newborns, information that could accelerate the development of treatments.

The study, led by TSRI Associate Professors Hyeryun Choe and Damon Page, was published recently in the journal Nature Scientific Reports.

In the new study, the scientists observed the virus’s effects in animal models at two different points—during early postnatal development, when the brain is growing rapidly, and at weaning, when the brain has largely reached adult size.

“In early postnatal Zika-infected models some brain areas and cell types showed particularly large increases in apoptosis [programmed cell death] that we did not observe in older animals,” Choe said.

The findings expand the current knowledge of cell types vulnerable to the effects of Zika infection to include not only neuron progenitor cells, but also post-mitotic neurons that have finished dividing but are still are undergoing rapid increases in cell size. These results are consistent with the theory that periods of rapid brain growth are especially susceptible to the damaging neurodevelopmental effects of Zika infection.

“An interesting aspect of the study is the comparison of the two time points,” Page noted. “There is neural cell death at both times, but it’s much greater when the brain is growing rapidly. We can take advantage of this strong effect to test potential treatments and to

August 30, 2017 30 understand whether some genetic backgrounds may confer enhanced susceptibility or resilience to Zika-induced microcephaly.”

Zika virus was first isolated in rhesus macaque monkeys in Uganda in 1947. Transmitted by mosquito, it is related to several other human pathogens, including West Nile virus, dengue, Japanese encephalitis virus and yellow . As widely reported, the 2015 Zika epidemic in Brazil coincided with a dramatic increase in cases of microcephalic newborns.

“Our findings establish a valuable model to investigate the mechanisms that underlie the horrific birth defects associated with Zika infection,” said TSRI Graduate Student Wen-Chin Huang, the first author of the study.

The team is continuing to build on this study to better understand and combat the virus.

In addition to Choe, Page and Huang, other authors of the study, “Zika Virus Infection During the Period Of Maximal Brain Growth Causes Microcephaly and Corticospinal Neuron Apoptosis in Wild Type Mice,” include Rachy Abraham and Byoung-Shik Shim of TSRI.

The study was supported by the National Institutes of Health (grant R01 AI110692) and by Ms. Nancy Lurie Marks.

International Team Unveils First Atomic-Level Image of the Human ‘Marijuana Receptor’

JUPITER, FL — October 20, 2016 – In a discovery that advances the understanding of how marijuana works in the human body, an international group of scientists, including those from the Florida campus of The Scripps Research Institute (TSRI), have for the first time created a three-dimensional atomic-level image of the molecular structure activated by tetrahydrocannabinol (THC), the active chemical in marijuana.

The new insights into the human cannabinoid receptor 1 (CB1) will provide an essential tool for understanding why some molecules related to THC have unexpectedly complex and sometimes harmful effects. The findings also have the potential to guide drug design for pain, inflammation, obesity, fibrosis and other indications. The new study, published by the journal Cell, was led by a quartet of scientists: TSRI’s Laura Bohn, Northeastern University’s Alexandros Makriyannis, Shanghai Tech University’s Zhi-Jie Liu and Shanghai Tech and University of Southern California’s Raymond C. Stevens.

At the beginning of the study, the team struggled to produce a crystal form—needed to obtain data to recreate the high-resolution structure—of the receptor bound with AM6538, a stabilizing a molecule that blocks the receptor’s action.

“The CB1 receptor proved as challenging for crystallization as it did for understanding its functional regulation and signaling,” said Bohn, who is a professor in TSRI’s Department of Molecular Therapeutics.

August 30, 2017 31

When the scientists succeeded in crystalizing the receptor and collecting the data, the structure of the cannabinoid receptor complex revealed an expansive and complicated binding pocket network consisting of multiple sub-pockets and channels to various regions of the receptor.

Cannabinoid receptors are part of a large class of receptors known as G protein-coupled receptors (GPCR), which account for about 40 percent of all prescription pharmaceuticals on the market and play key roles in many physiological functions. When an outside substance binds to a GPCR, it activates a G protein inside the cell to release components and create a specific cellular response.

AM6538, is an antagonist/inverse agonist that binds tightly to the receptor; it has a long half- life, making it potentially useful as a treatment of addiction disorders.

“As marijuana continues to become more common in society, it is critical that we understand how it works in the human body,” said Liu, who is professor and deputy director of the iHuman Institute of Shanghai Tech and is also affiliated with the Chinese Academy of Sciences.

The first author of the Cell study, “Crystal Structure of the Human Cannabinoid Receptor CB1,” is Tian Hua of Shanghai Tech University and the Chinese Academy of Sciences (Beijing).

In addition to Stevens, Liu, Bohn, Makriyannis and Hua, other authors of the study are Mengchen Pu, Lu Qu, Yiran Wu, Suwen Zhao, Wenqing Shui, Shanshan Li of iHuman Institute at ShanghaiTech University; Nikolai Zvonok, Anisha Korde, Kiran Vemuri of Northeastern University; Gye Won Han of University of Southern California; Robert Laprairie, Edward Stahl, and Jo-Hao Ho (graduate student) of TSRI; Irina Kufareva of University of California, San Diego; and Michael Hanson of the GPCR Consortium.

The study was supported by the Ministry of Science and Technology of China (grants 2014CB910400 and 2015CB910104), National Nature Science Foundation of China (grant 31330019), National Institutes of Health (grants P01DA009158, R37DA023142 and R01AI118985), the National Science Foundation, Shanghai Municipal Government, ShanghaiTech University and the GPCR Consortium.

Scripps Florida Scientists Illuminate Key Molecular Player in Both Morphine Addiction and Rare Disease

JUPITER, FL – October 26, 2016 – In a remarkable “two for one” discovery, scientists from the Florida campus of The Scripps Research Institute (TSRI) have illuminated a key molecular player in the addictive effects of morphine in animal models.

Interestingly, the protein—known as neurofibromin 1 (NF1)—is also known to be disrupted in an inherited disorder called neurofibromatosis type 1 (also called von Recklinghausen's disease), in which patients suffer from the growth of benign tumors beneath the skin, chronic pain, mild learning disabilities and an elevated risk of developing cancers.

August 30, 2017 32

“We were searching for proteins that influence the long-term effects of opioids,” said TSRI Professor Kirill Martemyanov. “In the short-term, opioids kill pain, but we were curious about mechanisms that lead to long-term adaptations that ultimately result in addiction. We screened for proteins that might possibly be involved in this process and what emerged was NF1.”

The study, recently published online ahead of print in the journal Current Biology, describes how NF1 influences opioid response through its impact on a signaling protein known as Ras in a part of the brain called the striatum, which is involved in decision making and reward.

When the researchers deleted NF1 in striatal neurons of animal models, opioids failed to engage Ras and its downstream signaling reactions, dramatically diminishing the addictive effects of morphine.

“One of the peculiar symptoms of neurofibromatosis type 1 is that patients often suffer from chronic, unexplained pain,” said Martemyanov. “Given that the endogenous opioid system is involved in controlling pain sensitivity and the role of NF1 protein in mediating signaling of opioid receptors, it makes sense that if the mechanism is broken pain, might become an issue.”

He also noted the results suggest potential therapies for both conditions: “If you came up with a strategy to inhibit NF1 activity, perhaps the opioids won’t be as addictive; but for those suffering from the neurofibromatosis, bypassing NF1 to activate Ras by receptors may be interesting to explore.”

The first author of the study, “NF1 Is a Direct G Protein Effector Essential for Opioid Signaling to Ras in the Striatum,” is Keqiang Xie. Other authors include Maria T. Dao, Brian S. Muntean, Laurie P. Sutton, Cesare Orlandi, Chien-Cheng Shih, Baoji Xu and Roy G. Smith of TSRI; Lesley A. Colgan and Ryohei Yasuda the Max Planck Florida Institute for Neuroscience; and Sanford L. Boye, Shannon E. Boye and Yuqing Li of the University of Florida.

The study was supported by the National Institutes of Health (grants DA036082, MH080047, MH101954, NS82244, NS073930 and EY024280); and the Department of Defense (CDMRP grant W81XWH-14-1-0074).

What Does It Take to Make a Memory? Study Says New Proteins

JUPITER, FL – November 10, 2016 – While the romantic poets’ idea of memories being akin to spirits may have poetic merit, the scientists’ perspective is that memories are concrete, physical entities that can be visualized within various regions of the brain.

Scientists from the Florida campus of The Scripps Research Institute (TSRI) have now for the first time identified a sub-region in the brain that works to form a particular kind of memory: fear-associated with a specific environmental cue or “contextual fear memory.”

The study, recently published in the journal Biological Psychiatry Cognitive Neuroscience and Neuroimaging, was led by TSRI Associate Professor Sathyanarayanan V. Puthanveettil.

August 30, 2017 33

“Much is still unknown about the identities of proteins synthesized to produce long-term memory,” Puthanveettil said. “The most striking observation from the new study is that the medial prefrontal cortex is the site of this early protein synthesis. We have also identified what proteins are newly synthesized in the medial prefrontal cortex.”

In particular, the study showed new protein synthesis in a specific sub-region of the prefrontal cortex known in rodents as the prelimbic. In humans, this area corresponds to the anterior cortex, which has been linked to processing emotional responses.

Initially, Puthanveettil and his colleagues ignored the medial prefrontal cortex because no one believed that it had anything to do with early encoding of long term memories.

However, when they closely examined the effects on the brain of conditioning rodents with a mild foot shock, the scientists found several messenger RNAs recruited to polyribosomes in the medial prefrontal cortex—a clear indication of new protein synthesis there.

Puthanveettil and his colleagues also discovered that if they inhibited new protein synthesis in the prelimbic region right after fear conditioning took place, those memories did not form. But if the researchers waited just a few hours, inhibiting protein synthesis in prelimbic cortex had no impact and the memories took hold. There is temporal and spatial regulation of new protein synthesis in the medial prefrontal cortex.

“It may be that the first wave of protein synthesis is critical for encoding contextual fear memory, while second wave in other sub-regions is important for memory storage,” he said.

It remains to be determined if other sub-regions of the cortex are also be involved in the synthesis of memory proteins.

“The medial prefrontal cortex has many sub-regions,” said TSRI Senior Research Associate Bindu L. Raveendra, co-first author of the study with Valerio Rizzo, Khalid Touzani and Supriya Swarnkar, all of TSRI at the time of the study. “But the specific roles of these sub- regions in encoding, expression and retrieval, as well as their underlying molecular mechanisms, remain to be unraveled.”

Other authors of the study, “Encoding of Contextual Fear Memory Requires De Novo Proteins in The Prelimbic Cortex,” include Beena M. Kadakkuzha and Xin-An Liu of TSRI; Joan Lora and Robert W. Stackman of Florida Atlantic University; and Chao Zhang and Doron Betel of Weill Cornell Medical College.

The study was supported by the Whitehall Foundation, the National Institutes of Health (grant number 1R21MH096258) and the State of Florida.

Scripps Florida Scientists Discover Clues to Altered Brain Wiring in Autism

JUPITER, FL – November 15, 2016 – Autism is an agonizing puzzle, a complex mixture of genetic and environmental factors. One piece of this puzzle that has emerged in recent years is

August 30, 2017 34 a biochemical cascade called the mTOR pathway that regulates growth in the developing brain. A mutation in one of the genes that controls this pathway, PTEN (also known as phosphatase and tensin homolog), can cause a particular form of autism called macrocephaly/autism syndrome.

Using an animal model of this syndrome, scientists from the Florida campus of The Scripps Research Institute (TSRI) have discovered that mutations in PTEN affect the assembly of connections between two brain areas important for the processing of social cues: the prefrontal cortex, an area of the brain associated with complex cognitive processes such as moderating social behavior, and the amygdala, which plays a role in emotional processing.

“When PTEN is mutated, we find that neurons that project from the prefrontal cortex to the amygdala are overgrown and make more synapses,” said TSRI Associate Professor Damon Page. “In this case, more synapses are not necessary a good thing because this contributes to abnormal activity in the amygdala and deficits in social behavior.”

The study was published on November 15 by the journal Nature Communications. The study also showed that targeting the activity of the mTOR pathway shortly after birth, a time when neurons are forming connections between these brain areas, can block the emergence of abnormal amygdala activity and social behavioral deficits. Likewise, reducing activity neurons that project between these areas in adulthood can also reverse these symptoms.

"Given that the functional connectivity between the prefrontal cortex and amygdala is largely conserved between mice and humans,” said TSRI Graduate Student Wen-Chin Huang, the first author of the study, “we anticipate the therapeutic strategies suggested here may be relevant for individuals on the autism spectrum.”

Although caution is warranted in extrapolating findings from animal models to humans, these findings have implications for individualized approaches to treating autism. “Even within individuals exposed to the same risk factor, different strategies may be appropriate to treat the symptoms of autism in early development versus maturity,” said Page.

In addition to Page and Huang, Youjun Chen of TSRI was an author of the Nature Communications study, “Hyperconnectivity of Prefrontal Cortex to Amygdala Projections in a Mouse Model of Macrocephaly/Autism Syndrome.”

The research was supported by the National Institutes of Health (grants R01MH105610 and R01MH108519), Ms. Nancy Lurie Marks, The American Honda and Children’s Healthcare Charity Inc., Fraternal Order of Eagles, the Simons Foundation (grant 360712) and an anonymous donor.

Scripps Florida Scientists Pinpoint Regulator of Amphetamine-Induced Motor Activity

JUPITER, FL – November 15, 2016 – In new findings that could have an impact on the development of therapies for a number of currently untreatable brain disorders such as Parkinson’s and Huntington’s diseases, scientists from the Florida campus of The Scripps

August 30, 2017 35

Research Institute (TSRI) have found, for the first time, that a specific signaling circuit in the brain is deeply involved in motor activity.

The study, which was led by TSRI’s Associate Professor Srinivasa Subramaniam, was published November 15 in the journal Science Signaling.

Despite many advances, the precise signaling mechanisms that regulate motor function in the striatum, that part of the brain responsible for motor activity, remain unknown. The new study identified for the first time a protein interaction network that helps control these functions by inhibiting the signaling of dopamine, a neurotransmitter involved in regulating movement.

“A pair of proteins operates through a protein-protein interaction network—what we call a 'Rhesactome'—in the striatum,” Subramaniam said. “This may have much broader implications in neurological, psychiatric and addictive disorders. Drugs that bind to either of these proteins may have therapeutic benefits for the diseases that affect this part of the brain.”

The study focused on amphetamine-induced activity affected by what is known as RasGRP1- Rhes signaling circuitry. Drugs like amphetamine, which trigger dopamine release in the striatum, enhance locomotor activity. Rhes acts as a kind of brake on the amphetamine-induced locomotion; in order for normal motor activity to occur, the RasGRP1 and other protein partners in the Rhesactome network induced by amphetamine have to block Rhes. It is the calibrated interaction of Rhes with the protein RasGRP1 that adjusts striatal control of motor functions.

In the study, the researchers succeeded in using RasGRP1 to inhibit Rhes-mediated control of striatal motor activity in animal models. Animal models that were Rhes-deficient had a much stronger active behavioral response to amphetamines. But all that changed if RasGRP1 was depleted.

“It’s a delicate and highly complex relationship,” Subramaniam said. “Imagine that you are running. This protein complex carefully controls that motor function by modulating the effect of Rhes. That’s why you need to have the double control elements of both RasGRP1 and Rhes to fine-tune those motor functions. Our study captures this dynamic complex, so that now for the first time we can biochemically visualize it at the network level.”

What remains unknown at this point is how RasGRP1 actually modulates Rhes.

“We speculate that both transcriptional and post-transcriptional mechanisms are involved,” said TSRI Staff Scientist Neelam Shahani, the first author of the study. “Considering that the Rhes protein is enhanced predominantly at synaptic locations, one intriguing possibility is that RasGRP1 regulates local translation of Rhes messenger RNA at the synapse.”

In addition to Subramaniam and Shahani, other authors of the study, “RasGRP1 Promotes Amphetamine-Induced Motor Behavior through a Rhes Interaction Network (“Rhesactome”) in the Striatum,” are Supriya Swarnkar, Vincenzo Giovinazzo, Jenny Morgenweck, Laura M.

August 30, 2017 36

Bohn, Catherina Scharager-Tapia, Bruce Pascal and Pablo Martinez-Acedo of TSRI; and Kshitij Khare of the University of Florida, Gainesville.

The study was supported by the National Institutes of Health (grant R01-NS087019).

Scripps Florida Scientists Create Innovative Drug Design Strategy to Improve Breast Cancer Treatment

JUPITER, FL – November 21, 2016 – While there have been advances in the treatment of hormone-driven breast cancer, resistance to these therapies remains a significant problem. Side effects, including an increased risk of uterine cancer among postmenopausal women, also severely curtail their use for cancer prevention.

However, a new study by scientists from the Florida campus of The Scripps Research Institute (TSRI) offers a novel structure-based drug design strategy aimed at altering the basic landscape of this type of breast cancer treatment.

The findings show that the current approach is not the only, or even the best way, to block the estrogen receptor.

“We have created a different approach that gives us a mechanism to produce new types of therapeutic molecules,” said TSRI Associate Professor Kendall Nettles. “There are a lot of ways to avoid resistance and other cancer risks, and this gives us a tool box full of alternative approaches that could limit or eliminate those effects.”

“With the standard method, no one understands the structural basis,” he continued. “With our approach we know exactly how we did it. If you can see the shape of the receptor protein and see how the drug works on it, that makes the development process that much faster.”

The findings were published November 21, 2016, by the journal Nature Chemical Biology.

Seeing Is Believing

The current method of creating this class of drugs, which includes tamoxifen, involves attaching a bulky cluster of atoms with a chainlike structure (called, appropriately, a side chain) to molecules that disrupt the estrogen receptor binding site.

The team’s new strategy taps a technique called X-ray crystallography to visualize the drug candidate as it binds to the receptor. This image is used to guide the production of estrogen receptor degraders that also lack the side chain, helping to reduce the risk of resistance and the development of other cancers.

“Our structure-trapping approach to X-ray crystallography provides a molecular snapshot of how subtle changes to a compound series generate a range of graded activity profiles,” said Research Associate Jerome C. Nwachukwu, who was co-first author with Research Associate Sathish Srinivasan. “This structurally distinct mechanism, acting indirectly rather than

August 30, 2017 37 involvement of the typical side chain, provides a new way to design biologically distinct molecules for breast cancer prevention and treatment.”

The new method also makes it possible to identify structural rules for how the molecules interact.

“This is the first example of a structure-based design strategy targeting the estrogen receptor where there is a clear correlation between the chemistry, crystal structure and activity, which is another big advance that will be of broad interest to the cancer community,” Srinivasan said. “We show that indirect antagonism can result in inhibition of proliferation in a predictive fashion.”

In addition to Nettles, Srinivasan and Nwachukwu, other authors of the study, “Full Antagonism of the Estrogen Receptor without a Prototypical Ligand Side Chain,” include Nelson E. Bruno, Venkatasubramanian Dharmarajan, Devrishi Goswami, Scott Novick, Jason Nowak and Patrick R Griffin of TSRI; Irida Kastrati, Nittaya Boonmuen, Yuechao Zhao, Benita S. Katzenellenbogen and John A. Katzenellenbogen, Jian Min and Jonna Frasor of the University of Illinois; Hai-Bing Zhou of Wuhan University (China).

The study was supported by the National Institutes of Health (PHS 5R37DK015556, 5R33CA132022, 5R01DK077085, 1U01GM102148 and 5R01CA130932); the Breast Cancer Research Foundation, BallenIsles Men’s Golf Association, Frenchman’s Creek Women for Cancer Research, Susan G. Komen for the Cure® (Grant PDF12229484); the National Natural Science Foundation of China (81172935, 81373255, 81573279) and Hubei Province’s Outstanding Medical Academic Leader Program.

Scripps Florida Scientists Find Surprising Answers to ‘Food Coma’ Conundrum

JUPITER, FL – November 22, 2016 – Anyone who has drifted into a fuzzy-headed stupor after a large holiday meal is familiar with the condition commonly known as a “food coma.” Now scientists from the Florida campus of The Scripps Research Institute (TSRI), Florida Atlantic University and Bowling Green State University may have finally found a reason for the phenomenon.

Until recently, there has been little more than anecdotal evidence to suggest that “food coma” is an actual physical condition—and the scientific evidence that does exist is unable to explain why some people fall asleep immediately after eating, some later and some not at all.

“Different foods play different roles in mammalian physiology, but there have been very few studies on the immediate effects of eating on sleep,” said TSRI’s Associate Professor William Ja, who led the study, published today in the online journal eLife. Ja and his colleagues used Drosophila, the common fruit fly, as a model, due to the well- documented sleep-metabolism interaction in which flies suppress sleep or increase locomotion when starved. They created a system called the Activity Recording CAFE (ARC), a small plastic chamber that allowed them to record fly activity before and after feeding.

August 30, 2017 38

Researchers found that after a meal, flies increased sleep for a short period before returning to a normal state of wakefulness. Their response varied according to food intake—flies that ate more also slept more. Further investigation of specific food components showed that while protein, salt and the amount eaten promoted sleep, sugar had no effect.

“The protein link to post-meal sleep has been mostly anecdotal, too, so to have it turn up in the study was remarkable,” Ja said. “In humans, high sugar consumption provides a quick boost to blood glucose followed by a crash, so its effect on sleep might only be observed beyond the 20 to 40 minute food coma window.”

The fact that larger-sized meals increased sleep in fruit flies may also have parallels in human behavior—it’s known that electrical activity increases in the brain with meal size and during certain stages of sleep. Salt consumption also influences sleep in mammals.

Unpublished data suggest that the “food coma” condition might be a way to maximize gut absorption of protein and salt, two nutrients that flies might prioritize or find limited in nature, Ja added.

“Using an animal model, we’ve learned there is something to the food coma effect, and we can now start to study the direct relationship between food and sleep in earnest," Ja said. "This behavior seems conserved across species, so it must be valuable to animals for some reason.”

The study also found some intriguing physiological reasons behind after-meal fly napping.

“By turning on and off neurons in the fly brain, we identified several circuits dedicated to controlling postprandial sleep,” said TSRI Graduate Student Keith Murphy, the first author of the study. “Some of these circuits responded to protein and others to circadian rhythm, demonstrating that the behavior has a diversity of inputs.”

In addition to Ja and Murphy, the other authors of the study, “Postprandial Sleep Mechanics in Drosophila,” are Sonali A. Deshpande, James P. Quinn, Jennifer L. Weissbach and Seth M. Tomchik of TSRI; Maria E. Yurgel, Alex C. Keene and Ken Dawson-Scully of Florida Atlantic University; and Robert Huber of Bowling Green State University.

This work was supported by the National Institutes of Health (grant R21DK092735), an Ellison Medical Foundation New Scholar in Aging Award and a Glenn Foundation for Medical Research Award for Research in Biological Mechanisms of Aging.

Scripps Florida Scientists Identify Novel Compound to Alleviate Pain and Itch While Avoiding Common Side Effects

JUPITER, FL– December 1, 2016 – In a new study, scientists from the Florida campus of The Scripps Research Institute (TSRI) have identified a possible drug candidate that suppresses pain and itch in animal models. Their new approach also reduces the potential for drug abuse and avoids the most common side effects—sedation and anxiety—of drugs designed to target the nervous system’s kappa opioid receptors (KORs).

August 30, 2017 39

“The most significant aspect of the study is that we can preserve itch and pain treatment qualities in a KOR agonist that we developed—triazole 1.1—while avoiding the euphoria associated with narcotic opioids and the dysphoria associated with some other selective KOR agonists,” said TSRI Professor Laura Bohn, senior author of the new study.

The research was published this week online ahead of print in the journal Science Signaling. Circumventing Side Effects

KORs help regulate the release of the neurotransmitter dopamine. Drugs that target KORs have shown promise as therapeutic candidates because of their efficacy for treating chronic itch and relieving pain. Unlike opioid narcotics that target other opioid receptors, these compounds do not produce a “high” or increased risk of overdose; however, they can deplete the body’s supply of dopamine and produce dysphoria and sedation, side effects that have limited their clinical development.

Bohn’s laboratory has pioneered the concept that KOR signaling can be fine-tuned to preferentially activate certain pathways over others so that the receptor signals through G proteins rather than through a protein called β-arrestin2.

In the new study, the researchers used rodent models to compare this kind of “biased” KOR agonist, called triazole 1.1, and a conventional KOR agonist.

They found that triazole 1.1 could indeed circumvent the two side effects of previously developed KOR compounds without decreasing dopamine levels, a property associated with dysphoria and sedation.

“This adds to the mounting evidence that shows analgesic effects can be separated from the sedative and dysphoric effects by altering how the agonist engages the receptor,” said TSRI Research Associate Tarsis Brust, first author of the study.

Bohn said the new findings clearly demonstrate that the strategy of developing biased KOR agonists offers a promising new way to treat pain and intractable itch without the potential for abuse.

In addition to Bohn and Brust, other authors of the study, “Biased Agonists of the Kappa Opioid Receptor Suppress Pain and Itch without Causing Sedation and Dysphoria,” include TSRI’s Jenny Morgenweck, Lei Zhou, Edward L. Stahl, Cullen L. Schmid and Michael D. Cameron; Susy A. Kim, Jamie H. Rose, Jason Locke, Sara L. Jones, and Thomas J. Martin of Wake Forrest University; and Sarah M. Scarry and Jeffrey Aubé of the University of North Carolina at Chapel Hill.

This research was supported by the National Institutes of Health (grants P01GM113852, P50DA006634, R01DA014030, U01AA014091 and R01DA031297).

August 30, 2017 40

Scripps Florida Scientists Uncover Potential Driver of Age- and Alzheimer’s-Related Memory Loss

JUPITER, FL – December 7, 2016 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have made an important discovery toward the development of drugs to treat age-related memory loss in diseases like Alzheimer’s. They found that reduced levels of a protein called Rheb result in spontaneous symptoms of memory loss in animal models and are linked to increased levels of another protein known to be elevated in the brains of Alzheimer’s disease patients.

Their study, led by TSRI Associate Professor Srinivasa Subramaniam, was published recently online ahead of print in the journal Neurobiology of Aging.

Link to Known Alzheimer’s Trigger

In the new study, Subramaniam’s group investigated the link between Rheb and an important enzyme called BACE1, which is elevated in older adults and people with Alzheimer’s disease.

“We know that Rheb regulates BACE1, which is a major drug target in Alzheimer’s disease,” Subramaniam said. “Studies of the autopsied brains of Alzheimer’s patients have found a significant reduction in Rheb, so it is possible that an increase in Rheb could reverse the buildup of amyloid plaque or help reduce or even reverse age-related memory loss.”

To uncover the impact of eliminating Rheb, Subramaniam and his colleagues put genetically altered mice through a battery of behavior tests beginning at around six months of age.

While Rheb depletion did not affect the overall body weight or motor activity of the animals, it did have subtle and selective effects on certain memory tasks they performed, such as navigating a maze and memory recall. The researchers compared these symptoms to memory deficits that occur in humans with Alzheimer’s disease and related dementia.

They also found that Rheb depletion increased BACE1 levels, which was consistent with previous research showing that higher BACE1 levels may be a contributing factor for memory deficits.

The fact that some research shows that Rheb messenger RNA is induced during protein starvation in fruit flies, led Subramaniam and his colleagues to theorize that a high-protein diet in humans might be a risk factor for decreasing Rheb levels with age, resulting in mild-to- severe cognitive deficits, as seen in animal models.

“This is an indication that nutrient signaling might regulate cognitive functions in mammals through alteration of Rheb–BACE1 pathway activity,” Subramaniam said.

“Overall, our study demonstrates that forebrain Rheb depletion promotes aging-associated cognitive defects,” said TSRI Staff Scientist Neelam Shahani, the first author of the study.

August 30, 2017 41

“Targeting the Rheb pathway may offer some therapeutic potential for aging- or Alzheimer’s disease-associated memory deficits.”

In addition to Subramaniam and Shahani, other authors of the study, “Forebrain Depletion of Rheb GTPase Elicits Spatial Memory Deficits in Mice,” are TSRI’s Wen-Chin Huang, Megan Varnum and Damon T. Page. This work was supported by the National Institutes of Health (grants NIH/NINDS R01- NS087019, NIH/NINDS R01-NS094577, and NIH/NIMH R01-MH105610), Ms. Nancy Lurie Marks and the Alzheimer’s Disease Association (grant 2015-NIRG-344356).

Ancient Enzyme Morphed Shape to Carry Out New Functions in Humans

LA JOLLA, CA & JUPITER, FL – December 9, 2016 – New research led by scientists at The Scripps Research Institute (TSRI) reveals that a human enzyme has changed little from its days as a bacterial enzyme. In fact, the enzyme appears to be unique in its ability to change its shape—and its job in cells—without overhauling its basic architecture.

“This work illustrates nature’s efficiency—how it can take one thing and convert it to another, with a tweak here and a tweak there,” said Paul Schimmel, professor at TSRI and senior author of the new study.

The findings were published recently in the journal Proceedings of the National Academy of Sciences.

A Unique Enzyme

Schimmel and his colleagues focused on a member of a family of enzymes called aminoacyl tRNA synthetases. These enzymes originated in ancient bacteria, where they decode genetic information to help produce amino acids. Over time, the enzymes have evolved to carry out even more functions in complex lifeforms, such as humans.

“Aminoacyl tRNA synthetases are associated with—and we believe needed for—the building of organismal complexity, such as making tissues and organs in humans,” Schimmel explained.

These new functions are controlled by “decorations,” or additions to the aminoacyl tRNA synthetase architecture, which are generally lacking in bacteria; however, there is one aminoacyl tRNA synthetase, called AlaRS, that lacks any new decorations. AlaRS somehow carries out new roles in humans using a preexisting bacterial domain in its structure.

“AlaRS presented an exception to what we thought was a ‘rule,’ ” said Schimmel.

AlaRS’s Secret

The scientists in the new study took a closer look at AlaRS using two imaging techniques: X- ray crystallography and small-angle X-ray scattering. The images, combined with functional

August 30, 2017 42 analysis, showed that a domain of AlaRS’s structure, called C-Ala, had been reshaped to take on a new role in humans. The end result is the same as if AlaRS had gained a new decoration.

Schimmel compared it to reshaping an airplane’s wing to serve as the airplane’s tail instead. “Nature has provided ways for reshaping objects, like C-Ala, and when that happens, new functions occur,” he said.

Schimmel said the next step is to figure out the function of human C-Ala. This work may shed light on diseases linked to mutations in aminoacyl tRNA synthetases, such as the neurodegenerative disease Charcot-Marie-Tooth.

In addition to Schimmel, authors of the study, “Two Crystal Structures Reveal Design for Repurposing the C-Ala Domain of Human AlaRS,” are Litao Sun (first author), Youngzee Song, David Blocquel and Xiang-Lei Yang of TSRI.

This work was supported by the National Cancer Institute (grant CA92577), the National Foundation for Cancer Research and the National Institutes of Health (grant R01 NS085092).

TSRI Scientists Devise New Approaches to Personalized Medicines

Researchers Open New Avenues for the Development of Precision Therapeutics Targeting RNA

JUPITER, FL – December 12, 2016 – Scientists on the Florida campus of The Scripps Research Institute (TSRI) have developed broad methods to design precision medicines against currently incurable diseases caused by RNA.

RNA carries out thousands of essential functions in cells, but many RNAs can act in uncontrolled ways and cause disease. For decades, scientists have tried to develop drug candidates that target human RNAs, but they have been hampered by an inability to achieve sufficient selectivity (to reduce the potential of side effects) and potency (ensuring effectiveness).

In a study published today online ahead of press in the journal Nature Chemical Biology, researchers—led by TSRI Professor Matthew Disney and Research Associate Suzanne Rzuczek, with important contributions from Professor Ryohei Yasuda and Research Associate Lesley Colgan of the Max Planck Florida Institute for Neuroscience—have disclosed several approaches to overcome these hurdles.

“This study reads like science fiction,” Disney said. “We present for the first time multiple solutions to this long-standing problem. With the precision of a surgeon’s scalpel, we have shown that small molecules can be designed to seek out and destroy only disease-causing RNAs. Further, we developed novel chemical approaches to use a disease-causing RNA to help make its own drug by using that RNA as a catalyst for drug synthesis at the needed site. It is like having your physician place a drug at the right place without exposing healthy cells.”

August 30, 2017 43

Although these studies have broad implications for RNA diseases in general, they were demonstrated on myotonic dystrophy type 1, an incurable inherited disorder that involves progressive muscle wasting and weakness. It is caused by an RNA defect known as a “triplet repeat,” a series of three nucleotides repeated more times than normal in an individual’s genetic code, in this case, a cytosine-uracil-guanine (CUG) triplet.

In many genetic diseases, there are two copies of the problem gene—a mutant copy that causes a disease and a normal copy that a cell needs to survive. Selective recognition of the diseased gene product has not been possible before.

This new study demonstrates that designer small molecules can selectively recognize larger, disease-associated repeats (alleles) over shorter, normal ones.

“We developed several approaches to create allele-selective small molecules that seek out only the disease-causing gene product, including covalent binding, cleavage and imaging,” said TSRI Research Associate Suzanne G. Rzuczek, first author of the study. “All approaches show precise recognition of toxic r(CUG) repeats and, more importantly, they showed that the mutant repeat is the sole target.”

The work also offers an innovative way to track the movement of RNA in a diseased cell via imaging. “We have brought RNAs out of the darkness and into the light by developing a chemical flare that goes off when a drug targets the RNA in a diseased cell and then continues to track the RNA’s movement,” Disney added.

"We probed disease-causing RNA using a technique called fluorescence lifetime imaging—a sensitive technique to measure fluorophore binding,” said Max Planck’s Ryohei Yasuda. “We were very excited when we observed a huge difference in signal from their probes between disease cells and normal cells under our microscope technique."

Max Planck’s Lesley Colgan added, "The combination of cutting-edge chemistry and microscopy techniques developed in Florida is a powerful approach to identify new methods to probe and manipulate (and kill) disease-causing RNA in cells."

Both Disney and Yasuda were 2015 recipients of the National Institutes of Health (NIH) Director’s Pioneer Award, which supports individual scientists of exceptional creativity who propose highly innovative approaches with high-impact potential.

In addition to Disney, Rzuczek, Colgan and Yasuda, other authors of the study, “Precise Small Molecule Recognition of a Toxic RNA Repeat Expansion,” include Yoshio Nakai and Michael D. Cameron of TSRI and Denis Furling of Sorbonne Universités (Paris).

The study was supported by the NIH (grants DP1NS096898 and DP1NS096787), the Muscular Dystrophy Association (grant 380467) and the Myotonic Dystrophy Foundation.

August 30, 2017 44

Scripps Florida Scientists Discover New Natural Source of Potent Anti-Cancer Drugs

JUPITER, FL – December 20, 2016 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have developed an efficient process to rapidly discover new “enediyne natural products” from soil microbes that could be further developed into extremely potent anticancer drugs.

The study highlights microbial natural products as abundant sources of new drug leads. The researchers’ discovery process involves prioritizing the microbes from the TSRI strain collection and focusing on the ones that are genetically predisposed to produce specific families of natural products. The scientists say this process saves time and resources in comparison to the traditional approaches used to identify these rare molecules.

The study, led by TSRI Professor Ben Shen, was published today in the journal mBio.

Shen and his colleagues uncovered a new family of enediyne natural products, called tiancimycins, (TNMs) which kill selected cancer cells more rapidly and completely in comparison to toxic molecules used in FDA-approved antibody-drug conjugates (ADCs)— monoclonal antibodies attached to cytotoxic drugs that target only cancer cells.

The scientists also discovered several new producers of C-1027, an antitumor antibiotic currently in clinical development, which can produce C-1027 at much higher levels.

It has been more than a decade since Shen first reported on the C-1027 enediyne biosynthetic machinery, and he speculated then that the knowledge obtained from studying biosynthesis of C-1027, and other enediynes, could be used for the discovery of novel enediyne natural products.

“The enediynes represent one of the most fascinating families of natural products for their extraordinary biological activities,” Shen said. “By surveying 3,400 strains from the TSRI collection, we were able to identify 81 strains that harbor genes encoding enediynes. With what we know, we can predict novel structural insights that can be exploited to radically accelerate enediyne-based drug discovery and development.”

“The work described by the Shen group is an excellent example of what can be achieved by coupling state of the art genomic analyses of potential biosynthetic clusters and modern physicochemical techniques,” said David J. Newman, retired chief of the National Cancer Institute’s Natural Products Branch. “As a result of their work, the potential number of enediynes has significantly increased.”

Shen’s method of strain prioritization and genome mining means a far more efficient use of resources involved in the discovery process, targeting only those strains that look to produce the most important natural compounds.

“This study shows that the potential to rapidly discover new enediyne natural products from a large strain collection is within our reach,” said TSRI Research Associate Xiaohui Yan, one of

August 30, 2017 45 four first authors of the study. “We also show the feasibility of manipulating tiancimycin biosynthesis in vivo, which means that sufficient quantities of these precious natural products can be reliably produced by microbial fermentation for drug development and eventual commercialization.”

In addition to Shen and Yan, first authors of the study, “Strain Prioritization and Genome Mining for Enediyne Natural Products,” include TSRI’s Huiming Ge, Tingting Huang and Hindra. Other authors include Dong Yang, Qihui Teng, Ivana Crnovčić, Xiuling Li, Jeffrey D. Rudolf, Jeremy R. Lohman and Christoph Rader of TSRI; Yannick Gansemans and Filip Van Nieuwerburgh of Ghent University, Belgium; Yanwen Duan, Xiangcheng Zhu and Yong Huang of Xiangya International Academy of Translational Medicine, Central South University, China; Li-Xing Zhao and Yi Jiang of Yunnan University, China.

The study was supported in part by the Chinese Ministry of Education (111 Project B08034), National High Technology Joint Research Program of China (grant 2011ZX09401-001), National High Technology Research and Development Program of China (grant 2012AA02A705), the National Institutes of Health (grants CA78747 and GM115575), the German Research Foundation and the Arnold and Mabel Beckman Foundation.

Scripps Florida Scientists Uncover Cellular Process Behind Premature Aging

JUPITER, FL – December 21, 2016 – In a new study, scientists from the Florida campus of The Scripps Research Institute (TSRI) have shown how two genes “balance” each other to maintain normal cell function. A disruption in one of the genes, called spns1, can induce degradation and premature “senescence”—or aging—while the other gene, called atp6v0ca, can jump in to suppress that degradation.

Their experiments in zebrafish suggest that these combined genetic disruptions can counteract premature aging and extend developmental lifespan.

“We found that the dual defects did indeed counteract senescence during development and extended the animal’s survival and life span,” said TSRI Associate Professor Shuji Kishi.

The findings, published recently in the journal Autophagy, could also guide future treatments for diseases that involve the body’s inability to degrade unwanted or harmful compounds.

A Closer Look at Lifespan

Cellular senescence is when cells stop dividing and is a normal part of aging. Interestingly, senescence is not only observed in later aging stages but is also detectable during embryonic development in vertebrates.

In the new study, the researchers took a closer look at the gene spns1. In vertebrates, such as zebrafish and humans, the protein encoded by spns1 is important in a cellular process called autophagy, when the cell moves unwanted material to a cellular structure called the lysosome.

August 30, 2017 46

Previous research had shown that defects in this gene can also cause senescence in the embryonic stage and premature aging symptoms in adulthood. However, Kishi and his colleagues found that a concurrent disruption of another gene— atp6v0ca, whose sole defect still causes senescence—led to suppression of the process induced by the defective spns1 gene.

“Our findings suggest that these two defects actually function at a balance point that is critically involved in the regulation of developmental senescence—and that balance allows for normal cell function,” said Kishi.

Restoring the Balance

The scientists are now considering ways to influence the balance between these genes as a strategy to treat lysosomal storage diseases such as Pompe disease, where the excessive buildup of a substance called glycogen results in severe muscle weakness. They believe there may also be applications in treating age-associated degenerative diseases linked to late-stage autophagy disruption.

“The use of appropriate inhibitors, selective for key steps in the biosynthesis of cellular macromolecules in general, may restore normal dynamics in the autolysosomal compartment and correct the pathological storage that is the ultimate cause of these types of disease,” said TSRI Research Associate Shanshan Lian, the co-first author of the study.

The findings may also lead to the development of tools to help identify new genes that affect the aging process without the need for performing lengthy adult lifespan analyses. This approach could be applied to the high-throughput identification of pharmacological agents that control aging and lifespan through enhanced resistance to various stressors, including oxygen radicals.

In addition to Kishi and Lian, other authors of the study, “Autolysosome Biogenesis and Developmental Senescence Are Regulated by Both Spns1 and V-Atpase,” include TSRI’s Tomoyuki Sasaki, a co-first author and Alam Khan; Jesse R. Llop, Andrew V. Samuelson of the University of Rochester; Wenbiao Chen of the Vanderbilt University; and Daniel J. Klionsky of the University of Michigan.

This study was supported by the National Institutes of Health (grants GM101508 and GM053396).

Scripps Florida Scientists Develop Drug Discovery Approach to Predict Health Impact of Endocrine-Disrupting Chemicals

JUPITER, FL – December 29, 2016 – Breast cancer researchers from the Florida campus of The Scripps Research Institute (TSRI) have developed a novel approach for identifying how chemicals in the environment—called environmental estrogens—can produce infertility, abnormal reproductive development, including “precocious puberty,” and promote breast cancer.

August 30, 2017 47

Environmental estrogens work by binding to the estrogen receptor, a protein in cells that guides sexual maturation and reproduction. The new research shows how high-resolution imaging techniques could give scientists a window into how exposure to these chemicals may impact public health.

This research method could also be used to speed up the discovery of new drugs for breast cancer and many other diseases, added study senior author Kendall Nettles, associate professor at TSRI.

The study was published online ahead of print in the journal Cell Chemical Biology.

Taking Snapshots of a Crucial Receptor

Environmental estrogens control the estrogen receptor’s activity by changing its shape, which can mimic the effects of estrogen. These chemicals can also block receptor activity, as seen with the breast cancer therapy tamoxifen.

These changes are often exceedingly small, in the sub-ångström range (one ångström is equal to one ten-billionth of a meter). In fact, changes to even a single atom in a chemical or endocrine therapy drug can often drive widely divergent outcomes, but the resolution of the typical crystal structure does not allow visualization of these small changes.

Nettles calls the approach in the new study “super-resolution x-ray crystallography.”

X-ray crystallography is a technique that produces a snapshot of the receptor’s 3-D atomic structure. Like the images produced by photography or microscopy, x-ray crystal structures have a certain resolution, or level of detail, that can be visualized. With optical microscopy, super-resolution imaging—a discovery for which researchers were awarded the Nobel Prize in 2014—can be achieved by combining many images to produce a sharper picture.

Nettles and his colleagues reasoned that they could use a similar approach with x-ray crystallography to compare molecular snapshots, or structures, and better understand how estrogenic chemicals control receptor activity.

Developing a Framework for Drug Design

They found that by combining data about structural disturbances from many structures with activity profiles of their ligands (the estrogenic chemicals), researchers can identify the sub- ångström details that determine endocrine disruption.

“Our novel approach offers a framework for understanding the diverse effects of environmental estrogens and other endocrine disruptors, furthering efforts to develop improved breast cancer therapies,” said TSRI Research Associate Jerome C. Nwachukwu, the first author of the study. “This approach can also be applied to other allosteric receptors.”

August 30, 2017 48

Super-resolution x-ray crystallography can also be applied to the large groups of receptors that comprise the targets for most known drugs. With structure-based drug design, chemists can use this new approach to visualize how to modify drug-candidate chemicals to promote the desirable therapeutic profile.

In addition to Nettles and Nwachukwu, other authors of the study, titled “Systems Structure Biology Analysis of Ligand Effects on ERa Predicts Cellular Response to Environmental Estrogens and Anti-Hormone Therapies,” are Sathish Srinivasan, Nelson E. Bruno, Jason Nowak, Nicholas J. Wright, Erumbi Rangarajan, Tina Izard and Douglas J. Kojetin of TSRI; Xin-Qui Yao and Barry J. Grant of the University of Michigan; Olivier Element of Weil Cornell Medical College; John A. Katzenellenbogen of the University of Illinois; and Filippo Minutolo of the Università di Pisa, Italy.

The study was supported by the National Institutes of Health (grant numbers PHS 5R37DK015556, 5R01DK077085 and 2R01GM070862-10A1), The BallenIsles Men’s Golf Association, The Frenchman’s Creek Women for Cancer Research, The National Science Foundation (NSF award 1359369) and the University of Michigan.

Scripps Florida Scientists Uncover New Way to Defeat Therapy-Resistant Prostate Cancer

JUPITER, FL – December 29, 2016 – A new study led by scientists from the Florida campus of The Scripps Research Institute (TSRI) sheds light on a signaling circuit in cells that drives therapy resistance in prostate cancer. The researchers found that targeting the components of this circuit suppresses advanced prostate cancer development.

The study, led by TSRI Associate Professor Jun-Li Luo, was published online ahead of print in the journal Molecular Cell.

A New Strategy to Fight Prostate Cancer

Prostate cancer—which, according to the American Cancer Society, affects one in six American men—is the second-leading cause of death after lung cancer in American men.

Currently, the most effective treatment for advanced prostate cancer is to deprive the cancer of what feeds it—androgen hormones, such as testosterone. Unfortunately, almost all patients eventually develop resistance to this therapy, leaving doctors with no options to counteract the inevitable.

The new study shows that a “constitutively active” signaling circuit can trigger cells to grow into tumors and drive therapy resistance in advanced prostate cancer. A cell signal pathway with constitutive activity requires no binding partner (ligand) to activate; instead, the signaling circuit continually activates itself.

This signaling circuit, which is composed of the protein complex IκBα/NF-κB (p65) and several other molecules, controls the expression of stem cell transcription factors (proteins that

August 30, 2017 49 guide the conversion of genetic information from DNA to RNA) that fuel the aggressive growth of these resistant cancer cells.

“The fact that the constitutive activation of NF-kB in the circuit is independent of traditional activation opens the door for potential treatment options,” said Luo.

Targeting Other Signaling Components Shows Promise

NF-kB plays important roles in cancer development, and it is regarded as one of the most important targets for cancer therapy. However, the use of NF-kB inhibitors in treating cancer is complicated by severe side effects related to immunosuppression caused by indiscriminate inhibition of NF-kB in normal immune cells.

Luo noted that targeting the other non-IκBα/NF-κB components in this signaling circuit would avoid the suppression of NF-κB in normal immune cells while keeping the potent anti-cancer efficacy.

In addition to IκBα/NF-κB, the signaling circuit includes the microRNA miR-196b-3p, Meis2 and PPP3CC. While miR-196b-3p promotes tumor development, Meis2, which is an essential developmental gene in mammals, can disrupt the circuit when overexpressed. The protein PPP3CC can inhibit NF-κB activity in prostate cancer cells.

“Disrupting this circuit by targeting any of its individual components blocks the expression of these transcription factors and significantly impairs therapy-resistant prostate cancer,” said TSRI Research Associate Ji-Hak Jeong, the first author of the study.

In addition to Luo and Jeong, other authors of the study, “A Constitutive Intrinsic Inflammatory Signaling Circuit Composed of miR-196b, Meis2, PPP3CC, and P65, Drives Prostate Cancer Castration Resistance,” are TSRI’s Sun-Jin Park and Shohreh Iravani Dickinson of the Moffitt Cancer Center, Tampa, FL.

The study was supported by the National Institutes of Health (grants 1R01CA140956, 1R01CA197944 and 1R21NS073098), the United States Department of Defense (grants W81XWH-09-1-0533, W81XWH-14-1-0051 and W81XWH-15-1-0235), The Florida Department of Health, Bankhead-Coley Cancer Research Program (grant 09BB-13), the ThinkPinkKids Foundation and the Frenchman’s Creek Women for Cancer Research.

Scripps Florida Scientists Expand Toolbox to Study Cellular Function

JUPITER, FL – January 4, 2017 – Scientists on the Florida campus of The Scripps Research Institute (TSRI) have developed a new tool for studying the molecular details of protein structure.

Their new study, published recently in the journal Proceedings of the National Academy of Sciences, explores how evolution can be used to discover new and useful enzyme tools, called proteases. Proteases cleave proteins into smaller peptide pieces that scientists can then analyze

August 30, 2017 50 to determine the identity of the protein and whether a cell has made chemical changes to the protein that might alter its function.

The new protease developed in the study helps shed light on these chemical changes, called post-translational modifications. Post-translational modifications are alterations made to proteins after the proteins are translated from RNA.

“We have to observe these protein modifications directly through chemical analysis; we can’t read them out of DNA sequence,” explained study senior author Brian M. Paegel, associate professor at TSRI.

These modifications can dramatically alter a protein’s stability and function, and unregulated modification can lead to disease, such as cancer. Therefore, understanding the nature and location of these modifications can be critical in the early phases of drug discovery.

Scientists currently rely on a technique called mass spectrometry to study post-translational modifications. Mass spectrometry analyzes peptides to see if their mass changes—a bit like zooming in on that protein to see hidden details. An unexpected change in mass can indicate the occurrence of a post-translational modification.

Many scientists today use a protease called trypsin to break proteins into peptides. Because there are few other proteases available for mass spectrometry, trypsin has become the workhorse of the field. However, Paegel explained, it’s luck of the draw if trypsin generates a peptide with a modified site. So Paegel and co-workers thought it would be useful to have a new tool that cleaved directly at the modified site.

To solve this problem, Paegel developed a new trypsin “mutant” using a technique called “directed evolution.” The scientists created many thousands of trypsin mutants and tested each mutant for its ability to cut a protein at modified sites. They discovered a mutant that could cut proteins at citrulline, which is one type of modification.

Paegel believes this new approach could be useful for mapping a wider range of post- translational modifications, and he hopes to use directed evolution to discover proteases that target many other post-translational modifications. “I think we’re on the brink of an explosion of new tools for mass spectrometry,” he said.

In addition to Paegel, authors of the study, “Evolution of a mass spectrometry-grade protease with PTM-directed specificity,” were Duc. T. Tran (first author), Valerie Cavett, Vuong Q. Dang and Héctor L. Torres of TSRI.

This study was supported by a National Institutes of Health’s Director’s New Innovator Award (grant OD008535) and a National Science Foundation Research Experiences for Undergraduates Grant (1359369).

August 30, 2017 51

TSRI Researchers Discover Surprising Process Behind Sense of Touch

JUPITER, FL – January 5, 2017 – Biologists on the Florida campus of The Scripps Research Institute (TSRI) have discovered a new mechanism that likely underlies how we feel force or touch. Their study suggests that “rafts” of fatty lipids on the cell surface act as compartments to keep certain enzymes from mixing with their binding partners. Disrupt these rafts through touch—also called mechanosensation—and the enzymes will mix with their partners and react, triggering a signal that communicates the touch to responsive proteins in the cell.

“This is really different from how anybody thinks about mechanosensation,” said TSRI biologist Scott Hansen, who supervised the study, published recently in the journal Nature Communications.

Hansen compared lipid rafts to compartments in a glow stick. Crack the barrier between the two sides of a glow stick and the components mix and start a chemical reaction to produce light. On the cell membrane, the mixing of enzymes and their partners produces a signaling lipid phosphatidic acid that starts the chain of events to create the sense of touch.

Release the Enzymes!

Hansen and his colleagues conducted this research with support from a National Institutes of Health Director’s New Innovator Award, which encourages scientists to pursue high-risk, high- reward translational research. Their mission was to shed light on the complicated cellular mechanisms behind touch with the hope that this work could lead to new ways to address chronic pain and other conditions where the sense of touch goes haywire.

The team focused on lipid rafts, small clumps of lipids and proteins on the cell membrane. Hansen said previous work had explained lipid rafts as “pedestals” of activity, like islands where the cell can carry out important activities. But there was also a question of whether rafts can act like holding pens, keeping enzymes from meeting their binding partners, or substrates, until the right moment.

The researchers answered this question by first observing lipid rafts in real time, using a technique called super-resolution imaging. They found that lipid rafts on live cells did indeed assemble and disassemble frequently, creating opportunities for enzymes to mix with their substrates. Next, the researchers determined that mechanical force, aka touch, can trigger rafts to mix, allowing enzymes to meet their substrates and release a signal.

While more research is needed, the researchers believe this signal may be picked up by other proteins in the membrane that carry on this message to sense touch. Hansen said it’s also likely that other enzymes in lipid rafts are mixing with their substrates in a similar way to regulate cellular responses to many stimuli outside touch. “This is just the first step in understanding this process,” said Hansen.

The researchers credited the NIH Director’s award program and the resources at TSRI for making this surprising finding possible. “This research probably wouldn’t have been funded

August 30, 2017 52 through normal mechanisms—it’s outside the box,” said Hansen. “But TSRI is the type of place where these things are done.”

In addition to Hansen, authors of the study, “Kinetic disruption of lipid rafts is a mechanosensor for phospholipase D,” were first author E. Nicholas Petersen, Hae-Won Chung and Arman Nayebosadri of TSRI.

This work was supported by a Director’s New Innovator Award (grant 1DP2NS087943-01) from the National Institutes of Health and a graduate fellowship from the Joseph B. Scheller & Rita P. Scheller Charitable Foundation.

Scripps Florida Scientists Find Clue to Why Zika, But Not Its Close Relatives, Causes Birth Defects

JUPITER, FL – February 6, 2017 – The most frightening aspect of Zika virus has been its ability to produce severe fetal birth defects during pregnancy, especially microcephaly—a small head.

Now, scientists from the Florida campus of The Scripps Research Institute (TSRI) have uncovered the details behind the virus’s unique ability to cross the placental barrier and expose the fetus to a range of birth defects that often go beyond microcephaly to include eye and joint injury, and even other types of brain damage.

The new study, led by TSRI Associate Professor Hyeryun Choe, was published online ahead of print this week in the journal Proceedings of the National Academy of Sciences.

How Zika virus crosses the placental barrier, while other closely related viruses in the flavivirus family including dengue and West Nile viruses do not, has puzzled researchers since the crisis began some two years ago in Brazil.

Obstacles to reaching the fetal brain are substantial—a virus must move from the mother’s blood into fetal circulation, which is separated by placental barrier cells designed to prevent that very occurrence.

The researchers found that human umbilical endothelial cells, derived from four donors in the study, proved far more susceptible to Zika infection than to other viruses, with viral counts as much as a hundred or thousand times higher than West Nile or dengue virus. The new research also suggests that Zika virus learned to exploit something of a secret passage, a cell surface molecule known as AXL, while West Nile and dengue viruses did not.

“Zika uses AXL to efficiently slip past one of the major barrier cell types in the placenta: fetal endothelial cells, which are the gateway to access fetal circulation,” said Choe.

What may help make the Zika virus particularly infectious in cells that other flaviviruses can't infect, said TSRI Research Associate Audrey Richard, a first author of the study, is that it profits from the built-in function of AXL. “The physiological function of AXL is to quench

August 30, 2017 53 activated immune reactions, including the antiviral interferon response,” said Richard. “By using AXL, Zika virus catches two birds with one stone; it enters cells and also gains favorable environment for its replication inside the cells.”

Zika is able to take advantage of AXL by binding to an intermediate molecule known as Gas6, which is present in blood and other bodily fluids. Gas6 acts as an active bridge between the virus and AXL by binding AXL on one end and the virus membrane on the other, helping the virus utilize AXL and gain entry to host cells.

These differences may help explain why, among related viruses, only Zika can efficiently access and infect the fetal bloodstream.

“We don’t yet understand why Zika virus uses AXL and the others don’t,” Choe added. “The common belief is that all flaviviruses have similar structures, but our findings suggest that Zika virus may have a different average population structure than others. This has significant scientific and clinical implications.”

“Structural studies show that most of the infectious virion membrane is completely covered with viral proteins, which makes it difficult for Gas6 to bind to the Zika virus membrane underneath the protein shell,” said TSRI Research Associate Byoung-Shik Shim, the study’s other first author. “However, flavivirus particles assume many asymmetric shapes and are in continuous dynamic motion, which likely exposes patches of the virion membrane. Our study suggests that Zika virus exposes enough membrane for Gas6 binding, whereas West Nile and dengue viruses do not.”

The researchers also speculated on Zika virus’ pathology. AXL is also present in the blood- brain barrier, the eye-blood barrier and the testes—where it maintains integrity of the blood vessels and the functions of the testes. It may be used by Zika virus to infect those cells and may explain Zika virus’ ability to infect the fetal brain and eye and to transmit sexually.

In addition to Choe, Richard and Shim, other authors of the study, “AXL-Dependent Infection of Human Fetal Endothelial Cells Distinguishes Zika Virus from Other Pathogenic Flaviviruses,” include Young-Chan Kwon, Yuka Otsuka, Kimberly Schmitt and Fatma Berri of TSRI; Rong Zhang and Michael S. Diamond of Washington University School of Medicine, St. Louis, MO.

The study was supported by start-up funds from the state of Florida, the National Institutes of Health (grants R01 AI110692, R01 AI073755 and R01AI101400).

Scripps Florida Scientists Take Aim at Obesity-Linked Protein

JUPITER, FL – February 15, 2017 – Scientists are working to understand the mechanisms that make weight loss so complicated. Exercise burns calories, of course, but scientists are also looking at how the body burns more energy to stay warm in cold temperatures.

Is there a way to get metabolism to ramp up—even when it’s not cold out?

August 30, 2017 54

TSRI Assistant Professor Anutosh Chakraborty is on a mission to answer this question. His past research revealed a new therapeutic target in this battle—a protein that actually promotes fat accumulation in animal models by slowing stored energy (fat) breakdown and encouraging weight gain.

Now, in a study recently published online in the journal Molecular Metabolism, Chakraborty and his colleagues have shown that deleting the gene for this protein, known as IP6K1, protects animal models from both obesity and diabetes. This protective effect is seen regardless of diet, even at what’s known as a thermoneutral temperature (around 86⁰F). This means inhibiting IP6K1 should help animals burn more energy, regardless of outside conditions.

“In genetically altered animal models that lack IP6K1, we found that deletion dramatically protects these knock-out mice from diet-induced obesity and insulin resistance regardless of the temperature in the environment,” Chakraborty said. “When we inhibited the enzyme with chemical compounds, the results were similar.”

Why Temperature Matters

Temperature is important in the study of obesity because an animal in lower temperatures will rapidly lose weight as it burns more energy to try to maintain core body temperature.

Because humans can maintain their body temperatures in a number of ways—clothing, for example—any pathway that reduces body weight at higher temperatures is a highly encouraging target in human obesity.

The new study suggests a future pharmaceutical may be able to target IP6K1 to mimic the energy burning seen at relatively lower temperatures.

“If we delete IP6K1, the animals gain less body weight because they simply expend more energy—regardless of temperature. That’s important because blocking weight gain by enhancing energy expenditure in a thermoneutral environment is harder and thus, targeting IP6K1 is expected to be successful in ameliorating obesity in humans,” said Chakraborty.

“If you’re developing an anti-obesity drug based on inhibiting IP6K1, our new findings shows that there are potentially very few restrictions for its use—a subject would lose weight even on a high-fat diet, and nobody would have to sit in a refrigerator to make it work,” he added.

The first author of the study, “Global IP6K1 deletion enhances temperature modulated energy expenditure which reduces carbohydrate and fat induced weight gain,” is TSRI’s Qingzhang Zhu. Other authors are Sarbani Ghoshal, at TSRI at the time of the study, now at the Saint Louis University School of Medicine, and Richa Tyagi of the Johns Hopkins University School of Medicine.

This work is supported by the National Institutes of Health (grant R01DK103746) and the state of Florida.

August 30, 2017 55

Fighting Blindness: TSRI Scientists Bring a Key Protein into Focus

JUPITER, FL – March 6, 2017 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have discovered how a protein called α2δ4 establishes proper vision. Their research helps explain why mutations in the gene encoding α2δ4 lead to retinal dystrophy, a disease characterized by defective color vision and night blindness.

To study how this protein supports vision, the researchers modeled retinal dystrophy in mice. Like humans, mice lacking α2δ4 succumbed to the disease and their vision was compromised.

“Much of our work is driven by desire to understand what goes awry in a range of blinding conditions,” explained TSRI Professor Kirill A. Martemyanov, senior author of the new study. “Now we have found a molecule that plays a key role in allowing photoreceptors to plug into the neural circuit and transmit the light signals they receive to the brain.”

The study was published online recently in the journal Neuron.

A Secret Ingredient for Vision

Our vision depends on two types of photoreceptors in the light-sensitive layer of eye called the retina. Rods photoreceptors detect photons at the lowest levels of light and support night vision, and cone photoreceptors sense bright light and discriminate between colors. Both rods and cones must wire into a neural circuit of the retina to send information to the brain.

Martemyanov and his colleagues are studying the neural connections that make vision possible. In a previous study, the researchers identified a novel cell-adhesion protein called ELFN1 that rods use for making contacts with their partners, called bipolar neurons. However, how ELFN1 accomplishes the task of photoreceptor wiring was not clear.

In the new study, experiments spearheaded by TSRI Research Associate Yuchen Wang of the Martemyanov laboratory showed that this connectivity requires α2δ4 to join a structure, called a higher order macromolecular complex, with ELFN1 and other proteins called calcium channels. These calcium channels trigger the release of the chemical messenger glutamate, which photoreceptors use for communicating with bipolar neurons.

In short, Wang explained, without both α2δ4 and the other calcium channels in the macromolecular complex, rods cannot connect to the neural circuit. “We found that α2δ4 is essential for organizing the presynaptic compartment of rod photoreceptors,” he said.

Strikingly, eliminating the corresponding gene for α2δ4 in a mouse model interrupted the transmission of light signals from photoreceptors to the brain without affecting the ability to detect light. “It’s like you are trying to make a phone call—and your phone is fully functional—but you are not heard because there is no signal,” Martemyanov said.

Cones seemed to handle the lack of α2δ4 only slightly better. Without the α2δ4, mice failed to see under dim light conditions and could not navigate a maze in low light due to their

August 30, 2017 56 dysfunctional rods. Their cones were affected too, but they could still send some weak signals through to the brain.

“Their dim-light vision was completely abolished,” said Martemyanov. “And the signal from the cones could barely make it.”

Wang said the researchers are doing more research now to account for this difference between rods and cones.

A Potential Way to Keep Eyes Healthy

Going forward, Martemyanov and his team plan to study whether manipulating α2δ4 could help photoreceptors transmit their signals and maintain connectivity to stay functional longer in models of age-related vision loss, a major blinding condition in humans.

“If we can entice dying photoreceptors to augment their communication with the retina circuitry and preserve the connections they make, we can likely delay the loss of vision in degenerative conditions like age-related macular degeneration,” Martemyanov said.

The researchers also think that wiring factors such as α2δ4 and ELFN1 could also help researchers address a current challenge in using stem cells to correct vision loss.

Martemyanov explained that current efforts of many laboratories are currently directed towards replacing dead photoreceptor cells with stem cell-derived rods and cones as a strategy to restore vision; however, integrating the new photoreceptors into the retina circuit has been a challenge. The new study suggests that α2δ4 may be the secret ingredient for getting these new cells to properly wire into the neural circuit.

In addition to Martemyanov and Wang, authors of the study, “The Auxiliary Calcium Channel Subunit α2δ4 Is Required for Axonal Elaboration, Synaptic Transmission, and Wiring of Rod Photoreceptors,” were Yan Cao, Ignacio Sarria, Ben Throesch and Kristin Baldwin also of TSRI; Katherine E. Fehlhaber, Norianne T. Ingram and Alapakkam P. Sampath of the Jules Stein Eye Institute, University of California, Los Angeles; Debbie Guerrero-Given and Naomi Kamasawa of the Max Planck Florida Institute for Neuroscience; and Toshihisa Ohtsuka of the University of Yamanashi.

The study was supported by the National Institutes of Health (grants EY018139, EY017606 and EY000331) and an Unrestricted Grant from Research to Prevent Blindness to the Stein Eye Institute, University of California, Los Angeles.

Potential Drug Candidates Halt Prostate and Breast Cancer Growth

JUPITER, FL – March 9, 2017 – Scientists on the Florida campus of The Scripps Research Institute (TSRI) have designed two new drug candidates to target prostate and triple negative breast cancers.

August 30, 2017 57

The new research, published recently as two separate studies in ACS Central Science and the Journal of the American Chemical Society, demonstrates that a new class of drugs called small molecule RNA inhibitors can successfully target and kill specific types of cancer.

“This is like designing a scalpel to precisely seek out and destroy a cancer—but with a pill and without surgery,” said TSRI Professor Matthew Disney, senior author of both studies.

A Tool to Fight Prostate Cancer

RNAs are molecules that translate our genetic code into proteins. RNA defects can lead to cancers, amyotrophic lateral sclerosis (ALS), myotonic dystrophy and many other diseases.

In their ACS Central Science study, Disney and his colleagues used DNA sequencing to evaluate thousands of small molecules as potential drug candidates. The researchers were on the lookout for molecules that could bind precisely with defective RNAs—like keys fitting in the right locks.

This strategy led them to a compound that targets the precursor molecule to an RNA called microRNA-18a. This RNA had caught the attention of scientists who found that mature microRNA-18a inhibits a protein that suppresses cancer. When microRNA-18a is overexpressed, cancers just keep growing.

Disney and his team tested their compound, called Targapremir-18a, and found that it could target microRNA-18a and trigger prostate cancer cell death.

“Since microRNA-18a is overexpressed in cancer cells and helps to maintain them as cancerous, application of Targapremir-18a to cancer cells causes them to kill themselves,” Disney said.

Disney said the precise binding of Targapremir-18a to microRNA-18a means a cancer drug that follows this strategy would be likely to kill prostate cancer cells without causing the broad side effects seen with many other cancer therapies.

And there may be even bigger implications. “We could apply the strategy used in this study to quickly identify and design small molecule drugs for other RNA-associated diseases,” explained study first author Sai Velagapudi, a research associate in the Disney lab.

Testing the Strategy in Breast Cancer

The same screening strategy led the researchers to a drug candidate to target triple negative breast cancer, as reported in the Journal of the American Chemical Society.

Triple negative breast cancer is especially hard to treat because it lacks the receptors, such as the estrogen receptor, targeted with other cancer drugs. The Disney lab aimed to get around this problem by instead targeting an RNA called microRNA-210, which is overexpressed in solid breast cancer tumors.

August 30, 2017 58

The researchers tested their drug compound, Targapremir-210, in mouse models of triple negative breast cancer. They found that the therapy significantly slowed down tumor growth. In fact, a single dose decreased tumor size by 60 percent over a three-week period. The researchers analyzed these smaller tumors and discovered that they also expressed less microRNA-210 compared with untreated tumors.

Targapremir-210 appears to work by reversing a circuit that tells cells to “survive at all costs” and become cancerous. With microRNA-210 in check, cells regain their normal function and cancer cannot grow.

“We believe Targapremir-210 can provide a potentially more precise, targeted therapy that would not harm healthy cells,” said study first author TSRI Graduate Student Matthew G. Costales.

Next, the researchers plan to further develop their molecule-screening strategy into a platform to test molecules against any form of RNA defect-related disease.

Additional authors of the ACS Central Science study, “Defining RNA−Small Molecule Affinity Landscapes Enables Design of a Small Molecule Inhibitor of an Oncogenic Noncoding RNA,” were first author Sai Pradeep Velagapudi, Yiling Luo, Tuan Tran, Hafeez S. Haniff, Yoshio Nakai, Mohammad Fallahi, Gustavo J. Martinez and Jessica L. Childs-Disney of TSRI.

The research was funded by the National Institutes of Health (grant R01-GM097455).

Additional authors of the Journal of the American Chemical Society study, “Small Molecule Inhibition of microRNA-210 Reprograms an Oncogenic Hypoxic Circuit,” were Christopher L. Haga, Sai Pradeep Velagapudi, Jessica L. Childs-Disney and Donald G. Phinney of TSRI.

The study was supported by the National Institutes of Health (grant R01GM9455) and a Scheller Graduate Student Fellowship.

Scripps Florida Scientists Develop New Drug Delivery Method for Cancer Therapy

JUPITER, FL – March 16, 2017 – Scientists from the Florida campus of The Scripps Research Institute (TSRI) have developed a new drug delivery method that produces strong results in treating cancers in animal models, including some hard-to-treat solid and liquid tumors.

The study, led by TSRI Associate Professor Christoph Rader, was published March 16, 2017, online ahead of print in the journal Cell Chemical Biology.

The new method involves a class of pharmaceuticals known as antibody-drug conjugates (ADCs), which include some of the most promising next-generation antibody therapeutics for cancer. ADCs can deliver a cytotoxic payload in a way that is remarkably tumor-selective. So far, three ADCs have been approved by the U.S. Food and Drug Administration (FDA), but neither attaches the drug to a defined site on the antibody.

August 30, 2017 59

“We’ve been working on this technology for some time,” Rader said. “It’s based on the rarely used natural amino acid selenocysteine, which we insert into our antibodies. We refer to these engineered antibodies as selenomabs.”

Antibodies are large immune system proteins that recognize unique molecular markers on tumor cells called antigens. On their own, Rader noted, antibodies are usually not potent enough to eradicate cancer. However, their high specificity for antigens makes them ideal vehicles for drug delivery straight to tumor cells.

“We now show for the first time that selenomab-drug conjugates, which are ADCs that utilize the unique reactivity of selenocysteine for drug attachment, are highly precise, stable and potent compositions and promise broad utility for cancer therapy.”

Along with its potency, Rader noted, the ADC’s stability is critical to its effectiveness. The researchers found that their new ADCs showed excellent stability in human blood in vitro and in circulating blood in animal models. Moreover, the new ADCs were highly effective against HER2 breast cancer, a particularly difficult cancer to treat, and against CD138 multiple myeloma. Importantly, the ADCs did not harm healthy cells and tissues.

“The selenomab-drug conjugate significantly inhibited the growth of an aggressive breast cancer,” said TSRI Research Associate Xiuling Li, first author of the study. “Four of the five mice tested were tumor-free at the end of the experiment, a full six weeks after their last treatment.”

The researchers plan to investigate similar ADCs going forward. Rader, along with TSRI Professor Ben Shen, was recently awarded $3.3 million from the National Cancer Institute of the National Institutes of Health to test highly cytotoxic natural products discovered in the Shen lab using selenomabs as drug delivery vehicles.

In addition to Rader and Li, authors of the study, “Stable and Potent Selenomab-Drug Conjugates,” were Tina Moroni, Pablo Martinez-Acedo and Alex R. Nanna of TSRI; Christopher G. Nelson, David Hymel and Terrence R. Burke Jr. of the National Cancer Institute; and Rajesh R. Nair and Lori Hazlehurst of the H. Lee Moffitt Cancer Center.

The study was supported by the National Institutes of Health (grants U01 CA174844 and R01 CA181258), the Intramural Research Program of the National Cancer Institute, the Lymphoma Research Foundation, the Klorfine Foundation and the Holm Charitable Trust.

Researchers Discover Key to Drug Resistance in Common Breast Cancer Treatment

JUPITER, FL – March 20, 2017 – Three-quarters of all breast cancer tumors are driven by the hormone estrogen. These tumors are frequently treated with drugs to suppress estrogen receptor activity, but unfortunately, at least half of patients do not respond to these treatments, leaving them with drug-resistant tumors and few options.

August 30, 2017 60

Now, scientists from the Florida campus of The Scripps Research Institute (TSRI), the University of California (UC), San Diego and the University of Illinois have found that two immune system molecules may be key to the development of drug resistance in estrogen-driven breast cancers.

The researchers believe this finding may open the door to novel therapeutic approaches and influence treatment decisions for the tens of thousands of patients who suffer from estrogen- driven breast cancers.

These molecules, which are called interleukin 1 beta (IL1β) and tumor necrosis factor alpha (TNFα), had previously been linked to the spread of drug-resistant cancer, but scientists were unsure of the exact mechanisms that led these molecules to drive drug resistance.

The new study, published online ahead of print in the journal Molecular Cell, reveals that IL1β and TNFα turn on pathways that modify the actual shape of the estrogen receptor. This phenomenon appears to drive resistance to the common anti-cancer drug tamoxifen.

“Cytokines change the shape of the estrogen receptor, and that change overrides the inhibitory effects of tamoxifen and leads to drug resistance,” said TSRI Associate Professor Kendall Nettles, who led the new study alongside senior author Christopher K. Glass and study first author Joshua D. Stender of UC San Diego. “These findings dramatically alter our understanding of the biological actions of pro-inflammatory cytokines in breast cancer cells.”

Striking Back at Drug Resistance

Using a combination of genomic, cellular, biochemical and structural approaches, the researchers found that the way these cytokines alter the estrogen receptor are sufficient to induce growth of breast cancer cells in the absence of estrogen, precisely what happens when breast cancer is initially treated with an endocrine therapy like tamoxifen. Scientists found that in addition to reversing tamoxifen suppression of growth, activation of the estrogen receptor also enhanced the invasive properties of a specific line of human breast cancer cells known as MCF-7, the most studied human breast cancer cell in in the world.

Using x-ray crystallography, Nettles and his colleagues developed an atomic snapshot of the estrogen receptor to show how these shape changes occur and how the process might be blocked.

Nettles pointed out that both inflammation and immune cells are known causes of resistance, but if that inflammation can be blocked, resistance can be reduced or eliminated.

“These tumors can reprogram the immune cells to their advantage so that the cells become tumor supportive,” Kettles said. “We think we can produce hormone therapies that can, in essence, re-reprogram the immune system or prevent it from altering the receptor in the first place, which is an obvious strategy for blocking these adverse effects. Importantly, our atomic snapshot of the receptor showed that the same mechanism can explain how Her2Neu or other

August 30, 2017 61 growth promoting factors, as well as certain invasion and motility signals also cause resistance to anti-hormone therapies.”

This research was supported by the National Institutes of Health (grants R01DK091183, R01DK015556, R01CA17390 and P30 DK063491), Frenchman’s Creek Women for Cancer Research and The BallenIsles Men’s Golf Association.

Additional authors of the study, “Structural and Molecular Mechanisms of Cytokine – Mediated Endocrine Resistance in Breast Cancer,” included Tobias Strid and Maayan Yakir of UC San Diego; Jerome C. Jwachukwu, Sathish Srinivasan, Jason Nowak, Tina Izard and Rangarajan Erumbi of TSRI; Irida Kastrai, Yohan Kim, Jonna Frasor and John A. Katzenellenbogen of the University of Illinois; Xin-Qiuy Yao and Barry J. Grant of the University of Michigan Medical School; Hon S Leong of Western University, Ontario, Canada; and Chin-Yo Lin of the University of Houston.

Scientists Discover New Class of Anti-Diabetes Compounds that Reduce Liver Glucose Production

JUPITER, FL – March 27, 2017 – Scientists may have found a new tool for studying—and maybe even treating—Type 2 diabetes, the form of diabetes considered responsible for close to 95 percent of cases in the United States.

A team of scientists from the Florida campus of The Scripps Research Institute (TSRI), Dana- Farber Cancer Institute, Harvard Medical School and the School of Medicine, among others, have identified a new class of compounds that reduce production of glucose in the liver. One of these compounds, designed and optimized by TSRI scientists, significantly improves the health of diabetic animal models by reducing glucose levels in the blood, increasing insulin sensitivity and improving glucose balance.

The study, published recently in the journal Cell, was led by Pere Puigsever of Harvard Medical School and the Dana-Farber Cancer Institute and included Patrick Griffin, co-chair of the TSRI Department of Molecular Medicine, and Theodore Kamenecka, TSRI Associate Professor of Molecular Medicine.

The compound they identified, called SR-18292, modifies a protein known as PGC-1α. This protein plays a pivotal role in energy balance and helps control genes involved in energy metabolism. When cells overexpress PGC-1, during fasting or starvation, for example, glucose production in the liver soars. But when scientists modify PGC-1α function through a process called acetylation, glucose production declines.

“This protein was generally considered non-druggable,” said Griffin. “But the team approached the problem through the process of acetylation, which means we can influence the protein’s behavior indirectly. SR-18292 increases acetylation of PGC-1, which in turn shuts down glucose production in liver cells.”

August 30, 2017 62

Suppressing this overproduction makes PGC-1αa target ripe for exploitation in anti-diabetes treatments.

“After the screening process found several potential candidates, the TSRI team designed derivatives of those initial hits,” Griffin said. “We selected this compound based on its ability to induce acetylation and the fact that it had good pharmaceutical properties–so we could use it in animal models of Type 2 diabetes.”

While it isn’t known at this point what protein or enzyme is directly targeted by SR-18292, Griffin explained, this new compound, plus several others we’ve made, can be used as chemical tools to study the regulation of glucose metabolism. The researchers added that these same small molecules could one day be developed as either a single agent to treat diabetes, or used in combination with current anti-diabetic drugs.

The first author of the study, “Small Molecule Screen Identifies Selective PGC-1α Gluconeogenic Inhibitors that Ameliorate Type 2 Diabetes,” is Kfir Sharabi of Dana-Farber and Harvard Medical School. Other authors include Hua Lin of TSRI; Clint D. J. Tavares, John E. Dominy and Amy K. Rines of Dana-Farber and Harvard Medical School; Mark P. Jedrychowski and Steve P. Gygi of Harvard Medical School; Joao Paulo Camporez and Rachel J. Perry of Howard Hughes Medical Institute and Yale University; Jaemin Lee and Umut Ozcan of Boston Children's Hospital, Harvard Medical School; Marc Hickey, Melissa Bennion, Michelle Palmer, Partha P. Nag, Josh A. Bittker and José Perez of the Broad Institute of MIT and Harvard University.

The study was supported by the National Institutes of Health (grants F32 DK102293-01, U54HG005032, R24DK080261, R01DK-409369, 2U2CDK059635, R03DA032468 and R01 DK069966), the American Heart Association (15POST22880002) and the American Diabetes Association (1-16-PDF-111 and 7-12-MN-68).

Scientists Unravel How Protein Impacts Intellectual Disability

JUPITER, FL – April 25, 2017 – Your brain needs just the right balance between excitatory “on” signals and inhibitory “calm down” signals. Now scientists from the Florida campus of The Scripps Research Institute (TSRI) have shown that a protein helps balance nerve cell communication.

The new study, published recently online in the journal Cell Reports, could have implications for potential treatments of intellectual disability and other neurodevelopmental disorders. “This paper adds a new dimension to our understanding of the molecular mechanisms that impact intellectual disability,” said Brock Grill, a TSRI associate professor in the Department of Neuroscience. “Our study is the first to identify a defect in neuron communication caused by altering the activity of a gene called HUWE1, which causes intellectual disability, including Juberg-Marsidi-Brooks syndrome.”

Studying neuronal communication is important because the brain needs to balance excitatory neurotransmitters (to increase signal transmission) and inhibitory neurotransmitters (to calm

August 30, 2017 63 nerve cells down). An imbalance in the excitatory/inhibitory ratio is a central feature of many neurodevelopmental disorders—which occurs through gene overexpression or a loss of gene function.

For the study, Grill and his colleagues investigated neuronal communication balance using a simple model circuit in the nematode C. elegans, a small, transparent worm. Despite its small size, this worm shares half its genetic make-up with humans, which makes it an ideal model to study the genetics of neuron function. The researchers took a close look at GABA, the principal inhibitory neurotransmitter in C. elegans and the human brain. In C. elegans, the protein responsible for regulating GABA transmission is called EEL-1; in humans, the equivalent protein is known as HUWE1.

The researchers studied the function of EEL-1/HUWE1 in the worm motor circuit and found that decreasing or increasing the protein alters GABA transmission, upending the excitatory/inhibitory balance, a shift that leads to impaired locomotion and increased sensitivity to electroshock-induced seizure.

“Using a simple model circuit, we’ve identified a key player required to achieve a balance of excitation and inhibition,” Grill said. “This opens up a new concept for why HUWE1 causes intellectual disability."

HUWE1 affects only the release of the GABA neurotransmitter, not the levels or function of the GABA receptor, Grill noted. He said more research is needed into how this actually affects the brain.

“The paper is an important step in understanding how increased or decreased activity of HUWE1 can alter circuit function and lead to intellectual disability,” said TSRI Research Assistant Karla Opperman, first author of the study.

The study represents important progress in understanding the molecular underpinnings of intellectual disability. In particular, results from the study show for the first time that mutations that cause Juberg-Marsidi-Brooks syndrome result in loss of HUWE1 function and can impair nerve cell function.

In addition to Grill and Opperman, other authors of the study, “The HECT Family Ubiquitin Ligase EEL-1 Regulates Neuronal Function and Development,” are Andrew Giles and Rayna L. Birnbaum of TSRI; Ben Mulcahy and Mei Zhen of the Lunenfeld-Tannenbaum Research Institute, Mt. Sinai Hospital, Toronto; Monica G. Risley and Ken Dawson-Scully of Florida Atlantic University; and Erik D. Tulgren of the University of Minnesota.

The study was supported by the National Institutes of Health (grant 2R01 NS072129) and the National Science Foundation (grant IOS-1121095).

August 30, 2017 64

Scientists Solve Major Cancer Protein Conundrum

JUPITER, FL – May 4, 2017 – Despite intense research, there’s been much confusion regarding the exact role of a protein in a critical cancer-linked pathway. On one hand, the protein is described as a cell proliferation inhibitor, on the other, a cell proliferation activator, a duality that has caused a great deal of scientific head scratching.

Now scientists from the Florida campus of The Scripps Research Institute (TSRI) have solved the conundrum, uncovering the regulatory machinery underlying the function of a protein, called angiomotin.

The researchers found that angiomotin’s activities depend on a process called phosphorylation—when a phosphate group is added to its structure at a specific location. Add a phosphate group, and the protein can inhibit cell proliferation. But remove a phosphate group from its normal makeup, and the protein promotes cell proliferation, encouraging cancer cell growth.

The study, led by Joseph Kissil, associate professor in the Department of Molecular Medicine at TSRI, was recently published in the journal eLife.

Protein’s Position in Cells is Key

The new study sheds light on signaling pathway in cells called the Hippo-YAP pathway.

YAP’s involvement in cancer has been demonstrated in several tissues, including liver, intestine, heart, pancreas and brain. Recent studies show YAP plays a key role in developing drug resistance in lung and colon cancer cells and promoting cancer in some colon and pancreatic cancers. Hippo regulates cell proliferation and programmed cell death, which often become corrupted in diseases like cancer.

Whether the Hippo-YAP pathway can be altered by the protein angiomotin is not in question. But while some studies give angiomotin a YAP-inhibitory function, others indicate that the protein is required for YAP activity.

Kissil and his colleagues discovered what lies at the heart of those seemingly contradictory reports. They found that YAP forms a complex with angiomotin and another protein called Merlin. When angiomotin is phosphorylated, that changes the localization of this complex to the cell plasma membrane where it prevents cells from proliferating.

“The relocation of the protein complex out of the nucleus to the plasma membrane prevents YAP from operating as a growth-promoting transcriptional activator,” explained TSRI Graduate Student Sany Hoxha, co-first author of the study.

Conversely, when angiomotin is less than fully phosphorylated, the complex is localized in the nucleus, where it promotes YAP-dependent cell proliferation.

August 30, 2017 65

“Since this is a major pathway for diseases like cancer and fibrosis, our findings add a brand- new layer of valuable information,” said Kissil.

In addition to Kissil and Hoxha, the other first author of the study, “Regulation of Localization and Function of the Transcriptional Co-Activator YAP by Angiomotin,” is Susana Moleirinho. Other authors include Vinay Mandati, Graziella Curtale and Scott Troutman of TSRI; and Ursula Ehmer of Technische Universität München, Munich, Germany.

The study was supported by the National Institutes of Health (grants NS077952 and CA124495) and the Children’s Tumor Foundation.

Scientific Trailblazers to Address Historic Graduating Class at TSRI

TSRI Graduate Program Marks 25th Commencement with Two Honorary Degree Recipients

LA JOLLA, CA & JUPITER, FL – May 15, 2017 – In recognition of their career-long contributions to science and public health, renowned chemist Christopher Walsh and public health advocate Gerald Chan will receive honorary doctoral degrees from The Scripps Research Institute’s Graduate Program on Friday, May 19. During the Institute’s 25th Commencement Ceremony, TSRI board members Walsh and Chan will join the graduates of the top-10 ranked program in celebrating academic achievement and individual contributions to scientific research.

“TSRI is privileged to honor two distinguished leaders in the fields of biomedical research and public health,” said TSRI President Peter G. Schultz. “The broad impact of Chris and Gerald’s achievements should serve as an inspiration to our graduating class as they embark on the next phase of their careers and apply their own learnings to advancing scientific discovery around the world.”

Chan is the co-founder of Morningside, a private equity and venture capital firm. His investments include support for infectious disease research, vaccines, antivirals, metabolic diseases, autoimmune diseases and rare orphan diseases.

Reflecting on the historic nature of this commencement, Chan will address the legacy of science in the United States. He will touch on the United States’ leadership in science following World War II, a tradition he hopes this era’s researchers can carry on with the support of government leaders. “If we care about science in America, we should care about the research institutions where some of the best science is done,” said Chan in a preview of his upcoming speech.

Walsh is a renowned chemist and academic leader who currently serves as a consulting professor to the Stanford University department of chemistry. He has been involved in a variety of venture-based biotechnology companies since 1981 and currently serves on several boards of directors, including the TSRI affiliate, the California Institute for Biomedical Research (Calibr).

August 30, 2017 66

Reflecting on his own career in chemistry, Walsh will advise on the importance of intellectual curiosity. “You should use your talent to address and solve important problems, not just fill in the pages of defined projects,” said Walsh, previewing his remarks.

TSRI’s rigorous doctoral program, which takes an average of five years to complete, consists of a year of classwork and rotation through laboratories, customizable to each student’s interests and long-term goals, followed by work on individual research projects under the guidance of a faculty mentor. U.S. News & World Report currently ranks TSRI’s graduate program as second in country in the specialty of biochemistry, sixth in the specialty of organic chemistry, seventh overall in chemistry and ninth overall in the biological sciences, based on a survey of department heads, deans, directors of graduate studies, and other academics in each discipline.

The 25th Commencement Ceremony will take place at TSRI’s Hazen Campus in La Jolla, CA, on Friday, May 19, 2017, at 10 AM.

Scripps Florida Scientist Named Finalist for Blavatnik National Awards for Young Scientists

Matthew Disney, a professor in the Department of Chemistry on the Florida campus of The Scripps Research Institute (TSRI) has been named one of 30 national finalists for the Blavatnik National Awards for Young Scientists in the Chemistry category.

The annual Blavatnik Awards, established in 2007 by the Blavatnik Family Foundation and administered by the New York Academy of Sciences, recognize exceptional young researchers who will “drive the next generation of innovation by answering today’s most complex and intriguing scientific questions.”

“I am very grateful for our group's recognition from the Blavatnik Foundation and what it represents—that the group has been doing important science and changing what the world views as druggable targets,” Disney said. “It is also important recognition for my great co- workers and team members that have tackled important problems that many thought could not be solved.”

“We look forward to learning of the directions that the pioneering work of the 2017 National Finalists will take in the coming years,” said Len Blavatnik, Founder and Chairman of Access Industries, head of the Blavatnik Family Foundation and an Academy Board Governor in a statement.

The finalists are considered 30 of America’s most important young scientific researchers aged 42 years and younger. They were selected from 308 nominations of faculty-rank researchers working at 149 of the nation’s top academic and research institutions, representing 41 states and the District of Columbia. From these finalists, three Laureates, from the disciplines of Life Sciences, Chemistry and Physical Sciences & Engineering, will be announced in late June 2017. Each Laureate will receive an unrestricted cash prize of $250,000—the largest prize of its kind for early career scientists.

August 30, 2017 67

In 2016, TSRI Professor Phil Baran, Darlene Shiley Chair in Chemistry and a TSRI alumnus, was named a Blavatnik Laureate in Chemistry.

Firefly Gene Illuminates Ability of Optimized CRISPR-Cpf1 to Efficiently Edit Human Genome

JUPITER, FL – June 19, 2017 – Scientists on the Florida campus of The Scripps Research Institute (TSRI) have improved a state-of-the-art gene-editing technology to advance the system’s ability to target, cut and paste genes within human and animal cells—and broadening the ways the CRISPR-Cpf1 editing system may be used to study and fight human diseases.

Professor Michael Farzan, co-chair of TSRI's Department of Immunology and Microbiology, and TSRI Research Associate Guocai Zhong improved the efficiency of the CRISPR-Cpf1 gene editing system by incorporating guide RNAs with "multiplexing" capability. Guide RNAs are short nucleic acid strings that lead the CRISPR molecular scissors to their intended gene targets. The TSRI discovery means multiple genetic targets in a cell may be hit by each CRISPR-Cpf1 complex.

"This system simplifies and significantly improves the efficiency of simultaneous editing of multiple genes, or multiple sites of a single gene," Zhong said. "This could be very useful when multiple disease-related genes or multiple sites of a disease-related gene need to be targeted."

"This approach improves gene editing for a number of applications," Farzan added. "The system makes some applications more efficient and other applications possible."

This study was published as an advanced online paper in the journal Nature Chemical Biology on June 19, 2017.

TSRI Advance Makes CRISPR More Efficient

Short for "Clustered Regularly Interspaced Short Palindromic Repeat," the CRISPR gene editing system exploits an ancient bacterial immune defense process. Some microbes thwart viral infection by sequestering a piece of a virus' foreign genetic material within its own DNA, to serve as a template. The next time the viral sequence is encountered by the microbe, it's recognized immediately and cut up for disposal with the help of two types of RNA. Molecules called guide RNAs provide the map to the invader, and CRISPR effector proteins act as the scissors that cut it apart.

Over the last five years, the CRISPR gene editing system has revolutionized microbiology and renewed hopes that genetic engineering might eventually become a useful treatment for disease. But time has revealed the technology's limitations. For one, gene therapy currently requires using a viral shell to serve as the delivery package for the therapeutic genetic material. The CRISPR molecule is simply too large to fit with multiple guide RNAs into the most popular and useful viral packaging system.

August 30, 2017 68

The new study from Farzan and colleagues helps solve this problem by letting scientists package multiple guide RNAs.

This advance could be important if gene therapy is to treat diseases such as hepatitis B, Farzan said. After infection, hepatitis B DNA sits in liver cells, slowly directing the production of new viruses, ultimately leading to liver damage, cirrhosis and even cancer. The improved CRISPR- Cpf1 system, with its ability to ‘multiplex,’ could more efficiently digest the viral DNA, before the liver is irrevocably damaged, he said.

"Efficiency is important. If you modify 25 cells in the liver, it is meaningless. But if you modify half the cells in the liver, that is powerful," Farzan said. "There are other good cases— say muscular dystrophy—where if you can repair the gene in enough muscle cells, you can restore the muscle function."

Two types of these molecular scissors are now being widely used for gene editing purposes: Cas9 and Cpf1. Farzan said he focused on Cpf1 because it is more precise in mammalian cells. The Cpf1 molecule they studied was sourced from two types of bacteria, Lachnospiraceae bacterium and Acidaminococus sp., whose activity has been previously studied in E. coli. A key property of these molecules is they are able to grab their guide RNAs out of a long string of such RNA; but it was not clear that it would work with RNA produced from mammalian cells. Guocai tested this idea by editing a firefly bioluminescence gene into the cell’s chromosome. The modified CRISPR-Cpf1 system worked as anticipated.

"This means we can use simpler delivery systems for directing the CRISPR effector protein plus guide RNAs," Farzan said. "It's going to make the CRISPR process more efficient for a variety of applications."

Looking forward, Farzan said the Cpf1 protein needs to be more broadly understood so that its utility in delivering gene therapy vectors can be further expanded.

In addition to Farzan and Zhong, the authors of the study, “Cpf1 proteins excise CRISPR RNAs from mRNA transcripts in mammalian cells,” included Haimin Wang, and Mai H. Tran of TSRI; and Yujun Li of TSRI and Wu Lien-Teh Institute, Harbin Medical University, Harbin, China.

The study was supported by the National Institutes of Health (grants R37 AI091476 and P01 AI100263).

Scripps Florida Scientists Awarded $2 Million to Develop ‘Industrial Level’ Screening for Treatments of Autism-Related Intellectual Disability

JUPITER, FL – July 5, 2017 – A team of scientists from the Florida campus of The Scripps Research Institute (TSRI) have been awarded nearly $2 million from the National Institute of Mental Health of the National Institutes of Health to develop an ‘industrial level’ high throughput screening platform that could lead to new treatments for a number of childhood brain disorders.

August 30, 2017 69

The three-year-grant will be overseen by Gavin Rumbaugh, a TSRI associate professor in the Department of Neuroscience, and Associate Professor Louis Scampavia and Assistant Professor Timothy Spicer, co-directors of the campus’ High Throughput Screening facility.

Over the past several years, the three scientists have worked as a collaborative group to create a cost-effective screening platform that can support medium-scale screening in neuronal networks. The new grant will allow them to optimize their current efforts to support screening campaigns of a much larger scale, which is expected to increase the chances of a successful screen.

The scientists are focused on mutations in the Syngap1 gene that can damage development of specific types of neurons in the brain and lead to intellectual disability. Problems with higher cognitive processes, such as language, reasoning and memory arise in children with these mutations.

These rare genetic brain disorders offer the greatest potential for new therapeutics because the disease mechanism is generally straight forward – basically low expression of a single protein in this case.

“To improve conditions in these patients, the most rational strategy is to treat them with ‘magic bullet’ compounds that replace those missing functional proteins,” Rumbaugh said. “However, searching for the right compounds can only be done in neurons because this gene is not expressed well outside the brain.

Currently, ultra high-throughput screening, where tens-of-thousands of compounds are tested, is not easily performed in neurons,” Rumbaugh noted.

“This grant will help support new technologies that will streamline screening in neurons,” he said. “One added benefit of this project is that it’s expected to stimulate the creation of screening assays for other brain disorders.”

Earlier studies done by the Rumbaugh lab demonstrated that if Syngap1 protein expression can be fixed by a form of gene therapy in an animal model, then neuronal function and intellectual ability in the animal model were also fixed.

Unfortunately, this type of gene therapy is not yet feasible in humans. However, animal studies suggest that identification of Syngap1-regulating compounds by ultra high-throughput screening may lead to therapies that target the disease mechanism in this genetic disorder.

Damaging mutations in Syngap1 that reduce the number of functional proteins are one of the most common causes of sporadic intellectual disability and are associated with schizophrenia and autism spectrum disorder. Some estimates suggest that these non-inherited genetic mutations account for two to eight percent of these intellectual disability cases.

The number of the grant is 1R01MH113648-01.

August 30, 2017 70

Scripps Florida Scientists Identify Protein Deficiency Involved in Childhood Anemia and Cancers

JUPITER, FL – July 31, 2017 – Scientists from the Florida campus of The Scripps Research Institute have shown how the lack of a specific protein may be a significant contributing factor to a devastating childhood anemia that has been closely associated with a 30-to-40-fold increase in the incidence of colon cancer, osteosarcoma and leukemia in those who suffer from it.

The new research, which was published in the journal Nature Structural and Molecular Biology, was led by TSRI Associate Professor Katrin Karbstein.

These new findings provide the first molecular understanding of the pathology of the disease known as Diamond-Blackfan anemia – and offer a potential target for developing new therapeutic approaches.

Karbstein’s research focuses on the assembly of ribosomes—large macromolecular machines that produce proteins by decoding information carried in messenger RNA (mRNA) and then turning that code into proteins, assembling them from amino acids. Problems in the assembly process or in messenger RNA (mRNA) can lead to serious problems, including birth defects like Diamond Blackfan Anemia and cancers.

In the new study, Karbstein and her colleagues show that the reduced levels of a protein known as Rps26, which are caused by Diamond-Blackfan anemia, lead to the formation of RPS26- deficient ribosomes. These Rps26 deficient ribosomes disrupt the translation of essential mRNAs, leading to reduced overall ribosome production, which helps drive the disease in the first place. The result, Karbstein said, is something of a paradox.

“Diamond-Blackfan patients tend to develop very specific cancers, far more than normal people,” she said. “But cancer cells need lots of ribosomes, which is the exact opposite of what is happening in Rps26-deficiency. The cancer might arise because of the new functions that the Rps26 deficient ribosomes develop.”

While this population of deficient ribosomes is capable of binding mRNA, they actually prefer a different population of transcripts – single strands of RNA -- than their normal counterparts, distinguished by a specific arrangement of nucleotides on the mRNA molecule known as Kozak sequences that help efficient translation.

As a general rule, mRNAs with a good Kozak sequence get translated more readily than others because they produce proteins with critical cellular functions. But Rps26 deficient ribosomes don’t care that these normal mRNAs are supposed to be privileged– its popular democracy in action. “They like all ribosomes,” Karbstein said. “So the privileged ones are doing less well, and the translation of what you might call the less privileged increases. That’s the critical difference. This increase perturbs the homeostasis and disease follows soon after.”

August 30, 2017 71

What causes this deficit in the first place? Besides the genetic causes, which lead to inherited disease, the study also revealed the surprising finding that exposure to high salt and pH stress can lead to the formation of Rps26-deficient ribosomes.

“We have long suspected that Rps26-deficient ribosomes might be the cause of diseases like Diamond Blackfan Anemia,” Karbstein said, “but we haven’t known that they have physiological roles in the response to environmental stresses. This is likely the reason that they are not discarded by cellular quality control pathways in the first place.”

“We found that ribosomes deficient in the protein Rps26 preferentially translate mRNAs from specific stress response pathways,” said Max B. Ferretti, the first author of the study. “Further, we show that yeast exposed to these stresses respond by removing Rps26 from their ribosomes, which shows how a general ribosomal defect dysregulates translation. “ In addition to Karbstein and Ferretti, other authors of the study, “Rps26 Directs mRNA-Specific Translation by Recognition of Kozak Sequence Elements,” are Homa Ghalei, Ethan A. Ward and Elizabeth L. Potts of TSRI.

The study was supported by the National Institutes of Health (Grant numbers R01-GM086451 and F31-GM116406), the PGA National Women’s Cancer Awareness Fellowship, the Richard & Helen DeVos Graduate Fellowship and the Howard Hughes Medical Institute.

A Perspective on the Biology of Forgetting

JUPITER, FL – August 3, 2017 – Ron Davis, Ph.D., Professor and Chair of Neuroscience at The Scripps Research Institute (TSRI) in Jupiter, Florida, teamed up with Yi Zhong, Ph.D., Professor of Life Sciences at Tsinghua University in Beijing, China, to pen a call to action published this week in the journal Neuron. The article reveals surprising new insights into how the brain manages memory storage.

Like many neuroscientists, the researchers have spent years working to understand the molecular mechanisms behind learning and memory, but about five years ago, they shifted their focus to the mechanisms behind forgetting. In the review, the pair presents evidence that our brains actively use molecular mechanisms to erode memories. “We're laying out to the scientific community the evidence that we have and the rationale for the brain being designed to forget,” says Davis.

While most scientists agree that our brains use molecular mechanisms that alter the structure and function of certain neurons in order to learn and store memories, some researchers are skeptical that our brains would eventually alter those neurons in such a way that could effectively erase those memories. T Many psychologists believe that the memory engram—the set of cells and molecules involved in encoding a given memory—never goes away, but that as memories are used less, people struggle to retrieve them. Davis and Zhong say that while there are instances in which people struggle to retrieve intact memory engrams, our brains also use active forgetting—a series of molecular changes that degrades a memory engram and gradually removes it completely—to make room for new memories.

August 30, 2017 72

The pair asserts that active forgetting is a mechanism for maintaining homeostasis in the brain. Our bodies employ homeostatic control mechanisms to maintain healthy internal temperatures, glucose levels and more. We are bombarded with information all day long, and there may be a limit to the resources the brain can use to store memories at a given time, and active forgetting of unused and unimportant memories frees up the resources needed to encode important new memories. Davis and Zhong believe disruption of this process could be involved in a variety of neuropsychiatric disorders, such as autism spectrum disorders and post-traumatic stress disorder.

There are likely dozens of different ways the brain approaches the task of molecularly erasing unused memories. In the article, the team describes what they and others have found about certain molecules involved in forgetting. The best-understood mechanism thus far is RAC1- dependent forgetting. They posit that the RAC1 molecule is involved in at least four different types of forgetting, which they have studied in the olfactory memories of Drosophila. RAC1 is known to be involved in chemical and structural changes that allow neurons to encode memories. The researchers develop arguments that when activated by a particular type of dopamine receptor, RAC1 is involved in a cascade that remodels the cell’s actin cytoskeleton to erode those memories. But this is only one piece of the puzzle—although the researchers have identified several other molecules involved in the RAC1-dependent mechanism, there are many steps left to elucidate in the process.

Scientists have a long way to go to fully understand how our brains forget unused and unimportant memories and make space for new ones. One of the main challenges, explains Davis, is the same obstacle that makes the research so exciting. There really isn’t a library of information on the topic, since so few researchers have studied active forgetting—meaning there aren’t a lot of clues to drive their questions. “It’s a whole new window into the learning and memory mechanisms,” he says. “We’re doing experiments that have never been done before and examining brain processes that have never been examined before.”

Research in the Zhong laboratory is supported by grants 2013cb835100 from the 973 program of the Ministry of Science and Technology of China, 91332207 from the National Science Foundation of China, and No. Z161100002616010 from Beijing Municipal Science & Technology Commission. Research on forgetting in the Davis laboratory is supported by grants 4R37NS019904, 5R01NS052351 and 1R35NS097224 from the NINDS.

TSRI Ranks No. 1 in Innovation Influence

LA JOLLA, CA—August 10, 2017—The Scripps Research Institute (TSRI) is the most influential research institution in the world, according to the Nature Index 2017 Innovation supplement, which sheds light on the impact of academic research on innovation. The ranking, released today by the journal Nature, analyzes data about research quality and the broad influence it has on inventions.

“This new ranking underscores the worldwide impact of TSRI scientists, who share a common goal of improving public health through scientific discovery, and, importantly, improving the way we make those discoveries,” said Jamie Williamson, TSRI’s Executive Vice President for

August 30, 2017 73

Research and Academic Affairs. “We are proud to be recognized for the profound influence our science has had on other researchers and laboratories around the world.”

The Normalized Lens Influence Metric used by the Nature Index measures the influence an institution’s research has had on innovation by calculating the citations of its research articles in patents owned by third parties, rather than those owned by institutions themselves.

According to this metric, TSRI ranks number one, above other internationally renowned research institutes such as the Rockefeller University in New York, the Massachusetts Institute of Technology and Stanford University.

“This analysis comes at a time when following the transfer of scientific knowledge into industry and the economy is a growing priority for governments and research funding agencies, said David Swinbanks, founder of the Nature Index. “For them, the need to demonstrate that publicly funded science is being used for society’s benefit is paramount,”

A key metric in this ranking is publications. More than 40 percent of all TSRI’s natural science articles appear in the Nature Index. The average across the 200 institutions listed is 21.9 percent.

TSRI scientists are also active in developing their own innovations, with more than 1,000 patents and seven FDA approved drugs to their credit. Additionally, the Institute’s recent alliance with the California Institute for Biomedical Research (Calibr) adds translational research to its capabilities, enabling a new bench-to-bedside model that will accelerate the translation of basic scientific discoveries into critically-needed new medicines for unmet medical needs.

New TSRI Research Reveals Pathway for Anti-Aging Therapies

LA JOLLA, CA, & JUPITER, FL – August 21, 2017 – Two new studies led by scientists at The Scripps Research Institute (TSRI) could guide future therapies to improve health and lifespan. Together, the studies in animal models shed light on how reducing calorie intake directly influences lifespan by also reducing body temperature. Importantly, the researchers also identified a molecule that responds to lower body temperatures to regulate lifespan in fruit flies, giving scientists a target for future pharmaceuticals that may increase longevity.

“This exciting discovery demonstrated that the main biochemical pathway that regulates aging acts by lowering body temperature,” said TSRI Professor Bruno Conti, who led the first study. “Flies also live longer in the cold and the mechanisms that regulate this response overlap with key aging pathways,” said TSRI Associate Professor William Ja, who led the second study on TSRI’s Florida campus.

The new research was published in the journal Proceedings of the National Academy of Sciences.

August 30, 2017 74

Study Sheds Light on How Calorie Restriction Extends Lifespan

Scientists have long known that reducing calories—without causing malnutrition—can extend lifespan in animals.

“Researchers had found that mice that eat 40 percent less than normal live much longer—and they actually look younger,” said Conti.

Calorie restriction can also lead to a lower core body temperature. This just makes sense, Conti explained. “If there’s no food, it’s smart to reduce body temperature. You’re reducing your energy expenditure,” he said. “It’s fundamental for the survival of the organism.”

The new study finally shows how these two observations are related. Conti and his colleagues found that it all comes down to how the body uses a molecule called the IGF1 receptor. Previous studies had shown that lowering the activity of the IGF1 receptor can extend lifespan.

Using genetically engineered mouse models, Conti’s team found that calorie restriction provides the signal to lower body temperature, and that this occurs by reducing the activity of the IGF1 receptor in the brain.

“For the first time, we showed that calorie restriction, body temperature and the IGF1 receptor are part of the same pathway,” said study first author Rigo Cintron-Colon, a graduate student in the Conti Lab.

TSRI Assistant Professor Manuel Sanchez-Alavez, who is a co-first author on the publication, emphasized that the research should not encourage people to try extreme diets or cool their body temperatures in the hope of living longer. “My advice is: Do not try this at home,” he said. He explained that the study is significant because a better understanding of the full pathway between calorie restriction and lifespan gives scientists several points where they could attempt to intervene with a pharmaceutical.

Inspired by observations that female mice seem more sensitive to calorie restriction, the researchers said their next step is to investigate whether male and female sex hormones affect the pathway.

Researchers Identify Potential Target for Anti-Aging Therapies

The second study, led by Ja, drilled even deeper into the relationship between temperature and lifespan.

Using fruit flies as a model, Ja and his colleagues found that colder ambient temperatures result in a “metabolic brake” to slow down the production of new proteins in the cell. A molecule called 4E-BP responds to cold by sparing select proteins and regulating longevity.

Although the Conti and Ja studies were conducted independently, they reveal another intriguing parallel. The biochemical pathways implicated by the two publications are related and interact

August 30, 2017 75 extensively. “Surprisingly, even though flies don’t maintain their body temperature like mammals do, organisms may use similar mechanisms to regulate lifespan in response to changes in temperature,” said Ja.

“Our results not only shed light on the temperature effect, but they also establish a parallel between temperature and diet, because 4E-BP is thought to play a role in diet-related life extension as well,” added Dr. Gil Carvalho who, along with Dr. Ilaria Drago, was a co-first author on the publication.

“We also show that the effects of diet on aging are cumulative,” Ja said. “Environmental factors such as temperature and diet seem to affect our health gradually over the years, so the sooner we implement healthy changes, the better off we're likely to be.” If the work in fruit flies holds true in humans, he noted, the research emphasizes the need to make healthy decisions from a young age. Ja hopes further studies into 4E-BP help scientists find pharmaceuticals that “tune” the body’s response to diet and temperature to regulate healthy aging.

The study led by Conti, titled “The insulin-like growth factor 1 regulates hypothermia during calorie restriction,” included William Nguyen and Simone Mori from TSRI, Ruben Gonzalez Rivera from the National Autonomous University of Mexico; Tiffany Lien from the University of California, San Diego; Tamas Bartfai from Stockholm University; and Saba Aïd, Jean- Christophe François and Martin Holzenberger from INSERM and Sorbonne Universities. The research was supported by the National Institutes of Health (grant GM113894), INSERM and The National Council of Science and Technology (CONACYT), Mexico (Postdoctoral Scholarship Award No. 37548).

The study led by Ja, titled “The 4E-BP growth pathway regulates the effect of ambient temperature on Drosophila metabolism and lifespan,” included authors from Florida Atlantic University, the University of Southern California, the University of Tokyo and the University of Colorado School of Medicine. The research was supported by the National Institutes of Health (grants R01GM113894, R01AG045036, and R21DK092735), the Glenn Foundation for Medical Research/American Federation for Aging Research and The Ellison Medical Foundation.

TSRI Researchers Develop Assay to Help Identify New Pain Medication Candidates

JUPITER, FL – August 23, 2017 – Scientists at the Scripps Research Institute (TSRI) in Jupiter, Florida, have developed a test that will help scientists identify new drug candidates for treating pain that have fewer side effects, like addiction. The study conducted by scientists in Scott Hansen’s lab at TSRI was recently published in Cell Reports.

Neurological drugs, like painkillers, are notoriously messy. They bind to targets throughout the brain and produce myriad undesirable side effects. One reason for this is that the tests, or assays, that researchers use to identify how molecules interact with certain receptors have been difficult to apply to ion channels, a type of structure that drives brain function. The Hansen Lab has overcome this limitation by developing an assay that will allow scientists to screen an ion channel, TREK-1, that is known to be involved in pain.

August 30, 2017 76

Ion channels are proteins situated in the membrane of a cell. When they open, they allow ions to flow into or out of the cell, altering the cell’s electrical charge and, ultimately, its behavior. While most scientists agree that lipids inside the cell’s membrane have important roles in the cell’s function, the interactions between lipids and ion channels have been difficult to study due to a lack of applicable assays.

“There’s this mystery about membranes and lipids,” said Scott Hansen, associate professor of molecular medicine at TSRI. “We’ve known that they are important regulators and potential targets [for drugs], and there just really wasn't an assay to screen them at the level needed.”

In order to conduct a high-throughput assay, scientists need to isolate the channels by solubilizing the environments surrounding them. Ion channels are found in the lipid membrane, which unfortunately is not soluble. Hansen explained that they’re like a clump of fat that won’t go down the sink when you’re trying to do your dishes.

Fortunately, Hansen and his team realized that like fat, once the lipid membrane could be broken up and solubilized with a little detergent, the channel could be used in a soluble assay. The team used the strategy in this study to show that a lipid called PIP2, which was thought to agonize the channel, is actually antagonizing—or blocking—it.

Next, the assay will be advanced to drug discovery and used to screen for possible drug candidates that will act on the receptor and may be useful in managing pain. TSRI is known for its pharmaceutical-grade drug-screening capabilities, and the technique is likely to be applicable for screening other ion channels.

In addition, the study provided evidence that these channels can be opened and closed by lipids, like PIP2. Scientists have long known that some ion channels are controlled by neurotransmitters binding to the membrane structure (ligand-gated ion channels). Hansen and his team have recently proposed one more way these channels are controlled. Lipids, which float throughout the lipid membrane itself, can bind to the receptors and cause them to open or close as well. Hansen calls these channels “lipid-gated ion channels.” This aspect of ion channel function opens a whole new set of questions about how our brains work.

In addition to Hansen, three other researchers worked on the study: Lead Researcher Cerrone Cabanos of TSRI, as well as Miao Wang and Xianlin Han of the Sanford Burnham Prebys Medical Discovery Institute in Orlando, Florida.

The work was supported by an NIH Director’s New Innovator Award (1DP2NS087943-01 to S.B.H.).

New Exploratory Grant Will Help Scripps Florida Scientists Advance Treatment Development

JUPITER, FL – August 24, 2017 – Scientists from the Florida campus of The Scripps Research Institute have been awarded nearly $1 million from the National Institute of Neurological Disorders and Stroke, part of the National Institutes of Health, to develop a genetically

August 30, 2017 77 engineered animal model of a debilitating inherited disease that could accelerate the development of potential treatments.

Joseph Kissil, a TSRI associate professor in the Department of Molecular Medicine, is the principal investigator for the three-year grant. This is a phased award; if certain benchmarks are met, the grant is extended for a third year.

Kissil has been studying Neurofibromatosis Type 2 (NF2) for a number of years. The disease is caused by mutations in the anti-tumor gene NF2, which leads to tumors of the auditory nerve that connects the inner ear to the brain and causes severe hearing loss and impaired balance and walking. A majority of patients develop additional tumors throughout the nervous system, which can lead to fluid buildup in the brain and seizures.

While knowledge of the molecular mechanisms underlying the disease has improved significantly over the past two decades, there are still no effective treatments. Current options are non-curative and include surgery, radiation therapy, as well as temporary interventions for symptom control.

“Clearly, there is an urgent need to develop better options for these patients,” Kissil said. “One of the main obstacles is the lack of animal models that can be used in pre-clinical testing of drug candidates. This grant will help us develop a genetically engineered animal model of the disease that accurately portrays the biology of tumor development in patients – and that can be used to evaluate drug candidate efficacy.”

In 2013, Kissil and his colleagues identified a new drug candidate to treat NF2. The compound —known as FRAX97—slowed the proliferation and progression of tumor cells in some preliminary animal models of the disease. Originally developed for neurodegenerative disease, the compound targets a protein family known as p21-activated kinases or PAKs. These kinases (enzymes that add a phosphate group to other proteins and change their function) play a critical role in the development of NF2.

“While the models that we used then offered a good approximation, they were just that – an approximation,” Kissil said. “There’s still a great need for models that more closely reflect what happens in patients and that the new model we’re working on does just that.”

The number of the grant is1R21NS099417-01A1.

TSRI Adds New Faculty to World-Class Institute

LA JOLLA, CA – August 24, 2017 – The Scripps Research Institute today announced the appointment of two new faculty members – Alexander Adibekian, Ph.D., who will join the Department of Chemistry in Jupiter, FL as associate professor, and Li Ye, Ph.D. who joins the Department of Neuroscience in La Jolla, Ca as an assistant professor.

“The appointments of Drs. Adibekian and Ye reflect TSRI’s commitment to hiring exceptional scientists with a passion for basic research,” said Jamie Williamson, TSRI’s Executive Vice President for Research and Academic Affairs. “Alex’s active program in chemical proteomics

August 30, 2017 78 and Li’s interdisciplinary approach to studying the link between the brain and metabolism are important new additions to the innovative research occurring on both campuses.”

Adibekian, a former member of Benjamin Cravatt’s laboratory, joins TSRI from the Department of Organic Chemistry at the University of Geneva, Switzerland, where he was an assistant professor. His research interests include the discovery of new cysteine-reactive small molecules and the identification of their proteomic targets, as well as the development of novel chemical strategies that allow rapid access to collections of structurally diverse cysteine- reactive small molecules.

Adibekian received his bachelor's and master’s degrees from the Leibniz Universität in Hanover, Germany, and his Ph.D. from ETH Zürich, Switzerland, where he conducted his doctoral research fellowship in the laboratory of Professor Peter H. Seeberger.

“I’m very excited to join the outstanding faculty at The Scripps Research Institute,” Adibekian said. “The research here is second to none, and the collaborative spirit among the faculty makes it a truly special place to conduct cutting edge research. I believe my work will flourish here.” Ye, who joins TSRI from Stanford University, is focused on the transcriptional control of peripheral metabolism through chemical biology and genetic approaches. His current research efforts aim to understand the central regulation of whole-body metabolism, especially in the context of obesity and insulin resistance.

“Having the opportunity to work with experts in high-throughput screens, molecular physiology, and behavioral neurosciences at TSRI is something I have looked forward to my whole career,” Ye said. “They are among the finest in the world in their fields and I look forward to learning from and collaborating with my new TSRI colleagues.”

Ye received his bachelor’s degree from Tsinghua University in Beijing and earned his Ph.D. at Harvard Medical School in Professor Bruce Spiegelman’s laboratory. He conducted his postdoctoral training in the lab of Professor Karl Deisseroth at Stanford University.

Adibekian and Ye will officially join TSRI’s ranks on November 1 and January 5, respectively.

Scripps Florida Scientists Identify Compounds That Significantly Increase Healthy Aging

JUPITER, FL – September 4, 2017 – Aging happens to everyone, but it may no longer mean a descent into disease or chronic disability. Recently, scientists from the Florida campus of The Scripps Research Institute (TSRI) identified a class of compounds, using animal models and human cells, that significantly delays the onset of several age-related symptoms and extends healthy aging—what has come to be known as “healthspan.”

The study, published in the journal Nature Communications, was led by TSRI Professor Paul Robbins and Associate Professor Laura Niedernhofer.

This newly identified class of drugs targets senescent cells—cells that have stopped replicating because of chromosome damage. As we get older, senescent cells accumulate, becoming a major contributor to age-related diseases.

August 30, 2017 79

Robbins and his colleagues developed a new screening platform to look for drugs that specifically affect senescent cells. They identified a class of compounds that inhibits a stabilizing protein as having significant anti-senescent activity.

“Our study documents that you can identify compounds that kill these damaged cells specifically,” Robbins added. “It also proves that this class of drugs can significantly extend healthspan.”

Animal models treated with one of these inhibitors reduced or delayed onset of multiple age- related symptoms, including osteoporosis and frailty, which led to a major improvement in overall healthspan. The inhibitors blocked the activity of a protein that plays a key role in survival of senescent cells, which led to their apoptosis or programmed cell death.

Although these inhibitors have anti-senescent activity in cell culture and in animal models, it is likely that they will be more effective in combination with other FDA-approved drugs that target other senescent cell pathways. More effective drug combinations, including ones with these inhibitors, could be used to extend healthspan in humans, according to the scientists.

“Our screening platform already has identified multiple classes of compounds that extend healthspan in animal models,” Robbins said. “Right now, we have a pipeline of compounds that should be even more effective than the ones in the study.”

Anti-senescent drugs may also prove useful in delaying, preventing, or treating age-related chronic diseases, as well as other disorders related to increased senescent cell accumulation, such as obesity with metabolic syndrome and osteoarthritis. Cancer survivors treated with irradiation or chemotherapy could also benefit.

“We found that there are at least two main groups of drugs that work as anti-senescent therapeutics: one that suppresses the senescent state, termed senomorphics, and one that kills senescent cells, termed senolytics,” said Heike Fuhrmann-Stroissnigg, the first author of the study. “So far, most of the drugs identified belong to this latter group—and some of these drugs are used as anti-cancer drugs too.”

In addition to Robbins, Niedernhofer and Fuhrmann-Stroissnigg, other authors of the study, “Identification of HSP90 Inhibitors as a Novel Class of Senolytics,” are Yuan Yuan Ling, Jing Zhao, Sara J. McGowan, Robert W. Brooks, Akaitz Dorronsoro, Lana Corbo, Priscilla Tang, Christina Bukata, Diego Grassi and Xuesen Li of TSRI; Siobhan Q. Gregg and Jennifer L. Stripay of the University of Pittsburgh School of Medicine; Yi Zhu, Tamara Tchkonia and James L. Kirkland of the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota; and Nadja Ring and Mauro Giacca of the International Centre for Genetic Engineering and Biotechnology, Trieste, Italy.

August 30, 2017 80 The study was supported by the National Institutes of Health (grant numbers AG043376, AG13925, DK50456), the Connor Group; the Noaber, Ted Nash, and Glenn Foundations; the American Federation for Aging Research; and Aldabra Biosciences.

Scripps Florida Scientists Confirm Quality Control Mechanism to Protect Protein Production

JUPITER, FL – September 7, 2017 – The devil, as always, is in the details. The production of proteins is a complex process, made up of a series of smaller intricate biochemical steps that succeeds because of an equally complex number of quality controls. Without them, the process would irreparably break down and diseases like cancer would run rampant.

Scientists from the Florida campus of The Scripps Research Institute (TSRI) have shown for the first time that the assembly of the cellular factories that produce proteins, the basic working units of any cell, also involves quality control steps.

The study, which was published September 7 online ahead of print in the journal Molecular Cell, was led by Katrin Karbstein, an associate professor in the Department of Integrative Structural and Computational Biology.

“With important cellular machines like ribosomes, it makes sense that mechanisms exist to make sure things work correctly,” Karbstein said. “We’ve confirmed for the first time that such a quality control function exists for these small ribosomal subunits that, in essence, do a test run but don’t produce a protein – without it, mistakes in RNA translation would produce dangerous mutated proteins.”

Ribosomes, which are large macromolecular machines required for cell growth in all organisms, catalyze the production of proteins in all cells. They read the genetic code carried by messenger RNA (mRNA), and then catalyze or translate that code into proteins within cells, assembling them from amino acids.

Understanding the process of ribosome assembly—which involves almost 200 essential proteins known as "assembly factors" in addition to the four RNA molecules and 78 ribosomal proteins that are part of the mature ribosome—is a potentially fruitful area of research because of the importance of ribosome assembly for cell growth.

“We used genetic and biochemical experiments to show that bypassing this step in translation- like cycle produces defects,” said Homa Ghalei, the first author of the study. “Our results show that this cycle is a quality control mechanism that ensures the fidelity of the cellular ribosome pool.”

The link between defects in ribosome assembly and cancer clearly points to this pathway as a new target for anti-cancer drugs. Ribosomes are the targets of many commercially used antibiotics and represent a promising area of research because of the importance of ribosome assembly and function for cell growth.

August 30, 2017 81

“We now understand how this complicated assembly process works and we can finally show that this really is a quality control mechanism – a significant advance over a discovery we made back in 2012, which we were never able to prove,” Karbstein said. “But even then, it seemed to be the most logical conclusion.”

The subunits in question are known as 40S ribosomal subunits that bind to other subunits to produce larger ribosomes – called 80S-like ribosomes because they do not produce any proteins.

In addition to Karbstein and Ghalei, other authors of the study, “The ATPase Fap7 Tests the Ability to Carry Out Translocation-like Conformational Changes and Releases Dim1 during 40S Ribosome Maturation,” are Juliette Trepreau, Jason C. Collins, Hari Bhaskaran, and Bethany S. Strunk, all of TSRI.

The study was supported by the National Institutes of Health (Grant R01-GM086451), the Howard Hughes Medical Institute and PGA National Gold Club.

Outside-In Reprogramming: Antibody Study Suggests a Better Way to Make Stem Cells

LA JOLLA, CA—Scientists at The Scripps Research Institute (TSRI) have found a new approach to the “reprogramming” of ordinary adult cells into stem cells.

In a study published today in an Advance Online paper in Nature Biotechnology, the TSRI scientists screened a library of 100 million antibodies and found several that can help reprogram mature skin-like cells into stem cells known as induced pluripotent stem cells (IPSCs).

Making IPSCs from more mature types of cells normally involves the insertions of four transcription factor genes into the DNA of those cells. The antibodies identified by the scientists can be applied to mature cells—where they bind to proteins on the cell surface—as a substitute for three of the standard transcription factor gene-insertions.

“This result suggests that ultimately we might be able to make IPSCs without putting anything in the cell nucleus, which potentially means that these stem cells will have fewer mutations and overall better properties,” said study senior author Kristin Baldwin, associate professor in TSRI’s department of neuroscience.

IPSCs can be made from patients’ own cells, and have a multitude of potential uses in personalized cell therapies and organ regeneration. However, none of IPSCs’ envisioned clinical uses has yet been realized, in part because of the risks involved in making them.

The standard IPSC induction procedure, developed a decade ago and known as OSKM, involves the insertion into adult cells of genes for four transcription factor proteins: Oct4, Sox2, Klf4 and c-Myc. With these genes added and active, the transcription factor proteins they encode are produced and in turn reprogram the cells to become IPSCs.

August 30, 2017 82 One problem with this procedure is that the viral insertion events or overproduction of the nuclear reprogramming factors may damage cell DNA in a way that turns the cell cancerous. Another is that this nuclear reprogramming typically yields a collection of IPSCs with variable properties. “This variability can be a problem even when we’re using IPSCs in the laboratory for studying diseases,” Baldwin said.

In contrast, during ordinary animal development, cell identity is altered by molecular signals that come in from outside the cell and induce changes in gene activity, without any risky insertions of DNA. To find natural pathways like these—through which ordinary cells could be turned into IPSCs—Baldwin and her laboratory teamed up with the TSRI laboratory of Richard Lerner, the Lita Annenberg Hazen Professor of Immunochemistry. Lerner has helped pioneer the development and screening of large libraries of human antibodies for finding new antibody- based drugs and scientific probes.

In this case, the team, including graduate student Joel W. Blanchard and postdoctoral research associate Jia Xie, who were lead authors, set up a library of about 100 million distinct antibodies and used it to find any that could substitute for OSKM transcription factors. In an initial set of experiments, the researchers tried to identify antibodies that can replace both Sox2 and c-Myc. They established a large population of mouse fibroblast cells—often used to make IPSCs in experiments—and inserted the genes for the other two transcription factors, Oct4 and Klf4. Next they added their huge library of antibody genes to the population of cells, such that each cell ended up containing the genes for one or more of the antibodies.

The scientists could then observe which of the cells began forming stem cell colonies— indicating that one of the antibodies produced by those cells had successfully replaced the functions of Sox2 and c-Myc and triggered the switch in cell identity. Sequencing the DNA of these cells allowed the researchers to determine the antibodies responsible.

In this way, the TSRI team discovered two antibodies that can be substituted for both Sox2 and c-Myc, and in a similar set of tests they found two antibodies that can replace a third transcription factor, Oct4. The scientists showed that instead of inserting these transcription factor genes they could simply supply the antibodies to the fibroblast cells in culture.

In this initial study, the scientists were unable to find antibodies that replace the function of the fourth OSKM transcription factor, Klf4. However, Baldwin expects that with more extensive screening she and her colleagues eventually will find antibody substitutes for Klf4 as well. “That one I think is going to take us a few more years to figure out,” she said.

The antibody-screening approach in principle allows scientists not only to find antibodies that can replace OSKM transcription factors, but also to study the natural signaling pathways through which these antibodies work.

In a proof of this principle, the scientists found that one of the Sox2-replacing antibodies binds to a protein on the cell membrane called Basp1. This binding event blocks Basp1’s normal activity and thus removes the restraints on WT1, a transcription factor protein that works in the cell nucleus. WT1, unleashed, then alters the activity of multiple genes, ultimately including

August 30, 2017 83 Sox2’s, to promote the stem cell state using a different order of events than when using the original reprogramming factors.

WT1 (Wilms tumor 1) is overproduced in some cancers and is considered an oncogene. That fact highlights an added value of such studies: to help scientists understand the relationship between cancer cell development and the stem cell state.

The TSRI researchers now plan larger, more complex antibody-screening studies using human cells rather than mouse cells.

Other co-authors of the paper, “Replacing reprogramming factors with antibodies selected from combinatorial antibody libraries,” were Nadja El-Mecharrafie, Simon Gross, and Sohyon Lee. The research was supported by the National Institute on Deafness and other Communication Disorders (DC012592), the National Institute of Mental Health (MH102698), the California Institute for Regenerative Medicine (RB3-02186), the Baxter Family and the Norris and Del Webb Foundations.

Scripps Florida Scientist Awarded $2.5 Million Collaborative Grant to Develop New Diabetes Treatment

JUPITER, FL – September 19, 2017 – Patrick R. Griffin, co-chair of the Department of Molecular Medicine on the Florida campus of The Scripps Research Institute (TSRI), has been awarded a $2.5 million collaborative grant with Brigham and Women’s Hospital by the National Institute of Diabetes and Digestive and Kidney Diseases, part of the National Institutes of Health (NIH).

The new five-year study seeks to confirm whether the inhibition of a particular protein that plays a role in regulating the body’s response to fat might be a viable target in the treatment of type 2 diabetes.

Griffin and Jorge Plutzky, MD, from Brigham and Women’s Hospital in Boston are co- principal investigators for the five-year multi-PI grant. TSRI’s Theodore Kamenecka, an associate professor in the Department of Molecular Medicine, is a co-investigator on the study.

“This new grant will let us focus on several lead candidates that inhibit a particular protein to confirm that this is a potential target to treat type 2 diabetes,” Griffin said. “These lead compounds came from a high-throughput screen run at Scripps Florida. Our strong preliminary evidence supports the idea of targeting fat directly.”

Griffin’s work covers a wide area of diseases ranging from cancer to diabetes to age-related bone loss. But they are all characterized by their impact on human metabolism – and the galaxy of disorders that more often than not occur in clusters: obesity, abnormal cholesterol, high blood pressure and insulin resistance.

“What we are looking for – what we’ve always looked for – is using chemical biology approaches to create novel therapeutics for the treatment of metabolic disease,” Griffin said.

August 30, 2017 84 “Our research efforts often complement others because in many metabolic syndromes, like obesity and diabetes, for example, the underlying aspects of the disorders overlap.”

What makes the new grant significant – and something of a milestone – is that Griffin now has five similar grants running simultaneously, several in collaboration with other institutes.

“Four of the five are highly collaborative, something we’re very good at – both within TSRI and with other research organizations,” he said. “It’s been one of the hallmarks of our research program.”

The latest study with Brigham and Women’s Health will focus on what is known as brown adipose tissue or brown fat, which serves as a kind of molecular furnace in humans, generating heat by burning large numbers of fat calories. Brown fat is loaded with mitochondria, the cell’s energy plant, which contain iron and give the fat its red-brown tint; mitochondria use nutrients to produce energy for the cell.

Brown fat is different than white adipose tissue (WAT), which stores energy and can lead to obesity. Studies have shown that brown fat characteristics can be induced in white fat through the inhibition of the protein retinaldehyde dehydrogenase 1 (ALDH1a1), which in turn protects against diet-induced obesity and diabetes.

The number of the grant is 1R01DK107239-01A1.

TSRI Researchers Explore Ways That a Drug Like Avandia Can Be Made Safer

JUPITER, FL – September 28, 2017 – With the heightened concerns over the dangerous side effects of the once-popular antidiabetic drug Avandia, researchers at The Scripps Research Institute (TSRI) in Jupiter, Florida, are working to understand how small molecules, like those in Avandia, can have such varied effects throughout the body. The insights could help researchers design new drugs with better efficacy and fewer side effects.

Douglas Kojetin, an associate professor at TSRI, and his team recently published a study in Structure, showing the ways that Avandia interacts with and changes the shape of a combination of proteins, receptors and DNA — called the “complex” — resulting in the drug’s effects. In addition to helping inform the design of future antidiabetic drugs, the study revealed that DNA plays an active role in determining the structure of the complex, a finding that has implications for understanding how any small molecule drug affects the body.

Researchers in Kojetin’s lab focus on nuclear receptors—proteins that can interact or “bind” to both genetic material, such as DNA and small molecules, as well as bind to other proteins called coregulatory proteins that impact gene expression. Avandia, for example, binds to PPARγ, a nuclear receptor, which binds to DNA sequences important in regulating fat storage and metabolism. But PPARγ does not work with Avandia alone. A second nuclear receptor, RXRα, interacts with PPARγ to form a “heterodimer”, a complex made up of two different proteins bound together that recruits a co-regulatory protein called Steroid Receptor

August 30, 2017 85 Coactivator-2 (SRC-2) to influence the activity of many different genes. As each piece binds, the complex changes shape, like pieces of wet clay mashing together to form a bowl.

The team set out to understand how specific sections of DNA affect the interaction between the nuclear receptor and coregulatory protein. For example, would this nuclear receptor interact with the coregulatory protein the same way if it weren’t bound to DNA?

Adding an extra layer of complexity, SRC-2 is an intrinsically disordered protein—a “floppy” protein without a consistent secondary structure that flops around like a loose spaghetti noodle. This means that popular methods that researchers would typically use to understand the protein’s structure, like x-ray crystallography, which requires a stable unfloppy sample, wouldn’t be able to tell the scientists very much about what this protein is doing.

Kojetin’s team used a combination of quantitative biochemical, biophysical and solution structural methods to form a detailed understanding of these molecular interactions. Each technique gave his team pieces of information that they used to build a picture of how all of these molecules interact.

“No one tool could have given us the answer. It was really the combination of all these tools that gave us a full picture of what was going on,” Kojetin said.

It turned out that what was going on was a “thermodynamic” mechanism by which binding to DNA caused the receptor heterodimer to change its shape and importantly stabilize its floppy regions.

Kojetin’s team showed that DNA interaction impacted the potency of Avandia and its ability to recruit the coregulatory protein. The receptors on their own are like two balloons randomly floating around in the wind, explained Kojetin. When it bound to the DNA, the receptors become stabilized, as if the balloons were tied down together, making it easier for SRC-2 to interact with it.

If the proteins had bound to another portion of DNA, the complex may have been able to form a different shape and exert a different, possibly even dangerous effect, or perhaps no effect at all. Different genes are exposed, or accessible, in different bodily tissues. This could help explain how drugs can have one effect in a certain area of the body and a different effect in other areas where the sections of accessible DNA are different.

“The drug does not control activity in the body by itself. When the receptor is bound to different DNA sequences, the activity of the drug could be changed.” Knowing this, said Kojetin, “is going to open up a lot of possibilities.”

In addition to Kojetin, other authors of the study titled, “Synergistic Regulation of Coregulator/Nuclear Receptor Interaction by Ligand and DNA,” include first author Ian de Vera, as well as Jie Zheng, Scott Novick, Jinsai Shang, Travis Hughes, Richard Brust, Paola Munoz-Tello, William Gardner, Jr., David Marciano, Xiangming Kong and Patrick Griffin – all of whom work at the Scripps Florida campus.

August 30, 2017 86 The study was supported in part by the James and Esther King Biomedical Research Program, Florida Department of Health (1KN-09); the William R. Kenan, Jr. Charitable Trust (TSRI High School Student Summer Internship Program); National Institutes of Health (NIH) grants DK101871 (DJK), DK105825 (PRG), F32DK097890 (TH), K99DK103116 (TH) and F32DK108442 (RB); and American Heart Association (AHA) fellowship awards 12POST12050025 (TH) and 16POST27780018 (RB).

HIVE Center Receives $27 Million in NIGMS Funding to Advance HIV Research

October 04, 2017 – The HIV Interactions in Viral Evolution (HIVE) Center at The Scripps Research Institute (TSRI) has been awarded nearly $27 million from the National Institute of General Medical Sciences (NIGMS) of the National Institutes of Health (NIH). This continues the HIVE Center’s crucial work for another five years studying at the atomic level the human immunodeficiency virus (HIV), which causes AIDS.

Viruses have complex interactions with the human host cells they infect, especially HIV given its capacity for utilizing human cell proteins, its ability to rapidly mutate to both immune and viral inhibitor-mediated pressures, and remain dormant in infected cells, noted HIVE Center Co-Director, Associate Professor Bruce Torbett. For the past five years, the NIGMS has funded five National Centers, of which the HIVE Center is one, dedicated to understanding how HIV functions in the body’s immune cells and utilizes human cellular components to evolve, adapt and evade treatment.

The Center’s highly collaborative researchers will continue to build on their past success of the initial HIVE funding period under Professor Arthur Olson. In the next five years, the goal of the Center’s researchers is to obtain at the atomic level an understanding of the structural and dynamic relationships between interacting human host cell and HIV macromolecules in the HIV life cycle. These insights will shed light as to how HIV usurps human host cell proteins to move through the cell undetected and how the virus utilizes host cell proteins to insert its DNA into cellular chromatin. Genomic and structural studies on the development of HIV inhibitor resistance should provide insights for HIV inhibitor resistance prediction and novel therapies. The information obtained from all studies will help to develop computational models to visualize HIV, which should allow an unprecedented view as to how HIV is formed and functions within host cells according to Torbett.

The Center includes scientists from both the California and Florida campuses of TSRI. On the California campus, Arthur Olson, K. Barry Sharpless, James Williamson, David Millar, Stefano Forli and David Goodsell, and on the Florida campus, Patrick Griffin and Douglas Kojetin.

Other scientists from across the country and abroad also will participate as part of the Center research effort which includes Stefan Sarafianos (Co-Director with Torbett) of Emory University, Karin Musier-Forsyth of Ohio State University, Dmitry Lyumkis of The Salk Insitute, Edward Arnold of Rutgers University, Ronald Levy of Temple University, Jason Okulicz of the Uniformed Services University of the Health Sciences, Joseph Marcotrigiano of National Institutes of Health, Alan Engelman of the Dana Farber Cancer Institute, Mamuka

August 30, 2017 87 Kvaratskhelia of University of Colorado Denver, Michael Malim of King’s College, London and Stephen Hughes of the National Cancer Institute-Frederick.

More details on the activities and resources of the HIVE Center can be found at: http://hivecenter.org.

August 30, 2017