The Neonatal Anti-Viral Response Fails to Control Measles Virus Spread in Neurons Despite Interferon- Gamma Expression and a Th1-Like Cytokine Profile Priya Ganesan

Total Page:16

File Type:pdf, Size:1020Kb

The Neonatal Anti-Viral Response Fails to Control Measles Virus Spread in Neurons Despite Interferon- Gamma Expression and a Th1-Like Cytokine Profile Priya Ganesan Duquesne University Duquesne Scholarship Collection Electronic Theses and Dissertations Fall 1-1-2017 The neonatal anti-viral response fails to control measles virus spread in neurons despite interferon- gamma expression and a Th1-like cytokine profile Priya Ganesan Follow this and additional works at: https://dsc.duq.edu/etd Part of the Developmental Neuroscience Commons, Immunology and Infectious Disease Commons, and the Virology Commons Recommended Citation Ganesan, P. (2017). The neonatal anti-viral response fails to control measles virus spread in neurons despite interferon-gamma expression and a Th1-like cytokine profile (Doctoral dissertation, Duquesne University). Retrieved from https://dsc.duq.edu/etd/ 230 This One-year Embargo is brought to you for free and open access by Duquesne Scholarship Collection. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of Duquesne Scholarship Collection. For more information, please contact [email protected]. THE NEONATAL ANTI-VIRAL RESPONSE FAILS TO CONTROL MEASLES VIRUS SPREAD IN NEURONS DESPITE INTERFERON-GAMMA EXPRESSION AND A TH1-LIKE CYTOKINE PROFILE A Dissertation Submitted to the Graduate School of Pharmaceutical Sciences Duquesne University In partial fulfillment of the requirements for the degree of Doctor of Philosophy By Priya Ganesan December 2017 Copyright by Priya Ganesan 2017 THE NEONATAL ANTI-VIRAL RESPONSE FAILS TO CONTROL MEASLES VIRUS SPREAD IN NEURONS DESPITE INTERFERON-GAMMA EXPRESSION AND A TH1-LIKE CYTOKINE PROFILE By Priya Ganesan Approved November 12, 2017 Lauren A. O’Donnell, Ph.D. Paula A. Witt-Enderby, Ph.D. Associate Professor of Pharmacology Professor of Pharmacology Graduate School of Pharmaceutical Sciences Graduate School of Pharmaceutical Sciences Duquesne University, Pittsburgh, PA Duquesne University, Pittsburgh, PA (Committee Chair) (Committee Member) Wilson S. Meng, Ph.D. Dr. Christopher Surratt Associate Professor of Pharmaceutics Professor of Pharmacology Graduate School of Pharmaceutical Sciences Graduate School of Pharmaceutical Sciences Duquesne University, Pittsburgh, PA Duquesne University, Pittsburgh, PA (Committee Member) (Committee Member) Dr. Kerry Empey James K. Drennen, III, Ph.D. Assistant Professor Associate Dean and Associate Professor Graduate School of Pharmaceutical Sciences of Pharmaceutics University of Pittsburgh, Pittsburgh, PA Duquesne University Graduate School of (Committee Member) Pharmaceutical Sciences J. Douglas Bricker, Ph.D. Dean, Duquesne University School of Pharmacy and the Graduate School of Pharmaceutical Sciences iii ABSTRACT THE NEONATAL ANTI-VIRAL RESPONSE FAILS TO CONTROL MEASLES VIRUS SPREAD IN NEURONS DESPITE INTERFERON-GAMMA EXPRESSION AND A TH1-LIKE CYTOKINE PROFILE By Priya Ganesan December 2017 Dissertation supervised by Dr. Lauren A. O’Donnell Neonates are highly susceptible to infections in the central nervous system (CNS) and have a greater risk of viral infections and encephalopathies. Neurotropic viral infections can lead to blindness, hearing loss and neurological deficiencies such as cognitive impairment, epilepsy, and even death in the neonatal and pediatric populations. Viral infections also are hypothesized to indirectly contribute to neurodegenerative and neuropsychiatric diseases such as Schizophrenia and Parkinson’s disease later in life due to early neuronal damage or stress. Many diverse viruses are capable of invading the neonatal CNS including Borna Disease Virus, Coxsackievirus (CV), Herpes simplex viruses (HSV), and measles virus. Although, we understand that many viruses can cause CNS disease, the mechanisms of viral pathogenesis in the brain and the character of the neonatal anti-viral immune response are not well understood. However, it is hypothesized that neurological damage results from the combined effect of the virus and the immune response. iv Therefore, it is critical to develop immune-mediated strategies to promote viral clearance from the CNS while preventing neuronal damage or loss. This remains a challenge during neonatal CNS infections because of the uniquely immature nature of the neonatal immune system and the sensitivity of developing neurons to inflammation. In order to better understand how the neonatal immune response behaves in the brain, we use neurotropic measles virus (MV) as a model to understand the deficits in the neonatal immunity. Measles is a single-stranded, negative-sense RNA virus that is highly contagious in humans. Typical infection involves inhalation of infected respiratory droplets, infection of dendritic cells and macrophages in the respiratory tract resulting in transient immunosuppression, and a characteristic fever and rash. However, in some cases, MV also causes severe neurological diseases such as Post-infectious encephalomyelitis (PIE), Subacute sclerosing panencephalitis (SSPE), and Measles inclusion body encephalitis (MIBE). Currently, there is no cure for these MV-related neurological conditions, which occur overwhelmingly in newborns and children. Thus, the goal of this project is to define how neonatal immunity responds to MV infection in the unique microenvironment of the brain. The role of interferon-gamma (IFN), a key anti-viral cytokine in controlling adult CNS infections, was explored during a neuronally-restricted MV infection in the neonatal brain. We hypothesized that neonatal mice would be deficient in either IFN production or in the infiltration of IFN-producing immune cells in the brain. In order to address this question, we utilized the CD46+ mouse model, in which the human CD46+ receptor for MV is expressed only in mature neurons of the CNS. We explored the differences in the neonatal immune response, where the mice succumb to MV infection, and compared that to the CD46+ adults, which successfully control MV and survive. Our findings suggest that IFN, which is critical for viral control and survival in v adults, only delays mortality in CD46+ neonates. The neonatal brains also show the infiltration of natural killer cells, neutrophils, infiltrating monocytes and T cells in an IFN-independent manner, all of which are capable of contributing to the IFN pool. However, neonates and adults differentially express pathogen recognition receptors (e.g. Toll-like receptors) and Type I interferons during infection in the CNS, which suggests that the initial recognition of the virus by the immune system may differ in an age-dependent manner. Both neonates and adults expressed IFN, CXCL10, IL-1, and IL-1RA, among other cytokines/chemokines. Regardless, CD46+ neonates succumb to infection despite mounting a Th1-like, but apparently defective, inflammatory response. We further explored whether there are age-dependent differences in IFN signalling given that both ages of mice expressed this critical cytokine. Both neonatal and adult CD46+ mice express similar levels of IFN but only adults show robust induction of the IFN- responsive genes CIITA and CXCL9. This suggests that IFN signaling may be defective in the induction of IFN-responsive genes in neonates compared to adults. To dissect the role of individual components of the immune system, we utilized CD46+ mice crossed to specific immune knockouts: CD46+/IFN-KO mice, which lack IFN, and CD46+/RAG2-KO mice, which lack mature B and T cells. We found that neonates lacking IFN succumbed more rapidly than wildtype CD46+ mice, while neonates lacking mature B and T cells showed delayed morbidity and mortality. Neonates without IFN show high infiltration of neutrophils and inflammatory monocytes but similar numbers of NK cell infiltration compared to CD46+ neonates. CD46+/IFN-KO neonatal brains also show high infiltration of CD4 and CD8 T cells at the later stages of MV infection. Additionally, IFNα is significantly upregulated in the absence of IFN in the brain post-MV infection. Thus, compensatory cytokines and high immune vi cell infiltration may contribute to uncontrolled inflammation and earlier death in CD46+/IFN-KO neonates. CD46+ mice deficient in T-cells and B-cells (CD46+/RAG2-KO) show prolonged survival and mount a robust IFNβ response post-MV infection. This suggests that the adaptive immune response may be detrimental during the neonatal period, potentially leading to greater tissue damage. Additionally, CD46+/RAG2-KO neonates alone upregulate unique genes such as bone morphogenetic proteins (BMPs), which may mediate neuroprotection. Thus, these results suggest age-dependent expression of cytokine profiles in the brain and distinct dynamic interplays between lymphocyte populations and cytokines/chemokines in MV-infected neonates. vii DEDICATION To my parents, Savithri and Ganesan Rajamani, and my sister Gowri Ganesan whose continuous support, sacrifices, love and strength throughout my life made this possible. viii ACKNOWLEDGEMENTS I thank Dr. Lauren A. O’Donnell, for giving me the opportunity to purse research in her lab, for her constant support, enthusiasm, and for being an amazing mentor. Your advice and patience have been pivotal to my journey as a researcher. I am indebted for all the lessons I learnt from you during this journey. I would like to thank my committee members Drs. Paula Witt-Endery, Wilson Meng, Chris Surratt, and Kerry Empey for their support and invaluable guidance. I want to especially thank Dr. Witt-Enderby and Dr. Meng for continuous encouragement during tough times and job search. I would like to thank Sanket Anaokar and Harsha Sree Pulugulla for their help with qRT-PCR and PCR. I also
Recommended publications
  • Replication by Iga Intraepithelial Cell Neutralization of HIV-1
    Intraepithelial Cell Neutralization of HIV-1 Replication by IgA Yung T. Huang, Alison Wright, Xing Gao, Lesya Kulick, Huimin Yan and Michael E. Lamm This information is current as of October 1, 2021. J Immunol 2005; 174:4828-4835; ; doi: 10.4049/jimmunol.174.8.4828 http://www.jimmunol.org/content/174/8/4828 Downloaded from References This article cites 57 articles, 24 of which you can access for free at: http://www.jimmunol.org/content/174/8/4828.full#ref-list-1 Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on October 1, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2005 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Intraepithelial Cell Neutralization of HIV-1 Replication by IgA1 Yung T. Huang,2*† Alison Wright,* Xing Gao,* Lesya Kulick,* Huimin Yan,3* and Michael E. Lamm* HIV is transmitted sexually through mucosal surfaces where IgA Abs are the first line of immune defense.
    [Show full text]
  • Characterization of the Relationship Between Measles Virus Fusion
    University of Massachusetts Medical School eScholarship@UMMS GSBS Dissertations and Theses Graduate School of Biomedical Sciences 2006-05-17 Characterization of the Relationship Between Measles Virus Fusion, Receptor Binding, and the Virus-Specific Interaction Between the Hemagglutinin and Fusion Glycoproteins: a Dissertation Elizabeth Ann Corey University of Massachusetts Medical School Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/gsbs_diss Part of the Amino Acids, Peptides, and Proteins Commons, Cells Commons, Chemical Actions and Uses Commons, Lipids Commons, and the Virus Diseases Commons Repository Citation Corey EA. (2006). Characterization of the Relationship Between Measles Virus Fusion, Receptor Binding, and the Virus-Specific Interaction Between the Hemagglutinin and Fusion Glycoproteins: a Dissertation. GSBS Dissertations and Theses. https://doi.org/10.13028/hg2y-0r35. Retrieved from https://escholarship.umassmed.edu/gsbs_diss/221 This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in GSBS Dissertations and Theses by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. CHARACTERIZATION OF THE RELATIONSHIP BETWEEN MEASLES VIRUS FUSION , RECEPTOR BINDING , AND THE VIRUS-SPECIFIC INTERACTION BETWEEN THE HEMAGGLUTININ AND FUSION GL YCOPROTEINS A Dissertation Presented Elizabeth Anne Corey Submitted to the Faculty of the University of Massachusetts Graduate
    [Show full text]
  • The Accessory Glycoprotein Gp3 of Canine Coronavirus Type 1
    The accessory glycoprotein gp3 of canine Coronavirus type 1 : investigations of sequence variability in feline host and of the basic features of the different variants Anne-Laure Pham-Hung d’Alexandry d’Orengiani To cite this version: Anne-Laure Pham-Hung d’Alexandry d’Orengiani. The accessory glycoprotein gp3 of canine Coro- navirus type 1 : investigations of sequence variability in feline host and of the basic features of the different variants. Virology. Université Paris Sud - Paris XI, 2014. English. NNT : 2014PA114831. tel-01089374 HAL Id: tel-01089374 https://tel.archives-ouvertes.fr/tel-01089374 Submitted on 1 Dec 2014 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. UNIVERSITÉ PARIS-SUD 11 ECOLE DOCTORALE : INNOVATION THÉRAPEUTIQUE : DU FONDAMENTAL A L’APPLIQUÉ PÔLE : MICROBIOLOGIE / THERAPEUTIQUES ANTIINFECTIEUSES DISCIPLINE : VIROLOGIE ANNÉE 2013 - 2014 SÉRIE DOCTORAT N° 1296 THÈSE DE DOCTORAT soutenue le 24/10/2014 par Anne-Laure Pham-Hung d’Alexandry d’Orengiani The accessory glycoprotein gp3 of canine Coronavirus type 1 : investigations
    [Show full text]
  • APICAL M2 PROTEIN IS REQUIRED for EFFICIENT INFLUENZA a VIRUS REPLICATION by Nicholas Wohlgemuth a Dissertation Submitted To
    APICAL M2 PROTEIN IS REQUIRED FOR EFFICIENT INFLUENZA A VIRUS REPLICATION by Nicholas Wohlgemuth A dissertation submitted to Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland October, 2017 © Nicholas Wohlgemuth 2017 All rights reserved ABSTRACT Influenza virus infections are a major public health burden around the world. This dissertation examines the influenza A virus M2 protein and how it can contribute to a better understanding of influenza virus biology and improve vaccination strategies. M2 is a member of the viroporin class of virus proteins characterized by their predicted ion channel activity. While traditionally studied only for their ion channel activities, viroporins frequently contain long cytoplasmic tails that play important roles in virus replication and disruption of cellular function. The currently licensed live, attenuated influenza vaccine (LAIV) contains a mutation in the M segment coding sequence of the backbone virus which confers a missense mutation (alanine to serine) in the M2 gene at amino acid position 86. Previously discounted for not showing a phenotype in immortalized cell lines, this mutation contributes to both the attenuation and temperature sensitivity phenotypes of LAIV in primary human nasal epithelial cells. Furthermore, viruses encoding serine at M2 position 86 induced greater IFN-λ responses at early times post infection. Reversing mutations such as this, and otherwise altering LAIV’s ability to replicate in vivo, could result in an improved LAIV development strategy. Influenza viruses infect at and egress from the apical plasma membrane of airway epithelial cells. Accordingly, the virus transmembrane proteins, HA, NA, and M2, are all targeted to the apical plasma membrane ii and contribute to egress.
    [Show full text]
  • The Nucleoprotein and Phosphoprotein of Peste Des Petits Ruminants Virus Inhibit Interferons Signaling by Blocking the JAK-STAT Pathway
    viruses Article The Nucleoprotein and Phosphoprotein of Peste des Petits Ruminants Virus Inhibit Interferons Signaling by Blocking the JAK-STAT Pathway 1,2, 2, 2 2 2 2 Pengfei Li y , Zixiang Zhu y, Xiangle Zhang , Wen Dang , Linlin Li , Xiaoli Du , Miaotao Zhang 1, Chunyan Wu 1, Qinghong Xue 3, Xiangtao Liu 2, Haixue Zheng 2,* and Yuchen Nan 1,* 1 Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China 2 State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China 3 China Institute of Veterinary Drug Control, Beijing100081, China * Correspondence: [email protected] (H.Z.); [email protected] (Y.N.) These authors contributed equally to this work. y Received: 17 June 2019; Accepted: 6 July 2019; Published: 8 July 2019 Abstract: Peste des petits ruminants virus (PPRV) is associated with global peste des petits ruminants resulting in severe economic loss. Peste des petits ruminants virus dampens host interferon-based signaling pathways through multiple mechanisms. Previous studies deciphered the role of V and C in abrogating IFN-β production. Moreover, V protein directly interacted with signal transducers and activators of transcription 1 (STAT1) and STAT2 resulting in the impairment of host IFN responses. In our present study, PPRV infection inhibited both IFN-β- and IFN-γ-induced activation of IFN-stimulated response element (ISRE) and IFN-γ-activated site (GAS) element, respectively. Both N and P proteins, functioning as novel IFN response antagonists, markedly suppressed IFN-β-induced ISRE and IFN-γ-induced GAS promoter activation to impair downstream upregulation of various interferon-stimulated genes (ISGs) and prevent STAT1 nuclear translocation.
    [Show full text]
  • Impact of RNA Virus Evolution on Quasispecies Formation and Virulence
    International Journal of Molecular Sciences Review Impact of RNA Virus Evolution on Quasispecies Formation and Virulence Madiiha Bibi Mandary, Malihe Masomian and Chit Laa Poh * Center for Virus and Vaccine Research, School of Science and Technology, Sunway University, Kuala Lumpur, Selangor 47500, Malaysia * Correspondence: [email protected]; Tel.: +60-3-7491-8622; Fax: +60-3-5635-8633 Received: 3 July 2019; Accepted: 26 August 2019; Published: 19 September 2019 Abstract: RNA viruses are known to replicate by low fidelity polymerases and have high mutation rates whereby the resulting virus population tends to exist as a distribution of mutants. In this review, we aim to explore how genetic events such as spontaneous mutations could alter the genomic organization of RNA viruses in such a way that they impact virus replications and plaque morphology. The phenomenon of quasispecies within a viral population is also discussed to reflect virulence and its implications for RNA viruses. An understanding of how such events occur will provide further evidence about whether there are molecular determinants for plaque morphology of RNA viruses or whether different plaque phenotypes arise due to the presence of quasispecies within a population. Ultimately this review gives an insight into whether the intrinsically high error rates due to the low fidelity of RNA polymerases is responsible for the variation in plaque morphology and diversity in virulence. This can be a useful tool in characterizing mechanisms that facilitate virus adaptation and evolution. Keywords: RNA viruses; quasispecies; spontaneous mutations; virulence; plaque phenotype 1. Introduction With their diverse differences in size, structure, genome organization and replication strategies, RNA viruses are recognized as being highly mutatable [1].
    [Show full text]
  • Anti-Measles Virus Monoclonal Antibody, Clone DW2,DW5 (DMABT-H21849) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Anti-Measles virus Monoclonal antibody, Clone DW2,DW5 (DMABT-H21849) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Product Overview Mouse Anti-Measles Blend Monoclonal Antibody Specificity A blend of two monoclonal antibodies that recognize the measles hemagglutinin protein by indirect immunofluorescence. Target Measles virus Immunogen Hemagglutinin protein obtained from Chicago wild type strain of measles Isotype IgG1 Source/Host Mouse Species Reactivity Measles virus Clone DW2,DW5 Conjugate Unconjugated Applications IF, IHC-P Format Ascites Size 100 μl Preservative None Storage Maintain at -20 °C in convenient aliquots for up to 12 months. BACKGROUND Introduction Hemagglutinin (HA) is a class I viral fusion protein from Influenza virus. It is a major glycoprotein, comprising over 80% of the envelope proteins present in the virus particle. HA binds to sialic acid-containing receptors on the cell surface, bringing about the attachment of the virus particle to the cell, and is responsible for penetration of the virus into the cell cytoplasm by mediating the fusion of the membrane of the endocytosed virus particle with the endosomal membrane. The extent of infection into host organism is determined by HA. In natural infection, inactive HA is 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved matured intoIB2and HA2 outside the cell by one or more trypsin-like, arginine-specific endoproteases secreted by the bronchial epithelial cells. The HA protein is a homotrimer of disulfide-linked HA1-HA2.
    [Show full text]
  • (21) International Application Number
    ( (51) International Patent Classification: EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV, C12N 15/86 (2006.0 1) A61K 9/51 (2006.0 1) MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, C07K 14/705 (2006.01) TR), OAPI (BF, BJ, CF, CG, Cl, CM, GA, GN, GQ, GW, KM, ML, MR, NE, SN, TD, TG). (21) International Application Number: PCT/US2020/049087 Declarations under Rule 4.17: (22) International Filing Date: — as to the applicant's entitlement to claim the priority of the 02 September 2020 (02.09.2020) earlier application (Rule 4.17(iii)) — of inventorship (Rule 4.17(iv)) (25) Filing Language: English Published: (26) Publication Language: English — with international search report (Art. 21(3)) (30) Priority Data: — with sequence listing part of description (Rule 5.2(a)) 62/895,454 03 September 2019 (03.09.2019) US 63/056,5 14 24 July 2020 (24.07.2020) US (71) Applicant: SANA BIOTECHNOLOGY, INC. [US/US]; 188 East Blaine Street, Suite 400, Seattle, Washington 98102 (US). (72) Inventors: EMMANUEL, Akinola Olumide; 188 East Blaine Street, Suite 400, Seattle, Washington 98102 (US). ENNAJDAOUI, Hanane; 188 East Blaine Street, Suite 400, Seattle, Washington 98102 (US). JAIN, Suvi; 188 East Blaine Street, Suite 400, Seattle, Washington 98102 (US). RUZO MATIAS, Alberto; 188 East Blaine Street, Suite 400, Seattle, Washington 98 102 (US). SHAH, Jagesh V.; 188 East Blaine Street, Suite 400, Seattle, Washington 98102 (US). (74) Agent: POTTER, Karen et al.; Morrison & Foerster LLP, 1253 1 High Bluff Drive, Suite 100, San Diego, California 92130-2040 (US).
    [Show full text]
  • Bioinformatic and Immunological Analysis Reveals Lack of Support For
    Polymeros et al. BMC Medicine 2014, 12:139 http://www.biomedcentral.com/1741-7015/12/139 RESEARCH ARTICLE Open Access Bioinformatic and immunological analysis reveals lack of support for measles virus related mimicry in Crohn’s disease Dimitrios Polymeros1, Zacharias P Tsiamoulos1*, Andreas L Koutsoumpas2, Daniel S Smyk2, Maria G Mytilinaiou2, Konstantinos Triantafyllou1, Dimitrios P Bogdanos2,3 and Spiros D Ladas4 Abstract Background: A link between measles virus and Crohn’s disease (CD) has been postulated. We assessed through bioinformatic and immunological approaches whether measles is implicated in CD induction, through molecular mimicry. Methods: The BLAST2p program was used to identify amino acid sequence similarities between five measles virus and 56 intestinal proteins. Antibody responses to measles/human mimics were tested by an in-house ELISA using serum samples from 50 patients with CD, 50 with ulcerative colitis (UC), and 38 matched healthy controls (HCs). Results: We identified 15 sets of significant (>70%) local amino acid homologies from two measles antigens, hemagglutinin-neuraminidase and fusion-glycoprotein, and ten human intestinal proteins. Reactivity to at least one measles 15-meric mimicking peptide was present in 27 out of 50 (54%) of patients with CD, 24 out of 50 (48%) with UC (CD versus UC, p = 0.68), and 13 out of 38 (34.2%) HCs (CD versus HC, p = 0.08). Double reactivity to at least one measles/human pair was present in four out of 50 (8%) patients with CD, three out of 50 (6%) with UC (p = 0.99), and in three out of 38 (7.9%) HCs (p >0.05 for all).
    [Show full text]
  • Molecular Requirements of Influenza Virus Hemagglutinin for Site-Specific
    Dissertation zur Erlangung des akademischen Grades d o c t o r r e r u m n a t u r a l i u m (Dr. rer. nat.) im Fach Biologie Eingereicht an der Lebenswissenschaftlichen Fakultät der Humboldt-Universität zu Berlin von Diplom-Biologin Katharina Brett Präsident der Humboldt-Universität zu Berlin Prof. Dr. Jan-Hendrik Olbertz Dekan der Lebenswissenschaftlichen Fakultät Prof. Dr. Richard Lucius Gutachter/Innen: 1. PD Dr. Michael Veit 2. Prof. Dr. Alexander Herrmann 3. Prof. Alexey Zaikin Tag der mündlichen Prüfung: 26.06.2015 Abbreviations ................................................................................................................................. i Zusammenfassung ........................................................................................................................ ii Summary ....................................................................................................................................... iii CHAPTER 1 – Introduction 1.1 Influenza Virus ........................................................................................................................1 1.1.1 Structure and composition of virus particles .............................................................2 1.1.2 Genetic variation between of influenza strains ..........................................................3 1.2 The Influenza spike protein Hemagglutinin 1.2.1 Molecular structure and biosynthesis .........................................................................4 1.2.2 Three-dimensional
    [Show full text]
  • Oncolytic Measles Virus Expressing the Sodium Iodide Symporter to Treat Drug-Resistant Ovarian Cancer Evanthia Galanis1,2, Pamela J
    Published OnlineFirst November 14, 2014; DOI: 10.1158/0008-5472.CAN-14-2533 Cancer Clinical Studies Research Oncolytic Measles Virus Expressing the Sodium Iodide Symporter to Treat Drug-Resistant Ovarian Cancer Evanthia Galanis1,2, Pamela J. Atherton3, Matthew J. Maurer3, Keith L. Knutson4,5, Sean C. Dowdy6, William A. Cliby6, Paul Haluska Jr1, Harry J. Long1, Ann Oberg3, Ileana Aderca2, Matthew S. Block1, Jamie Bakkum-Gamez6, Mark J. Federspiel2, Stephen J. Russell2, Kimberly R. Kalli1, Gary Keeney7, Kah Whye Peng2, and Lynn C. Hartmann1 Abstract Edmonstonvaccinestrainsofmeasles virus(MV)havesignificant other contemporary series. MV receptor CD46 and nectin-4 expres- antitumor activity in mouse xenograft models of ovarian cancer. sion was confirmed by immunohistochemistry in patient tumors. MV engineered to express the sodium iodide symporter gene (MV- Sodium iodide symporter expression in patient tumors after treat- NIS) facilitates localization of viral gene expression and offers a tool ment was confirmed in three patients by 123IuptakeonSPECT/CTs for tumor radiovirotherapy. Here, we report results from a clinical and was associated with long progression-free survival. Immune evaluation of MV-NIS in patients withtaxol- and platinum-resistant monitoring posttreatment showed an increase in effector T cells ovarian cancer. MV-NIS was given intraperitoneally every 4 weeks recognizing the tumor antigens IGFBP2 and FRa,indicatingthat for up to 6 cycles. Treatment was well tolerated and associated with MV-NIS treatment triggered cellular immunity against the patients' promising median overall survival in these patients with heavily tumor and suggesting that an immune mechanism mediating the pretreated ovarian cancer; no dose-limiting toxicity was observed in observed antitumor effect.
    [Show full text]
  • (12) United States Patent (10) Patent N0.: US 6,749,857 B1 Peters Et Al
    US006749857B1 (12) United States Patent (10) Patent N0.: US 6,749,857 B1 Peters et al. (45) Date of Patent: Jun. 15,2004 (54) RECOMBINANT DIMERIC ENVELOPE Primary Examiner—Laurie Scheiner VACCINE AGAINST FLAVIVIRAL Assistant Examiner—Jeffrey S. Parkin INFECTION (74) Attorney, Agent, or Firm—Morrison & Foerster LLP (75) Inventors: Iain D. Peters, BoZeman, MT (US); (57) ABSTRACT Beth-Ann G. Coller, WoluWe Saint Lambert (BE); Michael McDonell, The present invention discloses and claims vaccines Bogart, GA (US); John M. Ivy, College containing, as an active ingredient, a secreted recombinantly Station, TX (US); Kent Harada, produced dimeric form of truncated ?aviviral envelope Honolulu, HI (US) protein. The vaccines are capable of eliciting the production of neutralizing antibodies against ?aviviruses. The dimeric (73) Assignee: Hawaii Biotechnology Group, Inc., forms of truncated ?aviviral envelope protein are formed 1) Aiea, HI (US) by directly linking tWo tandem copies of 80% E in a head to ( * ) Notice: Subject to any disclaimer, the term of this tail fashion via a ?exible tether; 2) via the formation of a patent is extended or adjusted under 35 leucine Zipper domain through the homodimeric association U.S.C. 154(b) by 0 days. of tWo leucine Zipper helices each fused to the carboxy terminus of an 80% E molecule; or 3) via the formation of (21) Appl. No.: 09/376,463 a non-covalently associated four-helix bundle domain (22) Filed: Aug. 18, 1999 formed upon association of tWo helix-turn-helix moieties each attached to the carboxy terminus of an 80% E mol Related US. Application Data ecule.
    [Show full text]