I the Role of SHP-1 in Regulating the T Cell Response to Suppression By

Total Page:16

File Type:pdf, Size:1020Kb

I the Role of SHP-1 in Regulating the T Cell Response to Suppression By i The role of SHP-1 in regulating the T cell response to suppression by regulatory T cells Emily Rose Mercadante Succasunna, NJ M.S., University of Virginia, 2012 B.S., Ursinus College, 2010 A Dissertation presented to the Graduate Faculty of the University of Virginia in Candidacy for the Degree of Doctor of Philosophy Department of Microbiology, Immunology, and Cancer Biology University of Virginia June 2017 ii ABSTRACT The balance between activation of T cells and their suppression by regulatory T cells (Treg) is dysregulated in autoimmune diseases and cancer. In the past decade, evidence has accumulated to suggest that autoimmune diseases not only feature defective Tregs, but also feature T cells that are resistant to suppression by Tregs. On the other hand, in cancer, T cells are unable to mount anti-tumor responses due to the Treg- enriched suppressive microenvironment. Much remains unknown about what regulates this interplay between T cells and Tregs. However, it has become clear that maintaining the tenuous balance between T cell activation and suppression by Tregs is essential for immune homeostasis and prevention of disease. In this work, we investigated the role of the protein tyrosine phosphatase SHP-1, in regulating T cell responses to T cell receptor (TCR) stimulation. SHP-1 has been defined as a negative regulator of TCR signaling based on studies in global SHP-1 knockout mice and immortalized cell lines. We expanded in-depth upon these findings by utilizing two mouse models in which SHP-1 is deleted specifically in T cells and a pharmacological inhibitor of SHP-1. With this approach, we show that SHP-1 limits the responsiveness of T cells to TCR stimulation in a cell-intrinsic manner. Furthermore, we identified a novel function of SHP-1 in regulating the susceptibility of T cells to Treg- mediated suppression. Thus, SHP-1 deficiency rendered naïve CD4+ and CD8+ T cells resistant to Treg-mediated suppression in vitro, and regulated in vivo CD4+ T cell susceptibility to Treg suppression under conditions of homeostatic expansion. Mechanistically, SHP-1-deficient T cells did not mediate resistance to Treg suppression via soluble factors in vitro. We specifically ruled out an influence of IL-4 iii signaling on Treg-resistance in SHP-1-deficient T cells, as previous work had shown that SHP-1-deficient T cells are hyper-sensitive to IL-4 and that IL-4 can induce wildtype T cells to resist suppression. Rather, SHP-1 controlled the activation of the PI3K/Akt pathway, the enhancement of which has been previously linked to the induction of T cell resistance to Treg suppression. Collectively, these data establish SHP-1 as a critical player in setting the threshold downstream of TCR signaling and identify a novel function of SHP-1 as a regulator of T cell susceptibility to Treg-mediated suppression in vitro and in vivo. Thus, SHP-1 and the PI3K/Akt pathway could represent potential immunotherapeutic targets to modulate susceptibility of T cells to Treg suppression. In addition to investigating the role of SHP-1 in T cells, we also preliminarily assessed the regulatory function of SHP-1 in Tregs. Previous work from our lab established that Tregs from global knockout SHP-1 mice were more suppressive than wildtype Tregs. Thus, we generated a mouse model in which SHP-1 is deleted specifically in Tregs to assess the Treg cell-intrinsic function of SHP-1. Preliminary results herein recapitulate the inhibitory role of SHP-1 on Treg function. Thus, SHP-1- deficient Tregs were more potently suppressive than wildtype Tregs. The dual role of SHP-1 in T cells and Tregs provides the possibility of inhibiting SHP-1 in Tregs for treatment of autoimmune disease, and inhibiting SHP-1 in T cells to boost anti-tumor responses. Overall, this work better informs immunotherapeutic strategies for autoimmune disease and cancer, and highlights the importance of targeted approaches to avoid counterproductive systemic effects. iv Acknowledgements I would like to express my gratitude to the many individuals who have provided support throughout the completion of my thesis work. First, I would like to thank my mentor, Dr. Ulrike Lorenz, for allowing me the opportunity to undertake this work and for her scientific insight and advice throughout. I would also like to thank my thesis committee members, Drs. Loren Erickson, Kenneth Tung, Kodi Ravichandran, Vic Engelhard, and Tim Bender for challenging me to think more critically and pushing me to become an expert in my work. I thank the administrative staff of the CIC, especially Peggy Morris and Michelle Butler, who always offered a friendly smile and answers to my questions. I also thank Sandy Weirich for her hard work keeping MIC students on track. Thank you to my friends in the Erickson lab: William Loo, Tim Rosean, Kelly Cox, Christine Coquery, Nekeithia Wade, and Fionna Surette, with whom I shared many frustrations, inspirations, and good conversations. I was so very fortunate to have Jessica Harakal as a lab neighbor. Jessica could be counted on for scientific help, a good laugh, or a hug. I was also lucky to find another Jersey Girl and best friend in Brittany Durgin. I am grateful to all my many friends here- Glen Hirsh, Tim Raines, Jon and Kasia Handing, Sowmya Narayanan, Monica Buckley, Katie Margulieux, Caitlin Naylor, Dylan and Natalie Kennedy, Amber Woods, and Jason True. And thank you to my friend Kari Duck, for her long-distance commiseration while we both completed our doctorates. My dog, April, whom I adopted four years ago, and whose love is too large to contain in one sentence, made sure I did not take things too seriously during my doctoral v work. Most importantly, words cannot express my gratitude for my parents’ support, without which I would not have succeeded in this process. My parents have always had faith in me and allowed me the space to find my own path and pursue my dreams, no matter how challenging. I dedicate this work to them, and their infinite patience and love. vi Table of Contents Title ……………………………………………………………………………...page i Abstract ………………………………………………………………………….page ii Acknowledgements ……………………………………………………………...page iv Table of Contents ………………………………………………………………...page vi List of Abbreviations …………………………………………………………….page vii List of Tables and Figures ………………………………………………………page xiii Chapter 1: Introduction ………………………………………………………….page 1 Chapter 2: Materials and Methods ………………………………………………page 42 Chapter 3: T cells deficient in the tyrosine phosphatase SHP-1 resist suppression by regulatory T cells ………………………………………………………………. page 53 Chapter 4: Additional models of SHP-1 deficiency in T cells: Studies in motheaten and CD4-Cre SHP-1f/f mice, and pharmacological inhibition of SHP-1 via sodium stibogluconate ……………………………………………………………………page 92 f/f f/f Chapter 5: SHP-1-deficient Tregs from dLck-Cre SHP-1 mice and Foxp3-Cre SHP-1 mice are more suppressive than wildtype Tregs …………………………………page 114 Chapter 6: Conclusions, Future Directions, and Significance …………………...page 122 References ………………………………………………………………………..page 151 Appendix A – Review Article ……………………………………………………page 181 vii List of Abbreviations 4-1BB Tumor necrosis factor receptor superfamily member 9 αCD3 anti-CD3 Ab Antibody(s) ACT Adoptive cell transfer Ag Antigen(s) Akt Protein kinase B ANOVA Analysis of variance AP-1 Activator protein 1 APC Antigen-presenting cell(s) BL6 C57BL/6 cAMP cyclic adenosine monophosphate CAR-T Chimeric antigen receptor T cell Casp-3 Caspase-3 Cbl-b/c-Cbl Casitas-B-lineage-lymphoma proteins CFSE Carboxyfluorescein succinimidyl ester CTL(s) Cytotoxic T lymphocyte(s) CTLA-4 Cytotoxic T-lymphocyte-associated protein 4 viii DC(s) Dendritic cell(s) dLck Distal promoter of Lck DN Double negative (CD4-CD8-) thymocyte DP Double positive (CD4+CD8+) thymocyte EAE Experimentally-induced autoimmune encephalomyelitis ELISA Enzyme-linked immunosorbent assay Erk Extracellular-regulated kinases FMO Fluorescence minus one Foxp3 Forkhead box P3 GITR Glucocorticoid-induced TNFR-related protein GVHD Graft-versus-host disease HCV Hepatitis C virus HIV Human immunodeficiency virus HLA Human leukocyte antigen HSV Herpes simplex virus IBD Inflammatory bowel disease ICAM-1 Intracellular adhesion molecule 1 IDO Indoleamine 2,3-dioxygenase ix IFNγ Interferon gamma IL Interleukin i.p. Intraperitoneal i.v. Intravenous IPEX Immunodysregulation polyendocrinopathy enteropathy X-linked syndrome ITAM(s) Immune tyrosine-based activation motif(s) Jak Janus kinase proteins JIA Juvenile idiopathic arthritis KD Knockdown KO Knockout LAG3 Lymphocyte-activation gene 3 LAT Linker for activation of T cells Lck Lymphocyte-specific protein tyrosine kinase LFA-1 Lymphocyte function-associated antigen 1 LN Lymph node(s) MACS Magnetic activated cell sorting me/me motheaten mice x MFI Mean fluorescence intensity MHC I/II Major histocompatibility complex class I or class II mRNA Messenger ribonucleic acid MS Multiple sclerosis mTOR Mechanistic (formerly mammalian) target of rapamycin mut Mutant Nedd4 Neural precursor cell expressed developmentally downregulated protein 4 NFAT Nuclear factor of activated T-cells NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells OVA Ovalbumin OX40 Tumor necrosis factor receptor superfamily member 4 p85 Regulatory subunit of PI3K pAkt phosphorylated Akt PBS Phosphate-buffered saline pDC(s) Plasmacytoid dendritic cell(s) PI3K Phosphoinositide 3-kinase PKCθ Protein kinase C theta PMA Phorbol 12-myristate 13-acetate xi pMHC peptide: Major histocompatibility complex PTEN Phosphatase and
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Application of RNA-Seq in Ecotoxicogenomics: Exploring the Effects of Ibuprofen Exposure on Rainbow Trout and C
    Application of RNA-Seq in Ecotoxicogenomics: Exploring the Effects of Ibuprofen Exposure on Rainbow Trout and C. elegans by Lorraine Lok-Lun Yu Brown B.Sc., University of British Columbia, 2007 Thesis Submitted in Partial Fulfillment of the Requirements for the Degree of Master of Science in the Department of Molecular Biology and Biochemistry Faculty of Science Lorraine Lok-Lun Yu Brown 2013 SIMON FRASER UNIVERSITY Fall 2013 Approval Name: Lorraine Lok-Lun Yu Brown Degree: Master of Science Title of Thesis: Application of RNA-Seq in Ecotoxicogenomics: Exploring the Effects of Ibuprofen Exposure on Rainbow Trout and C. elegans Examining Committee: Chair: Nicholas Harden Professor Fiona Brinkman Senior Supervisor Professor William Davidson Supervisor Professor Steven Jones Supervisor Professor Christopher Kennedy Internal Examiner Professor Department of Biological Sciences Date Defended/Approved: December 16, 2013 ii Partial Copyright Licence iii Ethics Statement iv Abstract RNA-Seq was applied in this ecotoxicogenomics study to investigate the effects of ibuprofen in two species, rainbow trout (Oncorhynchus mykiss), a fish routinely used in ecotoxicology tests, and Caenorhabditis elegans, a well-studied nematode with immense genomics information. Exposure to environmentally relevant levels of ibuprofen resulted in gene expression changes relating to stress, prostaglandin synthesis, reproduction and development in both species. In fish, we observed sex-dependent differences in vitellogenin and prostaglandin synthase gene expression, highlighting the importance of genetic sex determination of juvenile fish used in bioassays. In worms, we saw a decrease in progeny production count. Our results suggest that ibuprofen may have negative impacts on reproduction in both species but requires further investigation.
    [Show full text]
  • Protein-Coding Variants Implicate Novel Genes Related to Lipid Homeostasis Contributing to Body-Fat Distribution
    ARTICLES https://doi.org/10.1038/s41588-018-0334-2 Protein-coding variants implicate novel genes related to lipid homeostasis contributing to body-fat distribution Body-fat distribution is a risk factor for adverse cardiovascular health consequences. We analyzed the association of body-fat distribution, assessed by waist-to-hip ratio adjusted for body mass index, with 228,985 predicted coding and splice site variants available on exome arrays in up to 344,369 individuals from five major ancestries (discovery) and 132,177 European-ancestry individuals (validation). We identified 15 common (minor allele frequency, MAF ≥5%) and nine low-frequency or rare (MAF <5%) coding novel variants. Pathway/gene set enrichment analyses identified lipid particle, adiponectin, abnormal white adi- pose tissue physiology and bone development and morphology as important contributors to fat distribution, while cross-trait associations highlight cardiometabolic traits. In functional follow-up analyses, specifically in Drosophila RNAi-knockdowns, we observed a significant increase in the total body triglyceride levels for two genes (DNAH10 and PLXND1). We implicate novel genes in fat distribution, stressing the importance of interrogating low-frequency and protein-coding variants. entral body-fat distribution, as assessed by waist-to-hip ratio 47 assuming an additive model and one under a recessive model (WHR), is a heritable and a well-established risk factor for (Table 1 and Supplementary Figs. 1–4). Due to possible heteroge- Cadverse metabolic outcomes1–6. Lower values of WHR are neity, we also performed European-only meta-analysis. Here, four associated with lower risk of cardiometabolic diseases such as type additional coding variants were significant (three novel) assum- 2 diabetes7,8, or differences in bone structure and gluteal muscle ing an additive model (Table 1 and Supplementary Figs.
    [Show full text]
  • T Cell Self-Reactivity During Thymic Development Dictates the Timing of Positive Selection
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.18.427079; this version posted January 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Title: T cell self-reactivity during thymic development dictates the timing of positive 2 selection 3 4 5 Authors: 6 Lydia K. Lutes1, Zoë Steier2, Laura L. McIntyre1, Shraddha Pandey1, James Kaminski3,#, 7 Ashley R. Hoover1,§, Silvia Ariotti1,†, Aaron Streets2,3,4, Nir Yosef2,3,4,5,6,, Ellen A. Robey1‡ 8 9 Author Affiliations: 10 1 Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of 11 California, Berkeley 12 2 Department of Bioengineering, University of California, Berkeley 13 3 Center for Computational Biology, University of California, Berkeley 14 4 Chan Zuckerberg Biohub, San Francisco, California 15 5 Department of Electrical Engineering and Computer Sciences, University of California, Berkeley 16 6 Ragon Institute of MGH, MIT and Harvard 17 ‡ To whom correspondence may be addressed. E-mail: [email protected]. 18 19 Present addresses: # Division of Pediatric Hematology/Oncology, Boston Children’s Hospital; Department of 20 Medical Oncology, Dana Farber Cancer Institute, and Harvard Medical School, Boston, MA; § Oklahoma 21 Medical Research Foundation, Oklahoma, United States; † Instituto de Medicina Molecular, João Lobo Atunes, 22 Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisbon, 1649-028, Portugal. 23 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.01.18.427079; this version posted January 21, 2021.
    [Show full text]
  • Protein Dysregulation in Immune Cells of Multiple Sclerosis Patients
    Master’s Thesis 2020 60 ECTS Faculty of Chemistry, Biotechnology and Food Sciences Protein Dysregulation in Immune Cells of Multiple Sclerosis Patients Oda Glomstad Kråbøl Biotechnology Faculty of Chemistry, Biotechnology and Food Sciences Protein Dysregulation in Immune Cells of Multiple Sclerosis Patients Oda Glomstad Kråbøl Main supervisor: Tone Berge Co-supervisor: Chiara Cappelletti Internal supervisor: Siv Kjølsrud Bøhn Oslo University Hospital – University of Oslo Multiple Sclerosis Research Group and The Norwegian University of Life Sciences, Faculty of Chemistry, Biotechnology and Food Sciences June 2020 ©Oda Glomstad Kråbøl Protein Dysregulation in Immune Cells of Multiple Sclerosis Patients https://nmbu.brage.unit.no Acknowledgements This thesis was carried out at the MS research group in the Neuroscience Research Unit of Oslo University Hospital (OUS) from August 2019 to June 2020. The thesis is a part of a Master’s Degree in Biotechnology at the Norwegian University of Life Sciences (NMBU), main affiliation being the Faculty of Chemistry, Biotechnology and Food Sciences (KBM). Firstly, I would like to thank Professor Tone Berge for being my supervisor, and for arranging this interesting project. Her expertise and enthusiasm have given me great insights into the challenging yet fascinating multiple sclerosis research field. The guidance I have received throughout this time has been exceptional! I am likewise thankful for my co-supervisor Chiara Cappelletti for devoting her time supervising me in the lab and answering all my questions. She has provided me with protocols as well as theoretical and practical explanations for everything I have needed, which has been invaluable. I would also like to thank the MS research group members for making me feel welcome and included.
    [Show full text]
  • Protein-Coding Variants Implicate Novel Genes Related to Lipid Homeostasis Contributing to Body-Fat Distribution
    ARTICLES https://doi.org/10.1038/s41588-018-0334-2 Protein-coding variants implicate novel genes related to lipid homeostasis contributing to body-fat distribution Body-fat distribution is a risk factor for adverse cardiovascular health consequences. We analyzed the association of body-fat distribution, assessed by waist-to-hip ratio adjusted for body mass index, with 228,985 predicted coding and splice site variants available on exome arrays in up to 344,369 individuals from five major ancestries (discovery) and 132,177 European-ancestry individuals (validation). We identified 15 common (minor allele frequency, MAF ≥5%) and nine low-frequency or rare (MAF <5%) coding novel variants. Pathway/gene set enrichment analyses identified lipid particle, adiponectin, abnormal white adi- pose tissue physiology and bone development and morphology as important contributors to fat distribution, while cross-trait associations highlight cardiometabolic traits. In functional follow-up analyses, specifically in Drosophila RNAi-knockdowns, we observed a significant increase in the total body triglyceride levels for two genes (DNAH10 and PLXND1). We implicate novel genes in fat distribution, stressing the importance of interrogating low-frequency and protein-coding variants. entral body-fat distribution, as assessed by waist-to-hip ratio 47 assuming an additive model and one under a recessive model (WHR), is a heritable and a well-established risk factor for (Table 1 and Supplementary Figs. 1–4). Due to possible heteroge- Cadverse metabolic outcomes1–6. Lower values of WHR are neity, we also performed European-only meta-analysis. Here, four associated with lower risk of cardiometabolic diseases such as type additional coding variants were significant (three novel) assum- 2 diabetes7,8, or differences in bone structure and gluteal muscle ing an additive model (Table 1 and Supplementary Figs.
    [Show full text]
  • Jachetti Suppl Table 1 80814.Xlsx
    Uniprot name Protein name Gene name Peptides OT1 - TPIN-SC(CD62Lhi)/OT1 - TNE-SC(CD62Llo) OT1 - TPIN-SC(CD62Lhi)/OT1 - TPIN-SC(CD62Llo) Log2 (ratio) Counts Log10 (Summed Significance B Log2 (ratio) Counts Log10 (Summed Significance B Intensities) Intensites) O08797 Protein Serpinb9 Serpinb9 5 1,6950 8 6,3029 0,0000 -0,2205 7 6,7278 0,5314 E9Q5W7 L-selectin Sell 2 1,4661 2 5,6553 0,0000 0,9395 4 6,3054 0,3946 Q544R1 Proline synthase co-transcribeD Prosc 2 1,4208 2 6,1679 0,0000 0,5063 2 5,7559 0,7523 bacterial homolog protein O09159 Lysosomal alpha-mannosiDase Man2b1 3 1,1965 2 5,4603 0,0000 NaN 1 5,1854 1,0000 E9PUM4 Talin-2 Tln2 19 1,1659 2 7,2477 0,0000 NaN 0 NaN 1,0000 Q3V157 Signal transDucer anD activator of Stat4 5 0,9376 3 5,7622 0,0000 0,1255 10 6,6896 0,8677 transcription 4 A2RSY6 TRMT1-like protein Trmt1l 2 0,8718 2 5,6063 0,0000 NaN 0 NaN 1,0000 P07356 Annexin A2 Anxa2 16 0,8307 15 6,9356 0,0000 0,9899 19 7,4857 0,4938 Q3UD32 Cathepsin S Ctss 4 0,8148 6 6,4179 0,0000 0,5122 5 6,7013 0,7468 Q8BV49 Pyrin anD HIN Domain-containing Pyhin1 6 0,7389 4 6,3847 0,0001 0,0098 5 6,9368 0,7255 protein 1 A2A4L8 SWI/SNF-relateD matrix- Smarce1 2 0,7367 2 5,5580 0,0001 NaN 1 6,0717 1,0000 associateD actin-DepenDent regulator of chromatin subfamily E member 1 Q7TPR4 Alpha-actinin-1 Actn1 37 0,6753 16 6,8461 0,0000 1,0007 16 7,3022 0,4868 Q6IRU2 Tropomyosin alpha-4 chain Tpm4 8 0,6405 5 6,6265 0,0000 0,5613 5 6,8943 0,8061 E9Q2W5 YTH Domain family protein 3 YthDf2 2 0,6162 2 7,8124 0,0000 NaN 1 7,4648 1,0000 O08738 Caspase-6 Casp6 3 0,5895 2
    [Show full text]
  • Systematic Prioritization of Candidate Genes in Disease Loci Identifies TRAFD1 As a 2 Master Regulator of IFN Signalling in Celiac Disease
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.04.973487; this version posted March 5, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Systematic prioritization of candidate genes in disease loci identifies TRAFD1 as a 2 master regulator of IFN signalling in celiac disease # # 3 Adriaan van der Graaf1 , Maria Zorro1 , Annique Claringbould1, Urmo Vosa1,2, Raul Aguirre- 4 Gamboa1, Chan Li1,3, Joram Mooiweer1, Isis Ricano-Ponce1, Zuzanna Borek4,5, Frits 5 Koning6, Yvonne Kooy-Winkelaar6, Ludvig Sollid3, Shuo-Wang Qiao3, BIOS consortium, 6 Vinod Kumar1,7, Yang Li1,7,8, Lude Franke1, Sebo Withoff1, Cisca Wijmenga1,3, Serena # # 7 Sanna1,9 *, Iris Jonkers1,3 *. 8 Affiliations 9 1 Department of Genetics, University Medical Center Groningen, University of Groningen, 10 Groningen, the Netherlands 11 2 Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia 12 3 Department of Immunology, K.G. Jebsen Coeliac Disease Research Centre, University of 13 Oslo, Oslo, Norway 14 4 Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz 15 Association, Berlin, Germany 16 5 Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, 17 Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of 18 Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany 19 6 Department of Immunohematology and Blood Transfusion, Leiden University
    [Show full text]
  • Influence of Host Genetics in Shaping the Rumen Bacterial Community In
    www.nature.com/scientificreports OPEN Infuence of host genetics in shaping the rumen bacterial community in beef cattle Waseem Abbas1,4, Jeremy T. Howard1,4, Henry A. Paz1,2, Kristin E. Hales3, James E. Wells3, Larry A. Kuehn3, Galen E. Erickson1, Matthew L. Spangler1* & Samodha C. Fernando1* In light of recent host-microbial association studies, a consensus is evolving that species composition of the gastrointestinal microbiota is a polygenic trait governed by interactions between host genetic factors and the environment. Here, we investigated the efect of host genetic factors in shaping the bacterial species composition in the rumen by performing a genome-wide association study. Using a common set of 61,974 single-nucleotide polymorphisms found in cattle genomes (n = 586) and corresponding rumen bacterial community composition, we identifed operational taxonomic units (OTUs), Families and Phyla with high heritability. The top associations (1-Mb windows) were located on 7 chromosomes. These regions were associated with the rumen microbiota in multiple ways; some (chromosome 19; position 3.0–4.0 Mb) are associated with closely related taxa (Prevotellaceae, Paraprevotellaceae, and RF16), some (chromosome 27; position 3.0–4.0 Mb) are associated with distantly related taxa (Prevotellaceae, Fibrobacteraceae, RF16, RFP12, S24-7, Lentisphaerae, and Tenericutes) and others (chromosome 23; position 0.0–1.0) associated with both related and unrelated taxa. The annotated genes associated with identifed genomic regions suggest the associations observed are directed toward selective absorption of volatile fatty acids from the rumen to increase energy availability to the host. This study demonstrates that host genetics afects rumen bacterial community composition.
    [Show full text]
  • Can We Treat Congenital Blood Disorders by Transplantation Of
    Can we Treat Congenital Blood Disorders by Transplantation of Stem Cells, Gene Therapy to the Fetus? Panicos Shangaris University College London 2019 A thesis submitted for the degree of Doctor of Philosophy 1 Declaration I, Panicos Shangaris confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. 2 Abstract Congenital diseases such as blood disorders are responsible for over a third of all pediatric hospital admissions. In utero transplantation (IUT) could cure affected fetuses but so far in humans, successful IUT has been limited to fetuses with severe immunologic defects, due to the maternal immune system and a functionally developed fetal immune system. I hypothesised that using autologous fetal cells could overcome the barriers to engraftment. Previous studies show that autologous haematopoietic progenitors can be easily derived from amniotic fluid (AF), and they can engraft long term into fetal sheep. In normal mice, I demonstrated that IUT of mouse AFSC results in successful haematopoietic engraftment in immune-competent mice. Congenic AFSCs appear to have a significant advantage over allogenic AFSCs. This was seen both by their end haematopoietic potential and the immune response of the host. Expansion of haematopoietic stem cells (HSC) has been a complicated and demanding process. To achieve adequate numbers for autologous stem cells for IUT, HSCs need to be expanded efficiently. I expanded and compared AFSCs, fetal liver stem cells and bone marrow stem cells. Culturing and expanding fetal and adult stem cells in embryonic stem cell conditions maintained their haematopoietic potential.
    [Show full text]
  • 2015-04-10 Introduction
    UNIVERSITY OF CALIFORNIA, SAN DIEGO Enhancers and Transcriptional Regulation in CD4+ T Cells A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Bioinformatics and Systems Biology by Karmel Alon Allison Committee in charge: Professor Christopher K. Glass, Chair Professor Stephen M. Hedrick, Co-Chair Professor John T. Chang Professor Terry Gaasterland Professor Bing Ren 2015 ! Copyright Karmel Alon Allison, 2015 All rights reserved. ! The Dissertation of Karmel Alon Allison is approved, and it is acceptable in quality and form for publication on microfilm and electronically: Co-Chair Chair University of California, San Diego 2015 iii! ! DEDICATION To my husband, David, who loved, supported, and funded me throughout this journey, to my son, Ezra, who was a very good little boy and let Mommy work, and to God, who is and does all things. iv! ! TABLE OF CONTENTS Signature Page........................................................................................................... iii Dedication.................................................................................................................. iv Table of Contents....................................................................................................... v List of Figures............................................................................................................ vi Acknowledgements.................................................................................................... viii Vita............................................................................................................................
    [Show full text]