Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function

Total Page:16

File Type:pdf, Size:1020Kb

Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Tay, Rong En. 2019. Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function. Doctoral dissertation, Harvard University, Graduate School of Arts & Sciences. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:42029527 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function A dissertation presented by Rong En Tay to The Department of Medical Sciences in partial fulfilment of the requirements for the degree of Doctor of Philosophy in the subject of Immunology Harvard University Cambridge, Massachusetts April 2019 © 2019 Rong En Tay All rights reserved. Dissertation Advisor: Kai Wucherpfennig Rong En Tay Discovery of Novel Epigenetic Regulators of CD8+ T Cell Effector Function Abstract CD8+ cytotoxic T lymphocytes (CTLs) play a key role in acquired immunity by killing infected or cancerous cells. Upon antigen recognition and activation, the majority of naïve CD8+ T cells differentiate into potent but short-lived effector CTLs, while a small fraction generates long-lived memory cells that are poised to proliferate rapidly upon antigen re-encounter. While transcriptional control of CD8+ T cell differentiation and effector function has been extensively studied, little is known about epigenetic regulation of these processes. Here we use two screening approaches to uncover novel epigenetic regulators of CD8+ T cell effector function. We first engineered a CRISPR-Cas9 genetic screening platform to reveal epigenetic regulators in tumour-infiltrating CD8+ T cells that could potentially be targeted to improve CD8+ anti- tumour function. This approach identified CARM1 as a strong candidate epigenetic regulator suppressing the accumulation of tumor-specific CD8+ T cells in the pre-clinical B16 melanoma tumour model. We also used a functional pharmacological approach to screen for epigenetic regulators of CD8+ T cell effector function during in vitro T cell activation. We thus identified a novel role of the histone deacetylase HDAC3 as an epigenetic regulator of CD8+ T cell cytotoxicity and persistence. Hdac3-deficient CD8+ T cells transiently acquired augmented cytotoxicity that was associated with early resistance to chronic LCMV infection but did not confer durable disease protection. Mechanistically, HDAC3 inhibited a gene programme of cytotoxicity-associated genes and terminal effector differentiation beginning early during CD8+ T cell activation, and required the transcription factors Runx3 and Blimp-1 as key downstream iii mediators for its regulation of CD8+ T cell effector function. Targeting HDAC3 activity to epigenetically modulate the balance between a short-lived, potent cytotoxic effector state and a durable, long-lived response could potentially lead to the development of new approaches in adoptive T cell immunotherapy and in therapeutic vaccine development. iv Table of Contents Acknowledgements vi Dedication vii Chapter 1 – Introduction 1 Chapter 2 – Materials and Methods 7 Chapter 3 – Discovery of CARM1 as a suppressor of CD8+ T cell accumulation in 23 tumours using an in vivo CRISPR-Cas9 genetic screening platform Chapter 4 – Discovery of HDAC3 as an inhibitor of CD8+ T cell cytotoxicity in an in 28 vitro functional pharmacologic screen Chapter 5 – HDAC3 is required during T cell activation for persistence of antigen- 40 experienced CD8+ T cells Chapter 6 – Effects of loss of HDAC3 activity in CD8+ T cells in settings of chronic 51 antigen burden Chapter 7 – Transcriptional and epigenetic signatures of HDAC3 60 Chapter 8 – Uncovering potential mechanisms of HDAC3-mediated regulation of 71 CD8+ T cell effector function using genetic approaches Chapter 9 – Summary and Discussion 87 Appendix 99 References 106 v Acknowledgements I reverently thank the LORD my God for sustaining me through this long and difficult journey. By His grace have I come thus far while keeping my honour and integrity intact. I thank my PhD advisor, Kai Wucherpfennig, for his supervision and direction and for giving me the opportunity to work on an interesting project. I am deeply and forever grateful to Hye-Jung Kim for her unwavering support of me, both scientifically and personally. Thank you for being a true friend and mentor to me and for believing in me. I thank my collaborators Paloma Cejas, Henry Long, and Clifford Meyer (DFCI) for their invaluable help in performing the ChIP-seq analyses, and Peng Jiang (DFCI) for computational analysis of the in vivo screen. I am very grateful to my dissertation advisory committee members Ulrich von Andrian (HMS), Myles Brown (DFCI), and Nicholas Haining (DFCI) for their helpful guidance and constructive feedback. I thank Scott Hiebert (Vanderbilt), Christina Weng and David Fisher (MGH), Steven Elledge (HMS), and Ulrich von Andrian (HMS) for their kind gifts of tools and resources in support of my projects. Special thanks to Olamide Olawoyin for her incredible hard work and dedication during our time working together. It was an honour and privilege to teach one so deserving as yourself. I thank Sabrina Haag for her insightful comments and scientific input into my projects. I thank my fellowship sponsors, the Agency for Science, Technology, and Research (A*STAR, Singapore), for their generous financial support over the course of my doctoral work. I thank all other members of the Wucherpfennig lab for their collegiality. vi Dedication Dedicated to the LORD my God as a testimony of His faithfulness to me, to my loving family for their unwavering support and continual prayers for the safe return of their son and brother, to my dearest fiancée Kayla, for her patience and love during this period of separation, and to Kayla’s father Sam, for his faith that this day would come. Give thanks to the LORD, for He is good. His love endures forever. - Psalm 136:1 (NIV) vii Chapter 1 Introduction Introduction Upon antigen encounter during inflammation, naïve CD8+ T cells undergo phenotypic changes during activation to develop into effector cytotoxic T lymphocytes that mediate immunity via contact-dependent killing of infected or malignant transformed cells and by secreting effector cytokines such as IFN-γ and TNF-α. By the peak of the CD8+ T cell effector response to acute antigen exposure, activated CD8+ T cells are already committed to one of two cell fates – a short-lived terminally-differentiated state with potent effector function1,2, or a long-lived memory precursor phenotype with high proliferative potential but weak effector function3,4. After antigen clearance, most of the terminally-differentiated effector cells die off, resulting in a quantitative and qualitative contraction of the CD8+ T cell response, whereas memory precursor cells persist and subsequently differentiate into memory cells, which are poised to mount a robust and rapid defence upon secondary antigen encounter. All these differentiation processes require co-ordination between the molecular pathways integrating extracellular signalling inputs with cell- intrinsic programming for proper acquisition of full effector CD8+ T cell function as well as for proper control and homeostasis of the CD8+ T cell immune response5. The transcriptional regulation of the developmental processes following CD8+ T cell activation has been extensively described and studied. Advances in RNA-sequencing technology and analysis techniques have led to comprehensive profiling of the roles of such key transcription factors as T-bet, Eomes, Blimp-1, and Bcl-6 in regulating the acquisition of effector functions of CD8+ T cells, as well as in regulating the commitment of activated CD8+ T cells to one of the two phenotypically-distinct cell fates following acute antigen encounter5,6. These studies have been indispensable to our current understanding of CD8+ T cell differentiation as a process governed by interactions of key driver molecules within an intricately complex transcriptional network7. Each transcription factor governs a distinct suite of effector functions or 2 phenotypes (e.g. T-bet is required to potentiate IFN-γ secretion and cytotoxicity, as well as effector phenotype acquisition8), and the co-ordination of several of these transcription factors across time thus constitutes a fundamental molecular control mechanism governing the acquisition of effector function and cell fate commitment. Whereas control of CD8+ T cell function and differentiation is proximally mediated by transcription factors, it is the epigenetic landscape9 that provides the framework within which the gene regulatory network operates. Broadly speaking, epigenetic processes constrain the spectrum of potential phenotypes that a given cell can acquire by covalently modifying DNA bases or specific amino acid residues of chromatin- associated proteins such as histones. These modifications serve as beacons for ‘reader’ proteins that recognise and recruit specific transcription factors to the marked regions of chromatin. The patterns of epigenetic marks also generate regions of chromatin with differential conformational states and accessibility to transcriptional machinery (i.e. ‘open’ and ‘closed’
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Abstract Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • Rabbit Anti-SEN1/FITC Conjugated Antibody
    SunLong Biotech Co.,LTD Tel: 0086-571- 56623320 Fax:0086-571- 56623318 E-mail:[email protected] www.sunlongbiotech.com Rabbit Anti-SEN1/FITC Conjugated antibody SL7576R-FITC Product Name: Anti-SEN1/FITC Chinese Name: FITC标记的细胞衰老相关蛋白1抗体 Cell senescence related gene complementation group B; Cellular senescence related protein 1; CSR; CSRB; MORF 4; MORF4; MORF4 protein; Mortality factor 4; SEN 1; Alias: SEN; Senescence (cellular) related 1; Senescence related cellular 1; Transcription factor like protein MORF4; MO4L1_HUMAN; Mortality factor 4-like protein 1; MORF- related gene 15 protein; Protein MSL3-1; Transcription factor-like protein MRG15. Organism Species: Rabbit Clonality: Polyclonal React Species: Human,Mouse,Rat,Dog,Cow,Zebrafish, IF=1:50-200 Applications: not yet tested in other applications. optimal dilutions/concentrations should be determined by the end user. Molecular weight: 27kDa Form: Lyophilized or Liquid Concentration: 1mg/ml immunogen: KLH conjugated synthetic peptide derived from human Mortality factor 4-like protein 1 Lsotype: IgGwww.sunlongbiotech.com Purification: affinity purified by Protein A Storage Buffer: 0.01M TBS(pH7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol. Store at -20 °C for one year. Avoid repeated freeze/thaw cycles. The lyophilized antibody is stable at room temperature for at least one month and for greater than a year Storage: when kept at -20°C. When reconstituted in sterile pH 7.4 0.01M PBS or diluent of antibody the antibody is stable for at least two weeks at 2-4 °C. background: Cellular senescence, the terminal nondividing state that normal cells enter following completion of their proliferative potential, is the dominant phenotype in hybrids of Product Detail: normal and immortal cells.
    [Show full text]
  • In Vivo Studies Using the Classical Mouse Diversity Panel
    The Mouse Diversity Panel Predicts Clinical Drug Toxicity Risk Where Classical Models Fail Alison Harrill, Ph.D The Hamner-UNC Institute for Drug Safety Sciences 0 The Importance of Predicting Clinical Adverse Drug Reactions (ADR) Figure: Cath O’Driscoll Nature Publishing 2004 Risk ID PGx Testing 1 People Respond Differently to Drugs Pharmacogenetic Markers Identified by Genome-Wide Association Drug Adverse Drug Risk Allele Reaction (ADR) Abacavir Hypersensitivity HLA-B*5701 Flucloxacillin Hepatotoxicity Allopurinol Cutaneous ADR HLA-B*5801 Carbamazepine Stevens-Johnson HLA-B*1502 Syndrome Augmentin Hepatotoxicity DRB1*1501 Ximelagatran Hepatotoxicity DRB1*0701 Ticlopidine Hepatotoxicity HLA-A*3303 Average preclinical populations and human hepatocytes lack the diversity to detect incidence of adverse events that occur only in 1/10,000 people. Current Rodent Models of Risk Assessment The Challenge “At a time of extraordinary scientific progress, methods have hardly changed in several decades ([FDA] 2004)… Toxicologists face a major challenge in the twenty-first century. They need to embrace the new “omics” techniques and ensure that they are using the most appropriate animals if their discipline is to become a more effective tool in drug development.” -Dr. Michael Festing Quantitative geneticist Toxicol Pathol. 2010;38(5):681-90 Rodent Models as a Strategy for Hazard Characterization and Pharmacogenetics Genetically defined rodent models may provide ability to: 1. Improve preclinical prediction of drugs that carry a human safety risk 2.
    [Show full text]
  • BMC Genomics Biomed Central
    BMC Genomics BioMed Central Research article Open Access The functional modulation of epigenetic regulators by alternative splicing Sergio Lois1,2, Noemí Blanco1,2, Marian Martínez-Balbás*1,2 and Xavier de la Cruz*2,3 Address: 1Instituto de Biología Molecular de Barcelona. CID. Consejo Superior de Investigaciones Científicas (CSIC); 08028 Barcelona, Spain, 2Institut de Recerca Biomèdica-PCB; 08028 Barcelona, Spain and 3Institució Catalana de Recerca i Estudis Avançats (ICREA); Barcelona, Spain Email: Sergio Lois - [email protected]; Noemí Blanco - [email protected]; Marian Martínez-Balbás* - [email protected]; Xavier de la Cruz* - [email protected] * Corresponding authors Published: 25 July 2007 Received: 24 April 2007 Accepted: 25 July 2007 BMC Genomics 2007, 8:252 doi:10.1186/1471-2164-8-252 This article is available from: http://www.biomedcentral.com/1471-2164/8/252 © 2007 Lois et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: Epigenetic regulators (histone acetyltransferases, methyltransferases, chromatin- remodelling enzymes, etc) play a fundamental role in the control of gene expression by modifying the local state of chromatin. However, due to their recent discovery, little is yet known about their own regulation. This paper addresses this point, focusing on alternative splicing regulation, a mechanism already known to play an important role in other protein families, e.g. transcription factors, membrane receptors, etc.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • List of Genes Used in Cell Type Enrichment Analysis
    List of genes used in cell type enrichment analysis Metagene Cell type Immunity ADAM28 Activated B cell Adaptive CD180 Activated B cell Adaptive CD79B Activated B cell Adaptive BLK Activated B cell Adaptive CD19 Activated B cell Adaptive MS4A1 Activated B cell Adaptive TNFRSF17 Activated B cell Adaptive IGHM Activated B cell Adaptive GNG7 Activated B cell Adaptive MICAL3 Activated B cell Adaptive SPIB Activated B cell Adaptive HLA-DOB Activated B cell Adaptive IGKC Activated B cell Adaptive PNOC Activated B cell Adaptive FCRL2 Activated B cell Adaptive BACH2 Activated B cell Adaptive CR2 Activated B cell Adaptive TCL1A Activated B cell Adaptive AKNA Activated B cell Adaptive ARHGAP25 Activated B cell Adaptive CCL21 Activated B cell Adaptive CD27 Activated B cell Adaptive CD38 Activated B cell Adaptive CLEC17A Activated B cell Adaptive CLEC9A Activated B cell Adaptive CLECL1 Activated B cell Adaptive AIM2 Activated CD4 T cell Adaptive BIRC3 Activated CD4 T cell Adaptive BRIP1 Activated CD4 T cell Adaptive CCL20 Activated CD4 T cell Adaptive CCL4 Activated CD4 T cell Adaptive CCL5 Activated CD4 T cell Adaptive CCNB1 Activated CD4 T cell Adaptive CCR7 Activated CD4 T cell Adaptive DUSP2 Activated CD4 T cell Adaptive ESCO2 Activated CD4 T cell Adaptive ETS1 Activated CD4 T cell Adaptive EXO1 Activated CD4 T cell Adaptive EXOC6 Activated CD4 T cell Adaptive IARS Activated CD4 T cell Adaptive ITK Activated CD4 T cell Adaptive KIF11 Activated CD4 T cell Adaptive KNTC1 Activated CD4 T cell Adaptive NUF2 Activated CD4 T cell Adaptive PRC1 Activated
    [Show full text]
  • DNA Methylation Regulates Discrimination of Enhancers From
    Sharifi-Zarchi et al. BMC Genomics (2017) 18:964 DOI 10.1186/s12864-017-4353-7 RESEARCHARTICLE Open Access DNA methylation regulates discrimination of enhancers from promoters through a H3K4me1-H3K4me3 seesaw mechanism Ali Sharifi-Zarchi1,2,3,4†, Daniela Gerovska5†, Kenjiro Adachi6, Mehdi Totonchi3, Hamid Pezeshk7,8, Ryan J. Taft9, Hans R. Schöler6,10, Hamidreza Chitsaz2, Mehdi Sadeghi8,11, Hossein Baharvand3,12* and Marcos J. Araúzo-Bravo5,13,14* Abstract Background: DNA methylation at promoters is largely correlated with inhibition of gene expression. However, the role of DNA methylation at enhancers is not fully understood, although a crosstalk with chromatin marks is expected. Actually, there exist contradictory reports about positive and negative correlations between DNA methylation and H3K4me1, a chromatin hallmark of enhancers. Results: We investigated the relationship between DNA methylation and active chromatin marks through genome- wide correlations, and found anti-correlation between H3K4me1 and H3K4me3 enrichment at low and intermediate DNA methylation loci. We hypothesized “seesaw” dynamics between H3K4me1 and H3K4me3 in the low and intermediate DNA methylation range, in which DNA methylation discriminates between enhancers and promoters, marked by H3K4me1 and H3K4me3, respectively. Low methylated regions are H3K4me3 enriched, while those with intermediate DNA methylation levels are progressively H3K4me1 enriched. Additionally, the enrichment of H3K27ac, distinguishing active from primed enhancers, follows a plateau in the lower range of the intermediate DNA methylation level, corresponding to active enhancers, and decreases linearly in the higher range of the intermediate DNA methylation. Thus, the decrease of the DNA methylation switches smoothly the state of the enhancers from a primed to an active state.
    [Show full text]
  • Finding Drug Targeting Mechanisms with Genetic Evidence for Parkinson’S Disease
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.24.208975; this version posted July 24, 2020. 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-NC-ND 4.0 International license. Finding drug targeting mechanisms with genetic evidence for Parkinson’s disease Catherine S. Storm1,*, Demis A. Kia1, Mona Almramhi1, Sara Bandres-Ciga2, Chris Finan3, Aroon D. Hingorani3,4,5, International Parkinson’s Disease Genomics Consortium (IPDGC), Nicholas W. Wood1,6,* 1 Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom 2 Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, United States of America 3 Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, United Kingdom 4 University College London British Heart Foundation Research Accelerator Centre, New Delhi, India 5 Health Data Research UK, 222 Euston Road, London, United Kingdom 6 Lead Contact * Correspondence: [email protected] (CSS), [email protected] (NWW) Summary Parkinson’s disease (PD) is a neurodegenerative movement disorder that currently has no disease-modifying treatment, partly owing to inefficiencies in drug target identification and validation using human evidence. Here, we use Mendelian randomization to investigate more than 3000 genes that encode druggable proteins, seeking to predict their efficacy as drug targets for PD. We use expression and protein quantitative trait loci for druggable genes to mimic exposure to medications, and we examine the causal effect on PD risk (in two large case-control cohorts), PD age at onset and progression.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • Interoperability in Toxicology: Connecting Chemical, Biological, and Complex Disease Data
    INTEROPERABILITY IN TOXICOLOGY: CONNECTING CHEMICAL, BIOLOGICAL, AND COMPLEX DISEASE DATA Sean Mackey Watford A dissertation submitted to the faculty at the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Gillings School of Global Public Health (Environmental Sciences and Engineering). Chapel Hill 2019 Approved by: Rebecca Fry Matt Martin Avram Gold David Reif Ivan Rusyn © 2019 Sean Mackey Watford ALL RIGHTS RESERVED ii ABSTRACT Sean Mackey Watford: Interoperability in Toxicology: Connecting Chemical, Biological, and Complex Disease Data (Under the direction of Rebecca Fry) The current regulatory framework in toXicology is expanding beyond traditional animal toXicity testing to include new approach methodologies (NAMs) like computational models built using rapidly generated dose-response information like US Environmental Protection Agency’s ToXicity Forecaster (ToXCast) and the interagency collaborative ToX21 initiative. These programs have provided new opportunities for research but also introduced challenges in application of this information to current regulatory needs. One such challenge is linking in vitro chemical bioactivity to adverse outcomes like cancer or other complex diseases. To utilize NAMs in prediction of compleX disease, information from traditional and new sources must be interoperable for easy integration. The work presented here describes the development of a bioinformatic tool, a database of traditional toXicity information with improved interoperability, and efforts to use these new tools together to inform prediction of cancer and complex disease. First, a bioinformatic tool was developed to provide a ranked list of Medical Subject Heading (MeSH) to gene associations based on literature support, enabling connection of compleX diseases to genes potentially involved.
    [Show full text]
  • 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]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]