1
|
Borrego-Yaniz G, Ortiz-Fernández L, Madrid-Paredes A, Kerick M, Hernández-Rodríguez J, Mackie SL, Vaglio A, Castañeda S, Solans R, Mestre-Torres J, Khalidi N, Langford CA, Ytterberg S, Beretta L, Govoni M, Emmi G, Cimmino MA, Witte T, Neumann T, Holle J, Schönau V, Pugnet G, Papo T, Haroche J, Mahr A, Mouthon L, Molberg Ø, Diamantopoulos AP, Voskuyl A, Daikeler T, Berger CT, Molloy ES, Blockmans D, van Sleen Y, Iles M, Sorensen L, Luqmani R, Reynolds G, Bukhari M, Bhagat S, Ortego-Centeno N, Brouwer E, Lamprecht P, Klapa S, Salvarani C, Merkel PA, Cid MC, González-Gay MA, Morgan AW, Martin J, Márquez A. Risk loci involved in giant cell arteritis susceptibility: a genome-wide association study. THE LANCET. RHEUMATOLOGY 2024; 6:e374-e383. [PMID: 38734017 PMCID: PMC11108802 DOI: 10.1016/s2665-9913(24)00064-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Giant cell arteritis is an age-related vasculitis that mainly affects the aorta and its branches in individuals aged 50 years and older. Current options for diagnosis and treatment are scarce, highlighting the need to better understand its underlying pathogenesis. Genome-wide association studies (GWAS) have emerged as a powerful tool for unravelling the pathogenic mechanisms involved in complex diseases. We aimed to characterise the genetic basis of giant cell arteritis by performing the largest GWAS of this vasculitis to date and to assess the functional consequences and clinical implications of identified risk loci. METHODS We collected and meta-analysed genomic data from patients with giant cell arteritis and healthy controls of European ancestry from ten cohorts across Europe and North America. Eligible patients required confirmation of giant cell arteritis diagnosis by positive temporal artery biopsy, positive temporal artery doppler ultrasonography, or imaging techniques confirming large-vessel vasculitis. We assessed the functional consequences of loci associated with giant cell arteritis using cell enrichment analysis, fine-mapping, and causal gene prioritisation. We also performed a drug repurposing analysis and developed a polygenic risk score to explore the clinical implications of our findings. FINDINGS We included a total of 3498 patients with giant cell arteritis and 15 550 controls. We identified three novel loci associated with risk of giant cell arteritis. Two loci, MFGE8 (rs8029053; p=4·96 × 10-8; OR 1·19 [95% CI 1·12-1·26]) and VTN (rs704; p=2·75 × 10-9; OR 0·84 [0·79-0·89]), were related to angiogenesis pathways and the third locus, CCDC25 (rs11782624; p=1·28 × 10-8; OR 1·18 [1·12-1·25]), was related to neutrophil extracellular traps (NETs). We also found an association between this vasculitis and HLA region and PLG. Variants associated with giant cell arteritis seemed to fulfil a specific regulatory role in crucial immune cell types. Furthermore, we identified several drugs that could represent promising candidates for treatment of this disease. The polygenic risk score model was able to identify individuals at increased risk of developing giant cell arteritis (90th percentile OR 2·87 [95% CI 2·15-3·82]; p=1·73 × 10-13). INTERPRETATION We have found several additional loci associated with giant cell arteritis, highlighting the crucial role of angiogenesis in disease susceptibility. Our study represents a step forward in the translation of genomic findings to clinical practice in giant cell arteritis, proposing new treatments and a method to measure genetic predisposition to this vasculitis. FUNDING Institute of Health Carlos III, Spanish Ministry of Science and Innovation, UK Medical Research Council, and National Institute for Health and Care Research.
Collapse
Affiliation(s)
- Gonzalo Borrego-Yaniz
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Lourdes Ortiz-Fernández
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Adela Madrid-Paredes
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain; Department of Clinical Pharmacy, San Cecilio University Hospital, Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Granada, Spain
| | - Martin Kerick
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - José Hernández-Rodríguez
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Sarah L Mackie
- School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Augusto Vaglio
- Department of Biomedical Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy; Meyer Children's Hospital, Nephrology and Dialysis Unit, Florence, Italy
| | - Santos Castañeda
- Department of Rheumatology, Hospital de la Princesa, IIS-IP, Madrid, Spain
| | - Roser Solans
- Autoimmune Systemic Diseases Unit, Department of Internal Medicine, Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Jaume Mestre-Torres
- Autoimmune Systemic Diseases Unit, Department of Internal Medicine, Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Nader Khalidi
- Division of Rheumatology, McMaster University, Hamilton, ON, Canada
| | - Carol A Langford
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | | | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Marcello Govoni
- Department of Rheumatology, Azienda Ospedaliero Universitaria S Anna, University of Ferrara, Ferrara, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Florence, Italy; Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Marco A Cimmino
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genova, Italy
| | | | - Thomas Neumann
- Klinik für Innere Medizin III, University-Hospital Jena, Jena, Germany; Department of Rheumatology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Julia Holle
- Vasculitis Clinic, Klinikum Bad Bramstedt and University Hospital of Schleswig Holstein, Bad Bramstedt, Germany
| | - Verena Schönau
- Department of Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gregory Pugnet
- Department of Internal Medicine, Toulouse University Hospital Center, Toulouse, France
| | - Thomas Papo
- Hôpital Bichat, Université Paris-Cité, Service de Médecine Interne, Paris, France
| | - Julien Haroche
- Department of Internal Medicine and French Reference Center for Rare Auto-immune & Systemic Diseases, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alfred Mahr
- ECSTRRA Research Unit, Centre of Research in Epidemiology and Statistics, Sorbonne Paris Cité Research Center UMR 1153, Inserm, Paris, France
| | - Luc Mouthon
- Cochin Hospital, National Referral Center for Rare Autoimmune and Systemic Diseases, Université Paris Descartes, Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Øyvind Molberg
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | | | - Alexandre Voskuyl
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Thomas Daikeler
- Department of Rheumatology, University Hospital Basel and Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Christoph T Berger
- Department of Biomedicine and Department of Internal Medicine, Translational Immunology and Medical Outpatient Clinic, University Hospital Basel, Basel, Switzerland
| | - Eamonn S Molloy
- Department of Rheumatology, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Dublin, Ireland
| | - Daniel Blockmans
- Department of General Internal Medicine, University Hospital Gasthuisberg, Leuven, Belgium
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Mark Iles
- School of Medicine, University of Leeds, Leeds, UK; Leeds Institute for Data Analytics, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Louise Sorensen
- School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK; NIHR Leeds Medtech and In Vitro Diagnostics Co-Operative, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Raashid Luqmani
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Gary Reynolds
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Marwan Bukhari
- Rheumatology Department, University Hospitals of Morecambe Bay NHS Foundation Trust, Royal Lancaster Infirmary, Lancaster, UK; Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Shweta Bhagat
- West Suffolk NHS Foundation Trust, Bury Saint Edmunds, Bury St Edmunds, UK
| | - Norberto Ortego-Centeno
- Department of Medicine, University of Granada, Instituto de Investigación Biosanitaria de Granada ibs GRANADA, Granada, Spain
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Sebastian Klapa
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Carlo Salvarani
- Azienda USL-IRCCS di Reggio Emilia and Università di Modena e Reggio Emilia, Reggio Emilia, Italy
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine, and Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - María C Cid
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Miguel A González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, Spain; Department of Medicine, University of Cantabria, Santander, Spain
| | - Ann W Morgan
- School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK; NIHR Leeds Medtech and In Vitro Diagnostics Co-Operative, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Ana Márquez
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain.
| |
Collapse
|
2
|
Middha P, Thummalapalli R, Betti MJ, Yao L, Quandt Z, Balaratnam K, Bejan CA, Cardenas E, Falcon CJ, Faleck DM, Gubens MA, Huntsman S, Johnson DB, Kachuri L, Khan K, Li M, Lovly CM, Murray MH, Patel D, Werking K, Xu Y, Zhan LJ, Balko JM, Liu G, Aldrich MC, Schoenfeld AJ, Ziv E. Polygenic risk score for ulcerative colitis predicts immune checkpoint inhibitor-mediated colitis. Nat Commun 2024; 15:2568. [PMID: 38531883 PMCID: PMC10966072 DOI: 10.1038/s41467-023-44512-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 12/15/2023] [Indexed: 03/28/2024] Open
Abstract
Immune checkpoint inhibitor-mediated colitis (IMC) is a common adverse event of treatment with immune checkpoint inhibitors (ICI). We hypothesize that genetic susceptibility to Crohn's disease (CD) and ulcerative colitis (UC) predisposes to IMC. In this study, we first develop a polygenic risk scores for CD (PRSCD) and UC (PRSUC) in cancer-free individuals and then test these PRSs on IMC in a cohort of 1316 patients with ICI-treated non-small cell lung cancer and perform a replication in 873 ICI-treated pan-cancer patients. In a meta-analysis, the PRSUC predicts all-grade IMC (ORmeta=1.35 per standard deviation [SD], 95% CI = 1.12-1.64, P = 2×10-03) and severe IMC (ORmeta=1.49 per SD, 95% CI = 1.18-1.88, P = 9×10-04). PRSCD is not associated with IMC. Furthermore, PRSUC predicts severe IMC among patients treated with combination ICIs (ORmeta=2.20 per SD, 95% CI = 1.07-4.53, P = 0.03). Overall, PRSUC can identify patients receiving ICI at risk of developing IMC and may be useful to monitor patients and improve patient outcomes.
Collapse
Affiliation(s)
- Pooja Middha
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rohit Thummalapalli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael J Betti
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lydia Yao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zoe Quandt
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eduardo Cardenas
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christina J Falcon
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David M Faleck
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew A Gubens
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Scott Huntsman
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University of Medicine, Stanford, CA, USA
| | - Khaleeq Khan
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Min Li
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christine M Lovly
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Megan H Murray
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Kristin Werking
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luna Jia Zhan
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Temerty School of Medicine, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam J Schoenfeld
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Center for Genes, Environment and Health, University of California San Francisco, San Francisco, CA, USA.
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
He S, Lim GE. The Application of High-Throughput Approaches in Identifying Novel Therapeutic Targets and Agents to Treat Diabetes. Adv Biol (Weinh) 2023; 7:e2200151. [PMID: 36398493 DOI: 10.1002/adbi.202200151] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/04/2022] [Indexed: 11/19/2022]
Abstract
During the past decades, unprecedented progress in technologies has revolutionized traditional research methodologies. Among these, advances in high-throughput drug screening approaches have permitted the rapid identification of potential therapeutic agents from drug libraries that contain thousands or millions of molecules. Moreover, high-throughput-based therapeutic target discovery strategies can comprehensively interrogate relationships between biomolecules (e.g., gene, RNA, and protein) and diseases and significantly increase the authors' knowledge of disease mechanisms. Diabetes is a chronic disease primarily characterized by the incapacity of the body to maintain normoglycemia. The prevalence of diabetes in modern society has become a severe public health issue that threatens the well-being of millions of patients. Although a number of pharmacological treatments are available, there is no permanent cure for diabetes, and discovering novel therapeutic targets and agents continues to be an urgent need. The present review discusses the technical details of high-throughput screening approaches in drug discovery, followed by introducing the applications of such approaches to diabetes research. This review aims to provide an example of the applicability of high-throughput technologies in facilitating different aspects of disease research.
Collapse
Affiliation(s)
- Siyi He
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| |
Collapse
|
4
|
Immune and spermatogenesis-related loci are involved in the development of extreme patterns of male infertility. Commun Biol 2022; 5:1220. [PMID: 36357561 PMCID: PMC9649734 DOI: 10.1038/s42003-022-04192-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2022] Open
Abstract
We conducted a genome-wide association study in a large population of infertile men due to unexplained spermatogenic failure (SPGF). More than seven million genetic variants were analysed in 1,274 SPGF cases and 1,951 unaffected controls from two independent European cohorts. Two genomic regions were associated with the most severe histological pattern of SPGF, defined by Sertoli cell-only (SCO) phenotype, namely the MHC class II gene HLA-DRB1 (rs1136759, P = 1.32E-08, OR = 1.80) and an upstream locus of VRK1 (rs115054029, P = 4.24E-08, OR = 3.14), which encodes a protein kinase involved in the regulation of spermatogenesis. The SCO-associated rs1136759 allele (G) determines a serine in the position 13 of the HLA-DRβ1 molecule located in the antigen-binding pocket. Overall, our data support the notion of unexplained SPGF as a complex trait influenced by common variation in the genome, with the SCO phenotype likely representing an immune-mediated condition. A GWAS in a large case-control cohort of European ancestry identifies two genomic regions, the MHC class II gene HLA-DRB1 and an upstream locus of VRK1, that are associated with the most severe phenotype of spermatogenic failure.
Collapse
|
5
|
Kerick M, Acosta-Herrera M, Simeón-Aznar CP, Callejas JL, Assassi S, Proudman SM, Nikpour M, Hunzelmann N, Moroncini G, de Vries-Bouwstra JK, Orozco G, Barton A, Herrick AL, Terao C, Allanore Y, Fonseca C, Alarcón-Riquelme ME, Radstake TRDJ, Beretta L, Denton CP, Mayes MD, Martin J. Complement component C4 structural variation and quantitative traits contribute to sex-biased vulnerability in systemic sclerosis. NPJ Genom Med 2022; 7:57. [PMID: 36198672 PMCID: PMC9534873 DOI: 10.1038/s41525-022-00327-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Copy number (CN) polymorphisms of complement C4 play distinct roles in many conditions, including immune-mediated diseases. We investigated the association of C4 CN with systemic sclerosis (SSc) risk. Imputed total C4, C4A, C4B, and HERV-K CN were analyzed in 26,633 individuals and validated in an independent cohort. Our results showed that higher C4 CN confers protection to SSc, and deviations from CN parity of C4A and C4B augmented risk. The protection contributed per copy of C4A and C4B differed by sex. Stronger protection was afforded by C4A in men and by C4B in women. C4 CN correlated well with its gene expression and serum protein levels, and less C4 was detected for both in SSc patients. Conditioned analysis suggests that C4 genetics strongly contributes to the SSc association within the major histocompatibility complex locus and highlights classical alleles and amino acid variants of HLA-DRB1 and HLA-DPB1 as C4-independent signals.
Collapse
Affiliation(s)
- Martin Kerick
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain.
| | - Marialbert Acosta-Herrera
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain.
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs. GRANADA, Granada, Spain.
| | | | | | - Shervin Assassi
- Department of Rheumatology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Susanna M Proudman
- Rheumatology Unit, Royal Adelaide Hospital and University of Adelaide, Adelaide, SA, Australia
| | - Mandana Nikpour
- The University of Melbourne at St. Vincent's Hospital, Melbourne, VIC, Australia
| | | | - Gianluca Moroncini
- Department of Clinical and Molecular Science, Università Politecnica delle Marche e Ospedali Riuniti, Ancona, Italy
| | | | - Gisela Orozco
- Center for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Center, Manchester University NHS Foundation Trust, Manchester, Greater Manchester, UK
| | - Anne Barton
- Center for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Center, Manchester University NHS Foundation Trust, Manchester, Greater Manchester, UK
| | - Ariane L Herrick
- Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Northern care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yannick Allanore
- Department of Rheumatology A, Hospital Cochin, Paris, Île-de-France, France
| | - Carmen Fonseca
- Center for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Marta Eugenia Alarcón-Riquelme
- Center for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Christopher P Denton
- Center for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Maureen D Mayes
- Department of Rheumatology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Javier Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain.
| |
Collapse
|
6
|
Isaacs JD, Brockbank S, Pedersen AW, Hilkens C, Anderson A, Stocks P, Lendrem D, Tarn J, Smith GR, Allen B, Casement J, Diboll J, Harry R, Cooles FAH, Cope AP, Simpson G, Toward R, Noble H, Parke A, Wu W, Clarke F, Scott D, Scott IC, Galloway J, Lempp H, Ibrahim F, Schwank S, Molyneux G, Lazarov T, Geissmann F, Goodyear CS, McInnes IB, Donnelly I, Gilmour A, Virlan AT, Porter D, Ponchel F, Emery P, El-Jawhari J, Parmar R, McDermott MF, Fisher BA, Young SP, Jones P, Raza K, Filer A, Pitzalis C, Barnes MR, Watson DS, Henkin R, Thorborn G, Fossati-Jimack L, Kelly S, Humby F, Bombardieri M, Rana S, Jia Z, Goldmann K, Lewis M, Ng S, Barbosa-Silva A, Tzanis E, Gallagher-Syed A, John CR, Ehrenstein MR, Altobelli G, Martins S, Nguyen D, Ali H, Ciurtin C, Buch M, Symmons D, Worthington J, Bruce IN, Sergeant JC, Verstappen SMM, Stirling F, Hughes-Morley A, Tom B, Farewell V, Zhong Y, Taylor PC, Buckley CD, Keidel S, Cuff C, Levesque M, Long A, Liu Z, Lipsky S, Harvey B, Macoritto M, Hong F, Kaymakcalan S, Tsuji W, Sabin T, Ward N, Talbot S, Padhji D, Sleeman M, Finch D, Herath A, Lindholm C, Jenkins M, Ho M, Hollis S, Marshall C, Parker G, Page M, Edwards H, Cuza A, Gozzard N, Pandis I, Rowe A, Capdevila FB, Loza MJ, Curran M, Verbeeck D, Dan Baker, Mela CM, Vranic I, Mela CT, Wright S, Rowell L, Vernon E, Joseph N, Payne N, Rao R, Binks M, Belson A, Ludbrook V, Hicks K, Tipney H, Ellis J, Hasan S, Didierlaurent A, Burny W, Haynes A, Larminie C, Harris R, Dastros-Pitei D, Carini C, Kola B, Jelinsky S, Hodge M, Maciejewski M, Ziemek D, Schulz-Knappe P, Zucht HD, Budde P, Coles M, Butler JA, Read S. RA-MAP, molecular immunological landscapes in early rheumatoid arthritis and healthy vaccine recipients. Sci Data 2022; 9:196. [PMID: 35534493 PMCID: PMC9085807 DOI: 10.1038/s41597-022-01264-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/04/2022] [Indexed: 11/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder with poorly defined aetiology characterised by synovial inflammation with variable disease severity and drug responsiveness. To investigate the peripheral blood immune cell landscape of early, drug naive RA, we performed comprehensive clinical and molecular profiling of 267 RA patients and 52 healthy vaccine recipients for up to 18 months to establish a high quality sample biobank including plasma, serum, peripheral blood cells, urine, genomic DNA, RNA from whole blood, lymphocyte and monocyte subsets. We have performed extensive multi-omic immune phenotyping, including genomic, metabolomic, proteomic, transcriptomic and autoantibody profiling. We anticipate that these detailed clinical and molecular data will serve as a fundamental resource offering insights into immune-mediated disease pathogenesis, progression and therapeutic response, ultimately contributing to the development and application of targeted therapies for RA.
Collapse
|
7
|
Teruel M, Barturen G, Martínez-Bueno M, Castellini-Pérez O, Barroso-Gil M, Povedano E, Kerick M, Català-Moll F, Makowska Z, Buttgereit A, Pers JO, Marañón C, Ballestar E, Martin J, Carnero-Montoro E, Alarcón-Riquelme ME. Integrative epigenomics in Sjögren´s syndrome reveals novel pathways and a strong interaction between the HLA, autoantibodies and the interferon signature. Sci Rep 2021; 11:23292. [PMID: 34857786 PMCID: PMC8640069 DOI: 10.1038/s41598-021-01324-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Primary Sjögren's syndrome (SS) is a systemic autoimmune disease characterized by lymphocytic infiltration and damage of exocrine salivary and lacrimal glands. The etiology of SS is complex with environmental triggers and genetic factors involved. By conducting an integrated multi-omics study, we confirmed a vast coordinated hypomethylation and overexpression effects in IFN-related genes, what is known as the IFN signature. Stratified and conditional analyses suggest a strong interaction between SS-associated HLA genetic variation and the presence of Anti-Ro/SSA autoantibodies in driving the IFN epigenetic signature and determining SS. We report a novel epigenetic signature characterized by increased DNA methylation levels in a large number of genes enriched in pathways such as collagen metabolism and extracellular matrix organization. We identified potential new genetic variants associated with SS that might mediate their risk by altering DNA methylation or gene expression patterns, as well as disease-interacting genetic variants that exhibit regulatory function only in the SS population. Our study sheds new light on the interaction between genetics, autoantibody profiles, DNA methylation and gene expression in SS, and contributes to elucidate the genetic architecture of gene regulation in an autoimmune population.
Collapse
Affiliation(s)
- María Teruel
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Guillermo Barturen
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Manuel Martínez-Bueno
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Olivia Castellini-Pérez
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Miguel Barroso-Gil
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Elena Povedano
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Martin Kerick
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Francesc Català-Moll
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Zuzanna Makowska
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | - Anne Buttgereit
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | | | - Concepción Marañón
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Martin
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Elena Carnero-Montoro
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
| | - Marta E Alarcón-Riquelme
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
- Institute for Environmental Medicine, Karolinska Institutet, 171 67, Solna, Sweden.
| |
Collapse
|
8
|
Ferreiro-Iglesias A, McKay JD, Brenner N, Virani S, Lesseur C, Gaborieau V, Ness AR, Hung RJ, Liu G, Diergaarde B, Olshan AF, Hayes N, Weissler MC, Schroeder L, Bender N, Pawlita M, Thomas S, Pring M, Dudding T, Kanterewicz B, Ferris R, Thomas S, Brhane Y, Díez-Obrero V, Milojevic M, Smith-Byrne K, Mariosa D, Johansson MJ, Herrero R, Boccia S, Cadoni G, Lacko M, Holcátová I, Ahrens W, Lagiou P, Lagiou A, Polesel J, Simonato L, Merletti F, Healy CM, Hansen BT, Nygård M, Conway DI, Wright S, Macfarlane TV, Robinson M, Alemany L, Agudo A, Znaor A, Amos CI, Waterboer T, Brennan P. Germline determinants of humoral immune response to HPV-16 protect against oropharyngeal cancer. Nat Commun 2021; 12:5945. [PMID: 34642315 PMCID: PMC8511029 DOI: 10.1038/s41467-021-26151-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 09/13/2021] [Indexed: 12/03/2022] Open
Abstract
Although several oropharyngeal cancer (OPC) susceptibility loci have been identified, most previous studies lacked detailed information on human papillomavirus (HPV) status. We conduct a genome-wide analysis by HPV16 serology status in 4,002 oral cancer cases (OPC and oral cavity cancer (OCC)) and 5,256 controls. We detect four susceptibility loci pointing to a distinct genetic predisposition by HPV status. Our most notable finding in the HLA region, that is now confirmed to be specific of HPV(+)OPC risk, reveal two independent loci with strong protective effects, one refining the previously reported HLA class II haplotype association. Antibody levels against HPV16 viral proteins strongly implicate the protective HLA variants as major determinants of humoral response against L1 capsid protein or E6 oncoprotein suggesting a natural immune response against HPV(+)OPC promoted by HLA variants. This indicates that therapeutic vaccines that target E6 and attenuate viral response after established HPV infections might protect against HPV(+)OPC.
Collapse
Affiliation(s)
- Aida Ferreiro-Iglesias
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France.
| | - James D McKay
- Section of Genetics, Genetic Cancer Susceptibility Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Nicole Brenner
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shama Virani
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Corina Lesseur
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valerie Gaborieau
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Andy R Ness
- National Institute for Health Research (NIHR) Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, UK
- Bristol Dental School, University of Bristol, Bristol, UK
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute of Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - Brenda Diergaarde
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Andrew F Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Neil Hayes
- Division of Medical Oncology and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mark C Weissler
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, USA
| | - Lea Schroeder
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Noemi Bender
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Pawlita
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steve Thomas
- Bristol Dental School, University of Bristol, Bristol, UK
| | - Miranda Pring
- Bristol Dental School, University of Bristol, Bristol, UK
| | - Tom Dudding
- Bristol Dental School, University of Bristol, Bristol, UK
| | | | | | - Sera Thomas
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Yonathan Brhane
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Virginia Díez-Obrero
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Maja Milojevic
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Karl Smith-Byrne
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Daniela Mariosa
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Mattias J Johansson
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Rolando Herrero
- Section of Early Detection and Prevention, Prevention and Implementation Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Stefania Boccia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma, Italy
- Department of Woman and Child Health and Public Health - Public Health Area, Fondazione Policlinico Universitario A.Gemelli IRCCS, Roma, Italy
| | - Gabriella Cadoni
- Department of Head and Neck Surgery, Institute of Clinical Otorhinolaryngology, Università Cattolica del Sacro Cuore, Roma, Italy
- Istituto di Clinica Otorinolaringoiatrica, Fondazione Policlinico Universitario A.Gemelli IRCCS, Roma, Italy
| | - Martin Lacko
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ivana Holcátová
- Institute of Hygiene and Epidemiology, Prague, Czech Republic
| | | | - Pagona Lagiou
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Areti Lagiou
- School of Public Health, University of West Attica, Athens, Greece
| | | | | | | | - Claire M Healy
- Trinity College School of Dental Science, Dublin, Ireland
| | | | - Mari Nygård
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - David I Conway
- School of Medicine, Dentistry, and Nursing, University of Glasgow, Glasgow, UK
| | - Sylvia Wright
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | | | - Max Robinson
- Centre for Oral Health Research, Newcastle University, Newcastle, UK
| | - Laia Alemany
- Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública-CIBERESP, Madrid, Spain
| | - Antonio Agudo
- Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
| | - Ariana Znaor
- Cancer Surveillance Section, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Brennan
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France.
| |
Collapse
|
9
|
Acosta-Herrera M, Kerick M, Lopéz-Isac E, Assassi S, Beretta L, Simeón-Aznar CP, Ortego-Centeno N, Proudman SM, Hunzelmann N, Moroncini G, de Vries-Bouwstra JK, Orozco G, Barton A, Herrick AL, Terao C, Allanore Y, Brown MA, Radstake TR, Fonseca C, Denton CP, Mayes MD, Martin J. Comprehensive analysis of the major histocompatibility complex in systemic sclerosis identifies differential HLA associations by clinical and serological subtypes. Ann Rheum Dis 2021; 80:1040-1047. [PMID: 34096881 PMCID: PMC8292594 DOI: 10.1136/annrheumdis-2021-219884] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The greatest genetic effect reported for systemic sclerosis (SSc) lies in the major histocompatibility complex (MHC) locus. Leveraging the largest SSc genome-wide association study, we aimed to fine-map this region to identify novel human leucocyte antigen (HLA) genetic variants associated with SSc susceptibility and its main clinical and serological subtypes. METHODS 9095 patients with SSc and 17 584 controls genome-wide genotyped were used to impute and test single-nucleotide polymorphisms (SNPs) across the MHC, classical HLA alleles and their composite amino acid residues. Additionally, patients were stratified according to their clinical and serological status, namely, limited cutaneous systemic sclerosis (lcSSc), diffuse cutaneous systemic sclerosis (dcSSc), anticentromere (ACA), antitopoisomerase (ATA) and anti-RNApolIII autoantibodies (ARA). RESULTS Sequential conditional analyses showed nine SNPs, nine classical alleles and seven amino acids that modelled the observed associations with SSc. This confirmed previously reported associations with HLA-DRB1*11:04 and HLA-DPB1*13:01, and revealed a novel association of HLA-B*08:01. Stratified analyses showed specific associations of HLA-DQA1*02:01 with lcSSc, and an exclusive association of HLA-DQA1*05:01 with dcSSc. Similarly, private associations were detected in HLA-DRB1*08:01 and confirmed the previously reported association of HLA-DRB1*07:01 with ACA-positive patients, as opposed to the HLA-DPA1*02:01 and HLA-DQB1*03:01 alleles associated with ATA presentation. CONCLUSIONS This study confirms the contribution of HLA class II and reveals a novel association of HLA class I with SSc, suggesting novel pathways of disease pathogenesis. Furthermore, we describe specific HLA associations with SSc clinical and serological subtypes that could serve as biomarkers of disease severity and progression.
Collapse
Affiliation(s)
- Marialbert Acosta-Herrera
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Andalucía, Spain
| | - Martin Kerick
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Andalucía, Spain
| | - Elena Lopéz-Isac
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Andalucía, Spain
| | - Shervin Assassi
- Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | | | | | - Susanna M Proudman
- Department of Rheumatology, Royal Adelaide Hospital, Adelaide, Victoria, Australia
| | | | - Gianluca Moroncini
- Department of Clinical and Molecular Science, Università Politecnica delle Marche and Ospedali Riuniti, Ancona, Italy
| | | | - Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, Greater Manchester, UK
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, Greater Manchester, UK
| | - Ariane L Herrick
- Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yannick Allanore
- Department of Rheumatology A, Hospital Cochin, Paris, Île-de-France, France
| | - Matthew A Brown
- NIHR Biomedical Research Centre, Guy's and Saint Thomas' NHS Foundation Trust and King's College, London, UK
| | - Timothy Rdj Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carmen Fonseca
- Centre for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Christopher P Denton
- Centre for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Maureen D Mayes
- Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Andalucía, Spain
| |
Collapse
|
10
|
Jönsson E, Ljung L, Norrman E, Freyhult E, Ärlestig L, Dahlqvist J, Dahlqvist SR. Pulmonary fibrosis in relation to genetic loci in an inception cohort of patients with early rheumatoid arthritis from northern Sweden. Rheumatology (Oxford) 2021; 61:943-952. [PMID: 33993221 PMCID: PMC8889303 DOI: 10.1093/rheumatology/keab441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/12/2021] [Indexed: 11/14/2022] Open
Abstract
Objectives Pulmonary manifestations in RA are common comorbidities. Interstitial lung disease (ILD), both idiopathic and in RA, has been associated with several genetic variants. We assessed pulmonary fibrosis (PF) in an inception cohort of RA patients in relation to genetic variants and disease-related factors. Methods A total of 1466 early RA patients were consecutively included and followed prospectively from the index date until death or 31 December 2016. Clinical and laboratory data and treatment were continuously registered according to the Swedish Rheumatology Quality Register. DNA was available from 1184 patients and 571 151 genome-wide single-nucleotide polymorphisms (SNPs) were analysed. Thirteen identified genetic variants were extracted. At follow-up, the patients answered a questionnaire regarding disease progression and lung involvement that was validated by reviewing medical records and analysing radiological examinations. Results The prevalence of PF was 5.6% and the annualized incidence rate was 5.0/1000 (95% CI 3.80, 6.54). Four SNPs were associated with PF in RA: rs35705950 [MUC5B; OR 2.5 (95% CI 1.5, 4.0), adjusted P-value = 0.00016, q-value = 0.0021]; rs111521887 [TOLLIP; OR 1.9 (95% CI 1.3, 2.8), adjusted P-value = 0.0014, q-value = 0.0092]; rs2609255 [FAM13A; OR 1.7 (95% CI 1.1, 2.5), adjusted P-value = 0.013, q-value = 0.055] and rs2736100 [TERT; OR 1.5 (95% CI 1.0, 2.2), adjusted P-value = 0.046, q-value = 0.15]. Older age and RF positivity were associated with increased risk, while MTX treatment was associated with a lower risk of PF. Conclusions Development of PF in an inception cohort of RA patients was associated with 4 of 12 ILD risk genes. RA-related factors except for age at diagnosis and RF positivity were of limited importance in PF development.
Collapse
Affiliation(s)
- Elias Jönsson
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Lotta Ljung
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Eva Norrman
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Eva Freyhult
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Lisbeth Ärlestig
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Johanna Dahlqvist
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| | - Solbritt Rantapää Dahlqvist
- Department of Public Health and Medicine/Rheumatology, Umeå University, Umeå, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala.,Department of Medical Biochemistry and Microbiology, and Medical Sciences, Uppsala, University, Uppsala, Sweden
| |
Collapse
|
11
|
Cook S, Choi W, Lim H, Luo Y, Kim K, Jia X, Raychaudhuri S, Han B. Accurate imputation of human leukocyte antigens with CookHLA. Nat Commun 2021; 12:1264. [PMID: 33627654 PMCID: PMC7904773 DOI: 10.1038/s41467-021-21541-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
The recent development of imputation methods enabled the prediction of human leukocyte antigen (HLA) alleles from intergenic SNP data, allowing studies to fine-map HLA for immune phenotypes. Here we report an accurate HLA imputation method, CookHLA, which has superior imputation accuracy compared to previous methods. CookHLA differs from other approaches in that it locally embeds prediction markers into highly polymorphic exons to account for exonic variability, and in that it adaptively learns the genetic map within MHC from the data to facilitate imputation. Our benchmarking with real datasets shows that our method achieves high imputation accuracy in a wide range of scenarios, including situations where the reference panel is small or ethnically unmatched.
Collapse
Affiliation(s)
- Seungho Cook
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine & Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Wanson Choi
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyunjoon Lim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, South Korea
| | - Yang Luo
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kunhee Kim
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine & Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Xiaoming Jia
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Centre for Genetics and Genomics Versus Arthritis, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Buhm Han
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine & Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, South Korea.
| |
Collapse
|
12
|
A cross-disease meta-GWAS identifies four new susceptibility loci shared between systemic sclerosis and Crohn's disease. Sci Rep 2020; 10:1862. [PMID: 32024964 PMCID: PMC7002703 DOI: 10.1038/s41598-020-58741-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified a number of genetic risk loci associated with systemic sclerosis (SSc) and Crohn’s disease (CD), some of which confer susceptibility to both diseases. In order to identify new risk loci shared between these two immune-mediated disorders, we performed a cross-disease meta-analysis including GWAS data from 5,734 SSc patients, 4,588 CD patients and 14,568 controls of European origin. We identified 4 new loci shared between SSc and CD, IL12RB2, IRF1/SLC22A5, STAT3 and an intergenic locus at 6p21.31. Pleiotropic variants within these loci showed opposite allelic effects in the two analysed diseases and all of them showed a significant effect on gene expression. In addition, an enrichment in the IL-12 family and type I interferon signaling pathways was observed among the set of SSc-CD common genetic risk loci. In conclusion, through the first cross-disease meta-analysis of SSc and CD, we identified genetic variants with pleiotropic effects on two clinically distinct immune-mediated disorders. The fact that all these pleiotropic SNPs have opposite allelic effects in SSc and CD reveals the complexity of the molecular mechanisms by which polymorphisms affect diseases.
Collapse
|
13
|
Terao C, Brynedal B, Chen Z, Jiang X, Westerlind H, Hansson M, Jakobsson PJ, Lundberg K, Skriner K, Serre G, Rönnelid J, Mathsson-Alm L, Brink M, Dahlqvist SR, Padyukov L, Gregersen PK, Barton A, Alfredsson L, Klareskog L, Raychaudhuri S. Distinct HLA Associations with Rheumatoid Arthritis Subsets Defined by Serological Subphenotype. Am J Hum Genet 2019; 105:616-624. [PMID: 31474319 PMCID: PMC6731376 DOI: 10.1016/j.ajhg.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 07/29/2019] [Indexed: 01/12/2023] Open
Abstract
Rheumatoid arthritis (RA) is the most common immune-mediated arthritis. Anti-citrullinated peptide antibodies (ACPA) are highly specific to RA and assayed with the commercial CCP2 assay. Genetic drivers of RA within the MHC are different for CCP2-positive and -negative subsets of RA, particularly at HLA-DRB1. However, aspartic acid at amino acid position 9 in HLA-B (Bpos-9) increases risk to both RA subsets. Here we explore how individual serologies associated with RA drive associations within the MHC. To define MHC differences for specific ACPA serologies, we quantified a total of 19 separate ACPAs in RA-affected case subjects from four cohorts (n = 6,805). We found a cluster of tightly co-occurring antibodies (canonical serologies, containing CCP2), along with several independently expressed antibodies (non-canonical serologies). After imputing HLA variants into 6,805 case subjects and 13,467 control subjects, we tested associations between the HLA region and RA subgroups based on the presence of canonical and/or non-canonical serologies. We examined CCP2(+) and CCP2(−) RA-affected case subjects separately. In CCP2(−) RA, we observed that the association between CCP2(−) RA and Bpos-9 was derived from individuals who were positive for non-canonical serologies (omnibus_p = 9.2 × 10−17). Similarly, we observed in CCP2(+) RA that associations between subsets of CCP2(+) RA and Bpos-9 were negatively correlated with the number of positive canonical serologies (p = 0.0096). These findings suggest unique genetic characteristics underlying fine-specific ACPAs, suggesting that RA may be further subdivided beyond simply seropositive and seronegative.
Collapse
|
14
|
Vadva Z, Larsen CE, Propp BE, Trautwein MR, Alford DR, Alper CA. A New Pedigree-Based SNP Haplotype Method for Genomic Polymorphism and Genetic Studies. Cells 2019; 8:E835. [PMID: 31387299 PMCID: PMC6721696 DOI: 10.3390/cells8080835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) are usually the most frequent genomic variants. Directly pedigree-phased multi-SNP haplotypes provide a more accurate view of polymorphic population genomic structure than individual SNPs. The former are, therefore, more useful in genetic correlation with subject phenotype. We describe a new pedigree-based methodology for generating non-ambiguous SNP haplotypes for genetic study. SNP data for haplotype analysis were extracted from a larger Type 1 Diabetes Genetics Consortium SNP dataset based on minor allele frequency variation and redundancy, coverage rate (the frequency of phased haplotypes in which each SNP is defined) and genomic location. Redundant SNPs were eliminated, overall haplotype polymorphism was optimized and the number of undefined haplotypes was minimized. These edited SNP haplotypes from a region containing HLA-DRB1 (DR) and HLA-DQB1 (DQ) both correlated well with HLA-typed DR,DQ haplotypes and differentiated HLA-DR,DQ fragments shared by three pairs of previously identified megabase-length conserved extended haplotypes. In a pedigree-based genetic association assay for type 1 diabetes, edited SNP haplotypes and HLA-typed HLA-DR,DQ haplotypes from the same families generated essentially identical qualitative and quantitative results. Therefore, this edited SNP haplotype method is useful for both genomic polymorphic architecture and genetic association evaluation using SNP markers with diverse minor allele frequencies.
Collapse
Affiliation(s)
- Zareen Vadva
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Charles E Larsen
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Bennett E Propp
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Michael R Trautwein
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Dennis R Alford
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Chester A Alper
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
15
|
González-Serna D, López-Isac E, Yilmaz N, Gharibdoost F, Jamshidi A, Kavosi H, Poursani S, Farsad F, Direskeneli H, Saruhan-Direskeneli G, Vargas S, Sawalha AH, Brown MA, Yavuz S, Mahmoudi M, Martin J. Analysis of the genetic component of systemic sclerosis in Iranian and Turkish populations through a genome-wide association study. Rheumatology (Oxford) 2019; 58:289-298. [PMID: 30247649 DOI: 10.1093/rheumatology/key281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Indexed: 12/13/2022] Open
Abstract
Objectives SSc is an autoimmune disease characterized by alteration of the immune response, vasculopathy and fibrosis. Most genetic studies on SSc have been performed in European-ancestry populations. The aim of this study was to analyse the genetic component of SSc in Middle Eastern patients from Iran and Turkey through a genome-wide association study. Methods This study analysed data from a total of 834 patients diagnosed with SSc and 1455 healthy controls from Iran and Turkey. DNA was genotyped using high-throughput genotyping platforms. The data generated were imputed using the Michigan Imputation Server, and the Haplotype Reference Consortium as a reference panel. A meta-analysis combining both case-control sets was conducted by the inverse variance method. Results The highest peak of association belonged to the HLA region in both the Iranian and Turkish populations. Strong and independent associations between the classical alleles HLA-DRB1*11: 04 [P = 2.10 × 10-24, odds ratio (OR) = 3.14] and DPB1*13: 01 (P = 5.37 × 10-14, OR = 5.75) and SSc were observed in the Iranian population. HLA-DRB1*11: 04 (P = 4.90 × 10-11, OR = 2.93) was the only independent signal associated in the Turkish cohort. An omnibus test yielded HLA-DRB1 58 and HLA-DPB1 76 as relevant amino acid positions for this disease. Concerning the meta-analysis, we also identified two associations close to the genome-wide significance level outside the HLA region, corresponding to IRF5-TNPO3 rs17424921-C (P = 1.34 × 10-7, OR = 1.68) and NFKB1 rs4648133-C (P = 3.11 × 10-7, OR = 1.47). Conclusion We identified significant associations in the HLA region and suggestive associations in IRF5-TNPO3 and NFKB1 loci in Iranian and Turkish patients affected by SSc through a genome-wide association study and an extensive HLA analysis.
Collapse
Affiliation(s)
- David González-Serna
- Cell Biology and Immunology Department, Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena López-Isac
- Cell Biology and Immunology Department, Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Neslihan Yilmaz
- Department of Rheumatology, Istanbul Bilim University, Istanbul, Turkey
| | - Farhad Gharibdoost
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Kavosi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Poursani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Faraneh Farsad
- Department of Rheumatology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Sofia Vargas
- Cell Biology and Immunology Department, Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, Ann Arbor MI, USA
- Department of Computational Medicine and Bioinformatics, The Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI, USA
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Queensland, Australia
| | - Sule Yavuz
- Department of Rheumatology, Istanbul Bilim University, Istanbul, Turkey
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Javier Martin
- Cell Biology and Immunology Department, Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
16
|
Ferreiro-Iglesias A, Lesseur C, McKay J, Hung RJ, Han Y, Zong X, Christiani D, Johansson M, Xiao X, Li Y, Qian DC, Ji X, Liu G, Caporaso N, Scelo G, Zaridze D, Mukeriya A, Kontic M, Ognjanovic S, Lissowska J, Szołkowska M, Swiatkowska B, Janout V, Holcatova I, Bolca C, Savic M, Ognjanovic M, Bojesen SE, Wu X, Albanes D, Aldrich MC, Tardon A, Fernandez-Somoano A, Fernandez-Tardon G, Le Marchand L, Rennert G, Chen C, Doherty J, Goodman G, Bickeböller H, Wichmann HE, Risch A, Rosenberger A, Shen H, Dai J, Field JK, Davies M, Woll P, Teare MD, Kiemeney LA, van der Heijden EHFM, Yuan JM, Hong YC, Haugen A, Zienolddiny S, Lam S, Tsao MS, Johansson M, Grankvist K, Schabath MB, Andrew A, Duell E, Melander O, Brunnström H, Lazarus P, Arnold S, Slone S, Byun J, Kamal A, Zhu D, Landi MT, Amos CI, Brennan P. Fine mapping of MHC region in lung cancer highlights independent susceptibility loci by ethnicity. Nat Commun 2018; 9:3927. [PMID: 30254314 PMCID: PMC6156406 DOI: 10.1038/s41467-018-05890-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
The basis for associations between lung cancer and major histocompatibility complex genes is not completely understood. Here the authors further consider genetic variation within the MHC region in lung cancer patients and identify independent associations within HLA genes that explain MHC lung cancer associations in Europeans and Asian populations. Lung cancer has several genetic associations identified within the major histocompatibility complex (MHC); although the basis for these associations remains elusive. Here, we analyze MHC genetic variation among 26,044 lung cancer patients and 20,836 controls densely genotyped across the MHC, using the Illumina Illumina OncoArray or Illumina 660W SNP microarray. We impute sequence variation in classical HLA genes, fine-map MHC associations for lung cancer risk with major histologies and compare results between ethnicities. Independent and novel associations within HLA genes are identified in Europeans including amino acids in the HLA-B*0801 peptide binding groove and an independent HLA-DQB1*06 loci group. In Asians, associations are driven by two independent HLA allele sets that both increase risk in HLA-DQB1*0401 and HLA-DRB1*0701; the latter better represented by the amino acid Ala-104. These results implicate several HLA–tumor peptide interactions as the major MHC factor modulating lung cancer susceptibility.
Collapse
Affiliation(s)
- Aida Ferreiro-Iglesias
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France
| | - Corina Lesseur
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France
| | - James McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Sinai Health System, University of Toronto, Toronto, M5G 1X5, Canada
| | - Younghun Han
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Xuchen Zong
- Lunenfeld-Tanenbaum Research Institute of Sinai Health System, University of Toronto, Toronto, M5G 1X5, Canada
| | - David Christiani
- Department of Environmental Health, Harvard TH Chan School of Public Health, Massachusetts General Hospital/ Harvard Medical School, Boston, 02115, MA, USA
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France
| | - Xiangjun Xiao
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Yafang Li
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - David C Qian
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Xuemei Ji
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute of Sinai Health System, University of Toronto, Toronto, M5G 1X5, Canada
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892-9768, MD, USA
| | - Ghislaine Scelo
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France
| | - David Zaridze
- Russian N.N. Blokhin Cancer Research Centre, Moscow, 115478, Russian Federation
| | - Anush Mukeriya
- Russian N.N. Blokhin Cancer Research Centre, Moscow, 115478, Russian Federation
| | | | - Simona Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, 11070, Serbia
| | - Jolanta Lissowska
- M. Sklodowska-Curie Cancer Center, Institute of Oncology, Warsaw, 02-034, Poland
| | - Małgorzata Szołkowska
- Department of Pathology, National Tuberculosis and Lung Diseases Research Institute, Warsaw, 01-138, Poland
| | - Beata Swiatkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, 91-348, Poland
| | - Vladimir Janout
- Faculty of Medicine, University of Olomouc, Olomouc, 701 03, Czech Republic
| | - Ivana Holcatova
- 2nd Faculty of Medicine, Institute of Public Health and Preventive Medicine, Charles University, Prague, CZ 128 00, Czech Republic
| | - Ciprian Bolca
- Institute of Pneumology "Marius Nasta", Bucharest, RO-050159, Romania
| | - Milan Savic
- Department of Thoracic Surgery Clinical Center of Serbia Belgrade, Belgrade, 11000, Serbia
| | - Miodrag Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, 11070, Serbia
| | - Stig Egil Bojesen
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, 2730, Denmark.,Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2730, Denmark
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892-9768, MD, USA
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, 37232-4682, TA, USA
| | - Adonina Tardon
- University of Oviedo and CIBERESP, Faculty of Medicine, Oviedo, 33006, Spain
| | | | | | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, 96813, HI, USA
| | - Gadi Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, 3525433, Israel
| | - Chu Chen
- Department of Epidemiology, University of Washington School of Public Health and Community Medicine, Seattle, 98195, WA, USA
| | - Jennifer Doherty
- Department of Epidemiology, University of Washington School of Public Health and Community Medicine, Seattle, 98195, WA, USA.,Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | | | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, 37073, Germany
| | - H-Erich Wichmann
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig Maximilians University, Munich, D-85764, Germany.,Helmholtz Center Munich, Institute of Epidemiology 2, Munich, D-85764, Germany.,Institute of Medical Statistics and Epidemiology, Technical University Munich, Munich, D-80333, Germany
| | - Angela Risch
- University of Salzburg and Cancer Cluster Salzburg, Salzburg, 5020, Austria.,Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, 69120, Germany.,German Center for Lung Research (DZL), Heidelberg, 69121, Germany
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, 37073, Germany
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Jiangsu Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - John K Field
- Institute of Translational Medicine, University of Liverpool, Liverpool, L3 9TA, UK
| | - Michael Davies
- Institute of Translational Medicine, University of Liverpool, Liverpool, L3 9TA, UK
| | - Penella Woll
- Department of Oncology, University of Sheffield, Sheffield, S10 2RX, UK
| | - M Dawn Teare
- School of Health and Related Research, University Of Sheffield, England, S1 4DA, UK
| | | | | | - Jian-Min Yuan
- University of Pittsburgh Cancer Institute, Pittsburgh, 15232, PA, USA
| | - Yun-Chul Hong
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Aage Haugen
- National Institute of Occupational Health, Oslo, N-0033, Norway
| | | | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, V5Z 1M9, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada
| | - Mikael Johansson
- Department of Radiation Sciences, Umeå University, Umeå, 901 85, Sweden
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, 901 85, Sweden
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, 33612, FL, USA
| | - Angeline Andrew
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Eric Duell
- Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, 08908, Spain
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, 221 00, Sweden.,Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Hans Brunnström
- Laboratory Medicine Region Skåne, Department of Clinical Sciences Lund, Pathology, Lund University, Lund, 221 00, Sweden
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, 99202, WA, USA
| | - Susanne Arnold
- University of Kentucky, Markey Cancer Center, Lexington, 40536-0098, KY, USA
| | - Stacey Slone
- University of Kentucky, Markey Cancer Center, Lexington, 40536-0098, KY, USA
| | - Jinyoung Byun
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Ahsan Kamal
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Dakai Zhu
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892-9768, MD, USA
| | - Christopher I Amos
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03755, NH, USA
| | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 cedex 08, France.
| |
Collapse
|
17
|
Qin N, Wang C, Zhu M, Lu Q, Ma Z, Huang M, Dai J, Ma H, Jin G, Hu Z, Shen H. Fine-mapping the MHC region in Asian populations identified novel variants modifying susceptibility to lung cancer. Lung Cancer 2017; 112:169-175. [PMID: 29191591 DOI: 10.1016/j.lungcan.2017.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/04/2017] [Accepted: 08/16/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVES The polymorphic major histocompatibility complex (MHC) plays a vital role in the immune system and drives predisposition to multiple cancers. A number of lung cancer-related genetic variants in the MHC have been identified in recent genome-wide association studies; however, the causal variants remain unclear. MATERIALS AND METHODS In the present study, we conducted a large-scale fine-mapping study of lung cancer in the MHC region of 13,945 unrelated Asian individuals to search for potential causal variants. We used the recently constructed Pan-Asian panel as the reference and imputed eight HLA genes (HLA-A, HLC-B, HLA-C, HLA-DRB1, HLA-DQA1, HLA-DQB1, HLA-DPA1, and HLA-DPB1) using SNP2HLA software. RESULTS We identified one single nucleotide polymorphism, rs12333226 (OR=1.41, P=3.97×10-7), five HLA amino acid polymorphisms in HLA-DRB1 (OR=0.89, P=7.51×10-6-8.57×10-6), and one two-digit classic HLA allele HLA-A*11 (OR=0.87, P=9.68×10-6) that were strongly associated with the risk of lung cancer. Rs12333226 was an expression quantitative trait locus of HLA-A and HLA-H in circulating monocytes, and exerted effect on lung cancer risk especially in the younger. HLA-DRβ1 positions 10, 16, and 25 drove the effect of one reported SNP rs2395185. The peptide position analysis identified additional lung cancer susceptibility amino acid positions, including HLA-DRβ1 position 30 and 11 (Pomnibus=6.11×10-5 and 6.91×10-5), HLA-DQa1 47 and 76 (Pomnibus=3.96×10-4 and 1.41×10-2) and HLA-A 152 (Pomnibus=4.86×10-4). Most of the peptide positions were located in the peptide-binding grooves and seemed to affect antigen presentation. All the existing and novel variants explained approximately 2.37% of the phenotypic variances, while 21.10% was attributed to the variants identified in this study. CONCLUSION We identified seven novel bi-allelic variants and five polymorphic amino acid positions in HLA-DRβ1, HLA-DQα1, and HLA-A that confer a risk of lung cancer. This finding provides evidence for the substantial contributions of HLA class I and II molecules to lung cancer susceptibility.
Collapse
Affiliation(s)
- Na Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China; Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211116, China
| | - Meng Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Qun Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Zijian Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Mingtao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
18
|
Ludwig KU, Böhmer AC, Bowes J, Nikolic M, Ishorst N, Wyatt N, Hammond NL, Gölz L, Thieme F, Barth S, Schuenke H, Klamt J, Spielmann M, Aldhorae K, Rojas-Martinez A, Nöthen MM, Rada-Iglesias A, Dixon MJ, Knapp M, Mangold E. Imputation of orofacial clefting data identifies novel risk loci and sheds light on the genetic background of cleft lip ± cleft palate and cleft palate only. Hum Mol Genet 2017; 26:829-842. [PMID: 28087736 PMCID: PMC5409059 DOI: 10.1093/hmg/ddx012] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/09/2017] [Indexed: 12/22/2022] Open
Abstract
Nonsyndromic cleft lip with or without cleft palate (nsCL/P) is among the most common human birth defects with multifactorial etiology. Here, we present results from a genome-wide imputation study of nsCL/P in which, after adding replication cohort data, four novel risk loci for nsCL/P are identified (at chromosomal regions 2p21, 14q22, 15q24 and 19p13). On a systematic level, we show that the association signals within this high-density dataset are enriched in functionally-relevant genomic regions that are active in both human neural crest cells (hNCC) and mouse embryonic craniofacial tissue. This enrichment is also detectable in hNCC regions primed for later activity. Using GCTA analyses, we suggest that 30% of the estimated variance in risk for nsCL/P in the European population can be attributed to common variants, with 25.5% contributed to by the 24 risk loci known to date. For each of these, we identify credible SNPs using a Bayesian refinement approach, with two loci harbouring only one probable causal variant. Finally, we demonstrate that there is no polygenic component of nsCL/P detectable that is shared with nonsyndromic cleft palate only (nsCPO). Our data suggest that, while common variants are strongly contributing to risk for nsCL/P, they do not seem to be involved in nsCPO which might be more often caused by rare deleterious variants. Our study generates novel insights into both nsCL/P and nsCPO etiology and provides a systematic framework for research into craniofacial development and malformation.
Collapse
Affiliation(s)
- Kerstin U Ludwig
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Anne C Böhmer
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - John Bowes
- Arthritis Research UK Centre for Genetics and Genomics, The University of Manchester, Manchester M13 9PT, UK
| | - Miloš Nikolic
- Center for Molecular Medicine Cologne.,Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Nina Ishorst
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Niki Wyatt
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Nigel L Hammond
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Lina Gölz
- Department of Orthodontics, University of Bonn, Bonn 53111, Germany
| | - Frederic Thieme
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Sandra Barth
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Hannah Schuenke
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Johanna Klamt
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, RG Development and Disease, Berlin 14195, Germany.,Institute for Medical and Human Genetics.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Khalid Aldhorae
- Orthodontic Department, College of Dentistry, Thamar University, Thamar, Yemen
| | - Augusto Rojas-Martinez
- Tecnologico de Monterrey, School of Medicine, and Universidad Autonoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey 64460, Mexico
| | - Markus M Nöthen
- Institute of Human Genetics University of Bonn, Bonn 53127, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn 53127, Germany
| | - Alvaro Rada-Iglesias
- Center for Molecular Medicine Cologne.,Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Michael J Dixon
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Michael Knapp
- Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn 53127, Germany
| | | |
Collapse
|
19
|
Hinks A, Bowes J, Cobb J, Ainsworth HC, Marion MC, Comeau ME, Sudman M, Han B, Becker ML, Bohnsack JF, de Bakker PIW, Haas JP, Hazen M, Lovell DJ, Nigrovic PA, Nordal E, Punnaro M, Rosenberg AM, Rygg M, Smith SL, Wise CA, Videm V, Wedderburn LR, Yarwood A, Yeung RSM, Prahalad S, Langefeld CD, Raychaudhuri S, Thompson SD, Thomson W. Fine-mapping the MHC locus in juvenile idiopathic arthritis (JIA) reveals genetic heterogeneity corresponding to distinct adult inflammatory arthritic diseases. Ann Rheum Dis 2016; 76:765-772. [PMID: 27998952 PMCID: PMC5530326 DOI: 10.1136/annrheumdis-2016-210025] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/12/2016] [Accepted: 11/05/2016] [Indexed: 11/06/2022]
Abstract
Objectives Juvenile idiopathic arthritis (JIA) is a heterogeneous group of diseases, comprising seven categories. Genetic data could potentially be used to help redefine JIA categories and improve the current classification system. The human leucocyte antigen (HLA) region is strongly associated with JIA. Fine-mapping of the region was performed to look for similarities and differences in HLA associations between the JIA categories and define correspondences with adult inflammatory arthritides. Methods Dense genotype data from the HLA region, from the Immunochip array for 5043 JIA cases and 14 390 controls, were used to impute single-nucleotide polymorphisms, HLA classical alleles and amino acids. Bivariate analysis was performed to investigate genetic correlation between the JIA categories. Conditional analysis was used to identify additional effects within the region. Comparison of the findings with those in adult inflammatory arthritic diseases was performed. Results We identified category-specific associations and have demonstrated for the first time that rheumatoid factor (RF)-negative polyarticular JIA and oligoarticular JIA are genetically similar in their HLA associations. We also observe that each JIA category potentially has an adult counterpart. The RF-positive polyarthritis association at HLA-DRB1 amino acid at position 13 mirrors the association in adult seropositive rheumatoid arthritis (RA). Interestingly, the combined oligoarthritis and RF-negative polyarthritis dataset shares the same association with adult seronegative RA. Conclusions The findings suggest the value of using genetic data in helping to classify the categories of this heterogeneous disease. Mapping JIA categories to adult counterparts could enable shared knowledge of disease pathogenesis and aetiology and facilitate transition from paediatric to adult services.
Collapse
Affiliation(s)
- A Hinks
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK
| | - J Bowes
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK
| | - J Cobb
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - H C Ainsworth
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - M C Marion
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - M E Comeau
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - M Sudman
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - B Han
- Divisions of Genetics and Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Department of Convergence Medicine, University of Ulsan College of Medicine & Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | | | - M L Becker
- Division of Rheumatology and Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy-Kansas City, Kansas City, Missouri, USA
| | - J F Bohnsack
- Division of Allergy, Immunology and Paediatric Rheumatology, University of Utah, Salt Lake City, Utah, USA
| | - P I W de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J P Haas
- German Center for Pediatric and Adolescent Rheumatology, Garmisch-Partenkirchen, Germany
| | - M Hazen
- Division of Immunology, Department of Rheumatology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - D J Lovell
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - P A Nigrovic
- Division of Immunology, Department of Rheumatology, Boston Children's Hospital, Boston, Massachusetts, USA.,Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - E Nordal
- Department of Paediatrics, University Hospital of North Norway, and UIT The Arctic University of Norway, Tromsø, Norway
| | - M Punnaro
- Arthritis Clinic Texas Scottish Rite Hospital for Children, Dallas, Texas, USA.,Department of Paediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - A M Rosenberg
- Division of Rheumatology, Department of Paediatrics, University of Saskatchewan, Saskatoon, Canada
| | - M Rygg
- Department of Laboratory Medicine, Children's and Women's Health, NTNU - Norwegian University of Science and Technology, and St. Olavs University Hospital, Trondheim, Norway
| | - S L Smith
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK
| | - C A Wise
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA.,Department of Orthopaedic Surgery, Paediatrics, and McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, USA
| | - V Videm
- Department of Laboratory Medicine, Children's and Women's Health, NTNU - Norwegian University of Science and Technology, and St. Olavs University Hospital, Trondheim, Norway
| | - L R Wedderburn
- Arthritis Research UK Centre for Adolescent Rheumatology, UCL GOS Institute of Child Health, University College London, London, UK.,NIHR-Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - A Yarwood
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK
| | - R S M Yeung
- The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - S Prahalad
- Department of Paediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, USA
| | - C D Langefeld
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - S Raychaudhuri
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK.,Divisions of Genetics and Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, USA.,Department of Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - S D Thompson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - W Thomson
- Arthritis Research UK Centre for Genetics and Genomics, Manchester Academic Health Science Centre, University Of Manchester, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
20
|
Márquez A, Cordero-Coma M, Martín-Villa JM, Gorroño-Echebarría MB, Blanco R, Díaz Valle D, Del Rio MJ, Blanco A, Olea JL, Cordero Y, Capella MJ, Díaz-Llopis M, Ortego-Centeno N, Ruiz-Arruza I, Llorenç V, Adán A, Fonollosa A, Ten Berge J, Atan D, Dick AD, De Boer JH, Kuiper J, Rothova A, Martín J. New insights into the genetic component of non-infectious uveitis through an Immunochip strategy. J Med Genet 2016; 54:38-46. [PMID: 27609017 DOI: 10.1136/jmedgenet-2016-104144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/16/2016] [Accepted: 08/20/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Large-scale genetic studies have reported several loci associated with specific disorders involving uveitis. Our aim was to identify genetic risk factors that might predispose to uveitis per se, independent of the clinical diagnosis, by performing a dense genotyping of immune-related loci. METHODS 613 cases and 3693 unaffected controls from three European case/control sets were genotyped using the Immunochip array. Only patients with non-infectious non-anterior uveitis and without systemic features were selected. To perform a more comprehensive analysis of the human leucocyte antigen (HLA) region, SNPs, classical alleles and polymorphic amino acid variants were obtained via imputation. A meta-analysis combining the three case/control sets was conducted by the inverse variance method. RESULTS The highest peak belonged to the HLA region. A more detailed analysis of this signal evidenced a strong association between the classical allele HLA-A*2902 and birdshot chorioretinopathy (p=3.21E-35, OR=50.95). An omnibus test yielded HLA-A 62 and 63 as relevant amino acid positions for this disease. In patients with intermediate and posterior uveitis, the strongest associations belonged to the rs7197 polymorphism, within HLA-DRA (p=2.07E-11, OR=1.99), and the HLA-DR15 haplotype (DRB1*1501: p=1.16E-10, OR=2.08; DQA1*0102: p=4.37E-09, OR=1.77; DQB1*0602: p=7.26E-10, OR=2.02). Outside the HLA region, the MAP4K4/IL1R2 locus reached statistical significance (rs7608679: p=8.38E-07, OR=1.42). Suggestive associations were found at five other loci. CONCLUSIONS We have further interrogated the association between the HLA region and non-infectious non-anterior uveitis. In addition, we have identified a new non-HLA susceptibility factor and proposed additional risk loci with putative roles in this complex condition.
Collapse
Affiliation(s)
- Ana Márquez
- Instituto de Parasitología y Biomedicina "López-Neyra", CSIC, PTS Granada, Granada, Spain
| | - Miguel Cordero-Coma
- Ophthalmology Department, Hospital de León, IBIOMED, Universidad de León, León, Spain
| | | | | | - Ricardo Blanco
- Rheumatology Department, Hospital Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - David Díaz Valle
- Ophthalmology Department, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Ana Blanco
- Ophthalmology Department, Hospital Donostia, San Sebastián (Guipúzcoa), Spain
| | - Jose Luis Olea
- Ophthalmology Department, Hospital Son Espases, Palma de Mallorca, Spain
| | - Yolanda Cordero
- Ophthalmology Department, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - María José Capella
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Díaz-Llopis
- Ophthalmology Department, Hospital La Fe, Universidad de Valencia, Valencia, Spain
| | | | - Ioana Ruiz-Arruza
- Autoimmune Diseases Research Unit, Internal Medicine Department, BioCruces Health Research Institute, Hospital Universitario Cruces, Barakaldo, Spain
| | - Víctor Llorenç
- Ophthalmology Department, Hospital Clinic, Barcelona, Spain
| | - Alfredo Adán
- Ophthalmology Department, Hospital Clinic, Barcelona, Spain
| | - Alejandro Fonollosa
- Ophthalmology Department, BioCruces Health Research Institute, Hospital Universitario Cruces, Barakaldo, Spain
| | - Josianne Ten Berge
- Department of Ophthalmology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Denize Atan
- School of Clinical Sciences, Bristol Eye Hospital, Bristol, UK
| | - Andrew D Dick
- School of Clinical Sciences, Bristol Eye Hospital, Bristol, UK
| | - Joke H De Boer
- Department of Ophthalmology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jonas Kuiper
- Department of Ophthalmology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Aniki Rothova
- Department of Ophthalmology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Javier Martín
- Instituto de Parasitología y Biomedicina "López-Neyra", CSIC, PTS Granada, Granada, Spain
| |
Collapse
|
21
|
Ortiz-Fernández L, Carmona FD, Montes-Cano MA, García-Lozano JR, Conde-Jaldón M, Ortego-Centeno N, Castillo MJ, Espinosa G, Graña-Gil G, Sánchez-Bursón J, Juliá MR, Solans R, Blanco R, Barnosi-Marín AC, Gómez de la Torre R, Fanlo P, Rodríguez-Carballeira M, Rodríguez-Rodríguez L, Camps T, Castañeda S, Alegre-Sancho JJ, Martín J, González-Escribano MF. Genetic Analysis with the Immunochip Platform in Behçet Disease. Identification of Residues Associated in the HLA Class I Region and New Susceptibility Loci. PLoS One 2016; 11:e0161305. [PMID: 27548383 PMCID: PMC4993481 DOI: 10.1371/journal.pone.0161305] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
Behcet's disease (BD) is an immuno-mediated vasculitis in which knowledge of its etiology and genetic basis is limited. To improve the current knowledge, a genetic analysis performed with the Immunochip platform was carried out in a population from Spain. A discovery cohort comprising 278 BD cases and 1,517 unaffected controls were genotyped using the Immunochip platform. The validation step was performed on an independent replication cohort composed of 130 BD cases and 600 additional controls. The strongest association signals were observed in the HLA class I region, being HLA-B*51 the highest peak (overall P = 6.82E-32, OR = 3.82). A step-wise conditional logistic regression with classical alleles identified HLA-B*57 and HLA-A*03 as additional independent markers. The amino acid model that best explained the association, includes the position 97 of the HLA-B molecule and the position 66 of the HLA-A. Among the non-HLA loci, the most significant in the discovery analysis were: IL23R (rs10889664: P = 3.81E-12, OR = 2.00), the JRKL/CNTN5 region (rs2848479: P = 5.00E-08, OR = 1.68) and IL12A (rs1874886: P = 6.67E-08, OR = 1.72), which were confirmed in the validation phase (JRKL/CNTN5 rs2848479: P = 3.29E-10, OR = 1.66; IL12A rs1874886: P = 1.62E-08, OR = 1.61). Our results confirm HLA-B*51 as a primary-association marker in predisposition to BD and suggest additional independent signals within the class I region, specifically in the genes HLA-A and HLA-B. Regarding the non-HLA genes, in addition to IL-23R, previously reported in our population; IL12A, described in other populations, was found to be a BD susceptibility factor also in Spaniards; finally, a new associated locus was found in the JRKL/CNTN5 region.
Collapse
Affiliation(s)
- Lourdes Ortiz-Fernández
- Department of Immunology, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Sevilla, 41013, Spain
| | - Francisco-David Carmona
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, PTS Granada, Granada, 18016, Spain
| | - Marco-Antonio Montes-Cano
- Department of Immunology, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Sevilla, 41013, Spain
| | - José-Raúl García-Lozano
- Department of Immunology, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Sevilla, 41013, Spain
| | - Marta Conde-Jaldón
- Department of Immunology, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Sevilla, 41013, Spain
| | | | - María Jesús Castillo
- Department of Internal Medicine, Hospital Universitario Virgen del Rocío, Sevilla, 41003, Spain
| | - Gerard Espinosa
- Department Autoimmune Diseases, Hospital Universitari Clínic, Barcelona, 08036, Spain
| | - Genaro Graña-Gil
- Department of Rheumatology, Complejo Hospitalario Universitario A Coruña, A Coruña, 15006, Spain
| | - Juan Sánchez-Bursón
- Department of Rheumatology, Hospital Universitario de Valme, Sevilla, 41014, Spain
| | - María Rosa Juliá
- Department of Immunology, Hospital Universitari Son Espases, Palma de Mallorca, 07120, Spain
| | - Roser Solans
- Department of Internal Medicine, Autoimmune Systemic Diseases Unit, Hospital Vall d’Hebron, Universidad Autonoma de Barcelona, Barcelona, 08035, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008, Spain
| | | | | | - Patricia Fanlo
- Department of Internal Medicine, Hospital Virgen del Camino, Pamplona, 31008, Spain
| | | | | | - Teresa Camps
- Department of Internal Medicine, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain
| | - Santos Castañeda
- Department of Rheumatology, Hospital de la Princesa, IIS-Princesa, Madrid, 28006, Spain
| | | | - Javier Martín
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, PTS Granada, Granada, 18016, Spain
| | | |
Collapse
|
22
|
Pociot F, Kaur S, Nielsen LB. Effects of the genome on immune regulation in type 1 diabetes. Pediatr Diabetes 2016; 17 Suppl 22:37-42. [PMID: 27411435 DOI: 10.1111/pedi.12336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/12/2015] [Indexed: 12/26/2022] Open
Abstract
Type 1 diabetes (T1DM) is a complex disease, arising through the interaction of an incompletely defined combination of genetic susceptibility and environmental factors. It is well accepted that T1DM results from selective immune-mediated destruction of the insulin-producing β cells in the islets of langerhans. Genetic studies of T1DM have identified several regions of susceptibility and identified major networks and pathways contributing to risk. In this study, we have taken advantages of the Immunochip fine-mapping genotyping data to address different aspects of immune regulation in relation to T1DM. First, we confirm that dense single nucleotide polymorphism (SNP) genotyping of the major histocompatibility complex/human leukocyte antigen (MHC/HLA) region capture the complex genetic contribution of this region to disease risk. Furthermore, it is shown that Immunochip genotyping can translate into a limited number of DRB1 and DQB1 amino acid residues that account for most of the HLA-risk. Second, we use the Immunochip data to look for functional significance by correlation to circulating levels of chemokines and demonstrate that genetic variation at chromosome 2, 3, and 6 correlates with circulating CCL2 and CCL4 in recent onset T1DM patients. Finally, we report that genetic variants predict autoantibody positivity in T1DM cases.
Collapse
Affiliation(s)
- Flemming Pociot
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Simranjeet Kaur
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Lotte B Nielsen
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| |
Collapse
|
23
|
Akolkar B, Hilner J, Nierras CR. Design and Measurement of Nonislet-Specific Autoantibodies for the Type 1 Diabetes Genetics Consortium Autoantibody Workshop. Diabetes Care 2015; 38 Suppl 2:S4-7. [PMID: 26405071 PMCID: PMC4582913 DOI: 10.2337/dcs15-2002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Type 1 Diabetes Genetics Consortium (T1DGC) comprised groups of investigators from many countries throughout the world, with a common goal of identifying genes predisposing to type 1 diabetes. The T1DGC ascertained and collected samples from families with two or more affected siblings with type 1 diabetes and generated a broad array of clinical, genetic, and immunologic data. The T1DGC Autoantibody Workshop was designed to distribute data for analyses to discover genes associated with autoantibodies in those with type 1 diabetes. In the T1DGC-affected sibling pair families, three T1DGC Network laboratories measured antibodies to the islet autoantigens GAD65 and the intracellular portion of protein tyrosine phosphatase (IA-2A). The availability of extensive genetic data provided an opportunity to investigate the associations between type 1 diabetes and other autoimmune diseases for which autoantibodies could be measured. Measurements of additional nonislet autoantibodies, including thyroid peroxidase, tissue transglutaminase, 21-hydroxylase, and the potassium/hydrogen ion transporter H+/K+-ATPase, were performed by the T1DGC laboratory at the Barbara Davis Center for Childhood Diabetes, Aurora, CO. Measurements of all autoantibodies were transmitted to the T1DGC Coordinating Center, and the data were made available to members of the T1DGC Autoantibody Working Groups for analysis in conjunction with existing T1DGC genetic data. This article describes the design of the T1DGC Autoantibody Workshop and the quality-control procedures to maintain and monitor the performance of each laboratory and provides the quality-control results for the nonislet autoantibody measurements.
Collapse
Affiliation(s)
- Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Joan Hilner
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL
| | | |
Collapse
|
24
|
Additive and interaction effects at three amino acid positions in HLA-DQ and HLA-DR molecules drive type 1 diabetes risk. Nat Genet 2015; 47:898-905. [PMID: 26168013 DOI: 10.1038/ng.3353] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022]
Abstract
Variation in the human leukocyte antigen (HLA) genes accounts for one-half of the genetic risk in type 1 diabetes (T1D). Amino acid changes in the HLA-DR and HLA-DQ molecules mediate most of the risk, but extensive linkage disequilibrium complicates the localization of independent effects. Using 18,832 case-control samples, we localized the signal to 3 amino acid positions in HLA-DQ and HLA-DR. HLA-DQβ1 position 57 (previously known; P = 1 × 10(-1,355)) by itself explained 15.2% of the total phenotypic variance. Independent effects at HLA-DRβ1 positions 13 (P = 1 × 10(-721)) and 71 (P = 1 × 10(-95)) increased the proportion of variance explained to 26.9%. The three positions together explained 90% of the phenotypic variance in the HLA-DRB1-HLA-DQA1-HLA-DQB1 locus. Additionally, we observed significant interactions for 11 of 21 pairs of common HLA-DRB1-HLA-DQA1-HLA-DQB1 haplotypes (P = 1.6 × 10(-64)). HLA-DRβ1 positions 13 and 71 implicate the P4 pocket in the antigen-binding groove, thus pointing to another critical protein structure for T1D risk, in addition to the HLA-DQ P9 pocket.
Collapse
|
25
|
van Steenbergen HW, Raychaudhuri S, Rodríguez-Rodríguez L, Rantapää-Dahlqvist S, Berglin E, Toes REM, Huizinga TWJ, Fernández-Gutiérrez B, Gregersen PK, van der Helm-van Mil AHM. Association of valine and leucine at HLA-DRB1 position 11 with radiographic progression in rheumatoid arthritis, independent of the shared epitope alleles but not independent of anti-citrullinated protein antibodies. Arthritis Rheumatol 2015; 67:877-86. [PMID: 25580908 DOI: 10.1002/art.39018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/30/2014] [Indexed: 01/29/2023]
Abstract
OBJECTIVE For decades it has been known that the HLA-DRB1 shared epitope (SE) alleles are associated with an increased risk of development and progression of rheumatoid arthritis (RA). Recently, the following variations in the peptide-binding grooves of HLA molecules that predispose to RA development have been identified: Val and Leu at HLA-DRB1 position 11, Asp at HLA-B position 9, and Phe at HLA-DPB1 position 9. This study was undertaken to investigate whether these variants are also associated with radiographic progression in RA, independent of SE and anti-citrullinated protein antibody (ACPA) status. METHODS A total of 4,911 radiograph sets from 1,878 RA patients included in the Leiden Early Arthritis Clinic (The Netherlands), Umeå (Sweden), Hospital Clinico San Carlos-Rheumatoid Arthritis (Spain), and National Data Bank for Rheumatic Diseases (US) cohorts were studied. HLA was imputed using single-nucleotide polymorphism data from an Immunochip, and the amino acids listed above were tested in relation to radiographic progression per cohort using an additive model. Results from the 4 cohorts were combined in inverse-variance weighted meta-analyses using a fixed-effects model. Analyses were conditioned on SE and ACPA status. RESULTS Val and Leu at HLA-DRB1 position 11 were associated with more radiographic progression (meta-analysis P = 5.11 × 10(-7)); this effect was independent of SE status (meta-analysis P = 0.022) but not independent of ACPA status. Phe at HLA-DPB1 position 9 was associated with more severe radiographic progression (meta-analysis P = 0.024), though not independent of SE status. Asp at HLA-B position 9 was not associated with radiographic progression. CONCLUSION Val and Leu at HLA-DRB1 position 11 conferred a risk of a higher rate of radiographic progression independent of SE status but not independent of ACPA status. These findings support the relevance of these amino acids at position 11.
Collapse
|
26
|
Scott IC, Rijsdijk F, Walker J, Quist J, Spain SL, Tan R, Steer S, Okada Y, Raychaudhuri S, Cope AP, Lewis CM. Do Genetic Susceptibility Variants Associate with Disease Severity in Early Active Rheumatoid Arthritis? J Rheumatol 2015; 42:1131-40. [PMID: 25979711 DOI: 10.3899/jrheum.141211] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Genetic variants affect both the development and severity of rheumatoid arthritis (RA). Recent studies have expanded the number of RA susceptibility variants. We tested the hypothesis that these associated with disease severity in a clinical trial cohort of patients with early, active RA. METHODS We evaluated 524 patients with RA enrolled in the Combination Anti-Rheumatic Drugs in Early RA (CARDERA) trials. We tested validated susceptibility variants - 69 single-nucleotide polymorphisms (SNP), 15 HLA-DRB1 alleles, and amino acid polymorphisms in 6 HLA molecule positions - for their associations with progression in Larsen scoring, 28-joint Disease Activity Scores, and Health Assessment Questionnaire (HAQ) scores over 2 years using linear mixed-effects and latent growth curve models. RESULTS HLA variants were associated with joint destruction. The *04:01 SNP (rs660895, p = 0.0003), *04:01 allele (p = 0.0002), and HLA-DRβ1 amino acids histidine at position 13 (p = 0.0005) and valine at position 11 (p = 0.0012) significantly associated with radiological progression. This association was only significant in anticitrullinated protein antibody (ACPA)-positive patients, suggesting that while their effects were not mediated by ACPA, they only predicted joint damage in ACPA-positive RA. Non-HLA variants did not associate with radiograph damage (assessed individually and cumulatively as a weighted genetic risk score). Two SNP - rs11889341 (STAT4, p = 0.0001) and rs653178 (SH2B3-PTPN11, p = 0.0004) - associated with HAQ scores over 6-24 months. CONCLUSION HLA susceptibility variants play an important role in determining radiological progression in early, active ACPA-positive RA. Genome-wide and HLA-wide analyses across large populations are required to better characterize the genetic architecture of radiological progression in RA.
Collapse
Affiliation(s)
- Ian C Scott
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Frühling Rijsdijk
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Jemma Walker
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Jelmar Quist
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Sarah L Spain
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Rachael Tan
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Sophia Steer
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Yukinori Okada
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Soumya Raychaudhuri
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Andrew P Cope
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| | - Cathryn M Lewis
- From the Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, and Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, and Department of Rheumatology, King's College London, and Guy's Hospital, London, UK; Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.I.C. Scott, PhD, Clinical Research Fellow, Department of Medical and Molecular Genetics, and Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London, and Guy's Hospital; F. Rijsdijk, PhD, Reader, SGDP Centre, Institute of Psychiatry, King's College London; J. Walker, PhD, Statistical Geneticist; J. Quist, PhD, MSc, Student; S.L. Spain, PhD, Research Associate, Department of Medical and Molecular Genetics, King's College London, and Guy's Hospital; R. Tan, MRes, Core Medical Trainee, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; S. Steer, PhD, Consultant Rheumatologist, Department of Rheumatology, King's College Hospital; Y. Okada, PhD, Tenure Track Junior Associate Professor, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, and Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences; S. Raychaudhuri, PhD, Professor, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School; A.P. Cope, PhD, Professor, Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London; C.M. Lewis, PhD, Prof
| |
Collapse
|
27
|
Carmona F, Mackie S, Martín JE, Taylor J, Vaglio A, Eyre S, Bossini-Castillo L, Castañeda S, Cid M, Hernández-Rodríguez J, Prieto-González S, Solans R, Ramentol-Sintas M, González-Escribano M, Ortiz-Fernández L, Morado I, Narváez J, Miranda-Filloy J, Beretta L, Lunardi C, Cimmino MA, Gianfreda D, Santilli D, Ramirez GA, Soriano A, Muratore F, Pazzola G, Addimanda O, Wijmenga C, Witte T, Schirmer JH, Moosig F, Schönau V, Franke A, Palm Ø, Molberg Ø, Diamantopoulos AP, Carette S, Cuthbertson D, Forbess LJ, Hoffman GS, Khalidi NA, Koening CL, Langford CA, McAlear CA, Moreland L, Monach PA, Pagnoux C, Seo P, Spiera R, Sreih AG, Warrington KJ, Ytterberg SR, Gregersen PK, Pease CT, Gough A, Green M, Hordon L, Jarrett S, Watts R, Levy S, Patel Y, Kamath S, Dasgupta B, Worthington J, Koeleman BP, de Bakker PI, Barrett JH, Salvarani C, Merkel PA, González-Gay MA, Morgan AW, Martín J, Martínez-Berriochoa A, Unzurrunzaga A, Hidalgo-Conde A, Madroñero-Vuelta A, Fernández-Nebro A, Ordóñez-Cañizares M, Escalante B, Marí-Alfonso B, Sopeña B, Magro C, Raya E, Grau E, Román J, de Miguel E, López-Longo F, Martínez L, Gómez-Vaquero C, Fernández-Gutiérrez B, Rodríguez-Rodríguez L, Díaz-López J, Caminal-Montero L, Martínez-Zapico A, Monfort J, Tío L, Sánchez-Martín J, Alegre-Sancho J, Sáez-Comet L, Pérez-Conesa M, Corbera-Bellalta M, García-Villanueva M, Fernández-Contreras M, Sanchez-Pernaute O, Blanco R, Ortego-Centeno N, Ríos-Fernández R, Callejas J, Fanlo-Mateo P, Martínez-Taboada V. A large-scale genetic analysis reveals a strong contribution of the HLA class II region to giant cell arteritis susceptibility. Am J Hum Genet 2015; 96:565-80. [PMID: 25817017 DOI: 10.1016/j.ajhg.2015.02.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/12/2015] [Indexed: 02/08/2023] Open
Abstract
We conducted a large-scale genetic analysis on giant cell arteritis (GCA), a polygenic immune-mediated vasculitis. A case-control cohort, comprising 1,651 case subjects with GCA and 15,306 unrelated control subjects from six different countries of European ancestry, was genotyped by the Immunochip array. We also imputed HLA data with a previously validated imputation method to perform a more comprehensive analysis of this genomic region. The strongest association signals were observed in the HLA region, with rs477515 representing the highest peak (p = 4.05 × 10(-40), OR = 1.73). A multivariate model including class II amino acids of HLA-DRβ1 and HLA-DQα1 and one class I amino acid of HLA-B explained most of the HLA association with GCA, consistent with previously reported associations of classical HLA alleles like HLA-DRB1(∗)04. An omnibus test on polymorphic amino acid positions highlighted DRβ1 13 (p = 4.08 × 10(-43)) and HLA-DQα1 47 (p = 4.02 × 10(-46)), 56, and 76 (both p = 1.84 × 10(-45)) as relevant positions for disease susceptibility. Outside the HLA region, the most significant loci included PTPN22 (rs2476601, p = 1.73 × 10(-6), OR = 1.38), LRRC32 (rs10160518, p = 4.39 × 10(-6), OR = 1.20), and REL (rs115674477, p = 1.10 × 10(-5), OR = 1.63). Our study provides evidence of a strong contribution of HLA class I and II molecules to susceptibility to GCA. In the non-HLA region, we confirmed a key role for the functional PTPN22 rs2476601 variant and proposed other putative risk loci for GCA involved in Th1, Th17, and Treg cell function.
Collapse
|
28
|
|
29
|
Pillai NE, Okada Y, Saw WY, Ong RTH, Wang X, Tantoso E, Xu W, Peterson TA, Bielawny T, Ali M, Tay KY, Poh WT, Tan LWL, Koo SH, Lim WY, Soong R, Wenk M, Raychaudhuri S, Little P, Plummer FA, Lee EJD, Chia KS, Luo M, De Bakker PIW, Teo YY. Predicting HLA alleles from high-resolution SNP data in three Southeast Asian populations. Hum Mol Genet 2014; 23:4443-51. [DOI: 10.1093/hmg/ddu149] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
30
|
Han B, Diogo D, Eyre S, Kallberg H, Zhernakova A, Bowes J, Padyukov L, Okada Y, González-Gay M, Rantapää-Dahlqvist S, Martin J, Huizinga T, Plenge R, Worthington J, Gregersen P, Klareskog L, de Bakker P, Raychaudhuri S. Fine mapping seronegative and seropositive rheumatoid arthritis to shared and distinct HLA alleles by adjusting for the effects of heterogeneity. Am J Hum Genet 2014; 94:522-32. [PMID: 24656864 DOI: 10.1016/j.ajhg.2014.02.013] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/24/2014] [Indexed: 11/25/2022] Open
Abstract
Despite progress in defining human leukocyte antigen (HLA) alleles for anti-citrullinated-protein-autoantibody-positive (ACPA(+)) rheumatoid arthritis (RA), identifying HLA alleles for ACPA-negative (ACPA(-)) RA has been challenging because of clinical heterogeneity within clinical cohorts. We imputed 8,961 classical HLA alleles, amino acids, and SNPs from Immunochip data in a discovery set of 2,406 ACPA(-) RA case and 13,930 control individuals. We developed a statistical approach to identify and adjust for clinical heterogeneity within ACPA(-) RA and observed independent associations for serine and leucine at position 11 in HLA-DRβ1 (p = 1.4 × 10(-13), odds ratio [OR] = 1.30) and for aspartate at position 9 in HLA-B (p = 2.7 × 10(-12), OR = 1.39) within the peptide binding grooves. These amino acid positions induced associations at HLA-DRB1(∗)03 (encoding serine at 11) and HLA-B(∗)08 (encoding aspartate at 9). We validated these findings in an independent set of 427 ACPA(-) case subjects, carefully phenotyped with a highly sensitive ACPA assay, and 1,691 control subjects (HLA-DRβ1 Ser11+Leu11: p = 5.8 × 10(-4), OR = 1.28; HLA-B Asp9: p = 2.6 × 10(-3), OR = 1.34). Although both amino acid sites drove risk of ACPA(+) and ACPA(-) disease, the effects of individual residues at HLA-DRβ1 position 11 were distinct (p < 2.9 × 10(-107)). We also identified an association with ACPA(+) RA at HLA-A position 77 (p = 2.7 × 10(-8), OR = 0.85) in 7,279 ACPA(+) RA case and 15,870 control subjects. These results contribute to mounting evidence that ACPA(+) and ACPA(-) RA are genetically distinct and potentially have separate autoantigens contributing to pathogenesis. We expect that our approach might have broad applications in analyzing clinical conditions with heterogeneity at both major histocompatibility complex (MHC) and non-MHC regions.
Collapse
|
31
|
Mayes M, Bossini-Castillo L, Gorlova O, Martin J, Zhou X, Chen W, Assassi S, Ying J, Tan F, Arnett F, Reveille J, Guerra S, Teruel M, Carmona F, Gregersen P, Lee A, López-Isac E, Ochoa E, Carreira P, Simeón C, Castellví I, González-Gay M, Zhernakova A, Padyukov L, Alarcón-Riquelme M, Wijmenga C, Brown M, Beretta L, Riemekasten G, Witte T, Hunzelmann N, Kreuter A, Distler JH, Voskuyl AE, Schuerwegh AJ, Hesselstrand R, Nordin A, Airó P, Lunardi C, Shiels P, van Laar JM, Herrick A, Worthington J, Denton C, Wigley FM, Hummers LK, Varga J, Hinchcliff ME, Baron M, Hudson M, Pope JE, Furst DE, Khanna D, Phillips K, Schiopu E, Segal BM, Molitor JA, Silver RM, Steen VD, Simms RW, Lafyatis RA, Fessler BJ, Frech TM, AlKassab F, Docherty P, Kaminska E, Khalidi N, Jones HN, Markland J, Robinson D, Broen J, Radstake TR, Fonseca C, Koeleman BP, Martin J, Ortego-Centeno N, Ríos R, Callejas J, Navarrete N, García Portales R, Camps M, Fernández-Nebro A, González-Escribano M, Sánchez-Román J, García-Hernández F, Castillo M, Aguirre M, Gómez-Gracia I, Fernández-Gutiérrez B, Rodríguez-Rodríguez L, Vicente E, Andreu J, Fernández de Castro M, García de la Peña P, López-Longo F, Martínez L, Fonollosa V, Espinosa G, Tolosa C, Pros A, Rodríguez Carballeira M, Narváez F, Rubio Rivas M, Ortiz Santamaría V, Díaz B, Trapiella L, Freire M, Sousa A, Egurbide M, Fanlo Mateo P, Sáez-Comet L, Díaz F, Hernández V, Beltrán E, Román-Ivorra J, Grau E, Alegre Sancho J, Blanco García F, Oreiro N, Fernández Sueiro L. Immunochip analysis identifies multiple susceptibility loci for systemic sclerosis. Am J Hum Genet 2014; 94:47-61. [PMID: 24387989 DOI: 10.1016/j.ajhg.2013.12.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022] Open
Abstract
In this study, 1,833 systemic sclerosis (SSc) cases and 3,466 controls were genotyped with the Immunochip array. Classical alleles, amino acid residues, and SNPs across the human leukocyte antigen (HLA) region were imputed and tested. These analyses resulted in a model composed of six polymorphic amino acid positions and seven SNPs that explained the observed significant associations in the region. In addition, a replication step comprising 4,017 SSc cases and 5,935 controls was carried out for several selected non-HLA variants, reaching a total of 5,850 cases and 9,401 controls of European ancestry. Following this strategy, we identified and validated three SSc risk loci, including DNASE1L3 at 3p14, the SCHIP1-IL12A locus at 3q25, and ATG5 at 6q21, as well as a suggested association of the TREH-DDX6 locus at 11q23. The associations of several previously reported SSc risk loci were validated and further refined, and the observed peak of association in PXK was related to DNASE1L3. Our study has increased the number of known genetic associations with SSc, provided further insight into the pleiotropic effects of shared autoimmune risk factors, and highlighted the power of dense mapping for detecting previously overlooked susceptibility loci.
Collapse
|
32
|
Xie G, Roshandel D, Sherva R, Monach PA, Lu EY, Kung T, Carrington K, Zhang SS, Pulit SL, Ripke S, Carette S, Dellaripa PF, Edberg JC, Hoffman GS, Khalidi N, Langford CA, Mahr AD, St.Clair EW, Seo P, Specks U, Spiera RF, Stone JH, Ytterberg SR, Raychaudhuri S, de Bakker PIW, Farrer LA, Amos CI, Merkel PA, Siminovitch KA. Association of granulomatosis with polyangiitis (Wegener's) with HLA-DPB1*04 and SEMA6A gene variants: evidence from genome-wide analysis. ARTHRITIS AND RHEUMATISM 2013; 65:2457-68. [PMID: 23740775 PMCID: PMC4471994 DOI: 10.1002/art.38036] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 05/21/2013] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To identify genetic determinants of granulomatosis with polyangiitis (Wegener's) (GPA). METHODS We carried out a genome-wide association study (GWAS) of 492 GPA cases and 1,506 healthy controls (white subjects of European descent), followed by replication analysis of the most strongly associated signals in an independent cohort of 528 GPA cases and 1,228 controls. RESULTS Genome-wide significant associations were identified in 32 single-nucleotide polymorphic (SNP) markers across the HLA region, the majority of which were located in the HLA-DPB1 and HLA-DPA1 genes encoding the class II major histocompatibility complex (MHC) DPβ chain 1 and DPα chain 1 proteins, respectively. Peak association signals in these 2 genes, emanating from SNPs rs9277554 (for DPβ chain 1) and rs9277341 (DPα chain 1) were strongly replicated in an independent cohort (in the combined analysis of the initial cohort and the replication cohort, P = 1.92 × 10(-50) and 2.18 × 10(-39) , respectively). Imputation of classic HLA alleles and conditional analyses revealed that the SNP association signal was fully accounted for by the classic HLA-DPB1*04 allele. An independent single SNP, rs26595, near SEMA6A (the gene for semaphorin 6A) on chromosome 5, was also associated with GPA, reaching genome-wide significance in a combined analysis of the GWAS and replication cohorts (P = 2.09 × 10(-8) ). CONCLUSION We identified the SEMA6A and HLA-DP loci as significant contributors to risk for GPA, with the HLA-DPB1*04 allele almost completely accounting for the MHC association. These two associations confirm the critical role of immunogenetic factors in the development of GPA.
Collapse
Affiliation(s)
- Gang Xie
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Delnaz Roshandel
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Richard Sherva
- Boston University School of Medicine, Boston, Massachusetts
| | - Paul A. Monach
- Boston University School of Medicine, Boston, Massachusetts
| | - Emily Yue Lu
- University of Texas M. D. Anderson Cancer Center, Houston
| | - Tabitha Kung
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Keisha Carrington
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Steven S. Zhang
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Sara L. Pulit
- Brigham and Women’s Hospital and Broad Institute, Boston, Massachusetts, and University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Simon Carette
- Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | - Alfred D. Mahr
- Hôpital Saint-Louis and Université Paris 7–Diderot, Paris, France
| | | | - Philip Seo
- Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | - Soumya Raychaudhuri
- Brigham and Women’s Hospital, Broad Institute, and Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, and University of Manchester, Manchester, UK
| | - Paul I. W. de Bakker
- Brigham and Women’s Hospital and Broad Institute, Boston, Massachusetts, and University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Christopher I. Amos
- University of Texas M. D. Anderson Cancer Center, Houston, and Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Peter A. Merkel
- Boston University School of Medicine, Boston, Massachusetts, and University of Pennsylvania School of Medicine, Philadelphia
| | - Katherine A. Siminovitch
- Mount Sinai Hospital Samuel Lunenfeld Research Institute, Toronto General Research Institute, and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Zhao LP, Huang X. Recursive organizer (ROR): an analytic framework for sequence-based association analysis. Hum Genet 2013; 132:745-59. [PMID: 23494241 DOI: 10.1007/s00439-013-1285-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 03/03/2013] [Indexed: 12/13/2022]
Abstract
The advent of next-generation sequencing technologies affords the ability to sequence thousands of subjects cost-effectively, and is revolutionizing the landscape of genetic research. With the evolving genotyping/sequencing technologies, it is not unrealistic to expect that we will soon obtain a pair of diploidic fully phased genome sequences from each subject in the near future. Here, in light of this potential, we propose an analytic framework called, recursive organizer (ROR), which recursively groups sequence variants based upon sequence similarities and their empirical disease associations, into fewer and potentially more interpretable super sequence variants (SSV). As an illustration, we applied ROR to assess an association between HLA-DRB1 and type 1 diabetes (T1D), discovering SSVs of HLA-DRB1 with sequence data from the Wellcome Trust Case Control Consortium. Specifically, ROR reduces 36 observed unique HLA-DRB1 sequences into 8 SSVs that empirically associate with T1D, a fourfold reduction of sequence complexity. Using HLA-DRB1 data from Type 1 Diabetes Genetics Consortium as cases and data from Fred Hutchinson Cancer Research Center as controls, we are able to validate associations of these SSVs with T1D. Further, SSVs consist of nine nucleotides, and each associates with its corresponding amino acids. Detailed examination of these selected amino acids reveals their potential functional roles in protein structures and possible implication to the mechanism of T1D.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Mailstop M2-B500, P.O. Box 19024, Seattle, WA 98109-1024, USA.
| | | |
Collapse
|
34
|
Kim Y, Ripke S, Kirov G, Sklar P, Purcell SM, Owen MJ, O'Donovan MC, Sullivan PF. Non-random mating, parent-of-origin, and maternal-fetal incompatibility effects in schizophrenia. Schizophr Res 2013; 143:11-7. [PMID: 23177929 PMCID: PMC4197457 DOI: 10.1016/j.schres.2012.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 10/30/2012] [Accepted: 11/01/2012] [Indexed: 01/10/2023]
Abstract
Although the association of common genetic variation in the extended MHC region with schizophrenia is the most significant yet discovered, the MHC region is one of the more complex regions of the human genome, with unusually high gene density and long-range linkage disequilibrium. The statistical test on which the MHC association is based is a relatively simple, additive model which uses logistic regression of SNP genotypes to predict case-control status. However, it is plausible that more complex models underlie this association. Using a well-characterized sample of trios, we evaluated more complex models by looking for evidence for: (a) non-random mating for HLA alleles, schizophrenia risk profiles, and ancestry; (b) parent-of-origin effects for HLA alleles; and (c) maternal-fetal genotype incompatibility in the HLA. We found no evidence for non-random mating in the parents of individuals with schizophrenia in terms of MHC genotypes or schizophrenia risk profile scores. However, there was evidence of non-random mating that appeared mostly to be driven by ancestry. We did not detect over-transmission of HLA alleles to affected offspring via the general TDT test (without regard to parent of origin) or preferential transmission via paternal or maternal inheritance. We evaluated the hypothesis that maternal-fetal HLA incompatibility may increase risk for schizophrenia using eight classical HLA loci. The most significant alleles were in HLA-B, HLA-C, HLA-DQB1, and HLA-DRB1 but none was significant after accounting for multiple comparisons. We did not find evidence to support more complex models of gene action, but statistical power may have been limiting.
Collapse
Affiliation(s)
- Yunjung Kim
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Iliadis A, Anastassiou D, Wang X. A unified framework for haplotype inference in nuclear families. Ann Hum Genet 2012; 76:312-25. [PMID: 22607042 DOI: 10.1111/j.1469-1809.2012.00715.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many large genome-wide association studies include nuclear families with more than one child (trio families), allowing for analysis of differences between siblings (sib pair analysis). Statistical power can be increased when haplotypes are used instead of genotypes. Currently, haplotype inference in families with more than one child can be performed either using the familial information or statistical information derived from the population samples but not both. Building on our recently proposed tree-based deterministic framework (TDS) for trio families, we augment its applicability to general nuclear families. We impose a minimum recombinant approach locally and independently on each multiple children family, while resorting to the population-derived information to solve the remaining ambiguities. Thus our framework incorporates all available information (familial and population) in a given study. We demonstrate that using all the constraints in our approach we can have gains in the accuracy as opposed to breaking the multiple children families to separate trios and resorting to a trio inference algorithm or phasing each family in isolation. We believe that our proposed framework could be the method of choice for haplotype inference in studies that include nuclear families with multiple children. Our software (tds2.0) is downloadable from www.ee.columbia.edu/∼anastas/tds.
Collapse
Affiliation(s)
- Alexandros Iliadis
- Department of Electrical Engineering, Columbia University, New York, NY, USA
| | | | | |
Collapse
|
36
|
Ahn R, Ding YC, Murray J, Fasano A, Green PHR, Neuhausen SL, Garner C. Association analysis of the extended MHC region in celiac disease implicates multiple independent susceptibility loci. PLoS One 2012; 7:e36926. [PMID: 22615847 PMCID: PMC3355177 DOI: 10.1371/journal.pone.0036926] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/17/2012] [Indexed: 12/22/2022] Open
Abstract
Celiac disease is a common autoimmune disease caused by sensitivity to the dietary protein gluten. Forty loci have been implicated in the disease. All disease loci have been characterized as low-penetrance, with the exception of the high-risk genotypes in the HLA-DQA1 and HLA-DQB1 genes, which are necessary but not sufficient to cause the disease. The very strong effects from the known HLA loci and the genetically complex nature of the major histocompatibility complex (MHC) have precluded a thorough investigation of the region. The purpose of this study was to test the hypothesis that additional celiac disease loci exist within the extended MHC (xMHC). A set of 1898 SNPs was analyzed for association across the 7.6 Mb xMHC region in 1668 confirmed celiac disease cases and 517 unaffected controls. Conditional recursive partitioning was used to create an informative indicator of the known HLA-DQA1 and HLA-DQB1 high-risk genotypes that was included in the association analysis to account for their effects. A linkage disequilibrium-based grouping procedure was utilized to estimate the number of independent celiac disease loci present in the xMHC after accounting for the known effects. There was significant statistical evidence for four new independent celiac disease loci within the classic MHC region. This study is the first comprehensive association analysis of the xMHC in celiac disease that specifically accounts for the known HLA disease genotypes and the genetic complexity of the region.
Collapse
Affiliation(s)
- Richard Ahn
- Department of Epidemiology, University of California Irvine, Irvine, California, United States of America
| | - Yuan Chun Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Joseph Murray
- Department of Medicine and Immunology, The Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alessio Fasano
- Center for Celiac Research, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Peter H. R. Green
- Celiac Disease Center, Columbia University, New York, New York, United States of America
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Chad Garner
- Department of Epidemiology, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
37
|
Invernizzi P, Ransom M, Raychaudhuri S, Kosoy R, Lleo A, Shigeta R, Franke A, Bossa F, Amos CI, Gregersen PK, Siminovitch KA, Cusi D, de Bakker PIW, Podda M, Gershwin ME, Seldin MF. Classical HLA-DRB1 and DPB1 alleles account for HLA associations with primary biliary cirrhosis. Genes Immun 2012; 13:461-8. [PMID: 22573116 PMCID: PMC3423484 DOI: 10.1038/gene.2012.17] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Susceptibility to primary biliary cirrhosis (PBC) is strongly associated with HLA region polymorphisms. To determine if associations can be explained by classical HLA determinants we studied Italian 676 cases and 1440 controls with genotyped with dense single nucleotide polymorphisms (SNPs) for which classical HLA alleles and amino acids were imputed. Although previous genome-wide association studies and our results show stronger SNP associations near DQB1, we demonstrate that the HLA signals can be attributed to classical DRB1 and DPB1 genes. Strong support for the predominant role of DRB1 is provided by our conditional analyses. We also demonstrate an independent association of DPB1. Specific HLA-DRB1 genes (*08, *11 and *14) account for most of the DRB1 association signal. Consistent with previous studies, DRB1*08 (p = 1.59 × 10−11) was the strongest predisposing allele where as DRB1*11 (p = 1.42 × 10−10) was protective. Additionally DRB1*14 and the DPB1 association (DPB1*03:01) (p = 9.18 × 10−7) were predisposing risk alleles. No signal was observed in the HLA class 1 or class 3 regions. These findings better define the association of PBC with HLA and specifically support the role of classical HLA-DRB1 and DPB1 genes and alleles in susceptibility to PBC.
Collapse
Affiliation(s)
- P Invernizzi
- Department of Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Glisic S, Jailwala P. Interaction between Treg apoptosis pathways, Treg function and HLA risk evolves during type 1 diabetes pathogenesis. PLoS One 2012; 7:e36040. [PMID: 22563437 PMCID: PMC3338571 DOI: 10.1371/journal.pone.0036040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 03/29/2012] [Indexed: 11/23/2022] Open
Abstract
We have previously reported increased apoptosis of regulatory T cells (Tregs) in recent-onset Type 1 Diabetes subjects (RO T1D) in the honeymoon phase and in multiple autoantibody-positive (Ab+) subjects, some of which are developing T1D. We have also reported that increased Treg apoptosis was associated with High HLA risk and that it subsided with cessation of honeymoon period. In this report, we present results generated using genetics, genomics, functional cell-based assays and flow cytometry to assess cellular changes at the T-cell level during T1D pathogenesis. We measured ex vivo Treg apoptosis and Treg function, surface markers expression, expression of HLA class II genes, the influence of HLA risk on Treg apoptosis and function, and evaluated contribution of genes reported to be involved in the apoptosis process. This integrated comprehensive approach uncovered important information that can serve as a basis for future studies aimed to modulate Treg cell responsiveness to apoptotic signals in autoimmunity. For example, T1D will progress in those subjects where increased Treg apoptosis is accompanied with decreased Treg function. Furthermore, Tregs from High HLA risk healthy controls had increased Treg apoptosis levels and overexpressed FADD but not Fas/FasL. Tregs from RO T1D subjects in the honeymoon phase were primarily dying through withdrawal of growth hormones with contribution of oxidative stress, mitochondrial apoptotic pathways, and employment of TNF-receptor family members. Ab+ subjects, however, expressed high inflammation level, which probably contributed to Treg apoptosis, although other apoptotic pathways were also activated: withdrawal of growth hormones, oxidative stress, mitochondrial apoptosis and Fas/FasL apoptotic pathways. The value of these results lie in potentially different preventive treatment subjects would receive depending on disease progression stage when treated.
Collapse
Affiliation(s)
- Sanja Glisic
- Department of Pediatrics, Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America.
| | | |
Collapse
|
39
|
Achkar JP, Klei L, de Bakker PI, Bellone G, Rebert N, Scott R, Lu Y, Regueiro M, Brzezinski A, Kamboh MI, Fiocchi C, Devlin B, Trucco M, Ringquist S, Roeder K, Duerr RH. Amino acid position 11 of HLA-DRβ1 is a major determinant of chromosome 6p association with ulcerative colitis. Genes Immun 2012; 13:245-52. [PMID: 22170232 PMCID: PMC3341846 DOI: 10.1038/gene.2011.79] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/12/2011] [Accepted: 10/31/2011] [Indexed: 01/25/2023]
Abstract
The major histocompatibility complex (MHC) on chromosome 6p is an established risk locus for ulcerative colitis (UC) and Crohn's disease (CD). We aimed to better define MHC association signals in UC and CD by combining data from dense single-nucleotide polymorphism (SNP) genotyping and from imputation of classical human leukocyte antigen (HLA) types, their constituent SNPs and corresponding amino acids in 562 UC, 611 CD and 1428 control subjects. Univariate and multivariate association analyses were performed, controlling for ancestry. In univariate analyses, absence of the rs9269955 C allele was strongly associated with risk for UC (P = 2.67 × 10(-13)). rs9269955 is a SNP in the codon for amino acid position 11 of HLA-DRβ1, located in the P6 pocket of the HLA-DR antigen binding cleft. This amino acid position was also the most significantly UC-associated amino acid in omnibus tests (P = 2.68 × 10(-13)). Multivariate modeling identified rs9269955-C and 13 other variants in best predicting UC vs control status. In contrast, there was only suggestive association evidence between the MHC and CD. Taken together, these data demonstrate that variation at HLA-DRβ1, amino acid 11 in the P6 pocket of the HLA-DR complex antigen binding cleft is a major determinant of chromosome 6p association with UC.
Collapse
Affiliation(s)
- Jean-Paul Achkar
- Department of Gastroenterology & Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lambertus Klei
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Paul I.W. de Bakker
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Julius Center for Health Sciences and Primary Care, and Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gaia Bellone
- Department of Statistics, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Nancy Rebert
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Regan Scott
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ying Lu
- Division of Immunogenetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Miguel Regueiro
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Aaron Brzezinski
- Department of Gastroenterology & Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Claudio Fiocchi
- Department of Gastroenterology & Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Massimo Trucco
- Division of Immunogenetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Steven Ringquist
- Division of Immunogenetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Kathryn Roeder
- Department of Statistics, Carnegie Mellon University, Pittsburgh, Pennsylvania
- Lane Center for Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Richard H Duerr
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
40
|
Baschal EE, Jasinski JM, Boyle TA, Fain PR, Eisenbarth GS, Siebert JC. Congruence as a measurement of extended haplotype structure across the genome. J Transl Med 2012; 10:32. [PMID: 22369243 PMCID: PMC3310717 DOI: 10.1186/1479-5876-10-32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 02/27/2012] [Indexed: 02/01/2023] Open
Abstract
Background Historically, extended haplotypes have been defined using only a few data points, such as alleles for several HLA genes in the MHC. High-density SNP data, and the increasing affordability of whole genome SNP typing, creates the opportunity to define higher resolution extended haplotypes. This drives the need for new tools that support quantification and visualization of extended haplotypes as defined by as many as 2000 SNPs. Confronted with high-density SNP data across the major histocompatibility complex (MHC) for 2,300 complete families, compiled by the Type 1 Diabetes Genetics Consortium (T1DGC), we developed software for studying extended haplotypes. Methods The software, called ExHap (Extended Haplotype), uses a similarity measurement we term congruence to identify and quantify long-range allele identity. Using ExHap, we analyzed congruence in both the T1DGC data and family-phased data from the International HapMap Project. Results Congruent chromosomes from the T1DGC data have between 96.5% and 99.9% allele identity over 1,818 SNPs spanning 2.64 megabases of the MHC (HLA-DRB1 to HLA-A). Thirty-three of 132 DQ-DR-B-A defined haplotype groups have > 50% congruent chromosomes in this region. For example, 92% of chromosomes within the DR3-B8-A1 haplotype are congruent from HLA-DRB1 to HLA-A (99.8% allele identity). We also applied ExHap to all 22 autosomes for both CEU and YRI cohorts from the International HapMap Project, identifying multiple candidate extended haplotypes. Conclusions Long-range congruence is not unique to the MHC region. Patterns of allele identity on phased chromosomes provide a simple, straightforward approach to visually and quantitatively inspect complex long-range structural patterns in the genome. Such patterns aid the biologist in appreciating genetic similarities and differences across cohorts, and can lead to hypothesis generation for subsequent studies.
Collapse
Affiliation(s)
- Erin E Baschal
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Denver, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
41
|
Raychaudhuri S, Sandor C, Stahl EA, Freudenberg J, Lee HS, Jia X, Alfredsson L, Padyukov L, Klareskog L, Worthington J, Siminovitch KA, Bae SC, Plenge RM, Gregersen PK, de Bakker PI. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet 2012; 44:291-6. [PMID: 22286218 PMCID: PMC3288335 DOI: 10.1038/ng.1076] [Citation(s) in RCA: 672] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/12/2011] [Indexed: 12/16/2022]
Abstract
The genetic association of the major histocompatibility complex (MHC) to rheumatoid arthritis risk has commonly been attributed to alleles in HLA-DRB1. However, debate persists about the identity of the causal variants in HLA-DRB1 and the presence of independent effects elsewhere in the MHC. Using existing genome-wide SNP data in 5,018 individuals with seropositive rheumatoid arthritis (cases) and 14,974 unaffected controls, we imputed and tested classical alleles and amino acid polymorphisms in HLA-A, HLA-B, HLA-C, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1 and HLA-DRB1, as well as 3,117 SNPs across the MHC. Conditional and haplotype analyses identified that three amino acid positions (11, 71 and 74) in HLA-DRβ1 and single-amino-acid polymorphisms in HLA-B (at position 9) and HLA-DPβ1 (at position 9), which are all located in peptide-binding grooves, almost completely explain the MHC association to rheumatoid arthritis risk. This study shows how imputation of functional variation from large reference panels can help fine map association signals in the MHC.
Collapse
Affiliation(s)
- Soumya Raychaudhuri
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Partners HealthCare Center for Personalized Genetic Medicine, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Cynthia Sandor
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Partners HealthCare Center for Personalized Genetic Medicine, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Eli A. Stahl
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jan Freudenberg
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore–Long Island Jewish Health System, Manhasset, NY USA
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Xiaoming Jia
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Solna, Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jane Worthington
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Katherine A. Siminovitch
- Department of Medicine, University of Toronto, Mount Sinai Hospital and University Health Network, Toronto, Ontario, Canada
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Robert M. Plenge
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Peter K. Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore–Long Island Jewish Health System, Manhasset, NY USA
| | - Paul I.W. de Bakker
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
42
|
Abstract
Host genetic variation is presently estimated to account for about one-fourth of the observed differences in control of HIV across infected individuals. Genome-wide association studies have confirmed that polymorphism within the HLA class I locus is the primary host genetic contributor to determining outcome after infection. Here we progress beyond the genetic associations alone to consider the functional explanations for these correlations. In this process, the complex and multidimensional effects of HLA molecules in viral disease become apparent.
Collapse
Affiliation(s)
- Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC–Frederick, Inc., NCI–Frederick, Frederick, Maryland 20882;
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114;
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114;
| |
Collapse
|
43
|
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease affecting approximately one in 300 individuals in the United States. The majority of genetic research to date has focused on the heritability that predisposes to islet autoimmunity and T1DM. The evidence so far points to T1DM being a polygenic, common, complex disease with major susceptibility lying in the major histocompatibility complex (MHC) on chromosome 6 with other smaller effects seen in loci outside of the MHC. With recent advances in technology, novel means of exploring the human genome have given way to new information in the development of T1DM. The newest technologies, namely high-throughput polymorphism typing and sequencing, have led to a paradigm shift in studying common diseases such as T1DM. In this review we highlight the advances in genetic associations in T1DM in the last several decades and how they have led to a better understanding of T1DM pathogenesis.
Collapse
Affiliation(s)
- Peter R Baker
- The Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045-6511, USA
| | | |
Collapse
|
44
|
Baschal EE, Sarkar SA, Boyle TA, Siebert JC, Jasinski JM, Grabek KR, Armstrong TK, Babu SR, Fain PR, Steck AK, Rewers MJ, Eisenbarth GS. Replication and further characterization of a Type 1 diabetes-associated locus at the telomeric end of the major histocompatibility complex. J Diabetes 2011; 3:238-47. [PMID: 21631897 PMCID: PMC3610173 DOI: 10.1111/j.1753-0407.2011.00131.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND We recently reported an association between Type 1 diabetes and the telomeric major histocompatibility complex (MHC) single nucleotide polymorphism (SNP) rs1233478. As further families have been analyzed in the Type 1 Diabetes Genetics Consortium (T1DGC), we tested replication of the association and, with more data, analyzed haplotypic associations. METHODS An additional 2717 case and 1315 control chromosomes have been analyzed from the T1DGC, with human leukocyte antigen (HLA) typing and data for 2837 SNPs across the MHC region. RESULTS We confirmed the association of rs1233478 (new data only: P=2.2E-5, OR=1.4). We also found two additional SNPs nearby that were significantly associated with Type 1 diabetes (new data only rs3131020: P=8.3E-9, OR=0.65; rs1592410: P=2.2E-8, OR=1.5). For studies of Type 1 diabetes in the MHC region, it is critical to account for linkage disequilibrium with the HLA genes. Logistic regression analysis of these new data indicated that the effects of rs3131020 and rs1592410 on Type 1 diabetes risk are independent of HLA alleles (rs3131020: P=2.3E-3, OR=0.73; rs1592410: P=2.1E-3, OR=1.4). Haplotypes of 12 SNPs (including the three highly significant SNPs) stratify diabetes risk (high risk, protective, and neutral), with high-risk haplotypes limited to approximately 20,000 bp in length. The 20,000-bp region is telomeric of the UBD gene and contains LOC729653, a hypothetical gene. CONCLUSIONS We believe that polymorphisms of the telomeric MHC locus LOC729653 may confer risk for Type 1 diabetes.
Collapse
Affiliation(s)
- Erin E Baschal
- Barbara Davis Center for Childhood Diabetes, University of Colorado-Denver, 1775 Aurora Ct., Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Baschal EE, Baker PR, Eyring KR, Siebert JC, Jasinski JM, Eisenbarth GS. The HLA-B 3906 allele imparts a high risk of diabetes only on specific HLA-DR/DQ haplotypes. Diabetologia 2011; 54:1702-9. [PMID: 21533899 PMCID: PMC3110277 DOI: 10.1007/s00125-011-2161-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 03/29/2011] [Indexed: 12/05/2022]
Abstract
AIMS/HYPOTHESIS We investigated the risk associated with HLA-B*39 alleles in the context of specific HLA-DR/DQ haplotypes. METHODS We studied a readily available dataset from the Type 1 Diabetes Genetics Consortium that consists of 2,300 affected sibling pair families genotyped for both HLA alleles and 2,837 single nucleotide polymorphisms across the major histocompatibility complex region. RESULTS The B*3906 allele significantly enhanced the risk of type 1 diabetes when present on specific HLA-DR/DQ haplotypes (DRB1 0801-DQB1 0402: p = 1.6 × 10(-6), OR 25.4; DRB1 0101-DQB1 0501: p = 4.9 × 10(-5), OR 10.3) but did not enhance the risk of DRB1 0401-DQB1 0302 haplotypes. In addition, the B 3901 allele enhanced risk on the DRB1 1601-DQB1 0502 haplotype (p = 3.7 × 10(-3), OR 7.2). CONCLUSIONS/INTERPRETATION These associations indicate that the B 39 alleles significantly increase risk when present on specific HLA-DR/DQ haplotypes, and HLA-B typing in concert with specific HLA-DR/DQ genotypes should facilitate genetic prediction of type 1 diabetes, particularly in a research setting.
Collapse
Affiliation(s)
- E. E. Baschal
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Box B140, Aurora, CO 80045-6511 USA
| | - P. R. Baker
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Box B140, Aurora, CO 80045-6511 USA
| | - K. R. Eyring
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Box B140, Aurora, CO 80045-6511 USA
| | | | - J. M. Jasinski
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Box B140, Aurora, CO 80045-6511 USA
| | - G. S. Eisenbarth
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Box B140, Aurora, CO 80045-6511 USA
| |
Collapse
|
46
|
Baker PR, Baschal EE, Fain PR, Nanduri P, Triolo TM, Siebert JC, Armstrong TK, Babu SR, Rewers MJ, Gottlieb PA, Barker JM, Eisenbarth GS. Dominant suppression of Addison's disease associated with HLA-B15. J Clin Endocrinol Metab 2011; 96:2154-62. [PMID: 21565792 PMCID: PMC3135206 DOI: 10.1210/jc.2010-2964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Autoimmune Addison's disease (AD) is the major cause of primary adrenal failure in developed nations. Autoantibodies to 21-hydroxylase (21OH-AA) are associated with increased risk of progression to AD. Highest genetic risk is associated with the Major Histocompatibility region (MHC), specifically human leukocyte antigen (HLA)-DR3 haplotypes (containing HLA-B8) and HLA-DR4. OBJECTIVE The objective of the study was the further characterization of AD risk associated with MHC alleles. DESIGN, SETTING, AND PARTICIPANTS MHC genotypes were determined for HLA-DRB1, DQA1, DQB1, MICA, HLA-B, and HLA-A in 168 total individuals with 21OH-AA (85 with AD at referral and 83 with positive 21OH-AA but without AD at referral). MAIN OUTCOME MEASURE(S) Genotype was evaluated in 21OH-AA-positive individuals. Outcomes were compared with general population controls and type 1 diabetes patients. RESULTS In HLA-DR4+ individuals, HLA-B15 was found in only one of 55 (2%) with AD vs. 24 of 63 (40%) 21OH-AA-positive nonprogressors (P = 2 × 10(-7)) and 518 of 1558 (33%) HLA-DR4 patients with type 1 diabetes (P = 1 × 10(-8)). On prospective follow-up, none of the HLA-B15-positive, 21-hydroxylase-positive individuals progressed to AD vs. 25% non-HLA-B15 autoantibody-positive individuals by life table analysis (P = 0.03). Single nucleotide polymorphism analysis revealed the HLA-DR/DQ region associated with risk and HLA-B15 were separated by multiple intervening single-nucleotide polymorphism haplotypes. CONCLUSIONS HLA-B15 is not associated with protection from 21OH-AA formation but is associated with protection from progression to AD in 21OH-AA-positive individuals. To our knowledge, this is one of the most dramatic examples of genetic disease suppression in individuals who already have developed autoantibodies and of novel dominant suppression of an autoimmune disease by a class I HLA allele.
Collapse
Affiliation(s)
- Peter R Baker
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado 80045-6511, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- Andrew J McMichael
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OS3 9DS, UK.
| | | |
Collapse
|
48
|
Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, Jia X, McLaren PJ, Ripke S, Brumme CJ, Pulit SL, Telenti A, Carrington M, Kadie CM, Carlson JM, Heckerman D, de Bakker PI, Pereyra F, de Bakker PI, Graham RR, Plenge RM, Deeks SG, Walker BD, Gianniny L, Crawford G, Sullivan J, Gonzalez E, Davies L, Camargo A, Moore JM, Beattie N, Gupta S, Crenshaw A, Burtt NP, Guiducci C, Gupta N, Carrington M, Gao X, Qi Y, Yuki Y, Pereyra F, Piechocka-Trocha A, Cutrell E, Rosenberg R, Moss KL, Lemay P, O’Leary J, Schaefer T, Verma P, Toth I, Block B, Baker B, Rothchild A, Lian J, Proudfoot J, Alvino DML, Vine S, Addo MM, Allen TM, Altfeld M, Henn MR, Le Gall S, Streeck H, Walker BD, Haas DW, Kuritzkes DR, Robbins GK, Shafer RW, Gulick RM, Shikuma CM, Haubrich R, Riddler S, Sax PE, Daar ES, Ribaudo HJ, Agan B, Agarwal S, Ahern RL, Allen BL, Altidor S, Altschuler EL, Ambardar S, Anastos K, Anderson B, Anderson V, Andrady U, Antoniskis D, Bangsberg D, Barbaro D, Barrie W, Bartczak J, Barton S, Basden P, Basgoz N, Bazner S, Bellos NC, Benson AM, Berger J, Bernard NF, Bernard AM, Birch C, Bodner SJ, Bolan RK, Boudreaux ET, Bradley M, Braun JF, Brndjar JE, Brown SJ, Brown K, Brown ST, Burack J, Bush LM, Cafaro V, Campbell O, Campbell J, Carlson RH, Carmichael JK, Casey KK, Cavacuiti C, Celestin G, Chambers ST, Chez N, Chirch LM, Cimoch PJ, Cohen D, Cohn LE, Conway B, Cooper DA, Cornelson B, Cox DT, Cristofano MV, Cuchural G, Czartoski JL, Dahman JM, Daly JS, Davis BT, Davis K, Davod SM, Deeks SG, DeJesus E, Dietz CA, Dunham E, Dunn ME, Ellerin TB, Eron JJ, Fangman JJ, Farel CE, Ferlazzo H, Fidler S, Fleenor-Ford A, Frankel R, Freedberg KA, French NK, Fuchs JD, Fuller JD, Gaberman J, Gallant JE, Gandhi RT, Garcia E, Garmon D, Gathe JC, Gaultier CR, Gebre W, Gilman FD, Gilson I, Goepfert PA, Gottlieb MS, Goulston C, Groger RK, Gurley TD, Haber S, Hardwicke R, Hardy WD, Harrigan PR, Hawkins TN, Heath S, Hecht FM, Henry WK, Hladek M, Hoffman RP, Horton JM, Hsu RK, Huhn GD, Hunt P, Hupert MJ, Illeman ML, Jaeger H, Jellinger RM, John M, Johnson JA, Johnson KL, Johnson H, Johnson K, Joly J, Jordan WC, Kauffman CA, Khanlou H, Killian RK, Kim AY, Kim DD, Kinder CA, Kirchner JT, Kogelman L, Kojic EM, Korthuis PT, Kurisu W, Kwon DS, LaMar M, Lampiris H, Lanzafame M, Lederman MM, Lee DM, Lee JM, Lee MJ, Lee ET, Lemoine J, Levy JA, Llibre JM, Liguori MA, Little SJ, Liu AY, Lopez AJ, Loutfy MR, Loy D, Mohammed DY, Man A, Mansour MK, Marconi VC, Markowitz M, Marques R, Martin JN, Martin HL, Mayer KH, McElrath MJ, McGhee TA, McGovern BH, McGowan K, McIntyre D, Mcleod GX, Menezes P, Mesa G, Metroka CE, Meyer-Olson D, Miller AO, Montgomery K, Mounzer KC, Nagami EH, Nagin I, Nahass RG, Nelson MO, Nielsen C, Norene DL, O’Connor DH, Ojikutu BO, Okulicz J, Oladehin OO, Oldfield EC, Olender SA, Ostrowski M, Owen WF, Pae E, Parsonnet J, Pavlatos AM, Perlmutter AM, Pierce MN, Pincus JM, Pisani L, Price LJ, Proia L, Prokesch RC, Pujet HC, Ramgopal M, Rathod A, Rausch M, Ravishankar J, Rhame FS, Richards CS, Richman DD, Robbins GK, Rodes B, Rodriguez M, Rose RC, Rosenberg ES, Rosenthal D, Ross PE, Rubin DS, Rumbaugh E, Saenz L, Salvaggio MR, Sanchez WC, Sanjana VM, Santiago S, Schmidt W, Schuitemaker H, Sestak PM, Shalit P, Shay W, Shirvani VN, Silebi VI, Sizemore JM, Skolnik PR, Sokol-Anderson M, Sosman JM, Stabile P, Stapleton JT, Starrett S, Stein F, Stellbrink HJ, Sterman FL, Stone VE, Stone DR, Tambussi G, Taplitz RA, Tedaldi EM, Telenti A, Theisen W, Torres R, Tosiello L, Tremblay C, Tribble MA, Trinh PD, Tsao A, Ueda P, Vaccaro A, Valadas E, Vanig TJ, Vecino I, Vega VM, Veikley W, Wade BH, Walworth C, Wanidworanun C, Ward DJ, Warner DA, Weber RD, Webster D, Weis S, Wheeler DA, White DJ, Wilkins E, Winston A, Wlodaver CG, Wout AV, Wright DP, Yang OO, Yurdin DL, Zabukovic BW, Zachary KC, Zeeman B, Zhao M. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 2010; 330:1551-7. [PMID: 21051598 PMCID: PMC3235490 DOI: 10.1126/science.1195271] [Citation(s) in RCA: 919] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Infectious and inflammatory diseases have repeatedly shown strong genetic associations within the major histocompatibility complex (MHC); however, the basis for these associations remains elusive. To define host genetic effects on the outcome of a chronic viral infection, we performed genome-wide association analysis in a multiethnic cohort of HIV-1 controllers and progressors, and we analyzed the effects of individual amino acids within the classical human leukocyte antigen (HLA) proteins. We identified >300 genome-wide significant single-nucleotide polymorphisms (SNPs) within the MHC and none elsewhere. Specific amino acids in the HLA-B peptide binding groove, as well as an independent HLA-C effect, explain the SNP associations and reconcile both protective and risk HLA alleles. These results implicate the nature of the HLA-viral peptide interaction as the major factor modulating durable control of HIV infection.
Collapse
Affiliation(s)
| | | | - Florencia Pereyra
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaoming Jia
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
| | - Paul J. McLaren
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amalio Telenti
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Paul I.W. de Bakker
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Genetics, Division of Biomedical Genetics, University Medical Center Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
| | - Bruce D. Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - Xiaoming Jia
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
| | - Paul J. McLaren
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephan Ripke
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Center for Human Genetic Research, MGH, Harvard Medical School, Boston, MA, USA
| | - Chanson J. Brumme
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Sara L. Pulit
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amalio Telenti
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, NCI-Frederick, Frederick, MD, USA
| | | | | | | | - Paul I.W. de Bakker
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Genetics, Division of Biomedical Genetics, University Medical Center Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
| | | | - Florencia Pereyra
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul I.W. de Bakker
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Genetics, Division of Biomedical Genetics, University Medical Center Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
| | | | - Robert M. Plenge
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven G. Deeks
- University of California San Francisco, San Francisco, CA, USA
| | - Bruce D. Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | | | | | | | | | - Leela Davies
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Amy Camargo
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | | | - Supriya Gupta
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Noël P. Burtt
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Namrata Gupta
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, NCI-Frederick, Frederick, MD, USA
| | - Xiaojiang Gao
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, NCI-Frederick, Frederick, MD, USA
| | - Ying Qi
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, NCI-Frederick, Frederick, MD, USA
| | - Yuko Yuki
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, NCI-Frederick, Frederick, MD, USA
| | | | - Florencia Pereyra
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alicja Piechocka-Trocha
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Emily Cutrell
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Rachel Rosenberg
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Kristin L. Moss
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Paul Lemay
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Jessica O’Leary
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Todd Schaefer
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Pranshu Verma
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Ildiko Toth
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Brian Block
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Brett Baker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Alissa Rothchild
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Jeffrey Lian
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Jacqueline Proudfoot
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Donna Marie L. Alvino
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Seanna Vine
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Marylyn M. Addo
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Todd M. Allen
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Marcus Altfeld
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | - Sylvie Le Gall
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Hendrik Streeck
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Bruce D. Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - David W. Haas
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Daniel R. Kuritzkes
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Roy M. Gulick
- Weill Medical College of Cornell University, New York, NY, USA
| | - Cecilia M. Shikuma
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | | | | | - Paul E. Sax
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric S. Daar
- University of California Los Angeles, Los Angeles, CA, USA
| | - Heather J. Ribaudo
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | | | - Brian Agan
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | | | | | | | | | | - Kathryn Anastos
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ben Anderson
- St. Leonards Medical Centre, St. Leonards, Australia
| | | | | | | | - David Bangsberg
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
- MGH, Harvard Medical School, Boston, MA, USA
| | - Daniel Barbaro
- Tarrant County Infectious Disease Associates, Fort Worth, TX, USA
| | | | | | - Simon Barton
- Chelsea and Westminster Hospital, St. Stephen’s Centre, London, UK
| | | | | | - Suzane Bazner
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | | | | | - Nicole F. Bernard
- Research Institute, McGill University Health Centre, Montreal General Hospital, Montreal, Canada
| | | | - Christopher Birch
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | | | - Emilie T. Boudreaux
- Louisiana State University Health Sciences Center, University Medical Center East Clinic, Lafayatte, LA, USA
| | - Meg Bradley
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - James F. Braun
- Physicians’ Research Network, Callen-Lorde Community Health Center, New York, NY, USA
| | | | | | | | | | | | - Larry M. Bush
- University of Miami-Miller School of Medicine, Lake Worth, FL, USA
| | | | | | | | | | | | | | | | | | | | - Nancy Chez
- H.E.L.P./Project Samaritan, Bronx, NY, USA
| | - Lisa M. Chirch
- David E. Rogers Center for HIV/AIDS Care, Southampton, NY, USA
| | | | | | - Lillian E. Cohn
- 9th Street Internal Medicine Associates, Philadelphia, PA, USA
| | - Brian Conway
- University of British Columbia, Vancouver, Canada
| | - David A. Cooper
- National Centre in HIV Epidemiology and Clinical Research, Sydney, Australia
| | | | - David T. Cox
- Metro Infectious Disease Consultants, Indianapolis, IN, USA
| | | | | | | | | | - Jennifer S. Daly
- University of Massachusetts Memorial Medical Center, Worcester, MA, USA
| | | | - Kristine Davis
- University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Steven G. Deeks
- University of California San Francisco, San Francisco, CA, USA
| | | | - Craig A. Dietz
- The Kansas City Free Health Clinic, Kansas City, MO, USA
| | - Eleanor Dunham
- David E. Rogers Center for HIV/AIDS Care, Southampton, NY, USA
| | | | | | - Joseph J. Eron
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Claire E. Farel
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Helen Ferlazzo
- Visiting Nurse Association of Central New Jersey, Community Health Center, Asbury Park, NJ, USA
| | | | | | | | | | - Neel K. French
- Private Practice of Neel K. French, M.D., Chicago, IL, USA
| | | | | | | | - Joel E. Gallant
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Efrain Garcia
- Private Practice of Efrain Garcia, M.D., Miami, FL, USA
| | | | - Joseph C. Gathe
- Private Practice of Joseph C. Gathe Jr., M.D., Houston, TX, USA
| | | | | | | | - Ian Gilson
- Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | | | | - W. David Hardy
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - Sonya Heath
- University of Alabama, Birmingham, Birmingham, AL, USA
| | | | | | - Melissa Hladek
- The Catholic University of America, School of Nursing, Washington, DC, USA
| | | | | | - Ricky K. Hsu
- New York University Medical Center, New York, NY, USA
| | | | - Peter Hunt
- University of California San Francisco, San Francisco, CA, USA
| | - Mark J. Hupert
- Tarrant County Infectious Disease Associates, Fort Worth, TX, USA
| | | | - Hans Jaeger
- HIV Research and Clinical Care Centre, Munich, Germany
| | | | - Mina John
- Murdoch University, Murdoch, Australia
| | - Jennifer A. Johnson
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Heather Johnson
- Tarrant County Infectious Disease Associates, Fort Worth, TX, USA
| | - Kay Johnson
- University of Cincinnati, Cincinnati, OH, USA
| | - Jennifer Joly
- David E. Rogers Center for HIV/AIDS Care, Southampton, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Wayne Kurisu
- Sharp Rees Stealy Medical Center, San Diego, CA, USA
| | - Douglas S. Kwon
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | - Harry Lampiris
- University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Jean M.L. Lee
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Janice Lemoine
- Greater Lawrence Family Health Center, Lawrence, MA, USA
| | - Jay A. Levy
- University of California San Francisco, San Francisco, CA, USA
| | - Josep M. Llibre
- Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | - Anne Y. Liu
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Dawn Loy
- Infectious Disease Associates, Sarasota, FL, USA
| | | | - Alan Man
- Kaiser Permanente, Portland, OR, USA
| | | | | | - Martin Markowitz
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, USA
| | - Rui Marques
- Deruico Doencas Infecciosas, Porto, Portugal
| | | | | | | | | | | | | | - Katherine McGowan
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dawn McIntyre
- Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Gavin X. Mcleod
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Prema Menezes
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Greg Mesa
- Highland Medical Associates, Hendersonville, NC, USA
| | | | - Dirk Meyer-Olson
- Medizinische Hochschule, Abteilung Klinische Immunologie, Hannover, Germany
| | | | | | | | - Ellen H. Nagami
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Iris Nagin
- Lower East Side Service Center, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Eunice Pae
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | - Moti Ramgopal
- Midway Immunology and Research Center, Fort Pierce, FL, USA
| | - Almas Rathod
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | - J. Ravishankar
- State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | | | | | | | | | - Berta Rodes
- Fundacion para la Investigacion Biomedica del Hospital Carlos III, Madrid, Spain
| | | | | | | | | | - Polly E. Ross
- Western North Carolina Community Health Services, Asheville, NC, USA
| | - David S. Rubin
- New York Hospital Medical Center of Queens, Flushing, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Paul Stabile
- William F. Ryan Community Health Center, New York, NY, USA
| | | | | | - Francine Stein
- Visiting Nurse Association of Central New Jersey, Community Health Center, Asbury Park, NJ, USA
| | | | | | | | | | | | | | | | - Amalio Telenti
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | - William Theisen
- Department of Medicine, Division of Infectious Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Phuong D. Trinh
- Montgomery Infectious Disease Associates, Silver Spring, MD, USA
| | - Alice Tsao
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Peggy Ueda
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | | | - Emilia Valadas
- Hospital de Santa Maria, Faculdade de Medicina de Lisboa, Lisbon, Portugal
| | | | - Isabel Vecino
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | | | - Barbara H. Wade
- Infectious Diseases Associates of Northwest Florida, Pensacola, FL, USA
| | | | | | | | | | | | | | - Steve Weis
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - David A. Wheeler
- Clinical Alliance for Research and Education-Infectious Diseases, Annandale, VA, USA
| | - David J. White
- Hawthorn House, Birmingham Heartlands Hospital, Birmingham, UK
| | - Ed Wilkins
- North Manchester General Hospital, Manchester, UK
| | | | | | | | | | - Otto O. Yang
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | - Beth Zeeman
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology (MIT) and Harvard, Boston, MA, USA
| | - Meng Zhao
- United Health Services Hospitals, Binghamton, NY, USA
| |
Collapse
|
49
|
Conditional meta-analysis stratifying on detailed HLA genotypes identifies a novel type 1 diabetes locus around TCF19 in the MHC. Hum Genet 2010; 129:161-76. [PMID: 21076979 PMCID: PMC3020293 DOI: 10.1007/s00439-010-0908-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 10/26/2010] [Indexed: 10/25/2022]
Abstract
The human leukocyte antigen (HLA) class II genes HLA-DRB1, -DQA1 and -DQB1 are the strongest genetic factors for type 1 diabetes (T1D). Additional loci in the major histocompatibility complex (MHC) are difficult to identify due to the region's high gene density and complex linkage disequilibrium (LD). To facilitate the association analysis, two novel algorithms were implemented in this study: one for phasing the multi-allelic HLA genotypes in trio families, and one for partitioning the HLA strata in conditional testing. Screening and replication were performed on two large and independent datasets: the Wellcome Trust Case-Control Consortium (WTCCC) dataset of 2,000 cases and 1,504 controls, and the T1D Genetics Consortium (T1DGC) dataset of 2,300 nuclear families. After imputation, the two datasets have 1,941 common SNPs in the MHC, of which 22 were successfully tested and replicated based on the statistical testing stratifying on the detailed DRB1 and DQB1 genotypes. Further conditional tests using the combined dataset confirmed eight novel SNP associations around 31.3 Mb on chromosome 6 (rs3094663, p = 1.66 × 10(-11) and rs2523619, p = 2.77 × 10(-10) conditional on the DR/DQ genotypes). A subsequent LD analysis established TCF19, POU5F1, CCHCR1 and PSORS1C1 as potential causal genes for the observed association.
Collapse
|
50
|
Baker PR, Baschal EE, Fain PR, Triolo TM, Nanduri P, Siebert JC, Armstrong TK, Babu SR, Rewers MJ, Gottlieb PA, Barker JM, Eisenbarth GS. Haplotype analysis discriminates genetic risk for DR3-associated endocrine autoimmunity and helps define extreme risk for Addison's disease. J Clin Endocrinol Metab 2010; 95:E263-70. [PMID: 20631027 PMCID: PMC3050098 DOI: 10.1210/jc.2010-0508] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Multiple autoimmune disorders (e.g. Addison's disease, type 1 diabetes, celiac disease) are associated with HLA-DR3, but it is likely that alleles of additional genes in linkage disequilibrium with HLA-DRB1 contribute to disease. OBJECTIVE The objective of the study was to characterize major histocompatability complex (MHC) haplotypes conferring extreme risk for autoimmune Addison's disease (AD). DESIGN, SETTING, AND PARTICIPANTS Eighty-six 21-hydroxylase autoantibody-positive, nonautoimmune polyendocrine syndrome type 1, Caucasian individuals collected from 1992 to 2009 with clinical AD from 68 families (12 multiplex and 56 simplex) were genotyped for HLA-DRB1, HLA-DQB1, MICA, HLA-B, and HLA-A as well as high density MHC single-nucleotide polymorphism (SNP) analysis for 34. MAIN OUTCOME MEASURES AD and genotype were measured. RESULT Ninety-seven percent of the multiplex individuals had both HLA-DR3 and HLA-B8 vs. 60% of simplex AD patients (P = 9.72 × 10(-4)) and 13% of general population controls (P = 3.00 × 10(-19)). The genotype DR3/DR4 with B8 was present in 85% of AD multiplex patients, 24% of simplex patients, and 1.5% of control individuals (P = 4.92 × 10(-191)). The DR3-B8 haplotype of AD patients had HLA-A1 less often (47%) than controls (81%, P = 7.00 × 10(-5)) and type 1 diabetes patients (73%, P = 1.93 × 10(-3)). Analysis of 1228 SNPs across the MHC for individuals with AD revealed a shorter conserved haplotype (3.8) with the loss of the extended conserved 3.8.1 haplotype approximately halfway between HLA-B and HLA-A. CONCLUSION Extreme risk for AD, especially in multiplex families, is associated with haplotypic DR3 variants, in particular a portion (3.8) but not all of the conserved 3.8.1 haplotype.
Collapse
Affiliation(s)
- Peter R Baker
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado 80045-6511, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|