351
|
Castiblanco J, Arcos-Burgos M, Anaya JM. What is next after the genes for autoimmunity? BMC Med 2013; 11:197. [PMID: 24107170 PMCID: PMC3765994 DOI: 10.1186/1741-7015-11-197] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/12/2013] [Indexed: 11/28/2022] Open
Abstract
Clinical pathologies draw us to envisage disease as either an independent entity or a diverse set of traits governed by common physiopathological mechanisms, prompted by environmental assaults throughout life. Autoimmune diseases are not an exception, given they represent a diverse collection of diseases in terms of their demographic profile and primary clinical manifestations. Although they are pleiotropic outcomes of non-specific disease genes underlying similar immunogenetic mechanisms, research generally focuses on a single disease. Drastic technologic advances are leading research to organize clinical genomic multidisciplinary approaches to decipher the nature of human biological systems. Once the currently costly omic-based technologies become universally accessible, the way will be paved for a cleaner picture to risk quantification, prevention, prognosis and diagnosis, allowing us to clearly define better phenotypes always ensuring the integrity of the individuals studied. However, making accurate predictions for most autoimmune diseases is an ambitious challenge, since the understanding of these pathologies is far from complete. Herein, some pitfalls and challenges of the genetics of autoimmune diseases are reviewed, and an approximation to the future of research in this field is presented.
Collapse
Affiliation(s)
- John Castiblanco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 #63-C-69, Bogota, Colombia.
| | | | | |
Collapse
|
352
|
Chang CJ, Kuo HC, Chang JS, Lee JK, Tsai FJ, Khor CC, Chang LC, Chen SP, Ko TM, Liu YM, Chen YJ, Hong YM, Jang GY, Hibberd ML, Kuijpers T, Burgner D, Levin M, Burns JC, Davila S, International Kawasaki Disease Genetics Consortium ¶, Korean Kawasaki Disease Genetics Consortium ¶, Taiwan Kawasaki Disease Genetics Consortium ¶, Chen YT, Chen CH, Wu JY, Lee YC. Replication and meta-analysis of GWAS identified susceptibility loci in Kawasaki disease confirm the importance of B lymphoid tyrosine kinase (BLK) in disease susceptibility. PLoS One 2013; 8:e72037. [PMID: 24023612 PMCID: PMC3758326 DOI: 10.1371/journal.pone.0072037] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/04/2013] [Indexed: 12/18/2022] Open
Abstract
The BLK and CD40 loci have been associated with Kawasaki disease (KD) in two genome-wide association studies (GWAS) conducted in a Taiwanese population of Han Chinese ancestry (Taiwanese) and in Japanese cohorts. Here we build on these findings with replication studies of the BLK and CD40 loci in populations of Korean and European descent. The BLK region was significantly associated with KD susceptibility in both populations. Within the BLK gene the rs2736340-located linkage disequilibrium (LD ) comprising the promoter and first intron was strongly associated with KD, with the combined results of Asian studies including Taiwanese, Japanese, and Korean populations (2,539 KD patients and 7,021 controls) providing very compelling evidence of association (rs2736340, OR = 1.498, 1.354–1.657; P = 4.74×10−31). We determined the percentage of B cells present in the peripheral blood mononuclear cell (PBMC) population and the expression of BLK in the peripheral blood leukocytes (leukocytes) of KD patients during the acute and convalescent stages. The percentage of B cells in the PBMC population and the expression of BLK in leukocytes were induced in patients in the acute stage of KD. In B cell lines derived from KD patients, and in purified B cells from KD patients obtained during the acute stage, those with the risk allele of rs2736340 expressed significantly lower levels of BLK. These results suggest that peripheral B cells play a pathogenic role during the acute stage of KD. Decreased BLK expression in peripheral blood B cells may alter B cell function and predispose individuals to KD. These associative data suggest a role for B cells during acute KD. Understanding the functional implications may facilitate the development of B cell-mediated therapy for KD.
Collapse
Affiliation(s)
- Chia-Jung Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ho-Chang Kuo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jeng-Sheng Chang
- Department of Pediatrics, China Medical University and Hospital, Taichung, Taiwan
| | - Jong-Keuk Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chiea Chuen Khor
- Division of Human Genetics, Genome Institute of Singapore, Singapore
- Department of Ophthalmology, School of Medicine, National University of Singapore, Singapore
- Department of Paediatrics, School of Medicine, National University of Singapore, Singapore
| | - Li-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shih-Ping Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tai-Ming Ko
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Min Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ying-Ju Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University Hospital, Seoul, Korea
| | - Gi Young Jang
- Department of Pediatrics, Korea University Hospital, Ansan, Korea
| | | | - Taco Kuijpers
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children’s Hospital Academic Medical Center, Amsterdam, The Netherlands
| | - David Burgner
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Victoria, Australia
| | - Michael Levin
- Department of Pediatrics, Imperial College London, London, United Kingdom
| | - Jane C. Burns
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Sonia Davila
- Division of Human Genetics, Genome Institute of Singapore, Singapore
- School of Epidemiology and Public Health, National University of Singapore, Singapore ,¶ A complete list of members and affiliations appears in File S1
| | | | | | | | - Yuan-Tsong Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (YTC) (YC); (CHC); (JYW); (YCL)
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
- * E-mail: (YTC) (YC); (CHC); (JYW); (YCL)
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
- * E-mail: (YTC) (YC); (CHC); (JYW); (YCL)
| | - Yi-Ching Lee
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- * E-mail: (YTC) (YC); (CHC); (JYW); (YCL)
| |
Collapse
|
353
|
Urayama KY, Chokkalingam AP, Metayer C, Hansen H, May S, Ramsay P, Wiemels JL, Wiencke JK, Trachtenberg E, Thompson P, Ishida Y, Brennan P, Jolly KW, Termuhlen AM, Taylor M, Barcellos LF, Buffler PA. SNP association mapping across the extended major histocompatibility complex and risk of B-cell precursor acute lymphoblastic leukemia in children. PLoS One 2013; 8:e72557. [PMID: 23991122 PMCID: PMC3749982 DOI: 10.1371/journal.pone.0072557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 07/12/2013] [Indexed: 02/01/2023] Open
Abstract
The extended major histocompatibility complex (xMHC) is the most gene-dense region of the genome and harbors a disproportionately large number of genes involved in immune function. The postulated role of infection in the causation of childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL) suggests that the xMHC may make an important contribution to the risk of this disease. We conducted association mapping across an approximately 4 megabase region of the xMHC using a validated panel of single nucleotide polymorphisms (SNPs) in childhood BCP-ALL cases (n=567) enrolled in the Northern California Childhood Leukemia Study (NCCLS) compared with population controls (n=892). Logistic regression analyses of 1,145 SNPs, adjusted for age, sex, and Hispanic ethnicity indicated potential associations between several SNPs and childhood BCP-ALL. After accounting for multiple comparisons, one of these included a statistically significant increased risk associated with rs9296068 (OR=1.40, 95% CI=1.19-1.66, corrected p=0.036), located in proximity to HLA-DOA. Sliding window haplotype analysis identified an additional locus located in the extended class I region in proximity to TRIM27 tagged by a haplotype comprising rs1237485, rs3118361, and rs2032502 (corrected global p=0.046). Our findings suggest that susceptibility to childhood BCP-ALL is influenced by genetic variation within the xMHC and indicate at least two important regions for future evaluation.
Collapse
Affiliation(s)
- Kevin Y Urayama
- School of Public Health, University of California, Berkeley, Berkeley, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
354
|
Handel AE, Disanto G, Ramagopalan SV. Next-generation sequencing in understanding complex neurological disease. Expert Rev Neurother 2013; 13:215-27. [PMID: 23368808 DOI: 10.1586/ern.12.165] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Next-generation sequencing techniques have made vast quantities of data on human genomes and transcriptomes available to researchers. Huge progress has been made towards understanding the basis of many Mendelian neurological conditions, but progress has been considerably slower in complex neurological diseases (multiple sclerosis, migraine, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and so on). The authors review current next-generation sequencing methodologies and present selected studies illustrating how these have been used to cast light on the genetic etiology of complex neurological diseases with specific focus on multiple sclerosis. The authors highlight particular pitfalls in next-generation sequencing experiments and speculate on both clinical and research applications of these sequencing platforms for complex neurological disorders in the future.
Collapse
Affiliation(s)
- Adam E Handel
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| | | | | |
Collapse
|
355
|
Genetic insights into common pathways and complex relationships among immune-mediated diseases. Nat Rev Genet 2013; 14:661-73. [PMID: 23917628 DOI: 10.1038/nrg3502] [Citation(s) in RCA: 406] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Shared aetiopathogenic factors among immune-mediated diseases have long been suggested by their co-familiality and co-occurrence, and molecular support has been provided by analysis of human leukocyte antigen (HLA) haplotypes and genome-wide association studies. The interrelationships can now be better appreciated following the genotyping of large immune disease sample sets on a shared SNP array: the 'Immunochip'. Here, we systematically analyse loci shared among major immune-mediated diseases. This reveals that several diseases share multiple susceptibility loci, but there are many nuances. The most associated variant at a given locus frequently differs and, even when shared, the same allele often has opposite associations. Interestingly, risk alleles conferring the largest effect sizes are usually disease-specific. These factors help to explain why early evidence of extensive 'sharing' is not always reflected in epidemiological overlap.
Collapse
|
356
|
Brown CD, Mangravite LM, Engelhardt BE. Integrative modeling of eQTLs and cis-regulatory elements suggests mechanisms underlying cell type specificity of eQTLs. PLoS Genet 2013; 9:e1003649. [PMID: 23935528 PMCID: PMC3731231 DOI: 10.1371/journal.pgen.1003649] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 06/04/2013] [Indexed: 12/11/2022] Open
Abstract
Genetic variants in cis-regulatory elements or trans-acting regulators frequently influence the quantity and spatiotemporal distribution of gene transcription. Recent interest in expression quantitative trait locus (eQTL) mapping has paralleled the adoption of genome-wide association studies (GWAS) for the analysis of complex traits and disease in humans. Under the hypothesis that many GWAS associations tag non-coding SNPs with small effects, and that these SNPs exert phenotypic control by modifying gene expression, it has become common to interpret GWAS associations using eQTL data. To fully exploit the mechanistic interpretability of eQTL-GWAS comparisons, an improved understanding of the genetic architecture and causal mechanisms of cell type specificity of eQTLs is required. We address this need by performing an eQTL analysis in three parts: first we identified eQTLs from eleven studies on seven cell types; then we integrated eQTL data with cis-regulatory element (CRE) data from the ENCODE project; finally we built a set of classifiers to predict the cell type specificity of eQTLs. The cell type specificity of eQTLs is associated with eQTL SNP overlap with hundreds of cell type specific CRE classes, including enhancer, promoter, and repressive chromatin marks, regions of open chromatin, and many classes of DNA binding proteins. These associations provide insight into the molecular mechanisms generating the cell type specificity of eQTLs and the mode of regulation of corresponding eQTLs. Using a random forest classifier with cell specific CRE-SNP overlap as features, we demonstrate the feasibility of predicting the cell type specificity of eQTLs. We then demonstrate that CREs from a trait-associated cell type can be used to annotate GWAS associations in the absence of eQTL data for that cell type. We anticipate that such integrative, predictive modeling of cell specificity will improve our ability to understand the mechanistic basis of human complex phenotypic variation. When interpreting genome-wide association studies showing that specific genetic variants are associated with disease risk, scientists look for a link between the genetic variant and a biological mechanism behind that disease. One functional mechanism is that the genetic variant may influence gene transcription via a co-localized genomic regulatory element, such as a transcription factor binding site within an open chromatin region. Often this type of regulation occurs in some cell types but not others. In this study, we look across eleven gene expression studies with seven cell types and consider how genetic transcription regulators, or eQTLs, replicate within and between cell types. We identify pervasive allelic heterogeneity, or transcriptional control of a single gene by multiple, independent eQTLs. We integrate extensive data on cell type specific regulatory elements from ENCODE to identify general methods of transcription regulation through enrichment of eQTLs within regulatory elements. We also build a classifier to predict eQTL replication across cell types. The results in this paper present a path to an integrative, predictive approach to improve our ability to understand the mechanistic basis of human phenotypic variation.
Collapse
Affiliation(s)
- Christopher D. Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (CDB); (BEE)
| | | | - Barbara E. Engelhardt
- Biostatistics & Bioinformatics Department, Duke University, Durham, North Carolina, United States of America
- Department of Statistical Science, Duke University, Durham, North Carolina, United States of America
- Institute for Genome Sciences & Policy, Duke University, Durham, North Carolina, United States of America
- * E-mail: (CDB); (BEE)
| |
Collapse
|
357
|
Lamontagne M, Couture C, Postma DS, Timens W, Sin DD, Paré PD, Hogg JC, Nickle D, Laviolette M, Bossé Y. Refining susceptibility loci of chronic obstructive pulmonary disease with lung eqtls. PLoS One 2013; 8:e70220. [PMID: 23936167 PMCID: PMC3728203 DOI: 10.1371/journal.pone.0070220] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/14/2013] [Indexed: 01/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of mortality worldwide. Recent genome-wide association studies (GWAS) have identified robust susceptibility loci associated with COPD. However, the mechanisms mediating the risk conferred by these loci remain to be found. The goal of this study was to identify causal genes/variants within susceptibility loci associated with COPD. In the discovery cohort, genome-wide gene expression profiles of 500 non-tumor lung specimens were obtained from patients undergoing lung surgery. Blood-DNA from the same patients were genotyped for 1,2 million SNPs. Following genotyping and gene expression quality control filters, 409 samples were analyzed. Lung expression quantitative trait loci (eQTLs) were identified and overlaid onto three COPD susceptibility loci derived from GWAS; 4q31 (HHIP), 4q22 (FAM13A), and 19q13 (RAB4B, EGLN2, MIA, CYP2A6). Significant eQTLs were replicated in two independent datasets (n = 363 and 339). SNPs previously associated with COPD and lung function on 4q31 (rs1828591, rs13118928) were associated with the mRNA expression of HHIP. An association between mRNA expression level of FAM13A and SNP rs2045517 was detected at 4q22, but did not reach statistical significance. At 19q13, significant eQTLs were detected with EGLN2. In summary, this study supports HHIP, FAM13A, and EGLN2 as the most likely causal COPD genes on 4q31, 4q22, and 19q13, respectively. Strong lung eQTL SNPs identified in this study will need to be tested for association with COPD in case-control studies. Further functional studies will also be needed to understand the role of genes regulated by disease-related variants in COPD.
Collapse
Affiliation(s)
- Maxime Lamontagne
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Christian Couture
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Dirkje S. Postma
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, GRIAC Research Institute, Groningen, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, GRIAC Research Institute, Groningen, The Netherlands
| | - Don D. Sin
- University of British Columbia James Hogg Research Center, Center for Heart and Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Respiratory Division, Department of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter D. Paré
- University of British Columbia James Hogg Research Center, Center for Heart and Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Respiratory Division, Department of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - James C. Hogg
- University of British Columbia James Hogg Research Center, Center for Heart and Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - David Nickle
- Merck & Co. Inc., Rahway, New Jersey, United States of America
| | - Michel Laviolette
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Yohan Bossé
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Department of Molecular Medicine, Laval University, Québec, Canada
- * E-mail:
| |
Collapse
|
358
|
Plenge RM, Scolnick EM, Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov 2013; 12:581-94. [PMID: 23868113 DOI: 10.1038/nrd4051] [Citation(s) in RCA: 469] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than 90% of the compounds that enter clinical trials fail to demonstrate sufficient safety and efficacy to gain regulatory approval. Most of this failure is due to the limited predictive value of preclinical models of disease, and our continued ignorance regarding the consequences of perturbing specific targets over long periods of time in humans. 'Experiments of nature' - naturally occurring mutations in humans that affect the activity of a particular protein target or targets - can be used to estimate the probable efficacy and toxicity of a drug targeting such proteins, as well as to establish causal rather than reactive relationships between targets and outcomes. Here, we describe the concept of dose-response curves derived from experiments of nature, with an emphasis on human genetics as a valuable tool to prioritize molecular targets in drug development. We discuss empirical examples of drug-gene pairs that support the role of human genetics in testing therapeutic hypotheses at the stage of target validation, provide objective criteria to prioritize genetic findings for future drug discovery efforts and highlight the limitations of a target validation approach that is anchored in human genetics.
Collapse
Affiliation(s)
- Robert M Plenge
- Division of Rheumatology, Immunology and Allergy, Brigham And Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
359
|
Abstract
Over several decades, various forms of genomic analysis of the human major histocompatibility complex (MHC) have been extremely successful in picking up many disease associations. This is to be expected, as the MHC region is one of the most gene-dense and polymorphic stretches of human DNA. It also encodes proteins critical to immunity, including several controlling antigen processing and presentation. Single-nucleotide polymorphism genotyping and human leukocyte antigen (HLA) imputation now permit the screening of large sample sets, a technique further facilitated by high-throughput sequencing. These methods promise to yield more precise contributions of MHC variants to disease. However, interpretation of MHC-disease associations in terms of the functions of variants has been problematic. Most studies confirm the paramount importance of class I and class II molecules, which are key to resistance to infection. Infection is likely driving the extreme variation of these genes across the human population, but this has been difficult to demonstrate. In contrast, many associations with autoimmune conditions have been shown to be specific to certain class I and class II alleles. Interestingly, conditions other than infections and autoimmunity are also associated with the MHC, including some cancers and neuropathies. These associations could be indirect, owing, for example, to the infectious history of a particular individual and selective pressures operating at the population level.
Collapse
Affiliation(s)
- John Trowsdale
- Department of Pathology and Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 1QP, United Kingdom;
| | | |
Collapse
|
360
|
Forabosco P, Ramasamy A, Trabzuni D, Walker R, Smith C, Bras J, Levine AP, Hardy J, Pocock JM, Guerreiro R, Weale ME, Ryten M. Insights into TREM2 biology by network analysis of human brain gene expression data. Neurobiol Aging 2013; 34:2699-714. [PMID: 23855984 PMCID: PMC3988951 DOI: 10.1016/j.neurobiolaging.2013.05.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/01/2013] [Indexed: 11/29/2022]
Abstract
Rare variants in TREM2 cause susceptibility to late-onset Alzheimer's disease. Here we use microarray-based expression data generated from 101 neuropathologically normal individuals and covering 10 brain regions, including the hippocampus, to understand TREM2 biology in human brain. Using network analysis, we detect a highly preserved TREM2-containing module in human brain, show that it relates to microglia, and demonstrate that TREM2 is a hub gene in 5 brain regions, including the hippocampus, suggesting that it can drive module function. Using enrichment analysis we show significant overrepresentation of genes implicated in the adaptive and innate immune system. Inspection of genes with the highest connectivity to TREM2 suggests that it plays a key role in mediating changes in the microglial cytoskeleton necessary not only for phagocytosis, but also migration. Most importantly, we show that the TREM2-containing module is significantly enriched for genes genetically implicated in Alzheimer's disease, multiple sclerosis, and motor neuron disease, implying that these diseases share common pathways centered on microglia and that among the genes identified are possible new disease-relevant genes.
Collapse
|
361
|
Smith SL, Plant D, Eyre S, Barton A. The potential use of expression profiling: implications for predicting treatment response in rheumatoid arthritis. Ann Rheum Dis 2013; 72:1118-24. [PMID: 23486412 DOI: 10.1136/annrheumdis-2012-202743] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Whole genome expression profiling, or transcriptomics, is a high throughput technology with the potential for major impacts in both clinical settings and drug discovery and diagnostics. In particular, there is much interest in this technique as a mechanism for predicting treatment response. Gene expression profiling entails the quantitative measurement of messenger RNA levels for thousands of genes simultaneously with the inherent possibility of identifying biomarkers of response to a particular therapy or by singling out those at risk of serious adverse events. This technology should contribute to the era of stratified medicine, in which patient specific populations are matched to potentially beneficial drugs via clinical tests. Indeed, in the oncology field, gene expression testing is already recommended to allow rational use of therapies to treat breast cancer. However, there are still many issues surrounding the use of the various testing platforms available and the statistical analysis associated with the interpretation of results generated. This review will discuss the implications this promising technology has in predicting treatment response and outline the various advantages and pitfalls associated with its use.
Collapse
Affiliation(s)
- Samantha Louise Smith
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Sciences Centre, School of Translational Medicine, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
362
|
Bønnelykke K, Matheson MC, Pers TH, Granell R, Strachan DP, Alves AC, Linneberg A, Curtin JA, Warrington NM, Standl M, Kerkhof M, Jonsdottir I, Bukvic BK, Kaakinen M, Sleimann P, Thorleifsson G, Thorsteinsdottir U, Schramm K, Baltic S, Kreiner-Møller E, Simpson A, St Pourcain B, Coin L, Hui J, Walters EH, Tiesler CMT, Duffy DL, Jones G, Ring SM, McArdle WL, Price L, Robertson CF, Pekkanen J, Tang CS, Thiering E, Montgomery GW, Hartikainen AL, Dharmage SC, Husemoen LL, Herder C, Kemp JP, Elliot P, James A, Waldenberger M, Abramson MJ, Fairfax BP, Knight JC, Gupta R, Thompson PJ, Holt P, Sly P, Hirschhorn JN, Blekic M, Weidinger S, Hakonarsson H, Stefansson K, Heinrich J, Postma DS, Custovic A, Pennell CE, Jarvelin MR, Koppelman GH, Timpson N, Ferreira MA, Bisgaard H, Henderson AJ. Meta-analysis of genome-wide association studies identifies ten loci influencing allergic sensitization. Nat Genet 2013; 45:902-906. [PMID: 23817571 DOI: 10.1038/ng.2694] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 06/10/2013] [Indexed: 12/17/2022]
Abstract
Allergen-specific immunoglobulin E (present in allergic sensitization) has a central role in the pathogenesis of allergic disease. We performed the first large-scale genome-wide association study (GWAS) of allergic sensitization in 5,789 affected individuals and 10,056 controls and followed up the top SNP at each of 26 loci in 6,114 affected individuals and 9,920 controls. We increased the number of susceptibility loci with genome-wide significant association with allergic sensitization from three to ten, including SNPs in or near TLR6, C11orf30, STAT6, SLC25A46, HLA-DQB1, IL1RL1, LPP, MYC, IL2 and HLA-B. All the top SNPs were associated with allergic symptoms in an independent study. Risk-associated variants at these ten loci were estimated to account for at least 25% of allergic sensitization and allergic rhinitis. Understanding the molecular mechanisms underlying these associations may provide new insights into the etiology of allergic disease.
Collapse
Affiliation(s)
- Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Melanie C Matheson
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, University of Melbourne, Melbourne, Australia
| | - Tune H Pers
- Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark.,Division of Endocrinology, Children's Hospital, Boston, USA.,Center for Basic and Translational Obesity Research, Children's Hospital, Boston, USA.,Broad Institute, Cambridge, USA
| | - Raquel Granell
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - David P Strachan
- Division of Population Health Sciences & Education, St George's, University of London, London, UK
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
| | - John A Curtin
- University of Manchester, Manchester Academic Health Science Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Nicole M Warrington
- School of Women's and Infants' Health, The University of Western Australia, Crawley, Australia
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Marjan Kerkhof
- Department of Epidemiology, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ingileif Jonsdottir
- deCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland.,University of Iceland, 101 Reykjavik, Iceland
| | - Blazenka K Bukvic
- General Hospital "Dr Josip Bencevic" Slavonski Brod, University of Osijek, Osijek, Croatia
| | - Marika Kaakinen
- Institute of Health Sciences, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Patrick Sleimann
- Center for Applied Genomics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Unnur Thorsteinsdottir
- deCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland.,University of Iceland, 101 Reykjavik, Iceland
| | - Katharina Schramm
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Svetlana Baltic
- Lung Institute of WA, University of WA, Perth, Western Australia, Australia.,Centre for Asthma, Allergy and Respiratory Research, University of WA, Perth, Western Australia, Australia
| | - Eskil Kreiner-Møller
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Angela Simpson
- University of Manchester, Manchester Academic Health Science Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Beate St Pourcain
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Lachlan Coin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia (WA), Nedlands, Australia.,School of Population Health, The University of WA, Nedlands, Australia.,School of Pathology and Laboratory Medicine, The University of WA, Nedlands, Australia.,Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | | | - Carla M T Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - David L Duffy
- Queensland Institute of Medical Research, Brisbane, Australia
| | - Graham Jones
- School of Science and Health, University of Western Sydney, Penrith, Australia
| | | | - Susan M Ring
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Wendy L McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, UK.,Division of Population Health Sciences & Education, St George's, University of London, London, UK
| | - Loren Price
- Lung Institute of WA, University of WA, Perth, Western Australia, Australia.,Centre for Asthma, Allergy and Respiratory Research, University of WA, Perth, Western Australia, Australia
| | - Colin F Robertson
- Respiratory Medicine, Murdoch Children's Research Institute, Melbourne, Australia
| | - Juha Pekkanen
- Department of Environmental Health, National Institute for Health and Welfare (THL), Kuopio, Finland.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Clara S Tang
- Queensland Institute of Medical Research, Brisbane, Australia
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Anna-Liisa Hartikainen
- Department of Clinical Sciences, Obstetrics and Gynecology, Institute of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Shyamali C Dharmage
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, University of Melbourne, Melbourne, Australia
| | - Lise L Husemoen
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - John P Kemp
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Paul Elliot
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Alan James
- Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of WA, Nedlands, Australia.,Department of Pulmonary Physiology, West Australian Sleep Disorders Research Institute, Nedlands, Australia
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael J Abramson
- Department of Epidemiology & Preventive Medicine, Monash University, Melbourne, Australia
| | - Benjamin P Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ramneek Gupta
- Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - Philip J Thompson
- Lung Institute of WA, University of WA, Perth, Western Australia, Australia.,Centre for Asthma, Allergy and Respiratory Research, University of WA, Perth, Western Australia, Australia
| | - Patrick Holt
- Telethon Institute for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter Sly
- Queensland Children's Medical Research Institute, University of Queensland, World Health Organization (WHO) Collaborating Centre for Research on Children's Environmental Health, Australia
| | - Joel N Hirschhorn
- Broad Institute, Cambridge, USA.,Division of Genetics, Children's Hospital, Boston, USA.,Division of Endocrinology, Children's Hospital, Boston, USA.,Department of Genetics, Harvard Medical School, Boston, USA
| | - Mario Blekic
- General Hospital "Dr Josip Bencevic" Slavonski Brod, University of Osijek, Osijek, Croatia
| | - Stephan Weidinger
- Department of Dermatology, Allergology, and Venerology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Hakon Hakonarsson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kari Stefansson
- deCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland.,University of Iceland, 101 Reykjavik, Iceland
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Dirkje S Postma
- Department of Pulmonology, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adnan Custovic
- University of Manchester, Manchester Academic Health Science Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Craig E Pennell
- School of Women's and Infants' Health, The University of Western Australia, Crawley, Australia
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Department of Epidemiology and Biostatistics, School of Public Health, MRC-HPA Centre for Environment and Health, Imperial College London, UK.,National Institute of Health and Welfare, Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology , Beatrix Children's Hospital, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nicholas Timpson
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - A John Henderson
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
363
|
Li S, Nakaya HI, Kazmin DA, Oh JZ, Pulendran B. Systems biological approaches to measure and understand vaccine immunity in humans. Semin Immunol 2013; 25:209-18. [PMID: 23796714 DOI: 10.1016/j.smim.2013.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 05/09/2013] [Indexed: 02/01/2023]
Abstract
Recent studies have demonstrated the utility of using systems approaches to identify molecular signatures that can be used to predict vaccine immunity in humans. Such approaches are now being used extensively in vaccinology, and are beginning to yield novel insights about the molecular networks driving vaccine immunity. In this review, we present a broad review of the methodologies involved in these studies, and discuss the promise and challenges involved in this emerging field of "systems vaccinology."
Collapse
Affiliation(s)
- Shuzhao Li
- Emory Vaccine Center, Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA
| | | | | | | | | |
Collapse
|
364
|
Fusco DN, Brisac C, John SP, Huang YW, Chin CR, Xie T, Zhao H, Zhang L, Chevalier S, Wambua D, Lin W, Peng L, Chung RT, Brass AL. A genetic screen identifies interferon-α effector genes required to suppress hepatitis C virus replication. Gastroenterology 2013; 144:1438-49, 1449.e1-9. [PMID: 23462180 PMCID: PMC3665646 DOI: 10.1053/j.gastro.2013.02.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 01/15/2013] [Accepted: 02/12/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV) infection is a leading cause of end-stage liver disease. Interferon-α (IFNα) is an important component of anti-HCV therapy; it up-regulates transcription of IFN-stimulated genes, many of which have been investigated for their antiviral effects. However, all of the genes required for the antiviral function of IFNα (IFN effector genes [IEGs]) are not known. IEGs include not only IFN-stimulated genes, but other nontranscriptionally induced genes that are required for the antiviral effect of IFNα. In contrast to candidate approaches based on analyses of messenger RNA (mRNA) expression, identification of IEGs requires a broad functional approach. METHODS We performed an unbiased genome-wide small interfering RNA screen to identify IEGs that inhibit HCV. Huh7.5.1 hepatoma cells were transfected with small interfering RNAs incubated with IFNα and then infected with JFH1 HCV. Cells were stained using HCV core antibody, imaged, and analyzed to determine the percent infection. Candidate IEGs detected in the screen were validated and analyzed further. RESULTS The screen identified 120 previously unreported IEGs. From these, we more fully evaluated the following: asparagine-linked glycosylation 10 homolog (yeast, α-1,2-glucosyltransferase); butyrylcholinesterase; dipeptidyl-peptidase 4 (CD26, adenosine deaminase complexing protein 2); glucokinase (hexokinase 4) regulator; guanylate cyclase 1, soluble, β 3; MYST histone acetyltransferase 1; protein phosphatase 3 (formerly 2B), catalytic subunit, β isoform; peroxisomal proliferator-activated receptor-γ-DBD-interacting protein 1; and solute carrier family 27 (fatty acid transporter), member 2; and demonstrated that they enabled IFNα-mediated suppression of HCV at multiple steps of its life cycle. Expression of these genes had more potent effects against flaviviridae because a subset was required for IFNα to suppress dengue virus but not influenza A virus. In addition, many of the host genes detected in this screen (92%) were not transcriptionally stimulated by IFNα; these genes represent a heretofore unknown class of non-IFN-stimulated gene IEGs. CONCLUSIONS We performed a whole-genome loss-of-function screen to identify genes that mediate the effects of IFNα against human pathogenic viruses. We found that IFNα restricts HCV via actions of general and specific IEGs.
Collapse
Affiliation(s)
- Dahlene N. Fusco
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Cynthia Brisac
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Sinu P. John
- Laboratory of Systems Biology, NIAID/NIH, Bethesda, MD
| | - Yi-Wen Huang
- Department of Internal Medicine, National Taiwan University College of, Medicine and Hospital, Liver Center, Cathay General Hospital Medical Center &, School of Medicine, Taipei Medical University, No. 280, Sec. 4, Jen-Ai Road, Taipei-10630, Taiwan
| | - Christopher R. Chin
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester MA 01605
| | - Tiao Xie
- Harvard Medical School Image and Data Analysis Core, 240 Longwood Avenue, Boston, MA 02115
| | - Hong Zhao
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China, 100034
| | - Leiliang Zhang
- MOH Key Laboratory of Systems Biology of Pathogens; Institute of Pathogen Biology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, China
| | - Stephane Chevalier
- Hospital University Henri Mondor, Department of Virology, Université Paris-Est, Créteil, France, Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Daniel Wambua
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Wenyu Lin
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Lee Peng
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Raymond T. Chung
- Gastrointestinal Unit, Massachusetts General Hospital, 55 Fruit Street Boston MA 02114
| | - Abraham L. Brass
- Ragon Institute, 149 13th Street Charlestown, MA 02129, Current Address: Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester MA 01605
| |
Collapse
|
365
|
Idaghdour Y, Awadalla P. Exploiting gene expression variation to capture gene-environment interactions for disease. Front Genet 2013; 3:228. [PMID: 23755064 PMCID: PMC3668192 DOI: 10.3389/fgene.2012.00228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/10/2012] [Indexed: 12/18/2022] Open
Abstract
Gene-environment interactions have long been recognized as a fundamental concept in evolutionary, quantitative, and medical genetics. In the genomics era, study of how environment and genome interact to shape gene expression variation is relevant to understanding the genetic architecture of complex phenotypes. While genetic analysis of gene expression variation focused on main effects, little is known about the extent of interaction effects implicating regulatory variants and their consequences on transcriptional variation. Here we survey the current state of the concept of transcriptional gene-environment interactions and discuss its utility for mapping disease phenotypes in light of the insights gained from genome-wide association studies of gene expression.
Collapse
Affiliation(s)
- Youssef Idaghdour
- Sainte-Justine Research Center, University of Montreal Montreal, QC, Canada
| | | |
Collapse
|
366
|
Abstract
Identification and functional interpretation of gene regulatory variants is a major focus of modern genomics. The application of genetic mapping to molecular and cellular traits has enabled the detection of regulatory variation on genome-wide scales and revealed an enormous diversity of regulatory architecture in humans and other species. In this review I summarise the insights gained and questions raised by a decade of genetic mapping of gene expression variation. I discuss recent extensions of this approach using alternative molecular phenotypes that have revealed some of the biological mechanisms that drive gene expression variation between individuals. Finally, I highlight outstanding problems and future directions for development.
Collapse
|
367
|
Li G, Diogo D, Wu D, Spoonamore J, Dancik V, Franke L, Kurreeman F, Rossin EJ, Duclos G, Hartland C, Zhou X, Li K, Liu J, De Jager PL, Siminovitch KA, Zhernakova A, Raychaudhuri S, Bowes J, Eyre S, Padyukov L, Gregersen PK, Worthington J, Rheumatoid Arthritis Consortium International (RACI), Gupta N, Clemons PA, Stahl E, Tolliday N, Plenge RM. Human genetics in rheumatoid arthritis guides a high-throughput drug screen of the CD40 signaling pathway. PLoS Genet 2013; 9:e1003487. [PMID: 23696745 PMCID: PMC3656093 DOI: 10.1371/journal.pgen.1003487] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/15/2013] [Indexed: 12/21/2022] Open
Abstract
Although genetic and non-genetic studies in mouse and human implicate the CD40 pathway in rheumatoid arthritis (RA), there are no approved drugs that inhibit CD40 signaling for clinical care in RA or any other disease. Here, we sought to understand the biological consequences of a CD40 risk variant in RA discovered by a previous genome-wide association study (GWAS) and to perform a high-throughput drug screen for modulators of CD40 signaling based on human genetic findings. First, we fine-map the CD40 risk locus in 7,222 seropositive RA patients and 15,870 controls, together with deep sequencing of CD40 coding exons in 500 RA cases and 650 controls, to identify a single SNP that explains the entire signal of association (rs4810485, P = 1.4×10−9). Second, we demonstrate that subjects homozygous for the RA risk allele have ∼33% more CD40 on the surface of primary human CD19+ B lymphocytes than subjects homozygous for the non-risk allele (P = 10−9), a finding corroborated by expression quantitative trait loci (eQTL) analysis in peripheral blood mononuclear cells from 1,469 healthy control individuals. Third, we use retroviral shRNA infection to perturb the amount of CD40 on the surface of a human B lymphocyte cell line (BL2) and observe a direct correlation between amount of CD40 protein and phosphorylation of RelA (p65), a subunit of the NF-κB transcription factor. Finally, we develop a high-throughput NF-κB luciferase reporter assay in BL2 cells activated with trimerized CD40 ligand (tCD40L) and conduct an HTS of 1,982 chemical compounds and FDA–approved drugs. After a series of counter-screens and testing in primary human CD19+ B cells, we identify 2 novel chemical inhibitors not previously implicated in inflammation or CD40-mediated NF-κB signaling. Our study demonstrates proof-of-concept that human genetics can be used to guide the development of phenotype-based, high-throughput small-molecule screens to identify potential novel therapies in complex traits such as RA. A current challenge in human genetics is to follow-up “hits” from genome-wide association studies (GWAS) to guide drug discovery for complex traits. Previously, we identified a common variant in the CD40 locus as associated with risk of rheumatoid arthritis (RA). Here, we fine-map the CD40 signal of association through a combination of dense genotyping and exonic sequencing in large patient collections. Further, we demonstrate that the RA risk allele is a gain-of-function allele that increases the amount of CD40 on the surface of primary human B lymphocyte cells from healthy control individuals. Based on these observations, we develop a high-throughput assay to recapitulate the biology of the RA risk allele in a system suitable for a small molecule drug screen. After a series of primary screens and counter screens, we identify small molecules that inhibit CD40-mediated NF-kB signaling in human B cells. While this is only the first step towards a more comprehensive effort to identify CD40-specific inhibitors that may be used to treat RA, our study demonstrates a successful strategy to progress from a GWAS to a drug screen for complex traits such as RA.
Collapse
Affiliation(s)
- Gang Li
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dorothée Diogo
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Di Wu
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
- Department of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
| | - Jim Spoonamore
- Chemical Biology Platform, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Vlado Dancik
- Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Fina Kurreeman
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Elizabeth J. Rossin
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
- Biological and Biomedical Sciences Program, Health Sciences and Technology Program, Harvard Medical School, Boston, Massachusetts, United States of America
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Grant Duclos
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cathy Hartland
- Chemical Biology Platform, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Xuezhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Kejie Li
- Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Jun Liu
- Department of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
| | - Philip L. De Jager
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Katherine A. Siminovitch
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Mount Sinai Hospital, Samuel Lunenfeld Research Institute and Toronto General Research Institute, Toronto, Ontario, Canada
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
- Department of Rheumatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - John Bowes
- Arthritis Research UK Epidemiology Unit, Musculoskeletal Research Group, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Steve Eyre
- Arthritis Research UK Epidemiology Unit, Musculoskeletal Research Group, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Peter K. Gregersen
- The Feinstein Institute for Medical Research, North Shore–Long Island Jewish Health System, Manhasset, New York, United States of America
| | - Jane Worthington
- Arthritis Research UK Epidemiology Unit, Musculoskeletal Research Group, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | | | - Namrata Gupta
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Paul A. Clemons
- Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Eli Stahl
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Nicola Tolliday
- Chemical Biology Platform, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Robert M. Plenge
- Division of Rheumatology, Immunology, and Allergy and Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Chemical Biology Program, Broad Institute, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
368
|
Lu M, Varley AW, Munford RS. Persistently active microbial molecules prolong innate immune tolerance in vivo. PLoS Pathog 2013; 9:e1003339. [PMID: 23675296 PMCID: PMC3649966 DOI: 10.1371/journal.ppat.1003339] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 03/18/2013] [Indexed: 01/01/2023] Open
Abstract
Measures that bolster the resolution phase of infectious diseases may offer new opportunities for improving outcome. Here we show that inactivation of microbial lipopolysaccharides (LPS) can be required for animals to recover from the innate immune tolerance that follows exposure to Gram-negative bacteria. When wildtype mice are exposed to small parenteral doses of LPS or Gram-negative bacteria, their macrophages become reprogrammed (tolerant) for a few days before they resume normal function. Mice that are unable to inactivate LPS, in contrast, remain tolerant for several months; during this time they respond sluggishly to Gram-negative bacterial challenge, with high mortality. We show here that prolonged macrophage reprogramming is maintained in vivo by the persistence of stimulatory LPS molecules within the cells' in vivo environment, where naïve cells can acquire LPS via cell-cell contact or from the extracellular fluid. The findings provide strong evidence that inactivation of a stimulatory microbial molecule can be required for animals to regain immune homeostasis following parenteral exposure to bacteria. Measures that disable microbial molecules might enhance resolution of tissue inflammation and help restore innate defenses in individuals recovering from many different infectious diseases. We showed previously that mice lacking acyloxyacyl hydrolase (AOAH), the host enzyme that inactivates Gram-negative bacterial lipopolysaccharides (LPS), are unable to regain normal immune responsiveness for many weeks/months after they are exposed in vivo to a small amount of LPS or Gram-negative bacteria. The many possible explanations for slow recovery included long-lasting epigenetic changes in macrophages or other host cells, chronically stimulated cells that produce certain mediators, and persistent signaling by internalized LPS within macrophages. Using several in vivo techniques to study peritoneal macrophages, we found that none of these mechanisms was correct. Rather, prolonged recovery is caused by intact LPS that remains in the environment where macrophages live and can pass from one cell to another in vivo. This is the first evidence that the persistence of a bioactive microbial agonist, per se, can prevent resolution of inflammation in vivo. It also identifies the stimulatory microbial molecule as a realistic target for intervention – in further support, we found that providing recombinant AOAH can be partially preventive. In a larger sense, showing that chemical inactivation of one important microbial signaling molecule is required for full recovery should encourage efforts to find out whether disabling other microbial agonists (chitin, lipopeptides, flagella, others) also benefits infected animals.
Collapse
Affiliation(s)
- Mingfang Lu
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | |
Collapse
|
369
|
Henig N, Avidan N, Mandel I, Staun-Ram E, Ginzburg E, Paperna T, Pinter RY, Miller A. Interferon-beta induces distinct gene expression response patterns in human monocytes versus T cells. PLoS One 2013; 8:e62366. [PMID: 23626809 PMCID: PMC3633862 DOI: 10.1371/journal.pone.0062366] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 03/21/2013] [Indexed: 12/31/2022] Open
Abstract
Background Monocytes, which are key players in innate immunity, are outnumbered by neutrophils and lymphocytes among peripheral white blood cells. The cytokine interferon-β (IFN-β) is widely used as an immunomodulatory drug for multiple sclerosis and its functional pathways in peripheral blood mononuclear cells (PBMCs) have been previously described. The aim of the present study was to identify novel, cell-specific IFN-β functions and pathways in tumor necrosis factor (TNF)-α-activated monocytes that may have been missed in studies using PBMCs. Methodology/Principal Findings Whole genome gene expression profiles of human monocytes and T cells were compared following in vitro priming to TNF-α and overnight exposure to IFN-β. Statistical analyses of the gene expression data revealed a cell-type-specific change of 699 transcripts, 667 monocyte-specific transcripts, 21 T cell-specific transcripts and 11 transcripts with either a difference in the response direction or a difference in the magnitude of response. RT-PCR revealed a set of differentially expressed genes (DEGs), exhibiting responses to IFN-β that are modulated by TNF-α in monocytes, such as RIPK2 and CD83, but not in T cells or PBMCs. Known IFN-β promoter response elements, such as ISRE, were enriched in T cell DEGs but not in monocyte DEGs. The overall directionality of the gene expression regulation by IFN-β was different in T cells and monocytes, with up-regulation more prevalent in T cells, and a similar extent of up and down-regulation recorded in monocytes. Conclusions By focusing on the response of distinct cell types and by evaluating the combined effects of two cytokines with pro and anti-inflammatory activities, we were able to present two new findings First, new IFN-β response pathways and genes, some of which were monocytes specific; second, a cell-specific modulation of the IFN-β response transcriptome by TNF-α.
Collapse
Affiliation(s)
- Noa Henig
- Division of Neuroimmunology and Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
- Department of Computer Science, Technion-Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nili Avidan
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ilana Mandel
- Division of Neuroimmunology and Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
| | - Elsebeth Staun-Ram
- Division of Neuroimmunology and Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Elizabeta Ginzburg
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Paperna
- Division of Neuroimmunology and Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ron Y. Pinter
- Department of Computer Science, Technion-Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ariel Miller
- Division of Neuroimmunology and Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine & Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
- * E-mail:
| |
Collapse
|
370
|
Civelek M, Hagopian R, Pan C, Che N, Yang WP, Kayne PS, Saleem NK, Cederberg H, Kuusisto J, Gargalovic PS, Kirchgessner TG, Laakso M, Lusis AJ. Genetic regulation of human adipose microRNA expression and its consequences for metabolic traits. Hum Mol Genet 2013; 22:3023-37. [PMID: 23562819 DOI: 10.1093/hmg/ddt159] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The genetics of messenger RNA (mRNA) expression has been extensively studied in humans and other organisms, but little is known about genetic factors contributing to microRNA (miRNA) expression. We examined natural variation of miRNA expression in adipose tissue in a population of 200 men who have been carefully characterized for metabolic syndrome (MetSyn) phenotypes as part of the Metabolic Syndrome in Men (METSIM) study. We genotyped the subjects using high-density single-nucleotide polymorphism microarrays and quantified the mRNA abundance using genome-wide expression arrays and miRNA abundance using next-generation sequencing. We reliably quantified 356 miRNA species that were expressed in human adipose tissue, a limited number of which made up most of the expressed miRNAs. We mapped the miRNA abundance as an expression quantitative trait and determined cis regulation of expression for nine of the miRNAs and of the processing of one miRNA (miR-28). The degree of genetic variation of miRNA expression was substantially less than that of mRNAs. For the majority of the miRNAs, genetic regulation of expression was independent of the expression of mRNA from which the miRNA is transcribed. We also showed that for 108 miRNAs, mapped reads displayed widespread variation from the canonical sequence. We found a total of 24 miRNAs to be significantly associated with MetSyn traits. We suggest a regulatory role for miR-204-5p which was predicted to inhibit acetyl coenzyme A carboxylase β, a key fatty acid oxidation enzyme that has been shown to play a role in regulating body fat and insulin resistance in adipose tissue.
Collapse
Affiliation(s)
- Mete Civelek
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
371
|
Gat-Viks I, Chevrier N, Wilentzik R, Eisenhaure T, Raychowdhury R, Steuerman Y, Shalek A, Hacohen N, Amit I, Regev A. Deciphering molecular circuits from genetic variation underlying transcriptional responsiveness to stimuli. Nat Biotechnol 2013; 31:342-9. [PMID: 23503680 PMCID: PMC3622156 DOI: 10.1038/nbt.2519] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 02/04/2013] [Indexed: 01/24/2023]
Abstract
Individual genetic variation affects gene responsiveness to stimuli, often by influencing complex molecular circuits. Here we combine genomic and intermediate-scale transcriptional profiling with computational methods to identify variants that affect the responsiveness of genes to stimuli (responsiveness quantitative trait loci or reQTLs) and to position these variants in molecular circuit diagrams. We apply this approach to study variation in transcriptional responsiveness to pathogen components in dendritic cells from recombinant inbred mouse strains. We identify reQTLs that correlate with particular stimuli and position them in known pathways. For example, in response to a virus-like stimulus, a trans-acting variant responds as an activator of the antiviral response; using RNA interference, we identify Rgs16 as the likely causal gene. Our approach charts an experimental and analytic path to decipher the mechanisms underlying genetic variation in circuits that control responses to stimuli.
Collapse
Affiliation(s)
- Irit Gat-Viks
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nicolas Chevrier
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Graduate Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA
- FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Roni Wilentzik
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Thomas Eisenhaure
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Raktima Raychowdhury
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Yael Steuerman
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alex Shalek
- Departments of Chemistry and Chemical Biology and of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, MA 02129, USA, and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ido Amit
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Aviv Regev
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02140, USA
| |
Collapse
|
372
|
Abstract
The molecular basis of adaptation--and, in particular, the relative roles of protein-coding versus gene expression changes--has long been the subject of speculation and debate. Recently, the genotyping of diverse human populations has led to the identification of many putative "local adaptations" that differ between populations. Here I show that these local adaptations are over 10-fold more likely to affect gene expression than amino acid sequence. In addition, a novel framework for identifying polygenic local adaptations detects recent positive selection on the expression levels of genes involved in UV radiation response, immune cell proliferation, and diabetes-related pathways. These results provide the first examples of polygenic gene expression adaptation in humans, as well as the first genome-scale support for the hypothesis that changes in gene expression have driven human adaptation.
Collapse
Affiliation(s)
- Hunter B Fraser
- Department of Biology, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
373
|
Couch FJ, Wang X, McGuffog L, Lee A, Olswold C, Kuchenbaecker KB, Soucy P, Fredericksen Z, Barrowdale D, Dennis J, Gaudet MM, Dicks E, Kosel M, Healey S, Sinilnikova OM, Lee A, Bacot F, Vincent D, Hogervorst FBL, Peock S, Stoppa-Lyonnet D, Jakubowska A, Investigators KC, Radice P, Schmutzler RK, SWE-BRCA, Domchek SM, Piedmonte M, Singer CF, Friedman E, Thomassen M, Ontario Cancer Genetics Network, Hansen TVO, Neuhausen SL, Szabo CI, Blanco I, Greene MH, Karlan BY, Garber J, Phelan CM, Weitzel JN, Montagna M, Olah E, Andrulis IL, Godwin AK, Yannoukakos D, Goldgar DE, Caldes T, Nevanlinna H, Osorio A, Terry MB, Daly MB, van Rensburg EJ, Hamann U, Ramus SJ, Ewart Toland A, Caligo MA, Olopade OI, Tung N, Claes K, Beattie MS, Southey MC, Imyanitov EN, Tischkowitz M, Janavicius R, John EM, Kwong A, Diez O, Balmaña J, Barkardottir RB, Arun BK, Rennert G, Teo SH, Ganz PA, Campbell I, van der Hout AH, van Deurzen CHM, Seynaeve C, Gómez Garcia EB, van Leeuwen FE, Meijers-Heijboer HEJ, Gille JJP, Ausems MGEM, Blok MJ, Ligtenberg MJL, Rookus MA, Devilee P, Verhoef S, van Os TAM, Wijnen JT, HEBON, EMBRACE, Frost D, Ellis S, Fineberg E, Platte R, Evans DG, Izatt L, Eeles RA, Adlard J, et alCouch FJ, Wang X, McGuffog L, Lee A, Olswold C, Kuchenbaecker KB, Soucy P, Fredericksen Z, Barrowdale D, Dennis J, Gaudet MM, Dicks E, Kosel M, Healey S, Sinilnikova OM, Lee A, Bacot F, Vincent D, Hogervorst FBL, Peock S, Stoppa-Lyonnet D, Jakubowska A, Investigators KC, Radice P, Schmutzler RK, SWE-BRCA, Domchek SM, Piedmonte M, Singer CF, Friedman E, Thomassen M, Ontario Cancer Genetics Network, Hansen TVO, Neuhausen SL, Szabo CI, Blanco I, Greene MH, Karlan BY, Garber J, Phelan CM, Weitzel JN, Montagna M, Olah E, Andrulis IL, Godwin AK, Yannoukakos D, Goldgar DE, Caldes T, Nevanlinna H, Osorio A, Terry MB, Daly MB, van Rensburg EJ, Hamann U, Ramus SJ, Ewart Toland A, Caligo MA, Olopade OI, Tung N, Claes K, Beattie MS, Southey MC, Imyanitov EN, Tischkowitz M, Janavicius R, John EM, Kwong A, Diez O, Balmaña J, Barkardottir RB, Arun BK, Rennert G, Teo SH, Ganz PA, Campbell I, van der Hout AH, van Deurzen CHM, Seynaeve C, Gómez Garcia EB, van Leeuwen FE, Meijers-Heijboer HEJ, Gille JJP, Ausems MGEM, Blok MJ, Ligtenberg MJL, Rookus MA, Devilee P, Verhoef S, van Os TAM, Wijnen JT, HEBON, EMBRACE, Frost D, Ellis S, Fineberg E, Platte R, Evans DG, Izatt L, Eeles RA, Adlard J, Eccles DM, Cook J, Brewer C, Douglas F, Hodgson S, Morrison PJ, Side LE, Donaldson A, Houghton C, Rogers MT, Dorkins H, Eason J, Gregory H, McCann E, Murray A, Calender A, Hardouin A, Berthet P, Delnatte C, Nogues C, Lasset C, Houdayer C, Leroux D, Rouleau E, Prieur F, Damiola F, Sobol H, Coupier I, Venat-Bouvet L, Castera L, Gauthier-Villars M, Léoné M, Pujol P, Mazoyer S, Bignon YJ, GEMO Study Collaborators, Złowocka-Perłowska E, Gronwald J, Lubinski J, Durda K, Jaworska K, Huzarski T, Spurdle AB, Viel A, Peissel B, Bonanni B, Melloni G, Ottini L, Papi L, Varesco L, Tibiletti MG, Peterlongo P, Volorio S, Manoukian S, Pensotti V, Arnold N, Engel C, Deissler H, Gadzicki D, Gehrig A, Kast K, Rhiem K, Meindl A, Niederacher D, Ditsch N, Plendl H, Preisler-Adams S, Engert S, Sutter C, Varon-Mateeva R, Wappenschmidt B, Weber BHF, Arver B, Stenmark-Askmalm M, Loman N, Rosenquist R, Einbeigi Z, Nathanson KL, Rebbeck TR, Blank SV, Cohn DE, Rodriguez GC, Small L, Friedlander M, Bae-Jump VL, Fink-Retter A, Rappaport C, Gschwantler-Kaulich D, Pfeiler G, Tea MK, Lindor NM, Kaufman B, Shimon Paluch S, Laitman Y, Skytte AB, Gerdes AM, Pedersen IS, Moeller ST, Kruse TA, Jensen UB, Vijai J, Sarrel K, Robson M, Kauff N, Mulligan AM, Glendon G, Ozcelik H, Ejlertsen B, Nielsen FC, Jønson L, Andersen MK, Ding YC, Steele L, Foretova L, Teulé A, Lazaro C, Brunet J, Pujana MA, Mai PL, Loud JT, Walsh C, Lester J, Orsulic S, Narod SA, Herzog J, Sand SR, Tognazzo S, Agata S, Vaszko T, Weaver J, Stavropoulou AV, Buys SS, Romero A, de la Hoya M, Aittomäki K, Muranen TA, Duran M, Chung WK, Lasa A, Dorfling CM, Miron A, BCFR, Benitez J, Senter L, Huo D, Chan SB, Sokolenko AP, Chiquette J, Tihomirova L, Friebel TM, Agnarsson BA, Lu KH, Lejbkowicz F, James PA, Hall P, Dunning AM, Tessier D, Cunningham J, Slager SL, Wang C, Hart S, Stevens K, Simard J, Pastinen T, Pankratz VS, Offit K, Easton DF, Chenevix-Trench G, Antoniou AC, on behalf of CIMBA. Genome-wide association study in BRCA1 mutation carriers identifies novel loci associated with breast and ovarian cancer risk. PLoS Genet 2013; 9:e1003212. [PMID: 23544013 PMCID: PMC3609646 DOI: 10.1371/journal.pgen.1003212] [Show More Authors] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/14/2012] [Indexed: 12/25/2022] Open
Abstract
BRCA1-associated breast and ovarian cancer risks can be modified by common genetic variants. To identify further cancer risk-modifying loci, we performed a multi-stage GWAS of 11,705 BRCA1 carriers (of whom 5,920 were diagnosed with breast and 1,839 were diagnosed with ovarian cancer), with a further replication in an additional sample of 2,646 BRCA1 carriers. We identified a novel breast cancer risk modifier locus at 1q32 for BRCA1 carriers (rs2290854, P = 2.7 × 10(-8), HR = 1.14, 95% CI: 1.09-1.20). In addition, we identified two novel ovarian cancer risk modifier loci: 17q21.31 (rs17631303, P = 1.4 × 10(-8), HR = 1.27, 95% CI: 1.17-1.38) and 4q32.3 (rs4691139, P = 3.4 × 10(-8), HR = 1.20, 95% CI: 1.17-1.38). The 4q32.3 locus was not associated with ovarian cancer risk in the general population or BRCA2 carriers, suggesting a BRCA1-specific association. The 17q21.31 locus was also associated with ovarian cancer risk in 8,211 BRCA2 carriers (P = 2×10(-4)). These loci may lead to an improved understanding of the etiology of breast and ovarian tumors in BRCA1 carriers. Based on the joint distribution of the known BRCA1 breast cancer risk-modifying loci, we estimated that the breast cancer lifetime risks for the 5% of BRCA1 carriers at lowest risk are 28%-50% compared to 81%-100% for the 5% at highest risk. Similarly, based on the known ovarian cancer risk-modifying loci, the 5% of BRCA1 carriers at lowest risk have an estimated lifetime risk of developing ovarian cancer of 28% or lower, whereas the 5% at highest risk will have a risk of 63% or higher. Such differences in risk may have important implications for risk prediction and clinical management for BRCA1 carriers.
Collapse
Affiliation(s)
- Fergus J. Couch
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Xianshu Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Lesley McGuffog
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Andrew Lee
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Curtis Olswold
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karoline B. Kuchenbaecker
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Penny Soucy
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire de Québec and Laval University, Québec City, Canada
| | - Zachary Fredericksen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Daniel Barrowdale
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Mia M. Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, United States of America
| | - Ed Dicks
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Matthew Kosel
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sue Healey
- Genetics Department, Queensland Institute of Medical Research, Brisbane, Australia
| | - Olga M. Sinilnikova
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Hospices Civils de Lyon–Centre Léon Bérard, Lyon, France
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Adam Lee
- Department of Molecular Pharmacology and Experimental Therapeutics (MPET), Mayo Clinic, Rochester, Minnesota, United States of America
| | - François Bacot
- Centre d'Innovation Génome Québec et Université McGill, Montreal, Canada
| | - Daniel Vincent
- Centre d'Innovation Génome Québec et Université McGill, Montreal, Canada
| | | | - Susan Peock
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Dominique Stoppa-Lyonnet
- Institut Curie, Department of Tumour Biology, Paris, France
- Institut Curie, INSERM U830, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - kConFab Investigators
- Kathleen Cuningham Consortium for Research into Familial Breast Cancer–Peter MacCallum Cancer Center, Melbourne, Australia
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, Italy
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Rita Katharina Schmutzler
- Centre of Familial Breast and Ovarian Cancer, Department of Gynaecology and Obstetrics and Centre for Integrated Oncology (CIO), Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Cologne, Germany
| | - SWE-BRCA
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Susan M. Domchek
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marion Piedmonte
- Gynecologic Oncology Group Statistical and Data Center, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Christian F. Singer
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Thomas V. O. Hansen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Csilla I. Szabo
- Center for Translational Cancer Research, Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Ignacio Blanco
- Genetic Counseling Unit, Hereditary Cancer Program, IDIBELL–Catalan Institute of Oncology, Barcelona, Spain
| | - Mark H. Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, United States of America
| | - Beth Y. Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Judy Garber
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Catherine M. Phelan
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Jeffrey N. Weitzel
- Clinical Cancer Genetics (for the City of Hope Clinical Cancer Genetics Community Research Network), City of Hope, Duarte, California, United States of America
| | - Marco Montagna
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV–IRCCS, Padua, Italy
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Irene L. Andrulis
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, IRRP, National Centre for Scientific Research Demokritos, Aghia Paraskevi Attikis, Athens, Greece
| | - David E. Goldgar
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Trinidad Caldes
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, Madrid, Spain
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Ana Osorio
- Human Genetics Group, Spanish National Cancer Centre (CNIO), and Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, New York, United States of America
| | - Mary B. Daly
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | | | - Ute Hamann
- Molecular Genetics of Breast Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Susan J. Ramus
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, California, United States of America
| | - Amanda Ewart Toland
- Divison of Human Cancer Genetics, Departments of Internal Medicine and Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Maria A. Caligo
- Section of Genetic Oncology, Department of Laboratory Medicine, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Olufunmilayo I. Olopade
- Center for Clinical Cancer Genetics and Global Health, University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Nadine Tung
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Kathleen Claes
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Mary S. Beattie
- Departments of Medicine, Epidemiology, and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Parkville, Australia
| | | | - Marc Tischkowitz
- Program in Cancer Genetics, Departments of Human Genetics and Oncology, McGill University, Montreal, Quebec, Canada
| | - Ramunas Janavicius
- Vilnius University Hospital Santariskiu Clinics, Hematology, Oncology and Transfusion Medicine Center, Department of Molecular and Regenerative Medicine, Vilnius, Lithuania
| | - Esther M. John
- Department of Epidemiology, Cancer Prevention Institute of California, Fremont, Califoria, United States of America
| | - Ava Kwong
- The Hong Kong Hereditary Breast Cancer Family Registry, Cancer Genetics Center, Hong Kong Sanatorium and Hospital, Hong Kong, China
| | - Orland Diez
- Oncogenetics Laboratory, University Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Judith Balmaña
- Department of Medical Oncology, University Hospital, Vall d'Hebron, Barcelona, Spain
| | - Rosa B. Barkardottir
- Department of Pathology, Landspitali University Hospital and BMC, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Banu K. Arun
- Department of Breast Medical Oncology and Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gad Rennert
- Clalit National Israeli Cancer Control Center and Department of Community Medicine and Epidemiology, Carmel Medical Center and B. Rappaport Faculty of Medicine, Haifa, Israel
| | - Soo-Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre and University Malaya Cancer Research Institute, University of Malaya, Kuala Lumpur, Malaysia
| | - Patricia A. Ganz
- UCLA Schools of Medicine and Public Health, Division of Cancer Prevention and Control Research, Jonsson Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Ian Campbell
- VBCRC Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Carolien H. M. van Deurzen
- Department of Pathology, Family Cancer Clinic, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Caroline Seynaeve
- Department of Medical Oncology, Family Cancer Clinic, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Encarna B. Gómez Garcia
- Department of Clinical Genetics and GROW, School for Oncology and Developmental Biology, MUMC, Maastricht, The Netherlands
| | - Flora E. van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Johannes J. P. Gille
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Marinus J. Blok
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marjolijn J. L. Ligtenberg
- Department of Human Genetics and Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Matti A. Rookus
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter Devilee
- Department of Human Genetics and Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Senno Verhoef
- Family Cancer Clinic, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Theo A. M. van Os
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Juul T. Wijnen
- Department of Human Genetics and Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - HEBON
- The Hereditary Breast and Ovarian Cancer Research Group Netherlands, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - EMBRACE
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Steve Ellis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Elena Fineberg
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Radka Platte
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - D. Gareth Evans
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Louise Izatt
- Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Rosalind A. Eeles
- Oncogenetics Team, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Julian Adlard
- Yorkshire Regional Genetics Service, Leeds, United Kingdom
| | - Diana M. Eccles
- University of Southampton Faculty of Medicine, Southampton University Hospitals NHS Trust, Southampton, United Kingdom
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, United Kingdom
| | - Carole Brewer
- Department of Clinical Genetics, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Fiona Douglas
- Institute of Genetic Medicine, Centre for Life, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Shirley Hodgson
- Department of Clinical Genetics, St George's University of London, London, United Kingdom
| | - Patrick J. Morrison
- Northern Ireland Regional Genetics Centre, Belfast Health and Social Care Trust, and Department of Medical Genetics, Queens University Belfast, Belfast, United Kingdom
| | - Lucy E. Side
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Trust and Institute for Womens Health, University College London, London, United Kingdom
| | - Alan Donaldson
- Clinical Genetics Department, St Michael's Hospital, Bristol, United Kingdom
| | - Catherine Houghton
- Cheshire and Merseyside Clinical Genetics Service, Liverpool Women's NHS Foundation Trust, Liverpool, United Kingdom
| | - Mark T. Rogers
- All Wales Medical Genetics Services, University Hospital of Wales, Cardiff, United Kingdom
| | - Huw Dorkins
- North West Thames Regional Genetics Service, Kennedy-Galton Centre, Harrow, United Kingdom
| | - Jacqueline Eason
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Helen Gregory
- North of Scotland Regional Genetics Service, NHS Grampian and University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Emma McCann
- All Wales Medical Genetics Services, Glan Clwyd Hospital, Rhyl, United Kingdom
| | - Alex Murray
- All Wales Medical Genetics Services, Singleton Hospital, Swansea, United Kingdom
| | - Alain Calender
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Hospices Civils de Lyon–Centre Léon Bérard, Lyon, France
| | | | | | | | - Catherine Nogues
- Oncogénétique Clinique, Hôpital René Huguenin/Institut Curie, Saint-Cloud, France
| | - Christine Lasset
- Unité de Prévention et d'Epidémiologie Génétique, Centre Léon Bérard, Lyon, France
- Université Lyon 1, CNRS UMR5558, Lyon, France
| | - Claude Houdayer
- Institut Curie, Department of Tumour Biology, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Dominique Leroux
- Department of Genetics, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
- Institut Albert Bonniot, Université de Grenoble, Grenoble, France
| | - Etienne Rouleau
- Laboratoire d'Oncogénétique, Hôpital René Huguenin, Institut Curie, Saint-Cloud, France
| | - Fabienne Prieur
- Service de Génétique Clinique Chromosomique et Moléculaire, Centre Hospitalier Universitaire de St Etienne, St Etienne, France
| | - Francesca Damiola
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Hagay Sobol
- Département Oncologie Génétique, Prévention et Dépistage, INSERM CIC-P9502, Institut Paoli-Calmettes/Université d'Aix-Marseille II, Marseille, France
| | - Isabelle Coupier
- Unité d'Oncogénétique, CHU Arnaud de Villeneuve, Montpellier, France
- Unité d'Oncogénétique, CRLCC Val d'Aurelle, Montpellier, France
| | - Laurence Venat-Bouvet
- Department of Medical Oncology, Centre Hospitalier Universitaire Dupuytren, Limoges, France
| | | | | | - Mélanie Léoné
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Hospices Civils de Lyon–Centre Léon Bérard, Lyon, France
| | - Pascal Pujol
- Unité d'Oncogénétique, CHU Arnaud de Villeneuve, Montpellier, France
- INSERM 896, CRCM Val d'Aurelle, Montpellier, France
| | - Sylvie Mazoyer
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Yves-Jean Bignon
- Département d'Oncogénétique, Centre Jean Perrin, Université de Clermont-Ferrand, Clermont-Ferrand, France
| | - GEMO Study Collaborators
- National Cancer Genetics Network, UNICANCER Genetic Group, Centre de Recherche en Cancérologie de Lyon and Institut Curie Paris, Paris, France
| | | | - Jacek Gronwald
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Durda
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Jaworska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Tomasz Huzarski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Amanda B. Spurdle
- Genetics Department, Queensland Institute of Medical Research, Brisbane, Australia
| | - Alessandra Viel
- Division of Experimental Oncology 1, Centro di Riferimento Oncologico, IRCCS, Aviano, Italy
| | - Bernard Peissel
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, Istituto Europeo di Oncologia, Milan, Italy
| | - Giulia Melloni
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, Italy
| | - Laura Ottini
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Laura Papi
- Unit of Medical Genetics, Department of Clinical Physiopathology, University of Florence, Firenze, Italy
| | - Liliana Varesco
- Unit of Hereditary Cancer, Department of Epidemiology, Prevention and Special Functions, IRCCS AOU San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | - Paolo Peterlongo
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, Italy
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Sara Volorio
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare and Cogentech Cancer Genetic Test Laboratory, Milan, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumori (INT), Milan, Italy
| | - Valeria Pensotti
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare and Cogentech Cancer Genetic Test Laboratory, Milan, Italy
| | - Norbert Arnold
- University Hospital of Schleswig-Holstein/University Kiel, Kiel, Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | | | | | - Andrea Gehrig
- Institute of Human Genetics, University of Würzburg, Wurzburg, Germany
| | - Karin Kast
- Department of Gynaecology and Obstetrics, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Kerstin Rhiem
- Centre of Familial Breast and Ovarian Cancer, Department of Gynaecology and Obstetrics and Centre for Integrated Oncology (CIO), Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Cologne, Germany
| | - Alfons Meindl
- Department of Gynaecology and Obstetrics, Division of Tumor Genetics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dieter Niederacher
- Department of Obstetrics and Gynaecology, University Medical Center Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nina Ditsch
- Department of Gynaecology and Obstetrics, University of Munich, Munich, Germany
| | - Hansjoerg Plendl
- Institute of Human Genetics, University Hospital of Schleswig-Holstein, University of Kiel, Kiel, Germany
| | | | - Stefanie Engert
- Department of Gynaecology and Obstetrics, Division of Tumor Genetics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian Sutter
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | | | - Barbara Wappenschmidt
- Centre of Familial Breast and Ovarian Cancer, Department of Gynaecology and Obstetrics and Centre for Integrated Oncology (CIO), Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, Cologne, Germany
| | | | - Brita Arver
- Department of Oncology and Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Stenmark-Askmalm
- Division of Clinical Genetics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Niklas Loman
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Katherine L. Nathanson
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Timothy R. Rebbeck
- Abramson Cancer Center and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stephanie V. Blank
- NYU Women's Cancer Program, New York University School of Medicine, New York, New York, United States of America
| | - David E. Cohn
- Ohio State University, Columbus Cancer Council, Columbus, Ohio, United States of America
| | - Gustavo C. Rodriguez
- Division of Gynecologic Oncology, North Shore University Health System, University of Chicago, Evanston, Illinois, United States of America
| | - Laurie Small
- Maine Medical Center, Maine Women's Surgery and Cancer Centre, Scarborough, Maine, United States of America
| | - Michael Friedlander
- ANZ GOTG Coordinating Centre, Australia New Zealand GOG, Camperdown, Australia
| | - Victoria L. Bae-Jump
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anneliese Fink-Retter
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Rappaport
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daphne Gschwantler-Kaulich
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Georg Pfeiler
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Muy-Kheng Tea
- Department of Obstetrics and Gynecology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Noralane M. Lindor
- Department of Health Science Research, Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | | | | | | | | | | | - Inge Sokilde Pedersen
- Section of Molecular Diagnostics, Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | | | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Joseph Vijai
- Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Kara Sarrel
- Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Mark Robson
- Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Noah Kauff
- Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Anna Marie Mulligan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine, and the Keenan Research Centre of the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Canada
| | - Gord Glendon
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Hilmi Ozcelik
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Bent Ejlertsen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Finn C. Nielsen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars Jønson
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette K. Andersen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yuan Chun Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Linda Steele
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Alex Teulé
- Genetic Counseling Unit, Hereditary Cancer Program, IDIBELL–Catalan Institute of Oncology, Barcelona, Spain
| | - Conxi Lazaro
- Molecular Diagnostic Unit, Hereditary Cancer Program, IDIBELL–Catalan Institute of Oncology, Barcelona, Spain
| | - Joan Brunet
- Genetic Counseling Unit, Hereditary Cancer Program, IDIBGI–Catalan Institute of Oncology, Girona, Spain
| | - Miquel Angel Pujana
- Translational Research Laboratory, Breast Cancer and Systems Biology Unit, IDIBELL–Catalan Institute of Oncology, Barcelona, Spain
| | - Phuong L. Mai
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jennifer T. Loud
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, United States of America
| | - Christine Walsh
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sandra Orsulic
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Steven A. Narod
- Women's College Research Institute, University of Toronto, Toronto, Canada
| | - Josef Herzog
- Clinical Cancer Genetics (for the City of Hope Clinical Cancer Genetics Community Research Network), City of Hope, Duarte, California, United States of America
| | - Sharon R. Sand
- Clinical Cancer Genetics (for the City of Hope Clinical Cancer Genetics Community Research Network), City of Hope, Duarte, California, United States of America
| | - Silvia Tognazzo
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV–IRCCS, Padua, Italy
| | - Simona Agata
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV–IRCCS, Padua, Italy
| | - Tibor Vaszko
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Joellen Weaver
- Biosample Repository, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Alexandra V. Stavropoulou
- Molecular Diagnostics Laboratory, IRRP, National Centre for Scientific Research Demokritos, Aghia Paraskevi Attikis, Athens, Greece
| | - Saundra S. Buys
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Atocha Romero
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, Madrid, Spain
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, Madrid, Spain
| | - Kristiina Aittomäki
- Department of Clinical Genetics, Helsinki University Central Hospital, Helsinki, Finland
| | - Taru A. Muranen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Mercedes Duran
- Institute of Biology and Molecular Genetics, Universidad de Valladolid (IBGM–UVA), Valladolid, Spain
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, Columbia University, New York, New York, United States of America
| | - Adriana Lasa
- Genetics Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Alexander Miron
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - BCFR
- Breast Cancer Family Registry, Cancer Prevention Institute of California, Fremont, California, United States of America
| | - Javier Benitez
- Human Genetics Group and Genotyping Unit, Spanish National Cancer Centre (CNIO), and Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Leigha Senter
- Divison of Human Genetics, Department of Internal Medicine, The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Dezheng Huo
- Center for Clinical Cancer Genetics and Global Health, University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Salina B. Chan
- Cancer Risk Program, Helen Diller Family Cancer Center, University of California San Francisco, San Francisco, California, United States of America
| | | | - Jocelyne Chiquette
- Unité de Recherche en Santé des Populations, Centre des Maladies du Sein Deschênes-Fabia, Centre de Recherche FRSQ du Centre Hospitalier Affilié Universitaire de Québec, Québec, Canada
| | | | - Tara M. Friebel
- University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Bjarni A. Agnarsson
- Landspitali University Hospital and University of Iceland School of Medicine, Reykjavik, Iceland
| | - Karen H. Lu
- Department of Breast Medical Oncology and Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Flavio Lejbkowicz
- Clalit National Israeli Cancer Control Center and Department of Community Medicine and Epidemiology, Carmel Medical Center, Haifa, Israel
| | - Paul A. James
- Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alison M. Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Tessier
- Centre d'Innovation Génome Québec et Université McGill, Montreal, Canada
| | - Julie Cunningham
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Susan L. Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Steven Hart
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kristen Stevens
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jacques Simard
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire de Québec and Laval University, Québec City, Canada
| | - Tomi Pastinen
- Department of Human Genetics, McGill University and Génome Québec Innovation Centre, McGill University, Montréal, Canada
| | - Vernon S. Pankratz
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kenneth Offit
- Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
374
|
Raine EVA, Dodd AW, Reynard LN, Loughlin J. Allelic expression analysis of the osteoarthritis susceptibility gene COL11A1 in human joint tissues. BMC Musculoskelet Disord 2013; 14:85. [PMID: 23497244 PMCID: PMC3599795 DOI: 10.1186/1471-2474-14-85] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 02/26/2013] [Indexed: 12/27/2022] Open
Abstract
Background The single nucleotide polymorphism (SNP) rs2615977 is associated with osteoarthritis (OA) and is located in intron 31 of COL11A1, a strong candidate gene for this degenerative musculoskeletal disease. Furthermore, the common non-synonymous COL11A1 SNP rs1676486 is associated with another degenerative musculoskeletal disease, lumbar disc herniation (LDH). rs1676486 is a C-T transition mediating its affect on LDH susceptibility by modulating COL11A1 expression. The risk T-allele of rs1676486 leads to reduced expression of the COL11A1 transcript, a phenomenon known as allelic expression imbalance (AEI). We were keen therefore to assess whether the effect that rs1676486 has on COL11A1 expression in LDH is also observed in OA and whether the rs2615977 association to OA also marked AEI. Methods Using RNA from OA cartilage, we assessed whether either SNP correlated with COL11A1 AEI by 1) measuring COL11A1 expression and stratifying the data by genotype at each SNP; and 2) quantifying the mRNA transcribed from each allele of the two SNPs. We also assessed whether rs1676486 was associated with OA susceptibility using a case–control cohort of over 18,000 individuals. Results We observed significant AEI at rs1676486 (p < 0.0001) with the T-allele correlating with reduced COL11A1 expression. This corresponded with observations in LDH but the SNP was not associated with OA. We did not observe AEI at rs2615977. Conclusions COL11A1 is subject to AEI in OA cartilage. AEI at rs1676486 is a risk factor for LDH, but not for OA. These two diseases therefore share a common functional phenotype, namely AEI of COL11A1, but this appears to be a disease risk only in LDH. Other functional effects on COL11A1 presumably account for the OA susceptibility that maps to this gene.
Collapse
Affiliation(s)
- Emma V A Raine
- Newcastle University, Musculoskeletal Research Group, Institute of Cellular Medicine, 4th Floor Catherine Cookson Building, The Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | | | | | | |
Collapse
|
375
|
Zhao SX, Liu W, Zhan M, Song ZY, Yang SY, Xue LQ, Pan CM, Gu ZH, Liu BL, Wang HN, Liang L, Liang J, Zhang XM, Yuan GY, Li CG, Chen MD, Chen JL, Gao GQ, Song HD, The China Consortium for the Genetics of Autoimmune Thyroid Disease. A refined study of FCRL genes from a genome-wide association study for Graves' disease. PLoS One 2013; 8:e57758. [PMID: 23505439 PMCID: PMC3591391 DOI: 10.1371/journal.pone.0057758] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 01/24/2013] [Indexed: 02/05/2023] Open
Abstract
To pinpoint the exact location of the etiological variant/s present at 1q21.1 harboring FCRL1-5 and CD5L genes, we carried out a refined association study in the entire FCRL region in 1,536 patients with Graves' disease (GD) and 1,516 sex-matched controls by imputation analysis, logistic regression, and cis-eQTL analysis. Among 516 SNPs with P<0.05 in the initial GWAS scan, the strongest signals associated with GD and correlated to FCRL3 expression were located at a cluster of SNPs including rs7528684 and rs3761959. And the allele-specific effects for rs3761959 and rs7528684 on FCRL3 expression level revealed that the risk alleles A of rs3761959 and C of rs7528684 were correlated with the elevated expression level of FCRL3 whether in PBMCs or its subsets, especially in CD19(+) B cells and CD8(+) T subsets. Next, the combined analysis with 5,300 GD cases and 4,916 control individuals confirmed FCRL3 was a susceptibility gene of GD in Chinese Han populations, and rs3761959 and rs7528684 met the genome-wide association significance level (P(combined) = 2.27×10(-12) and 7.11×10(-13), respectively). Moreover, the haplotypes with the risk allele A of rs3761959 and risk allele C of rs7528684 were associated with GD risk. Finally, our epigenetic analysis suggested the disease-associated C allele of rs7528684 increased affinity for NF-KB transcription factor. Above data indicated that FCRL3 gene and its proxy SNP rs7528684 may be involved in the pathogenesis of GD by excessive inhibiting B cell receptor signaling and the impairment of suppressing function of Tregs.
Collapse
Affiliation(s)
- Shuang-Xia Zhao
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Institute of Endocrinology and Metabolism, Ruijin Hospital Affiliated to SJTU School of Medicine, Shanghai, China
| | - Wei Liu
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Ming Zhan
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Zhi-Yi Song
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Shao-Ying Yang
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Li-Qiong Xue
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Chun-Ming Pan
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Zhao-Hui Gu
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
- Shanghai Center for Systems Biomedicine, SJTU, Shanghai, China
| | - Bing-Li Liu
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Hai-Ning Wang
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
| | - Liming Liang
- Department of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou Affiliated to Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Xiao-Mei Zhang
- Department of Endocrinology, The First Hospital Affiliated to Bengbu Medical College, Bengbu, Anhui Province, China
| | - Guo-Yue Yuan
- Department of Endocrinology, The Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chang-Gui Li
- Department of Endocrinology, Gout Laboratory, Medical School Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Ming-Dao Chen
- Department of Endocrinology, Shanghai Institute of Endocrinology and Metabolism, Ruijin Hospital Affiliated to SJTU School of Medicine, Shanghai, China
| | - Jia-Lun Chen
- Department of Endocrinology, Shanghai Institute of Endocrinology and Metabolism, Ruijin Hospital Affiliated to SJTU School of Medicine, Shanghai, China
| | - Guan-Qi Gao
- Department of Endocrinology, Linyi People’s Hospital, Linyi, Shandong Province, China
| | - Huai-Dong Song
- State Key Laboratory of Medical Genomics, Molecular Medicine Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University (SJTU) School of Medicine, Shanghai, China
- Department of Endocrinology, Shanghai Institute of Endocrinology and Metabolism, Ruijin Hospital Affiliated to SJTU School of Medicine, Shanghai, China
| | | |
Collapse
|
376
|
Satoh JI, Tabunoki H. Molecular network of chromatin immunoprecipitation followed by deep sequencing-based vitamin D receptor target genes. Mult Scler 2013; 19:1035-45. [PMID: 23401126 DOI: 10.1177/1352458512471873] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Vitamin D is a liposoluble vitamin essential for calcium metabolism. The ligand-bound vitamin D receptor (VDR), heterodimerized with retinoid X receptor, interacts with vitamin D response elements (VDREs) to regulate gene expression. Vitamin D deficiency due to insufficient sunlight exposure confers an increased risk for multiple sclerosis (MS). OBJECTIVE To study a protective role of vitamin D in multiple sclerosis (MS), it is important to characterize the global molecular network of VDR target genes (VDRTGs) in immune cells. METHODS We identified genome-wide VDRTGs collectively from two distinct chromatin immunoprecipitation followed by deep sequencing (ChIP-Seq) datasets of VDR-binding sites derived from calcitriol-treated human cells of B cell and monocyte origins. We mapped short reads of next generation sequencing (NGS) data on hg19 with Bowtie, detected the peaks with Model-based Analysis of ChIP-Seq (MACS), and identified genomic locations by GenomeJack, a novel genome viewer for NGS platforms. RESULTS We found 2997 stringent peaks distributed on protein-coding genes, chiefly located in the promoter and the intron on VDRE DR3 sequences. However, the corresponding transcriptome data verified calcitriol-induced upregulation of only a small set of VDRTGs. The molecular network of 1541 calcitriol-responsive VDRTGs showed a significant relationship with leukocyte transendothelial migration, Fcγ receptor-mediated phagocytosis, and transcriptional regulation by VDR, suggesting a pivotal role of genome-wide VDRTGs in immune regulation. CONCLUSION These results suggest the working hypothesis that persistent deficiency of vitamin D might perturb the complex network of VDRTGs in immune cells, being responsible for induction of an autoimmune response causative for MS.
Collapse
Affiliation(s)
- Jun-ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan.
| | | |
Collapse
|
377
|
Abstract
Our understanding of immunity has historically been informed by studying heritable mutations in both the adaptive and innate immune responses, including primary immunodeficiency and autoimmune diseases. Recent advances achieved through the application of genomic and epigenomic approaches are reshaping the study of immune dysfunction and opening up new avenues for therapeutic interventions. Moreover, applying genomic techniques to resolve functionally important genetic variation between individuals is providing new insights into immune function in health. This review describes progress in the study of rare variants and primary immunodeficiency diseases arising from whole-exome sequencing (WES), and discusses the application, success, and challenges of applying genome-wide association studies (GWAS) to disorders of immune function and how they may inform more rational use of therapeutics. In addition, the application of expression quantitative-trait mapping to immune phenotypes, progress in understanding MHC disease associations, and insights into epigenetic mechanisms at the interface of immunity and the environment are reviewed.
Collapse
Affiliation(s)
- Julian C Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| |
Collapse
|
378
|
Identification of single nucleotide polymorphisms regulating peripheral blood mRNA expression with genome-wide significance: an eQTL study in the Japanese population. PLoS One 2013; 8:e54967. [PMID: 23359819 PMCID: PMC3554677 DOI: 10.1371/journal.pone.0054967] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/18/2012] [Indexed: 12/02/2022] Open
Abstract
Several recent studies have reported that expression quantitative trait loci (eQTLs) may affect gene expression in a cell-dependent manner. In the current study, a genome-wide eQTL analysis was performed in whole blood samples collected from 76 Japanese subjects. RNA microarray analysis was performed for 3 independent sample groups that were genotyped in a genome-wide scan. The correlations between the genotypes of 534,404 autosomal single nucleotide polymorphisms (SNPs) and the expression levels of 30,465 probes were examined for each sample group. The SNP-probe pairs with combined correlation coefficients of all 3 sample groups corresponding to P<3.1×10−12 (i.e., Bonferroni-corrected P<0.05) were considered significant. SNP-probe pairs with a high likelihood of cross-hybridization and SNP-in-probe effects were excluded to avoid false positive results. We identified 102 cis-acting and 5 trans-acting eQTL regions. The cis-eQTL regions were widely distributed both upstream and downstream of the gene, as well as within the gene. The eQTL SNPs identified were examined for their influence on the expression levels in lymphoblastoid cell lines by using a public database. The results showed that genetic variants affecting expression levels in whole blood may have different effects on gene expression in lymphoblastoid cell lines. Further studies are required to clarify how SNPs function in affecting the expression levels in whole blood as well as in other tissues.
Collapse
|
379
|
|
380
|
Guo Y, Lanktree MB, Taylor KC, Hakonarson H, Lange LA, Keating BJ, The IBC 50K SNP array BMI Consortium. Gene-centric meta-analyses of 108 912 individuals confirm known body mass index loci and reveal three novel signals. Hum Mol Genet 2013; 22:184-201. [PMID: 23001569 PMCID: PMC3522401 DOI: 10.1093/hmg/dds396] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 08/04/2012] [Accepted: 09/06/2012] [Indexed: 12/18/2022] Open
Abstract
Recent genetic association studies have made progress in uncovering components of the genetic architecture of the body mass index (BMI). We used the ITMAT-Broad-Candidate Gene Association Resource (CARe) (IBC) array comprising up to 49 320 single nucleotide polymorphisms (SNPs) across ~2100 metabolic and cardiovascular-related loci to genotype up to 108 912 individuals of European ancestry (EA), African-Americans, Hispanics and East Asians, from 46 studies, to provide additional insight into SNPs underpinning BMI. We used a five-phase study design: Phase I focused on meta-analysis of EA studies providing individual level genotype data; Phase II performed a replication of cohorts providing summary level EA data; Phase III meta-analyzed results from the first two phases; associated SNPs from Phase III were used for replication in Phase IV; finally in Phase V, a multi-ethnic meta-analysis of all samples from four ethnicities was performed. At an array-wide significance (P < 2.40E-06), we identify novel BMI associations in loci translocase of outer mitochondrial membrane 40 homolog (yeast) - apolipoprotein E - apolipoprotein C-I (TOMM40-APOE-APOC1) (rs2075650, P = 2.95E-10), sterol regulatory element binding transcription factor 2 (SREBF2, rs5996074, P = 9.43E-07) and neurotrophic tyrosine kinase, receptor, type 2 [NTRK2, a brain-derived neurotrophic factor (BDNF) receptor gene, rs1211166, P = 1.04E-06] in the Phase IV meta-analysis. Of 10 loci with previous evidence for BMI association represented on the IBC array, eight were replicated, with the remaining two showing nominal significance. Conditional analyses revealed two independent BMI-associated signals in BDNF and melanocortin 4 receptor (MC4R) regions. Of the 11 array-wide significant SNPs, three are associated with gene expression levels in both primary B-cells and monocytes; with rs4788099 in SH2B adaptor protein 1 (SH2B1) notably being associated with the expression of multiple genes in cis. These multi-ethnic meta-analyses expand our knowledge of BMI genetics.
Collapse
Affiliation(s)
- Yiran Guo
- Center for Applied Genomics, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Suite 1014H, Philadelphia 19104, PA, USA
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Matthew B. Lanktree
- Department of Medicine and
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Kira C. Taylor
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY 40292, USA and
- Epidemiology and
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Suite 1014H, Philadelphia 19104, PA, USA
| | - Leslie A. Lange
- Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brendan J. Keating
- Center for Applied Genomics, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Suite 1014H, Philadelphia 19104, PA, USA
| | | |
Collapse
|
381
|
Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat Genet 2012; 45:124-30. [PMID: 23263488 DOI: 10.1038/ng.2504] [Citation(s) in RCA: 447] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 11/28/2012] [Indexed: 02/07/2023]
Abstract
If trait-associated variants alter regulatory regions, then they should fall within chromatin marks in relevant cell types. However, it is unclear which of the many marks are most useful in defining cell types associated with disease and fine mapping variants. We hypothesized that informative marks are phenotypically cell type specific; that is, SNPs associated with the same trait likely overlap marks in the same cell type. We examined 15 chromatin marks and found that those highlighting active gene regulation were phenotypically cell type specific. Trimethylation of histone H3 at lysine 4 (H3K4me3) was the most phenotypically cell type specific (P < 1 × 10(-6)), driven by colocalization of variants and marks rather than gene proximity (P < 0.001). H3K4me3 peaks overlapped with 37 SNPs for plasma low-density lipoprotein concentration in the liver (P < 7 × 10(-5)), 31 SNPs for rheumatoid arthritis within CD4(+) regulatory T cells (P = 1 × 10(-4)), 67 SNPs for type 2 diabetes in pancreatic islet cells (P = 0.003) and the liver (P = 0.003), and 14 SNPs for neuropsychiatric disease in neuronal tissues (P = 0.007). We show how cell type-specific H3K4me3 peaks can inform the fine mapping of associated SNPs to identify causal variation.
Collapse
|
382
|
Tsuchiya N. Genetics of ANCA-associated vasculitis in Japan: a role for HLA-DRB1*09:01 haplotype. Clin Exp Nephrol 2012. [PMID: 23180035 DOI: 10.1007/s10157-012-0691-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The epidemiology of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is considerably different between European and Asian populations. Whereas granulomatosis with polyangiitis is the most common form of AAV in northern European populations, microscopic polyangiitis (MPA) accounts for the majority of AAV in Japan. This difference may at least in part derive from the difference in genetic background. In this review, I focus on our observation on HLA, an obvious candidate gene for immune disorders, and discuss its potential implication. In Japanese AAV, significant association was detected with HLA-DRB1*09:01, the carrier frequency of which was increased in MPA [P=0.0087, odds ratio (OR) 1.90, 95% confidence interval (CI) 1.17-3.08] and in myeloperoxidase (MPO)-ANCA-positive AAV (P=0.0016, OR 2.05, 95% CI 1.31-3.23) when compared with healthy Japanese controls. HLA-DRB1*09:01 is one of the most common HLA-DRB1 alleles in Asians but is rare in Caucasian populations. Interestingly, HLA-DRB1*09:01 has been shown to be associated with multiple autoimmune diseases, including type 1 diabetes, rheumatoid arthritis, and systemic lupus erythematosus, suggesting that either HLA-DRB1*09:01 itself or other genes in tight linkage disequilibrium may play a role in a molecular pathway shared by various autoimmune diseases in Japanese and possibly in other Asian populations.
Collapse
Affiliation(s)
- Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
383
|
de Bakker PIW, Raychaudhuri S. Interrogating the major histocompatibility complex with high-throughput genomics. Hum Mol Genet 2012; 21:R29-36. [PMID: 22976473 PMCID: PMC3459647 DOI: 10.1093/hmg/dds384] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 09/06/2012] [Indexed: 12/11/2022] Open
Abstract
The major histocompatibility complex (MHC) region on the short arm of chromosome 6 harbors the largest number of replicated associations across the human genome for a wide range of diseases, but the functional basis for these associations is still poorly understood. One fundamental challenge in fine-mapping associations to functional alleles is the enormous sequence diversity and broad linkage disequilibrium of the MHC, both of which hamper the cost-effective interrogation in large patient samples and the identification of causal variants. In this review, we argue that there is now a valuable opportunity to leverage existing genome-wide association study (GWAS) datasets for in-depth investigation to identify independent effects in the MHC. Application of imputation to GWAS data facilitates comprehensive interrogation of the classical human leukocyte antigen (HLA) loci. These datasets are, in many cases, sufficiently large to give investigators the ability to disentangle effects at different loci. We also explain how querying variation at individual amino acid positions for association can be powerful and expand traditional analyses that focus only on the classical HLA types.
Collapse
Affiliation(s)
- Paul I W de Bakker
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
384
|
Stranger BE, De Jager PL. Coordinating GWAS results with gene expression in a systems immunologic paradigm in autoimmunity. Curr Opin Immunol 2012; 24:544-51. [PMID: 23040211 DOI: 10.1016/j.coi.2012.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 08/31/2012] [Accepted: 09/05/2012] [Indexed: 11/28/2022]
Abstract
There has been considerable progress in our understanding of the genetic architecture of susceptibility to inflammatory diseases in recent years: several hundred susceptibility loci have been discovered in genome-wide association studies (GWAS) of human populations. This success has created an important challenge in identifying the functional consequences of these risk-associated variants and in elucidating how the repercussions of individual susceptibility loci integrate to yield dysregulation of immune pathways and, ultimately, syndromic clinical phenotypes. The integration of GWAS association signals with high-resolution transcriptome and other genomic data that capture the dynamics of cellular state and function in the context of individual's collection of susceptibility alleles has proven to be a successful avenue of investigation. The rapid pace of methodological development in this area has been coupled with an accumulation of experimental data that makes the elucidation of complex biological networks underlying susceptibility to these common inflammatory diseases a reasonable goal in the near future.
Collapse
Affiliation(s)
- Barbara E Stranger
- Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
385
|
|
386
|
Abstract
Type 1 diabetes (T1D) is a multi-factorial, organ-specific autoimmune disease in genetically susceptible individuals, which is characterized by a selective and progressive loss of insulin-producing β-cells. Cells mediating innate as well as adaptive immunity infiltrate pancreatic islets, thereby generating an aberrant inflammatory process called insulitis that can be mirrored by a pathologic autoantibody production and autoreactive T-cells. In tight cooperation with infiltrating innate immune cells, which secrete high levels of pro-inflammatory cytokines like IL-1β, TNFα, and INFγ effector T-cells trigger the fatal destruction process of β-cells. There is ongoing discussion on the contribution of inflammation in T1D pathogenesis, ranging from a bystander reaction of autoimmunity to a dysregulation of immune responses that initiate inflammatory processes and thereby actively promoting β-cell death. Here, we review recent advances in anti-inflammatory interventions in T1D animal models and preclinical studies and discuss their mode of action as well as their capacity to interfere with T1D development.
Collapse
Affiliation(s)
- Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert Einstein Allee 11, 89081, Ulm, Germany.
| | | | | |
Collapse
|
387
|
Lu Y, Vaarhorst A, Merry AHH, Dollé MET, Hovenier R, Imholz S, Schouten LJ, Heijmans BT, Müller M, Slagboom PE, van den Brandt PA, Gorgels APM, Boer JMA, Feskens EJM. Markers of endogenous desaturase activity and risk of coronary heart disease in the CAREMA cohort study. PLoS One 2012; 7:e41681. [PMID: 22911844 PMCID: PMC3402436 DOI: 10.1371/journal.pone.0041681] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/24/2012] [Indexed: 11/26/2022] Open
Abstract
Background Intakes of n-3 polyunsaturated fatty acids (PUFAs), especially EPA (C20∶5n-3) and DHA (C22∶6n-3), are known to prevent fatal coronary heart disease (CHD). The effects of n-6 PUFAs including arachidonic acid (C20∶4n-6), however, remain unclear. δ-5 and δ-6 desaturases are rate-limiting enzymes for synthesizing long-chain n-3 and n-6 PUFAs. C20∶4n-6 to C20∶3n-6 and C18∶3n-6 to C18∶2n-6 ratios are markers of endogenous δ-5 and δ-6 desaturase activities, but have never been studied in relation to incident CHD. Therefore, the aim of this study was to investigate the relation between these ratios as well as genotypes of FADS1 rs174547 and CHD incidence. Methods We applied a case-cohort design within the CAREMA cohort, a large prospective study among the general Dutch population followed up for a median of 12.1 years. Fatty acid profile in plasma cholesteryl esters and FADS1 genotype at baseline were measured in a random subcohort (n = 1323) and incident CHD cases (n = 537). Main outcome measures were hazard ratios (HRs) of incident CHD adjusted for major CHD risk factors. Results The AA genotype of rs174547 was associated with increased plasma levels of C204n-6, C20∶5n-3 and C22∶6n-3 and increased δ-5 and δ-6 desaturase activities, but not with CHD risk. In multivariable adjusted models, high baseline δ-5 desaturase activity was associated with reduced CHD risk (P for trend = 0.02), especially among those carrying the high desaturase activity genotype (AA): HR (95% CI) = 0.35 (0.15–0.81) for comparing the extreme quintiles. High plasma DHA levels were also associated with reduced CHD risk. Conclusion In this prospective cohort study, we observed a reduced CHD risk with an increased C20∶4n-6 to C20∶3n-6 ratio, suggesting that δ-5 desaturase activity plays a role in CHD etiology. This should be investigated further in other independent studies.
Collapse
Affiliation(s)
- Yingchang Lu
- Division of Human Nutrition, Wageningen University and Research Center, Wageningen, The Netherlands
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- * E-mail: (YL); (EF)
| | - Anika Vaarhorst
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Audrey H. H. Merry
- Department of Epidemiology, CAPHRI School for Public Health and Primary Care, Maastricht University, Maastricht, The Netherlands
| | - Martijn E. T. Dollé
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Robert Hovenier
- Division of Human Nutrition, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Sandra Imholz
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Leo J. Schouten
- Department of Epidemiology, GROW School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Bastiaan T. Heijmans
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael Müller
- Division of Human Nutrition, Wageningen University and Research Center, Wageningen, The Netherlands
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Piet A. van den Brandt
- Department of Epidemiology, CAPHRI School for Public Health and Primary Care, Maastricht University, Maastricht, The Netherlands
- Department of Epidemiology, GROW School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Anton P. M. Gorgels
- Department of Cardiology, University Hospital Maastricht, Maastricht, The Netherlands
| | - Jolanda M. A. Boer
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Edith J. M. Feskens
- Division of Human Nutrition, Wageningen University and Research Center, Wageningen, The Netherlands
- * E-mail: (YL); (EF)
| |
Collapse
|
388
|
Abstract
A new study reports the mapping of gene expression in primary immune cell subsets, showing the presence of cell type-specific cis and trans expression quantitative trait loci (eQTLs). The identification of cell type-specific trans-regulated networks can inform functional studies of susceptibility loci identified from genome-wide association studies for human complex diseases.
Collapse
Affiliation(s)
- Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, New York, USA.
| |
Collapse
|