101
|
Fine-mapping the human leukocyte antigen locus in rheumatoid arthritis and other rheumatic diseases: identifying causal amino acid variants? Curr Opin Rheumatol 2015; 27:256-61. [PMID: 25760279 DOI: 10.1097/bor.0000000000000165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To provide an update on and the context of the recent findings obtained with novel statistical methods on the association of the human leukocyte antigen (HLA) locus with rheumatic diseases. RECENT FINDINGS Novel single nucleotide polymorphism fine-mapping data obtained for the HLA locus have indicated the strongest association with amino acid positions 11 and 13 of HLA-DRB1 molecule for several rheumatic diseases. On the basis of these data, a dominant role for position 11/13 in driving the association with these diseases is proposed and the identification of causal variants in the HLA region in relation to disease susceptibility implicated. SUMMARY The HLA class II locus is the most important risk factor for several rheumatic diseases. Recently, new statistical approaches have identified previously unrecognized amino acid positions in the HLA-DR molecule that associate with anticitrullinated protein antibody-negative and anticitrullinated protein antibody-positive rheumatoid arthritis. Likewise, similar findings have been made for other rheumatic conditions such as giant-cell arteritis and systemic lupus erythematosus. Interestingly, all these studies point toward an association with the same amino acid positions: amino acid positions 11 and 13 of the HLA-DR β chain. As both these positions influence peptide binding by HLA-DR and have been implicated in antigen presentation, the novel fine-mapping approach is proposed to map causal variants in the HLA region relevant to rheumatoid arthritis and several rheumatic diseases. If these interpretations are correct, they would direct the biological research aiming to address the explanation for the HLA-disease association. Here, we provide an overview of the recent findings and evidence from literature that, although relevant new insights have been obtained on HLA-disease associations, the interpretation of the biological role of these amino acids as causal variants explaining that such associations should be taken with caution.
Collapse
|
102
|
Orent W, Mchenry AR, Rao DA, White C, Klein HU, Bassil R, Srivastava G, Replogle JM, Raj T, Frangieh M, Cimpean M, Cuerdon N, Chibnik L, Khoury SJ, Karlson EW, Brenner MB, De Jager P, Bradshaw EM, Elyaman W. Rheumatoid arthritis-associated RBPJ polymorphism alters memory CD4+ T cells. Hum Mol Genet 2015; 25:404-17. [PMID: 26604133 DOI: 10.1093/hmg/ddv474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/15/2015] [Indexed: 02/07/2023] Open
Abstract
Notch signaling has recently emerged as an important regulator of immune responses in autoimmune diseases. The recombination signal-binding protein for immunoglobulin kappa J region (RBPJ) is a transcriptional repressor, but converts into a transcriptional activator upon activation of the canonical Notch pathway. Genome-wide association studies of rheumatoid arthritis (RA) identified a susceptibility locus, rs874040(CC), which implicated the RBPJ gene. Here, chromatin state mapping generated using the chromHMM algorithm reveals strong enhancer regions containing DNase I hypersensitive sites overlapping the rs874040 linkage disequilibrium block in human memory, but not in naïve CD4(+) T cells. The rs874040 overlapping this chromatin state was associated with increased RBPJ expression in stimulated memory CD4(+) T cells from healthy subjects homozygous for the risk allele (CC) compared with memory CD4(+) T cells bearing the protective allele (GG). Transcriptomic analysis of rs874040(CC) memory T cells showed a repression of canonical Notch target genes IL (interleukin)-9, IL-17 and interferon (IFN)γ in the basal state. Interestingly, activation of the Notch pathway using soluble Notch ligand, Jagged2-Fc, induced IL-9 and IL-17A while delta-like 4Fc, another Notch ligand, induced higher IFNγ expression in the rs874040(CC) memory CD4(+) T cells compared with their rs874040(GG) counterparts. In RA, RBPJ expression is elevated in memory T cells from RA patients compared with control subjects, and this was associated with induced inflammatory cytokines IL-9, IL-17A and IFNγ in response to Notch ligation in vitro. These findings demonstrate that the rs874040(CC) allele skews memory T cells toward a pro-inflammatory phenotype involving Notch signaling, thus increasing the susceptibility to develop RA.
Collapse
Affiliation(s)
| | | | - Deepak A Rao
- Division of Rheumatology, Immunology and Allergy and
| | - Charles White
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Hans-Ulrich Klein
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | | | - Gyan Srivastava
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Joseph M Replogle
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Towfique Raj
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | | | - Maria Cimpean
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Nicole Cuerdon
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Lori Chibnik
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | | | | | - Philip De Jager
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Elizabeth M Bradshaw
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| | - Wassim Elyaman
- Ann Romney Center for Neurologic Diseases, Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Broad Institute at Harvard University and MIT, NRB-641, 77 Avenue Louis Pasteur, Boston, MA 02115, USA and
| |
Collapse
|
103
|
Wells A, Kopp N, Xu X, O'Brien DR, Yang W, Nehorai A, Adair-Kirk TL, Kopan R, Dougherty JD. The anatomical distribution of genetic associations. Nucleic Acids Res 2015; 43:10804-20. [PMID: 26586807 PMCID: PMC4678833 DOI: 10.1093/nar/gkv1262] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023] Open
Abstract
Deeper understanding of the anatomical intermediaries for disease and other complex genetic traits is essential to understanding mechanisms and developing new interventions. Existing ontology tools provide functional, curated annotations for many genes and can be used to develop mechanistic hypotheses; yet information about the spatial expression of genes may be equally useful in interpreting results and forming novel hypotheses for a trait. Therefore, we developed an approach for statistically testing the relationship between gene expression across the body and sets of candidate genes from across the genome. We validated this tool and tested its utility on three applications. First, we show that the expression of genes in associated loci from GWA studies implicates specific tissues for 57 out of 98 traits. Second, we tested the ability of the tool to identify novel relationships between gene expression and phenotypes. Specifically, we experimentally confirmed an underappreciated prediction highlighted by our tool: that white blood cell count--a quantitative trait of the immune system--is genetically modulated by genes expressed in the skin. Finally, using gene lists derived from exome sequencing data, we show that human genes under selective constraint are disproportionately expressed in nervous system tissues.
Collapse
Affiliation(s)
- Alan Wells
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nathan Kopp
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoxiao Xu
- The Preston M. Green Department of Electrical and Systems Engineering, Washington University, St. Louis, MO 63130, USA
| | - David R O'Brien
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Arye Nehorai
- The Preston M. Green Department of Electrical and Systems Engineering, Washington University, St. Louis, MO 63130, USA
| | - Tracy L Adair-Kirk
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raphael Kopan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - J D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
104
|
Messemaker TC, Huizinga TW, Kurreeman F. Immunogenetics of rheumatoid arthritis: Understanding functional implications. J Autoimmun 2015. [DOI: 10.1016/j.jaut.2015.07.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
105
|
Zhou G, Wang Y, Fang Z, Liu R, Wang A, Zhao F, Chen L. CD40 -1C>T polymorphism and the risk of lung cancer in a Chinese population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15163-15169. [PMID: 26823861 PMCID: PMC4713647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/21/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND The co-stimulatory molecule CD40 plays an important role in anti-tumor responses by promoting cytotoxic T lymphocyte (CTL) activity and differentiation of helper T cells. Growing evidence suggests that single nucleotide polymorphisms (SNPs) in CD40 are associated with the susceptibility to cancer. This study investigated the association between the CD40 -1C/T SNP (rs1883832) and lung cancer in a Chinese population. METHODS We conducted a hospital-based case-control study including 105 lung cancer patients and 109 healthy control subjects. The -1C/T SNP in CD40 was genotyped by the polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP), and its association with lung cancer susceptibility was evaluated. RESULTS The distribution of the genotypes of CD40-1C/T was significantly different between lung cancer patients and controls. The frequency of the TT genotype (adjusted P = 0.017; overall risk [OR] = 2.94; 95% confidence interval [CI] = 1.21-7.13) and TT/CT genotype (adjusted P = 0.020; OR = 1.95; 95% CI = 1.11-3.43) were significantly higher in lung cancer patients than that in controls. When the cases were categorized by tumor histology, the TT genotype was associated with a significantly increased risk of squamous cell carcinoma (adjusted OR = 6.53; 95% CI = 1.97-21.61; P = 0.002). CONCLUSION Our findings suggest that the CD40 -1C/T SNP (rs1883832) is correlated with the susceptibility to lung cancer in Chinese, and the TT genotype may further increase the risk of lung cancer.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Immunology, Fourth Military Medical UniversityXi’an, China
| | - Ying Wang
- Department of Immunology, Fourth Military Medical UniversityXi’an, China
- Department of Stomatology, Affiliated Hospital of Academy of Military Medical SciencesBeijing, China
| | - Ziyao Fang
- Department of Immunology, Fourth Military Medical UniversityXi’an, China
| | - Rongrong Liu
- Department of Immunology, Fourth Military Medical UniversityXi’an, China
| | - Anhui Wang
- Department of Preventive Medicine and Health Statistics, Fourth Military Medical UniversityXi’an, China
| | - Feng Zhao
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical UniversityXi’an, China
| | - Lihua Chen
- Department of Immunology, Fourth Military Medical UniversityXi’an, China
| |
Collapse
|
106
|
Karaky M, Alcina A, Fedetz M, Barrionuevo C, Potenciano V, Delgado C, Izquierdo G, Matesanz F. The multiple sclerosis-associated regulatory variant rs10877013 affects expression of CYP27B1 and VDR under inflammatory or vitamin D stimuli. Mult Scler 2015; 22:999-1006. [DOI: 10.1177/1352458515610208] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/10/2015] [Indexed: 12/19/2022]
Abstract
Background: Vitamin D deficit is considered an important risk factor for many inflammatory and autoimmune diseases. Objective: To investigate the influence of the multiple sclerosis (MS)-associated regulatory variant rs10877013 on the expression of genes involved in vitamin D activation ( CYP27B1), vitamin D receptor ( VDR), and vitamin D degradation ( CYP24A1) under inflammatory environment or vitamin D. Methods: We used lipopolysaccharide and interferon-gamma (LPS+IFNγ) activated monocytes from 119 individuals and vitamin D-stimulated lymphoblastoid cell lines (LCLs, n = 109) of 1000 genomes to quantify the mRNA expression of vitamin D genes by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Results: We found that CYP27B1 mRNA expression level was associated with the rs10877013 genotypes ( p = 5.0E-6) in LPS+IFNγ treated monocytes, but not in vitamin D-stimulated LCLs. Inversely, rs10877013 genotypes were associated with VDR expression in LCLs ( p = 6.0E-4) but not in monocytes. Finally, CYP24A1 was highly induced by the active form of vitamin D and its expression correlated with the expression of VDR in LCLs but neither the MS-associated variant in the region (rs2248359) nor any other variant located in 1 Mb around CYP24A1 was associated with its expression. Conclusions: The MS-associated variant rs10877013 is a genetic determinant that affects the functioning of the vitamin D system linking environmental and genetic factors.
Collapse
Affiliation(s)
- Mohamad Karaky
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| | - Antonio Alcina
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| | - María Fedetz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| | - Cristina Barrionuevo
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| | - Victor Potenciano
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| | - Concepción Delgado
- Centro Regional de Transfusión Sanguínea Granada-Almería (CRTS), Granada, Spain
| | - Guillermo Izquierdo
- Unidad de Esclerosis Múltiple, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina “López Neyra” (IPBLN-CSIC), Granada, Spain
| |
Collapse
|
107
|
Brezar V, Tu WJ, Seddiki N. PKC-Theta in Regulatory and Effector T-cell Functions. Front Immunol 2015; 6:530. [PMID: 26528291 PMCID: PMC4602307 DOI: 10.3389/fimmu.2015.00530] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/28/2015] [Indexed: 01/20/2023] Open
Abstract
One of the major goals in immunology research is to understand the regulatory mechanisms that underpin the rapid switch on/off of robust and efficient effector (Teffs) or regulatory (Tregs) T-cell responses. Understanding the molecular mechanisms underlying the regulation of such responses is critical for the development of effective therapies. T-cell activation involves the engagement of T-cell receptor and co-stimulatory signals, but the subsequent recruitment of serine/threonine-specific protein Kinase C-theta (PKC-θ) to the immunological synapse (IS) is instrumental for the formation of signaling complexes, which ultimately lead to a transcriptional network in T cells. Recent studies demonstrated that major differences between Teffs and Tregs occurred at the IS where its formation induces altered signaling pathways in Tregs. These pathways are characterized by reduced recruitment of PKC-θ, suggesting that PKC-θ inhibits Tregs suppressive function in a negative feedback loop. As the balance of Teffs and Tregs has been shown to be central in several diseases, it was not surprising that some studies revealed that PKC-θ plays a major role in the regulation of this balance. This review will examine recent knowledge on the role of PKC-θ in T-cell transcriptional responses and how this protein can impact on the function of both Tregs and Teffs.
Collapse
Affiliation(s)
- Vedran Brezar
- INSERM U955, Équipe 16 and Faculté de Médecine, Université Paris Est , Créteil , France ; Vaccine Research Institute (VRI) , Créteil , France
| | - Wen Juan Tu
- Faculty of Education, Science, Technology and Maths, University of Canberra , Canberra, ACT , Australia
| | - Nabila Seddiki
- INSERM U955, Équipe 16 and Faculté de Médecine, Université Paris Est , Créteil , France ; Vaccine Research Institute (VRI) , Créteil , France
| |
Collapse
|
108
|
Association of CD40 polymorphisms and haplotype with risk of systemic lupus erythematosus. Rheumatol Int 2015; 36:45-52. [PMID: 26289938 DOI: 10.1007/s00296-015-3345-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/06/2015] [Indexed: 10/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease that is caused by genetic and environmental factors. Current evidence shows that the CD40-CD40L system plays a crucial role in the development, progression and outcome of SLE. CD40, which stimulates lymphocyte proliferation and differentiation, is an important immunomodulator and is expressed in the thyroid follicular cells as well as antigen-presenting cells. The aim of the present study was to investigate whether CD40 gene polymorphism confers susceptibility to SLE and its impact on CD40 expression in Chinese. We analyzed four single nucleotide polymorphisms of CD40 gene rs1883832C/T, rs13040307C/T, rs752118C/T, and rs3765459G/A in 205 patients with SLE and 220 age- and sex-matched controls, using Snapshot SNP genotyping assays and DNA sequencing method. Soluble CD40 (sCD40) levels were measured by ELISA. There were significant differences in the genotype and allele frequencies of CD40 gene rs1883832 C/T polymorphism between the group of patients with SLE and the control group (P < 0.05). sCD40 levels were increased in patients with SLE compared with controls (P < 0.01). Moreover, genotypes carrying the CD40 rs1883832 T variant allele were associated with increased CD40 levels compared with the homozygous wild-type genotype in patients with SLE. The rs1883832 C/T polymorphism of CD40 and its sCD40 levels were associated with SLE in the Chinese population. These data suggest that CD40 gene may play an essential role in the development of SLE.
Collapse
|
109
|
Zhou QM, Zheng Y, Chibnik LB, Karlson EW, Cai T. Assessing incremental value of biomarkers with multi-phase nested case-control studies. Biometrics 2015. [PMID: 26195245 DOI: 10.1111/biom.12344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Accurate risk prediction models are needed to identify different risk groups for individualized prevention and treatment strategies. In the Nurses' Health Study, to examine the effects of several biomarkers and genetic markers on the risk of rheumatoid arthritis (RA), a three-phase nested case-control (NCC) design was conducted, in which two sequential NCC subcohorts were formed with one nested within the other, and one set of new markers measured on each of the subcohorts. One objective of the study is to evaluate clinical values of novel biomarkers in improving upon existing risk models because of potential cost associated with assaying biomarkers. In this paper, we develop robust statistical procedures for constructing risk prediction models for RA and estimating the incremental value (IncV) of new markers based on three-phase NCC studies. Our method also takes into account possible time-varying effects of biomarkers in risk modeling, which allows us to more robustly assess the biomarker utility and address the question of whether a marker is better suited for short-term or long-term risk prediction. The proposed procedures are shown to perform well in finite samples via simulation studies.
Collapse
Affiliation(s)
- Qian M Zhou
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC, Canada, V5A1S6
| | - Yingye Zheng
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lori B Chibnik
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Tianxi Cai
- Department of Biostatistics, Harvard University, Boston, MA, USA
| |
Collapse
|
110
|
Pal LR, Moult J. Genetic Basis of Common Human Disease: Insight into the Role of Missense SNPs from Genome-Wide Association Studies. J Mol Biol 2015; 427:2271-89. [PMID: 25937569 PMCID: PMC4893807 DOI: 10.1016/j.jmb.2015.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/28/2015] [Accepted: 04/27/2015] [Indexed: 01/03/2023]
Abstract
Recent genome-wide association studies (GWAS) have led to the reliable identification of single nucleotide polymorphisms (SNPs) at a number of loci associated with increased risk of specific common human diseases. Each such locus implicates multiple possible candidate SNPs for involvement in disease mechanism. A variety of mechanisms may link the presence of an SNP to altered in vivo gene product function and hence contribute to disease risk. Here, we report an analysis of the role of one of these mechanisms, missense SNPs (msSNPs) in proteins in seven complex trait diseases. Linkage disequilibrium information was used to identify possible candidate msSNPs associated with increased disease risk at each of 356 loci for the seven diseases. Two computational methods were used to estimate which of these SNPs has a significant impact on in vivo protein function. 69% of the loci have at least one candidate msSNP and 33% have at least one predicted high-impact msSNP. In some cases, these SNPs are in well-established disease-related proteins, such as MST1 (macrophage stimulating 1) for Crohn's disease. In others, they are in proteins identified by GWAS as likely candidates for disease relevance, but previously without known mechanism, such as ADAMTS13 (ADAM metallopeptidase with thrombospondin type 1 motif, 13) for coronary artery disease. In still other cases, the missense SNPs are in proteins not previously suggested as disease candidates, such as TUBB1 (tubulin, beta 1, class VI) for hypertension. Together, these data support a substantial role for this class of SNPs in susceptibility to common human disease.
Collapse
Affiliation(s)
- Lipika R Pal
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| | - John Moult
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
111
|
Jiang DK, Ma XP, Yu H, Cao G, Ding DL, Chen H, Huang HX, Gao YZ, Wu XP, Long XD, Zhang H, Zhang Y, Gao Y, Chen TY, Ren WH, Zhang P, Shi Z, Jiang W, Wan B, Saiyin H, Yin J, Zhou YF, Zhai Y, Lu PX, Zhang H, Gu X, Tan A, Wang JB, Zuo XB, Sun LD, Liu JO, Yi Q, Mo Z, Zhou G, Liu Y, Sun J, Shugart YY, Zheng SL, Zhang XJ, Xu J, Yu L. Genetic variants in five novel loci including CFB and CD40 predispose to chronic hepatitis B. Hepatology 2015; 62:118-28. [PMID: 25802187 DOI: 10.1002/hep.27794] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
UNLABELLED Hepatitis B virus affects more than 2 billion people worldwide, 350 million of which have developed chronic hepatitis B (CHB). The genetic factors that confer CHB risk are still largely unknown. We sought to identify genetic variants for CHB susceptibility in the Chinese population. We undertook a genome-wide association study (GWAS) in 2,514 CHB cases and 1,130 normal controls from eastern China. We replicated 33 of the most promising signals and eight previously reported CHB risk loci through a two-stage validation totaling 6,600 CHB cases and 8,127 controls in four independent populations, of which two populations were recruited from eastern China, one from northern China and one from southern China. The joint analyses of 9,114 CHB cases and 9,257 controls revealed significant association of CHB risk with five novel loci. Four loci are located in the human leukocyte antigen (HLA) region at 6p21.3, including two nonsynonymous variants (rs12614 [R32W] in complement factor B [CFB], Pmeta =1.28 × 10(-34) ; and rs422951 [T320A] in NOTCH4, Pmeta = 5.33 × 10(-16) ); one synonymous variant (rs378352 in HLA-DOA corresponding to HLA-DOA*010101, Pmeta = 1.04 × 10(-23) ); and one noncoding variant (rs2853953 near HLA-C, Pmeta = 5.06 × 10(-20) ). Another locus is located at 20q13.1 (rs1883832 in the Kozak sequence of CD40, Pmeta = 2.95 × 10(-15) ). Additionally, we validated seven of eight previously reported CHB susceptibility loci (rs3130542 at HLA-C, rs1419881 at TCF19, rs652888 at EHMT2, rs2856718 at HLA-DQB1, rs7453920 at HLA-DQB2, rs3077 at HLA-DPA1, and rs9277535 at HLA-DPA2, which are all located in the HLA region, 9.84 × 10(-71) ≤ Pmeta ≤ 9.92 × 10(-7) ). CONCLUSION Our GWAS identified five novel susceptibility loci for CHB. These findings improve the understanding of CHB etiology and may provide new targets for prevention and treatment of this disease.
Collapse
Affiliation(s)
- De-Ke Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China.,Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Xiao-Pin Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hongjie Yu
- James D. Watson Institute of Genome Sciences, College of life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangwen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Dong-Lin Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Haitao Chen
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Hui-Xing Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yu-Zhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Xiao-Pan Wu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xi-Dai Long
- Department of Pathology, Youjiang Medical College for Nationalities, Guangxi, China
| | - Hongxing Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Youjie Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Yong Gao
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Tao-Yang Chen
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Wei-Hua Ren
- Luoyang Central Hospital, Affiliated to Zhengzhou University, Luoyang, China
| | - Pengyin Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Zhuqing Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Wei Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Bo Wan
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jianhua Yin
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Yuan-Feng Zhou
- Department of Pathology, Youjiang Medical College for Nationalities, Guangxi, China
| | - Yun Zhai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Pei-Xin Lu
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Hongwei Zhang
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Xiaoli Gu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Aihua Tan
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Jin-Bing Wang
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Xian-Bo Zuo
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Liang-Dan Sun
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Qing Yi
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.,Division of Cancer Medicine, and the Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zengnan Mo
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ying Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jielin Sun
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - S Lilly Zheng
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC.,Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL
| | - Xue-Jun Zhang
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Jianfeng Xu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China.,Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China.,Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Institute of Biomedical Science, Fudan University, Shanghai, China
| |
Collapse
|
112
|
The MS Risk Allele of CD40 Is Associated with Reduced Cell-Membrane Bound Expression in Antigen Presenting Cells: Implications for Gene Function. PLoS One 2015; 10:e0127080. [PMID: 26068105 PMCID: PMC4465929 DOI: 10.1371/journal.pone.0127080] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 04/10/2015] [Indexed: 12/31/2022] Open
Abstract
Human genetic and animal studies have implicated the costimulatory molecule CD40 in the development of multiple sclerosis (MS). We investigated the cell specific gene and protein expression variation controlled by the CD40 genetic variant(s) associated with MS, i.e. the T-allele at rs1883832. Previously we had shown that the risk allele is expressed at a lower level in whole blood, especially in people with MS. Here, we have defined the immune cell subsets responsible for genotype and disease effects on CD40 expression at the mRNA and protein level. In cell subsets in which CD40 is most highly expressed, B lymphocytes and dendritic cells, the MS-associated risk variant is associated with reduced CD40 cell-surface protein expression. In monocytes and dendritic cells, the risk allele additionally reduces the ratio of expression of full-length versus truncated CD40 mRNA, the latter encoding secreted CD40. We additionally show that MS patients, regardless of genotype, express significantly lower levels of CD40 cell-surface protein compared to unaffected controls in B lymphocytes. Thus, both genotype-dependent and independent down-regulation of cell-surface CD40 is a feature of MS. Lower expression of a co-stimulator of T cell activation, CD40, is therefore associated with increased MS risk despite the same CD40 variant being associated with reduced risk of other inflammatory autoimmune diseases. Our results highlight the complexity and likely individuality of autoimmune pathogenesis, and could be consistent with antiviral and/or immunoregulatory functions of CD40 playing an important role in protection from MS.
Collapse
|
113
|
Citro A, Scrivo R, Martini H, Martire C, De Marzio P, Vestri AR, Sidney J, Sette A, Barnaba V, Valesini G. CD8+ T Cells Specific to Apoptosis-Associated Antigens Predict the Response to Tumor Necrosis Factor Inhibitor Therapy in Rheumatoid Arthritis. PLoS One 2015; 10:e0128607. [PMID: 26061065 PMCID: PMC4465029 DOI: 10.1371/journal.pone.0128607] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/28/2015] [Indexed: 12/27/2022] Open
Abstract
CD8+ T cells specific to caspase-cleaved antigens derived from apoptotic T cells (apoptotic epitopes) represent a principal player in chronic immune activation, which is known to amplify immunopathology in various inflammatory diseases. The purpose of the present study was to investigate the relationship involving these autoreactive T cells, the rheumatoid arthritis immunopathology, and the response to tumor necrosis factor-α inhibitor therapy. The frequency of autoreactive CD8+ T cells specific to various apoptotic epitopes, as detected by both enzyme-linked immunospot assay and dextramers of major histocompatibility complex class I molecules complexed with relevant apoptotic epitopes, was longitudinally analyzed in the peripheral blood of rheumatoid arthritis patients who were submitted to etanercept treatment (or other tumor necrosis factor inhibitors as a control). The percentage of apoptotic epitope-specific CD8+ T cells was significantly higher in rheumatoid arthritis patients than in healthy donors, and correlated with the disease activity. More important, it was significantly more elevated in responders to tumor necrosis factor-α inhibitor therapy than in non-responders before the start of therapy; it significantly dropped only in the former following therapy. These data indicate that apoptotic epitope-specific CD8+ T cells may be involved in rheumatoid arthritis immunopathology through the production of inflammatory cytokines and that they may potentially represent a predictive biomarker of response to tumor necrosis factor-α inhibitor therapy to validate in a larger cohort of patients.
Collapse
Affiliation(s)
- Alessandra Citro
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Rossana Scrivo
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Helene Martini
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carmela Martire
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Paolo De Marzio
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Anna Rita Vestri
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza Università di Roma, Rome, Italy
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, San Diego, California 92121, United States of America
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, San Diego, California 92121, United States of America
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy
- * E-mail:
| | - Guido Valesini
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
114
|
Kirino Y, Remmers EF. Genetic architectures of seropositive and seronegative rheumatic diseases. Nat Rev Rheumatol 2015; 11:401-14. [PMID: 25907699 DOI: 10.1038/nrrheum.2015.41] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis and some other rheumatic diseases are genetically complex, with evidence of familial clustering, but not of Mendelian inheritance. These diseases are thought to result from contributions and interactions of multiple genetic and nongenetic risk factors, which have small effects individually. Genome-wide association studies (GWAS) of large collections of data from cases and controls have revealed many genetic factors that contribute to non-Mendelian rheumatic diseases, thus providing insights into associated molecular mechanisms. This Review summarizes methods for the identification of gene variants that influence genetically complex diseases and focuses on what we have learned about the rheumatic diseases for which GWAS have been reported. Our review of the disease-associated loci identified to date reveals greater sharing of risk loci among the groups of seropositive (diseases in which specific autoantibodies are often present) or seronegative diseases than between these two groups. The nature of the shared and discordant loci suggests important similarities and differences among these diseases.
Collapse
Affiliation(s)
- Yohei Kirino
- Yokohama City University Graduate School of Medicine, Department of Internal Medicine and Clinical Immunology, 3-9 Fukuura, Kanazawa-Ku, Yokohama 236-0004, Japan
| | - Elaine F Remmers
- National Institutes of Health, National Human Genome Research Institute, Inflammatory Disease Section, 10 Center Drive, MSC 1849, Bethesda, MD 20892, USA
| |
Collapse
|
115
|
Lee YH, Bae SC, Choi SJ, Ji JD, Song GG. Associations between the functional CD40 rs4810485 G/T polymorphism and susceptibility to rheumatoid arthritis and systemic lupus erythematosus: a meta-analysis. Lupus 2015; 24:1177-83. [DOI: 10.1177/0961203315583543] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/30/2015] [Indexed: 11/16/2022]
Abstract
Objective The aim of this study was to determine whether the functional CD40 rs4810485 G/T polymorphism is associated with susceptibility to rheumatoid arthritis (RA) or with susceptibility to systemic lupus erythematosus (SLE). Methods A series of meta-analyses were conducted to test for association between the CD40 rs4810485 G/T polymorphism and RA or SLE. Results A total of 21 comparisons involving 15,095 patients and 27,050 controls for RA, and 1353 patients and 2342 controls for SLE were considered. Meta-analysis showed a significant association between the CD40 rs4810485 T allele and RA in all subjects (odds ratio (OR) 0.890, 95% confidence interval (CI) 0.846–0.936, p = 5.5 × 10−7). After stratification by ethnicity, the CD40 T allele was found to be significantly associated with RA in Europeans (OR 0.879, 95% CI 0.848–0.901, p = 3.0 × 10−9). A similar pattern of association was observed between the CD40 T allele and RA when the analysis was performed using the recessive, dominant, and additive models. Meta-analysis also showed a significant association between the CD40 polymorphism and SLE in Europeans (OR for the T allele 0.715, 95% CI 0.641–0.832, p = 1.4 × 10−6). Conclusions Our meta-analyses confirm that the CD40 rs4810485 G/T polymorphism is associated with susceptibility to RA and SLE in Europeans.
Collapse
Affiliation(s)
- Y H Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - S-C Bae
- The Hospital for Rheumatic Diseases, Hanyang University Medical Center, Seoul, Korea
| | - S J Choi
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - J D Ji
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - G G Song
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
116
|
Terao C, Ohmura K, Kochi Y, Ikari K, Okada Y, Shimizu M, Nishina N, Suzuki A, Myouzen K, Kawaguchi T, Takahashi M, Takasugi K, Murasawa A, Mizuki S, Iwahashi M, Funahashi K, Natsumeda M, Furu M, Hashimoto M, Ito H, Fujii T, Ezawa K, Matsubara T, Takeuchi T, Kubo M, Yamada R, Taniguchi A, Yamanaka H, Momohara S, Yamamoto K, Mimori T, Matsuda F. Anti-citrullinated peptide/protein antibody (ACPA)-negative RA shares a large proportion of susceptibility loci with ACPA-positive RA: a meta-analysis of genome-wide association study in a Japanese population. Arthritis Res Ther 2015; 17:104. [PMID: 25927497 PMCID: PMC4431175 DOI: 10.1186/s13075-015-0623-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 04/08/2015] [Indexed: 11/16/2022] Open
Abstract
Introduction Although susceptibility genes for anti-citrullinated peptide/protein antibodies (ACPA)-positive rheumatoid arthritis (RA) have been successfully discovered by genome-wide association studies (GWAS), little is known about the genetic background of ACPA-negative RA. We intended to elucidate genetic background of ACPA-negative RA. Method We performed a meta-analysis of GWAS comprising 670 ACPA-negative RA and 16,891 controls for 1,948,138 markers, followed by a replication study of the top 35 single nucleotide polymorphisms (SNPs) using 916 cases and 3,764 controls. Inverse-variance method was applied to assess overall effects. To assess overlap of susceptibility loci between ACPA-positive and -negative RA, odds ratios (ORs) of the 21 susceptibility markers to RA in Japanese were compared between the two subsets. In addition, SNPs were stratified by the p-values in GWAS meta-analysis for either ACPA-positive RA or ACPA-negative RA to address the question whether weakly-associated genes were also shared. The correlations between ACPA-positive RA and the subpopulations of ACPA-negative RA (rheumatoid factor (RF)-positive and RF-negative subsets) were also addressed. Results Rs6904716 in LEMD2 of the human leukocyte antigen (HLA) locus showed a borderline association with ACPA-negative RA (overall p = 5.7 × 10−8), followed by rs6986423 in CSMD1 (p = 2.4 × 10−6) and rs17727339 in FCRL3 (p = 1.4 × 10−5). ACPA-negative RA showed significant correlations of ORs with ACPA-positive RA for the 21 susceptibility SNPs and non-HLA SNPs with p-values far from significance. These significant correlations with ACPA-positive RA were true for ACPA-negative RF-positive and ACPA-negative RF-negative RA. On the contrary, positive correlations were not observed between the ACPA-negative two subpopulations. Conclusion Many of the susceptibility loci were shared between ACPA-positive and -negative RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0623-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chikashi Terao
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan.
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | - Katsunori Ikari
- Institute of Rheumatology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Yukinori Okada
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Masakazu Shimizu
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Naoshi Nishina
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | - Keiko Myouzen
- Laboratory for Autoimmune Diseases, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Meiko Takahashi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | - Akira Murasawa
- Department of Rheumatology, Niigata Rheumatic Center, Niigata, Japan.
| | - Shinichi Mizuki
- The Centre for Rheumatic Diseases, Matsuyama Red Cross Hospital, Matsuyama, Japan.
| | | | - Keiko Funahashi
- Pharm C, Matsubara Mayflower Hospital, 944-25 Fujita, Kato City, Hyogo, Japan.
| | | | - Moritoshi Furu
- Department of the Control for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Motomu Hashimoto
- Department of the Control for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hiromu Ito
- Department of the Control for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Takao Fujii
- Department of the Control for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | - Tsukasa Matsubara
- Matsubara Mayflower Hospital, 944-25 Fujita, Kato City, Hyogo, Japan.
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Michiaki Kubo
- Laboratory for Autoimmune Diseases, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | - Ryo Yamada
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Atsuo Taniguchi
- Institute of Rheumatology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Hisashi Yamanaka
- Institute of Rheumatology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Shigeki Momohara
- Institute of Rheumatology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan.
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan. .,Institut National de la Sante et de la Recherche Medicale (INSERM) Unite U852, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
117
|
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder of unknown aetiology resulting in inflammation of the synovium, cartilage and bone. The disease has a heterogeneous character, consisting of clinical subsets of anti-citrullinated protein antibody (ACPA)-positive and APCA-negative disease. Although, the pathogenesis of RA is incompletely understood, genetic factors play a vital role in susceptibility to RA as the heritability of RA is between 50 and 60%, with the human leukocyte antigen (HLA) locus accounting for at least 30% of overall genetic risk. Non-HLA genes, i.e. tumour necrosis factor-α (TNF-α) within the MHC (major histocompatibility complex) have also been investigated for association with RA. Although, some contradictory results have originated from several studies on TNF-α gene, the data published so far indicate the possible existence of TNF-α gene promoter variants that act as markers for disease severity and response to treatment in RA. The correlation of HLA and non-HLA genes within MHC region is apparently interpreted. A considerable number of confirmed associations with RA and other autoimmune disease susceptibility loci including peptidylarginine deiminase type 4 (PADI4), protein tyrosine phosphatase non-receptor type 22 (PTPN22), signal transducer and activator of transcription (STAT4), cluster of differentiation 244 (CD244) and cytotoxic T lymphocyte-associated antigen 4 (CTLA4), located outside the MHC have been reported recently. In this review, we aim to give an update on recent progress in RA genetics, the importance of the combination of HLA-DRB1 alleles, non-HLA gene polymorphism, its detection and autoantibodies as susceptibility markers for early RA disease.
Collapse
|
118
|
Carmona F, Mackie S, Martín JE, Taylor J, Vaglio A, Eyre S, Bossini-Castillo L, Castañeda S, Cid M, Hernández-Rodríguez J, Prieto-González S, Solans R, Ramentol-Sintas M, González-Escribano M, Ortiz-Fernández L, Morado I, Narváez J, Miranda-Filloy J, Beretta L, Lunardi C, Cimmino MA, Gianfreda D, Santilli D, Ramirez GA, Soriano A, Muratore F, Pazzola G, Addimanda O, Wijmenga C, Witte T, Schirmer JH, Moosig F, Schönau V, Franke A, Palm Ø, Molberg Ø, Diamantopoulos AP, Carette S, Cuthbertson D, Forbess LJ, Hoffman GS, Khalidi NA, Koening CL, Langford CA, McAlear CA, Moreland L, Monach PA, Pagnoux C, Seo P, Spiera R, Sreih AG, Warrington KJ, Ytterberg SR, Gregersen PK, Pease CT, Gough A, Green M, Hordon L, Jarrett S, Watts R, Levy S, Patel Y, Kamath S, Dasgupta B, Worthington J, Koeleman BP, de Bakker PI, Barrett JH, Salvarani C, Merkel PA, González-Gay MA, Morgan AW, Martín J, Martínez-Berriochoa A, Unzurrunzaga A, Hidalgo-Conde A, Madroñero-Vuelta A, Fernández-Nebro A, Ordóñez-Cañizares M, Escalante B, Marí-Alfonso B, Sopeña B, Magro C, Raya E, Grau E, Román J, de Miguel E, López-Longo F, Martínez L, Gómez-Vaquero C, Fernández-Gutiérrez B, Rodríguez-Rodríguez L, Díaz-López J, Caminal-Montero L, Martínez-Zapico A, Monfort J, Tío L, Sánchez-Martín J, Alegre-Sancho J, Sáez-Comet L, Pérez-Conesa M, Corbera-Bellalta M, García-Villanueva M, Fernández-Contreras M, Sanchez-Pernaute O, Blanco R, Ortego-Centeno N, Ríos-Fernández R, Callejas J, Fanlo-Mateo P, Martínez-Taboada V. A large-scale genetic analysis reveals a strong contribution of the HLA class II region to giant cell arteritis susceptibility. Am J Hum Genet 2015; 96:565-80. [PMID: 25817017 DOI: 10.1016/j.ajhg.2015.02.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/12/2015] [Indexed: 02/08/2023] Open
Abstract
We conducted a large-scale genetic analysis on giant cell arteritis (GCA), a polygenic immune-mediated vasculitis. A case-control cohort, comprising 1,651 case subjects with GCA and 15,306 unrelated control subjects from six different countries of European ancestry, was genotyped by the Immunochip array. We also imputed HLA data with a previously validated imputation method to perform a more comprehensive analysis of this genomic region. The strongest association signals were observed in the HLA region, with rs477515 representing the highest peak (p = 4.05 × 10(-40), OR = 1.73). A multivariate model including class II amino acids of HLA-DRβ1 and HLA-DQα1 and one class I amino acid of HLA-B explained most of the HLA association with GCA, consistent with previously reported associations of classical HLA alleles like HLA-DRB1(∗)04. An omnibus test on polymorphic amino acid positions highlighted DRβ1 13 (p = 4.08 × 10(-43)) and HLA-DQα1 47 (p = 4.02 × 10(-46)), 56, and 76 (both p = 1.84 × 10(-45)) as relevant positions for disease susceptibility. Outside the HLA region, the most significant loci included PTPN22 (rs2476601, p = 1.73 × 10(-6), OR = 1.38), LRRC32 (rs10160518, p = 4.39 × 10(-6), OR = 1.20), and REL (rs115674477, p = 1.10 × 10(-5), OR = 1.63). Our study provides evidence of a strong contribution of HLA class I and II molecules to susceptibility to GCA. In the non-HLA region, we confirmed a key role for the functional PTPN22 rs2476601 variant and proposed other putative risk loci for GCA involved in Th1, Th17, and Treg cell function.
Collapse
|
119
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
120
|
Denninger KCM, Litman T, Marstrand T, Moller K, Svensson L, Labuda T, Andersson Å. Kinetics of gene expression and bone remodelling in the clinical phase of collagen-induced arthritis. Arthritis Res Ther 2015; 17:43. [PMID: 25889670 PMCID: PMC4391727 DOI: 10.1186/s13075-015-0531-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 01/19/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Pathological bone changes differ considerably between inflammatory arthritic diseases and most studies have focused on bone erosion. Collagen-induced arthritis (CIA) is a model for rheumatoid arthritis, which, in addition to bone erosion, demonstrates bone formation at the time of clinical manifestations. The objective of this study was to use this model to characterise the histological and molecular changes in bone remodelling, and relate these to the clinical disease development. Methods A histological and gene expression profiling time-course study on bone remodelling in CIA was linked to onset of clinical symptoms. Global gene expression was studied with a gene chip array system. Results The main histopathological changes in bone structure and inflammation occurred during the first two weeks following the onset of clinical symptoms in the joint. Hereafter, the inflammation declined and remodelling of formed bone dominated. Global gene expression profiling showed simultaneous upregulation of genes related to bone changes and inflammation in week 0 to 2 after onset of clinical disease. Furthermore, we observed time-dependent expression of genes involved in early and late osteoblast differentiation and function, which mirrored the histopathological bone changes. The differentially expressed genes belong to the bone morphogenetic pathway (BMP) and, in addition, include the osteoblast markers integrin-binding sialoprotein (Ibsp), bone gamma-carboxyglutamate protein (Bglap1), and secreted phosphoprotein 1 (Spp1). Pregnancy-associated protein A (Pappa) and periostin (Postn), differentially expressed in the early disease phase, are proposed to participate in bone formation, and we suggest that they play a role in early bone formation in the CIA model. Comparison to human genome-wide association studies (GWAS) revealed differential expression of several genes associated with human arthritis. Conclusions In the CIA model, bone formation in the joint starts shortly after onset of clinical symptoms, which results in bony fusion within one to two weeks. This makes it a candidate model for investigating the relationship between inflammation and bone formation in inflammatory arthritis. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0531-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katja C M Denninger
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, Ø DK-2100, Denmark. .,Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Thomas Litman
- Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Troels Marstrand
- Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Kristian Moller
- Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Lars Svensson
- Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Tord Labuda
- Disease Pharmacology/Molecular Biomedicine, LEO Pharma A/S, Industriparken 55, Ballerup, DK-2750, Denmark.
| | - Åsa Andersson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, Ø DK-2100, Denmark.
| |
Collapse
|
121
|
Saad MN, Mabrouk MS, Eldeib AM, Shaker OG. Identification of rheumatoid arthritis biomarkers based on single nucleotide polymorphisms and haplotype blocks: A systematic review and meta-analysis. J Adv Res 2015; 7:1-16. [PMID: 26843965 PMCID: PMC4703421 DOI: 10.1016/j.jare.2015.01.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/13/2015] [Accepted: 01/20/2015] [Indexed: 12/30/2022] Open
Abstract
Genetics of autoimmune diseases represent a growing domain with surpassing biomarker results with rapid progress. The exact cause of Rheumatoid Arthritis (RA) is unknown, but it is thought to have both a genetic and an environmental bases. Genetic biomarkers are capable of changing the supervision of RA by allowing not only the detection of susceptible individuals, but also early diagnosis, evaluation of disease severity, selection of therapy, and monitoring of response to therapy. This review is concerned with not only the genetic biomarkers of RA but also the methods of identifying them. Many of the identified genetic biomarkers of RA were identified in populations of European and Asian ancestries. The study of additional human populations may yield novel results. Most of the researchers in the field of identifying RA biomarkers use single nucleotide polymorphism (SNP) approaches to express the significance of their results. Although, haplotype block methods are expected to play a complementary role in the future of that field.
Collapse
Affiliation(s)
- Mohamed N Saad
- Biomedical Engineering Department, Faculty of Engineering, Misr University for Science and Technology, 6th of October City, Egypt
| | - Mai S Mabrouk
- Biomedical Engineering Department, Faculty of Engineering, Misr University for Science and Technology, 6th of October City, Egypt
| | - Ayman M Eldeib
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Giza, Egypt
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
122
|
Systematic confirmation study of GWAS-identified genetic variants for Kawasaki disease in a Chinese population. Sci Rep 2015; 5:8194. [PMID: 25645453 PMCID: PMC4314627 DOI: 10.1038/srep08194] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/12/2015] [Indexed: 12/16/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) associated with Kawasaki disease (KD). In this study, we replicated the associations of 10 GWAS-identified SNPs with KD in a Han Chinese population. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by logistic regression, and cumulative effect of non-risk genotypes were also performed. Although none of the SNPs reached the corrected significance level, 4 SNPs showed nominal associations with KD risk. Compared with their respective wild type counterparts, rs1801274 AG+GG genotypes and rs3818298 TC+CC genotypes were nominally associated with the reduced risk of KD (OR = 0.77, 95% CI = 0.59-0.99, P = 0.045; OR = 0.74, 95% CI = 0.56-0.98, P = 0.038). Meanwhile, rs1801274 GG genotype, rs2736340 CC genotype or rs4813003 TT genotype showed a reduced risk trend (OR = 0.57, 95% CI = 0.35-0.93, P = 0.024; OR = 0.46, 95% CI = 0.26-0.83, P = 0.010; OR = 0.64, 95% CI = 0.43-0.94, P = 0.022), compared with rs1801274 AG+AA genotypes, rs2736340 CT+TT genotypes or rs4813003 TC+CC genotypes, respectively. Furthermore, a cumulative effect was observed with the ORs being gradually decreased with the increasing accumulative number of non-risk genotypes (Ptrend<0.001). In conclusion, our study suggests that 4 GWAS-identified SNPs, rs2736340, rs4813003, rs3818298 and rs1801274, were nominally associated with KD risk in a Han Chinese population individually and jointly.
Collapse
|
123
|
Jiang X, Frisell T, Askling J, Karlson EW, Klareskog L, Alfredsson L, Källberg H. To What Extent Is the Familial Risk of Rheumatoid Arthritis Explained by Established Rheumatoid Arthritis Risk Factors? Arthritis Rheumatol 2015; 67:352-62. [DOI: 10.1002/art.38927] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/21/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Xia Jiang
- Karolinska InstitutetStockholmSweden
| | | | - Johan Askling
- Karolinska Institutet and Karolinska University Hospital SolnaStockholmSweden
| | | | | | - Lars Alfredsson
- Karolinska Institutet and Centre for Occupational and Environmental Medicine, Stockholm County CouncilStockholmSweden
| | | |
Collapse
|
124
|
The contribution of genetic factors to rheumatoid arthritis. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00089-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
125
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
126
|
THEMIS is required for pathogenesis of cerebral malaria and protection against pulmonary tuberculosis. Infect Immun 2014; 83:759-68. [PMID: 25452553 DOI: 10.1128/iai.02586-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We identify an N-ethyl-N-nitrosourea (ENU)-induced I23N mutation in the THEMIS protein that causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Themis(I23N) homozygous mice show reduced CD4(+) and CD8(+) T lymphocyte numbers. ECM resistance in P. berghei ANKA-infected Themis(I23N) mice is associated with decreased cerebral cellular infiltration, retention of blood-brain barrier integrity, and reduced proinflammatory cytokine production. THEMIS(I23N) protein expression is absent from mutant mice, concurrent with the decreased THEMIS(I23N) stability observed in vitro. Biochemical studies in vitro and functional complementation in vivo in Themis(I23N/+):Lck(-/+) doubly heterozygous mice demonstrate that functional coupling of THEMIS to LCK tyrosine kinase is required for ECM pathogenesis. Damping of proinflammatory responses in Themis(I23N) mice causes susceptibility to pulmonary tuberculosis. Thus, THEMIS is required for the development and ultimately the function of proinflammatory T cells. Themis(I23N) mice can be used to study the newly discovered association of THEMIS (6p22.33) with inflammatory bowel disease and multiple sclerosis.
Collapse
|
127
|
Kennedy JM, Fodil N, Torre S, Bongfen SE, Olivier JF, Leung V, Langlais D, Meunier C, Berghout J, Langat P, Schwartzentruber J, Majewski J, Lathrop M, Vidal SM, Gros P. CCDC88B is a novel regulator of maturation and effector functions of T cells during pathological inflammation. ACTA ACUST UNITED AC 2014; 211:2519-35. [PMID: 25403443 PMCID: PMC4267237 DOI: 10.1084/jem.20140455] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kennedy et al. identify a mutation in coiled-coil domain containing protein 88b (Ccdc88b) that confers protection against lethal neuroinflammation during experimental cerebral malaria. CCDC88B is expressed in immune cells and regulates T cell maturation and effector functions. In humans, the CCDC88B gene maps to a locus associated with susceptibility to several inflammatory and autoimmune disorders. We used a genome-wide screen in mutagenized mice to identify genes which inactivation protects against lethal neuroinflammation during experimental cerebral malaria (ECM). We identified an ECM-protective mutation in coiled-coil domain containing protein 88b (Ccdc88b), a poorly annotated gene that is found expressed specifically in spleen, bone marrow, lymph nodes, and thymus. The CCDC88B protein is abundantly expressed in immune cells, including both CD4+ and CD8+ T lymphocytes, and in myeloid cells, and loss of CCDC88B protein expression has pleiotropic effects on T lymphocyte functions, including impaired maturation in vivo, significantly reduced activation, reduced cell division as well as impaired cytokine production (IFN-γ and TNF) in response to T cell receptor engagement, or to nonspecific stimuli in vitro, and during the course of P. berghei infection in vivo. This identifies CCDC88B as a novel and important regulator of T cell function. The human CCDC88B gene maps to the 11q13 locus that is associated with susceptibility to several inflammatory and auto-immune disorders. Our findings strongly suggest that CCDC88B is the morbid gene underlying the pleiotropic effect of the 11q13 locus on inflammation.
Collapse
Affiliation(s)
- James M Kennedy
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Nassima Fodil
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Sabrina Torre
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Silayuv E Bongfen
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Jean-Frédéric Olivier
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Vicki Leung
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - David Langlais
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Charles Meunier
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Joanne Berghout
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Pinky Langat
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Jeremy Schwartzentruber
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Jacek Majewski
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Mark Lathrop
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Silvia M Vidal
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Philippe Gros
- Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada Department of Biochemistry, Department of Human Genetics, McGill and Genome Quebec Innovation Center, Complex Traits Group, McGill University, Montreal, Quebec H3A 0G4, Canada
| |
Collapse
|
128
|
İnal EE, Rüstemoğlu A, İnanır A, Ekinci D, Gül Ü, Yiğit S, Ateş Ö. Associations of rs4810485 and rs1883832 polymorphisms of CD40 gene with susceptibility and clinical findings of Behçet's disease. Rheumatol Int 2014; 35:837-43. [PMID: 25373542 DOI: 10.1007/s00296-014-3171-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/28/2014] [Indexed: 01/18/2023]
Abstract
There are evidences that besides geographic tendency, interactions between genetic and environmental factors play an essential role in the pathogenesis of Behçet's disease (BD). In this study, we have evaluated the associations between rs4810485 and rs1883832 single nucleotide polymorphism (SNP)s of CD40 gene with the susceptibility and clinical findings of BD. Two hundred and eighty-five patients with BD and 225 age-matched healthy controls were enrolled in this study. The clinical findings of patients were noted. The distributions of genotypes, alleles, combined genotypes and haplotypes of these two SNPs in BD patients were compared with those in healthy controls. In further evaluation, we evaluated the patients with and without any of clinical findings with regarding to distribution of genotypes and alleles of these two SNPs. There was no significant difference concerning frequencies of genotypes, alleles, combined genotypes and haplotypes of rs4810485 and rs1883832 between patients and controls (p > 0.05 for all). Frequency of GT genotype of CD40 rs4810485 polymorphism was found to be significantly higher in patients with skin lesions (p < 0.05, OR 1.65, 95 % CI 1.02-2.64). Also, we have found significantly higher frequencies of CC genotype and C allele of CD40 rs1883832 polymorphism in patients with genital ulcers (p < 0.05 for both, OR 2.30, 95 % CI 1.07-4.94 and OR 1.78, 95 % CI 1.06-2.97, respectively). However, these significances were disappeared after Bonferroni correction. We suggest that differences in the expression levels of CD40 because of different genotypes of these two SNPs may take part in the development of skin lesions or genital ulcers in patients with BD.
Collapse
Affiliation(s)
- Esra Erkol İnal
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey,
| | | | | | | | | | | | | |
Collapse
|
129
|
Shimabukuro-Vornhagen A, Zoghi S, Liebig TM, Wennhold K, Chemitz J, Draube A, Kochanek M, Blaschke F, Pallasch C, Holtick U, Scheid C, Theurich S, Hallek M, von Bergwelt-Baildon MS. Inhibition of protein geranylgeranylation specifically interferes with CD40-dependent B cell activation, resulting in a reduced capacity to induce T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 193:5294-305. [PMID: 25311809 DOI: 10.4049/jimmunol.1203436] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ab-independent effector functions of B cells, such as Ag presentation and cytokine production, have been shown to play an important role in a variety of immune-mediated conditions such as autoimmune diseases, transplant rejection, and graft-versus-host disease. Most current immunosuppressive treatments target T cells, are relatively unspecific, and result in profound immunosuppression that places patients at an increased risk of developing severe infections and cancer. Therapeutic strategies, which interfere with B cell activation, could therefore be a useful addition to the current immunosuppressive armamentarium. Using a transcriptomic approach, we identified upregulation of genes that belong to the mevalonate pathway as a key molecular event following CD40-mediated activation of B cells. Inhibition of 3-hydroxy-3-methylglutaryl CoA reductase, the rate-limiting enzyme of the mevalonate pathway, by lipophilic statins such as simvastatin and atorvastatin resulted in a specific inhibition of B cell activation via CD40 and impaired their ability to act as stimulatory APCs for allospecific T cells. Mechanistically, the inhibitory effect resulted from the inhibition of protein geranylgeranylation subsequent to the depletion of mevalonate, the metabolic precursor for geranylgeranyl. Thus, inhibition of geranylgeranylation either directly through geranylgeranyl transferase inhibitors or indirectly through statins represents a promising therapeutic approach for the treatment of diseases in which Ag presentation by B cells plays a role.
Collapse
Affiliation(s)
- Alexander Shimabukuro-Vornhagen
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany;
| | - Shahram Zoghi
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Tanja M Liebig
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Kerstin Wennhold
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Jens Chemitz
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Andreas Draube
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Matthias Kochanek
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| | - Florian Blaschke
- Department of Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; and Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Christian Pallasch
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Udo Holtick
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Christof Scheid
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Sebastian Theurich
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Michael Hallek
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| | - Michael S von Bergwelt-Baildon
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| |
Collapse
|
130
|
Thude H, Kramer K, Koch M, Peine S, Sterneck M, Nashan B. Lack of association between CD40 polymorphisms and acute rejection in German liver transplant recipients. Hum Immunol 2014; 75:1123-7. [PMID: 25305459 DOI: 10.1016/j.humimm.2014.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/15/2023]
Abstract
CD40 and its ligand, CD154, are major costimulatory molecules whose interactions are important in alloreactive transplant rejection. The aim of this study was to examine the association of CD40 polymorphisms with the susceptibility to acute rejection episodes in liver transplantation. In total, 112 liver transplant recipients with biopsy proven acute rejections (BPAR), 97 without BPAR (WBPAR), and 112 healthy control individuals were enrolled in the study. Two single nucleotide polymorphisms (SNPs) of CD40 gene (rs1883832 and rs4810485) were genotyped by polymerase chain reaction-allele specific restriction enzyme analysis (PCR-ASRA). Both SNPs has been tested for a recessive and a dominant model. No significant differences were found in the genotype and allele frequencies of the SNPs rs1883832 and rs4810485 between BPAR liver recipients and WBPAR recipients. Our results do not suggest an important role of tested CD40 SNPs in the susceptibility to acute liver transplant rejection in a Caucasian population.
Collapse
Affiliation(s)
- Hansjörg Thude
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Kathrin Kramer
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Martina Koch
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| | - Sven Peine
- University Medical Center Hamburg-Eppendorf, Institute for Transfusion Medicine, Martinistraße 52, 20246 Hamburg, Germany
| | - Martina Sterneck
- University Medical Center Hamburg-Eppendorf, Department of Medicine, Martinistraße 52, 20246 Hamburg, Germany
| | - Björn Nashan
- University Medical Center Hamburg-Eppendorf, Department of Hepatobiliary and Transplant Surgery, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
131
|
Kreft KL, van Meurs M, Wierenga-Wolf AF, Melief MJ, van Strien ME, Hol EM, Oostra BA, Laman JD, Hintzen RQ. Abundant kif21b is associated with accelerated progression in neurodegenerative diseases. Acta Neuropathol Commun 2014; 2:144. [PMID: 25274010 PMCID: PMC4207309 DOI: 10.1186/s40478-014-0144-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 09/14/2014] [Indexed: 01/29/2023] Open
Abstract
Kinesin family member 21b (kif21b) is one of the few multiple sclerosis (MS) risk genes with a presumed central nervous system function. Kif21b belongs to the kinesin family, proteins involved in intracellular transport of proteins and organelles. We hypothesised that kif21b is involved in the neurodegenerative component of MS and Alzheimer's (AD) disease. Post-mortem kinesin expression was assessed in 50 MS, 58 age and gender matched non-demented controls (NDC) and 50 AD. Kif21b expression was five-fold increased in AD compared to MS and NDC aged below 62 years (p = 8*10(-5)), three-fold between 62-72 years (p = 0.005) and not different above 72 years. No significant differences were observed between MS and NDC. In AD, kif21b expression was two-fold increased in Braak stage 6 (scoring for density of neurofibrillary tangles) compared with stage 5 (p = 0.003). In MS patients, kif21b correlated with the extent of grey matter demyelination (Spearman's rho = 0.31, p = 0.03). Abundant kif21b, defined as expression above the median, was associated with a two-fold accelerated development of the Kurtzke Expanded Disability Status Scale (EDSS) 6.0 (median time in low kif21b group 16 years vs. high kif21b 7.5 years, log-rank test p = 0.04) in MS. Given the genetic association of kif21b with MS, the results were stratified according to rs12122721[A] single nucleotide polymorphism (SNP). No association was found between kif21b expression or the time to EDSS 6 in kif21b risk SNP carriers compared to non-risk carriers. Kif21b was expressed in astrocytes in addition to neurons. Upon astrocyte activation, kif21b increased nine-fold. Abundant kif21b expression is associated with severe MS and AD pathology and with accelerated neurodegeneration independent of the kif21b risk SNP.
Collapse
Affiliation(s)
- Karim L Kreft
- Department Neurology Erasmus MC, University Medical Center, Room Ba 4.92, Rotterdam, 3000 CA The Netherlands
- Department Immunology Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marjan van Meurs
- Department Immunology Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Annet F Wierenga-Wolf
- Department Immunology Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marie-Jose Melief
- Department Immunology Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Miriam E van Strien
- Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elly M Hol
- Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ben A Oostra
- Department Clinical Genetics Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jon D Laman
- Department Immunology Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rogier Q Hintzen
- Department Neurology Erasmus MC, University Medical Center, Room Ba 4.92, Rotterdam, 3000 CA The Netherlands
- Department MS Center ErasMS Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW Autoimmune diseases such as rheumatoid arthritis (RA) pose an increasing, worldwide economic and health burden. Significantly, no cure exists for the majority of autoimmune diseases and consequently treatment is largely aimed at controlling disease symptoms. Therefore, there exists a critical need to develop new approaches that directly address the cause of disease, leading to disease remission and ultimately cure. RECENT FINDINGS The organs, cells and molecules involved in the breach of self-tolerance have been partially defined in experimental models of autoimmunity. However, the broad applicability of this dogma in clinical disease is only partially understood. This gap between analyses of established disease and investigating early disease pathogenesis argues for the need for complementary studies in mice and humans. SUMMARY Through a combination of clinical and experimental systems, novel autoantigens and neoepitopes involved in RA have been revealed. These have clear utility in predisease diagnosis and offer the possibility of antigen-specific immunotherapy. Ongoing experimental and clinical studies, for example using dendritic cell transfer, will facilitate a clearer understanding of the molecules, cells and organs that should be targeted to reinstate immunological tolerance. Antigen-specific immunotherapy therefore offers disease intervention without broad immunosuppression, and most importantly increases the likelihood of achieving true disease remission and cure.
Collapse
|
133
|
Korczowska I. Rheumatoid arthritis susceptibility genes: An overview. World J Orthop 2014; 5:544-549. [PMID: 25232530 PMCID: PMC4133460 DOI: 10.5312/wjo.v5.i4.544] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 05/29/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory autoimmune disease sustained by genetic factors. Various aspects of the genetic contribution to the pathogenetics and outcome of RA are still unknown. Several genes have been indicated so far in the pathogenesis of RA. Apart from human leukocyte antigen, large genome wide association studies have identified many loci involved in RA pathogenesis. These genes include protein tyrosine phosphatase, nonreceptor type 22, Peptidyl Arginine Deiminase type IV, signal transducer and activator of transcription 4, cytotoxic T-lymphocyte-associated protein 4, tumor necrosis factor-receptor associated factor 1/complement component 5, tumor necrosis factor and others. It is important to determine whether a combination of RA risk alleles are able to identify patients who will develop certain clinical outcomes, such myocardium infarction, severe infection or lymphoma, as well as to identify patients who will respond to biological medication therapy.
Collapse
|
134
|
Okada Y. From the era of genome analysis to the era of genomic drug discovery: a pioneering example of rheumatoid arthritis. Clin Genet 2014; 86:432-40. [PMID: 25060537 DOI: 10.1111/cge.12465] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 01/18/2023]
Abstract
Although we have obtained comprehensive catalogs of genetic risk loci that are linked to human diseases, little is known regarding how to devise a systematic strategy to integrate genetic study results with diverse biological resources. Such strategies will be crucial for providing novel insights into disease biology and for aiding drug discovery as an ultimate goal. Here we describe the current progress in this field using a pioneering example of large-scale genetic association studies on rheumatoid arthritis (RA), an autoimmune disease characterized by inflammation and destruction of joints. Through functional and bioinformatic annotations of risk single nucleotide polymorphisms (SNPs) and genes from >100 RA risk loci identified by genome-wide association study (GWAS) meta-analysis, we found novel biological insights into RA pathogenicity. Further, by integrating RA genetic findings with the complete catalog of approved drugs for RA and other diseases, we provide empirical data to indicate that human genetic-based approaches may be useful for supporting 'genetics-driven genomic drug discovery' efforts in complex human traits and suggest that further development of integrative approaches should be undertaken.
Collapse
Affiliation(s)
- Y Okada
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
135
|
Genetics, environment, and gene-environment interactions in the development of systemic rheumatic diseases. Rheum Dis Clin North Am 2014; 40:637-57. [PMID: 25437282 DOI: 10.1016/j.rdc.2014.07.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rheumatic diseases offer distinct challenges to researchers because of heterogeneity in disease phenotypes, low disease incidence, and geographic variation in genetic and environmental factors. Emerging research areas, including epigenetics, metabolomics, and the microbiome, may provide additional links between genetic and environmental risk factors in the pathogenesis of rheumatic disease. This article reviews the methods used to establish genetic and environmental risk factors and studies gene-environment interactions in rheumatic diseases, and provides specific examples of successes and challenges in identifying gene-environment interactions in rheumatoid arthritis, systemic lupus erythematosus, and ankylosing spondylitis. Emerging research strategies and future challenges are discussed.
Collapse
|
136
|
Snir O, Gomez-Cabrero D, Montes A, Perez-Pampin E, Gómez-Reino JJ, Seddighzadeh M, Klich KU, Israelsson L, Ding B, Catrina AI, Holmdahl R, Alfredsson L, Klareskog L, Tegnér J, Gonzalez A, Malmström V, Padyukov L. Non-HLA genes PTPN22, CDK6 and PADI4 are associated with specific autoantibodies in HLA-defined subgroups of rheumatoid arthritis. Arthritis Res Ther 2014; 16:414. [PMID: 25138370 PMCID: PMC4292996 DOI: 10.1186/s13075-014-0414-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/28/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Genetic susceptibility to complex diseases has been intensively studied during the last decade, yet only signals with small effect have been found leaving open the possibility that subgroups within complex traits show stronger association signals. In rheumatoid arthritis (RA), autoantibody production serves as a helpful discriminator in genetic studies and today anti-citrullinated cyclic peptide (anti-CCP) antibody positivity is employed for diagnosis of disease. The HLA-DRB1 locus is known as the most important genetic contributor for the risk of RA, but is not sufficient to drive autoimmunity and additional genetic and environmental factors are involved. Hence, we addressed the association of previously discovered RA loci with disease-specific autoantibody responses in RA patients stratified by HLA-DRB1*04. METHODS We investigated 2178 patients from three RA cohorts from Sweden and Spain for 41 genetic variants and four autoantibodies, including the generic anti-CCP as well as specific responses towards citrullinated peptides from vimentin, alpha-enolase and type II collagen. RESULTS Our data demonstrated different genetic associations of autoantibody-positive disease subgroups in relation to the presence of DRB1*04. Two specific subgroups of autoantibody-positive RA were identified. The SNP in PTPN22 was associated with presence of anti-citrullinated enolase peptide antibodies in carriers of HLA-DRB1*04 (Cochran-Mantel-Haenszel test P = 0.0001, P corrected <0.05), whereas SNPs in CDK6 and PADI4 were associated with anti-CCP status in DRB1*04 negative patients (Cochran-Mantel-Haenszel test P = 0.0004, P corrected <0.05 for both markers). Additionally we see allelic correlation with autoantibody titers for PTPN22 SNP rs2476601 and anti-citrullinated enolase peptide antibodies in carriers of HLA-DRB1*04 (Mann Whitney test P = 0.02) and between CDK6 SNP rs42041 and anti-CCP in non-carriers of HLA-DRB1*04 (Mann Whitney test P = 0.02). CONCLUSION These data point to alternative pathways for disease development in clinically similar RA subgroups and suggest an approach for study of genetic complexity of disease with strong contribution of HLA.
Collapse
|
137
|
Genome-wide association studies to advance our understanding of critical cell types and pathways in rheumatoid arthritis: recent findings and challenges. Curr Opin Rheumatol 2014; 26:85-92. [PMID: 24276088 DOI: 10.1097/bor.0000000000000012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW A significant number of loci implicated in rheumatoid arthritis (RA) susceptibility have been highlighted by genome-wide association studies (GWAS). Here, we review the recent advances of GWAS in understanding the genetic architecture of RA, and place these findings in the context of RA pathogenesis. RECENT FINDINGS Although the interpretation of GWAS findings in the context of the disease biology remains challenging, interesting observations can be highlighted. Integration of GWAS results with cell-type specific gene expression or epigenetic marks have highlighted regulatory T cells and CD4 memory T cells as critical cell types in RA. In addition, many genes in RA loci are involved in the nuclear factor-kappaB signaling pathway or the Janus kinase (JAK)-signal transducers and activators of transcription (STAT) signaling pathway. The observation that these pathways are targeted by several approved drugs used to treat the symptoms of RA highlights the promises of human genetics to provide insights in the disease biology, and help identify new therapeutic targets. SUMMARY These findings highlight the promises and need of future studies investigating causal genes and underlined mechanisms in GWAS loci to advance our understanding of RA.
Collapse
|
138
|
Platt AM, Benson RA, McQueenie R, Butcher JP, Braddock M, Brewer JM, McInnes IB, Garside P. The active metabolite of spleen tyrosine kinase inhibitor fostamatinib abrogates the CD4+ T cell-priming capacity of dendritic cells. Rheumatology (Oxford) 2014; 54:169-77. [DOI: 10.1093/rheumatology/keu273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
139
|
Reinards THCM, Albers HM, Brinkman DMC, Kamphuis SSM, van Rossum MAJ, Girschick HJ, Wouters C, Hoppenreijs EPAH, Saurenmann RK, Hinks A, Ellis JA, Bakker E, Verduijn W, Slagboom P, Huizinga TWJ, Toes REM, Houwing-Duistermaat JJ, ten Cate R, Schilham MW. CD226 (DNAM-1) is associated with susceptibility to juvenile idiopathic arthritis. Ann Rheum Dis 2014; 74:2193-8. [PMID: 25057181 DOI: 10.1136/annrheumdis-2013-205138] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 07/11/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Juvenile idiopathic arthritis (JIA) is considered a complex genetic autoimmune disease. We investigated the association of genetic variants previously implicated in JIA, autoimmunity and/or immunoregulation, with susceptibility to JIA. METHODS A genetic association study was performed in 639 JIA patients and 1613 healthy controls of northwest European descent. Ninety-three single nucleotide polymorphisms (SNP) were genotyped in a candidate gene approach. Results of the entire JIA patient group (all subtypes) were compared with results obtained, alternatively, with a clinically homogeneous patient group including only oligoarticular and rheumatoid factor (RF) negative polyarticular JIA patients (n=493). Meta-analyses were performed for all SNPs that have been typed in other Caucasian JIA cohorts before. RESULTS SNPs in or near PTPN22, VTCN1, the IL2-IL21 region, ANKRD55 and TNFA were confirmed to be associated with JIA (p<0.05), strengthening the evidence for involvement of these genes in JIA. In the majority of these replicated SNPs, effect sizes were larger when analysing a homogeneous patient cohort than when analysing all subtypes. We identified two novel associations with oligoarticular and RF-negative polyarticular JIA: CD226 rs763361 (OR 1.30, 95% CI 1.12 to 1.51, p=0.0006) and CD28 rs1980422 (OR 1.29, 95% CI 1.07 to 1.55, p=0.008). Meta-analyses including reported studies confirmed the association of both SNPs with susceptibility to JIA (OR 1.16, p=0.001 and OR 1.18, p=0.001, for rs763361 and rs1980422, respectively). CONCLUSIONS The CD226 gene has been identified as novel association with JIA, and a SNP near CD28 as a suggestive association. Both genes are probable candidate risk factors, since they are involved in costimulation of T cells.
Collapse
Affiliation(s)
- T H C M Reinards
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - H M Albers
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - D M C Brinkman
- Department of Pediatrics/Pediatric Rheumatology, Rijnland Hospital, Leiderdorp, The Netherlands
| | - S S M Kamphuis
- Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - M A J van Rossum
- Department of Pediatrics/Pediatric Rheumatology, Academic Medical Centre/Emma Children's Hospital and Reade (Jan van Breemen location), Amsterdam, The Netherlands
| | - H J Girschick
- Vivantes Children's Hospital, Berlin-Friedrichshain, Germany
| | - C Wouters
- University Hospital Gasthuisberg, Leuven, Belgium
| | - E P A H Hoppenreijs
- Department of Pediatrics/Pediatric Rheumatology, St Maartenskliniek and Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - R K Saurenmann
- Zürich University Children's Hospital, Zürich, Switzerland
| | - A Hinks
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - J A Ellis
- Department of Pediatrics, The University of Melbourne, Melbourne, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Australia
| | - E Bakker
- Centre for Human and Clinical Genetics/Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - W Verduijn
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - P Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - T W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - R ten Cate
- Department of Pediatrics/Pediatric Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - M W Schilham
- Department of Pediatrics/Laboratory for Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
140
|
Senapati S, Gutierrez-Achury J, Sood A, Midha V, Szperl A, Romanos J, Zhernakova A, Franke L, Alonso S, Thelma BK, Wijmenga C, Trynka G. Evaluation of European coeliac disease risk variants in a north Indian population. Eur J Hum Genet 2014; 23:530-5. [PMID: 25052311 PMCID: PMC4666579 DOI: 10.1038/ejhg.2014.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 06/10/2014] [Accepted: 06/18/2014] [Indexed: 01/06/2023] Open
Abstract
Studies in European populations have contributed to a better understanding of the genetics of complex diseases, for example, in coeliac disease (CeD), studies of over 23 000 European samples have reported association to the HLA locus and another 39 loci. However, these associations have not been evaluated in detail in other ethnicities. We sought to better understand how disease-associated loci that have been mapped in Europeans translate to a disease risk for a population with a different ethnic background. We therefore performed a validation of European risk loci for CeD in 497 cases and 736 controls of north Indian origin. Using a dense-genotyping platform (Immunochip), we confirmed the strong association to the HLA region (rs2854275, P=8.2 × 10−49). Three loci showed suggestive association (rs4948256, P=9.3 × 10−7, rs4758538, P=8.6 × 10−5 and rs17080877, P=2.7 × 10−5). We directly replicated five previously reported European variants (P<0.05; mapping to loci harbouring FASLG/TNFSF18, SCHIP1/IL12A, PFKFB3/PRKCQ, ZMIZ1 and ICOSLG). Using a transferability test, we further confirmed association at PFKFB3/PRKCQ (rs2387397, P=2.8 × 10−4) and PTPRK/THEMIS (rs55743914, P=3.4 × 10−4). The north Indian population has a higher degree of consanguinity than Europeans and we therefore explored the role of recessively acting variants, which replicated the HLA locus (rs9271850, P=3.7 × 10−23) and suggested a role of additional four loci. To our knowledge, this is the first replication study of CeD variants in a non-European population.
Collapse
Affiliation(s)
| | - Javier Gutierrez-Achury
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana, India
| | - Vandana Midha
- Department of Medicine, Dayanand Medical College and Hospital, Ludhiana, India
| | - Agata Szperl
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Jihane Romanos
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Santos Alonso
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Leioa, Spain
| | - B K Thelma
- Department of Genetics, University of Delhi South Campus, New Delhi, India
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Gosia Trynka
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| |
Collapse
|
141
|
Bentham J, Vyse TJ. The development of genome-wide association studies and their application to complex diseases, including lupus. Lupus 2014; 22:1205-13. [PMID: 24097992 DOI: 10.1177/0961203313492870] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In this review, we explain the motivation for carrying out genome-wide association studies (GWAS), contrasting the achievements of linkage-based experiments for Mendelian traits with the difficulties found when applying that type of experiment to complex diseases. We explain the technical and organizational developments that were required to make GWAS feasible, as well as some of the theoretical concerns that were raised during the design of these studies. We describe the impressive achievements of GWAS in lupus, and compare them with the experiences in three other genetically complex disorders: rheumatoid arthritis, type 1 diabetes and coronary heart disease. GWAS have been successful in identifying many new susceptibility loci for these four diseases, and have provided the motivation for novel immunological work. We conclude by describing preliminary steps that have been taken towards translating the results of GWAS into improvements in patient care, explaining some of the difficulties involved, as well as successes that have already been achieved.
Collapse
Affiliation(s)
- J Bentham
- Medical & Molecular Genetics, King's College London, UK
| | | |
Collapse
|
142
|
Protein kinase C inhibitors for immune disorders. Drug Discov Today 2014; 19:1217-21. [PMID: 24892801 DOI: 10.1016/j.drudis.2014.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/14/2014] [Indexed: 12/11/2022]
Abstract
Protein kinase C (PKC) proteins are a group of well-conserved, intracellular signaling enzymes expressed in all cells and tissues, including immune cells. Much of the molecular insight into PKC immunobiology has been gleaned from studies using PKC gene (Prkc) knockout mice and the analysis of different disease models in these animals. More-recent studies have revealed that PKCs also have crucial roles in the pathogenesis of human immune disorders. Therefore, strategies to modulate the functions of PKC enzymes could have a major impact on the treatment and therapies of autoimmune diseases and other immune disorders.
Collapse
|
143
|
Jiang L, Yin J, Ye L, Yang J, Hemani G, Liu AJ, Zou H, He D, Sun L, Zeng X, Li Z, Zheng Y, Lin Y, Liu Y, Fang Y, Xu J, Li Y, Dai S, Guan J, Jiang L, Wei Q, Wang Y, Li Y, Huang C, Zuo X, Liu Y, Wu X, Zhang L, Zhou L, Zhang Q, Li T, Chen L, Xu Z, Yang X, Qian F, Xie W, Liu W, Guo Q, Huang S, Zhao J, Li M, Jin Y, Gao J, Lv Y, Wang Y, Lin L, Guo A, Danoy P, Willner D, Cremin C, Hadler J, Zhang F, Zhao Y, Li M, Yue T, Fan X, Guo J, Mu R, Li J, Wu C, Zeng M, Wang J, Li S, Jin L, Wang B, Wang J, Ma X, Sun L, Zhang X, Brown MA, Visscher PM, Su DF, Xu H. Novel Risk Loci for Rheumatoid Arthritis in Han Chinese and Congruence With Risk Variants in Europeans. Arthritis Rheumatol 2014; 66:1121-32. [PMID: 24782177 DOI: 10.1002/art.38353] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 01/02/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Lei Jiang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Jian Yin
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Lingying Ye
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Jian Yang
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Gibran Hemani
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Ai-jun Liu
- Second Military Medical University; Shanghai China
| | - Hejian Zou
- Huashan Hospital and Fudan University; Shanghai China
| | | | | | - Xiaofeng Zeng
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College; Beijing China
| | - Zhanguo Li
- People's Hospital and Beijing University; Beijing China
| | - Yi Zheng
- Beijing Chaoyang Hospital and Capital Medical University; Beijing China
| | - Yiping Lin
- 202nd Hospital of People's Liberation Army; Shenyang China
| | - Yi Liu
- West China Hospital and Sichuan University; Chengdu China
| | - Yongfei Fang
- Southwest Hospital and Third Military Medical University; Chongqing China
| | - Jianhua Xu
- First Affiliated Hospital of Anhui Medical University; Hefei China
| | - Yinong Li
- People's Hospital of Fujian Province and Fujian University of Traditional Chinese Medicine; Fuzhou China
| | - Shengming Dai
- Shanghai Changhai Hospital and Second Military Medical University; Shanghai China
| | - Jianlong Guan
- Huadong Hospital and Fudan University; Shanghai China
| | - Lindi Jiang
- Zhongshan Hospital and Fudan University; Shanghai China
| | - Qianghua Wei
- Shanghai First People's Hospital Affiliated with Shanghai Jiaotong University; Shanghai China
| | - Yi Wang
- Lanzhou University Second Hospital; Lanzhou China
| | - Yang Li
- Second Affiliated Hospital of Harbin Medical University; Harbin China
| | | | - Xiaoxia Zuo
- Xiangya Hospital and Central South University; Changsha China
| | - Yu Liu
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Xin Wu
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Libin Zhang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Ling Zhou
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Qing Zhang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Ting Li
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Ling Chen
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Zhen Xu
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Xiaoping Yang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Feng Qian
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Weilin Xie
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Wei Liu
- Shanghai Changzheng Hospital and Second Military Medical University, Shanghai, China, and 442nd Hospital of Fuzhou General Hospital, Nanjing Military Region; Ningde China
| | - Qian Guo
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Shaolan Huang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Jing Zhao
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Mengmeng Li
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Yanhua Jin
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Jie Gao
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Ying Lv
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Yiwen Wang
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Li Lin
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Aihua Guo
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| | - Patrick Danoy
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Dana Willner
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Catherine Cremin
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Johanna Hadler
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Fengchun Zhang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College; Beijing China
| | - Yan Zhao
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College; Beijing China
| | - Mengtao Li
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College; Beijing China
| | - Tao Yue
- Guanghua Hospital; Shanghai China
| | | | - Jianping Guo
- People's Hospital and Beijing University; Beijing China
| | - Rong Mu
- People's Hospital and Beijing University; Beijing China
| | - Jingyi Li
- Southwest Hospital and Third Military Medical University; Chongqing China
| | - Chao Wu
- Third Military Medical University; Chongqing China
| | - Ming Zeng
- National Institute for Food and Drug Control; Beijing China
| | | | | | - Li Jin
- Fudan University; Shanghai China
| | - Binbin Wang
- National Research Institute for Family Planning; Beijing China
| | - Jing Wang
- National Research Institute for Family Planning and Capital Medical University; Beijing China
| | - Xu Ma
- National Research Institute for Family Planning; Beijing China
| | | | | | - Matthew A. Brown
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Peter M. Visscher
- Princess Alexandra Hospital and University of Queensland, Brisbane; Queensland Australia
| | - Ding-feng Su
- Second Military Medical University; Shanghai China
| | - Huji Xu
- Shanghai Changzheng Hospital and Second Military Medical University; Shanghai China
| |
Collapse
|
144
|
Hartkamp LM, Fine JS, van Es IE, Tang MW, Smith M, Woods J, Narula S, DeMartino J, Tak PP, Reedquist KA. Btk inhibition suppresses agonist-induced human macrophage activation and inflammatory gene expression in RA synovial tissue explants. Ann Rheum Dis 2014; 74:1603-11. [PMID: 24764451 DOI: 10.1136/annrheumdis-2013-204143] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 04/06/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Bruton's tyrosine kinase (Btk) is required for B lymphocyte and myeloid cell contributions to pathology in murine models of arthritis. Here, we examined the potential contributions of synovial Btk expression and activation to inflammation in rheumatoid arthritis (RA). MATERIALS AND METHODS Btk was detected by immunohistochemistry and digital image analysis in synovial tissue from biologically naive RA (n=16) and psoriatic arthritis (PsA) (n=12) patients. Cell populations expressing Btk were identified by immunofluorescent double labelling confocal microscopy, quantitative (q-) PCR and immunoblotting. The effects of a Btk-specific inhibitor, RN486, on gene expression in human macrophages and RA synovial tissue explants (n=8) were assessed by qPCR, ELISA and single-plex assays. RESULTS Btk was expressed at equivalent levels in RA and PsA synovial tissue, restricted to B lymphocytes, monocytes, macrophages and mast cells. RN486 significantly inhibited macrophage IL-6 production induced by Fc receptor and CD40 ligation. RN486 also reduced mRNA expression of overlapping gene sets induced by IgG, CD40 ligand (CD40L) and RA synovial fluid, and significantly suppressed macrophage production of CD40L-induced IL-8, TNF, MMP-1 and MMP-10, LPS-induced MMP-1, MMP-7 and MMP-10 production, and spontaneous production of IL-6, PDGF, CXCL-9 and MMP-1 by RA synovial explants. CONCLUSIONS Btk is expressed equivalently in RA and PsA synovial tissue, primarily in macrophages. Btk activity is needed to drive macrophage activation in response to multiple agonists relevant to inflammatory arthritis, and promotes RA synovial tissue cytokine and MMP production. Pharmacological targeting of Btk may be of therapeutic benefit in the treatment of RA and other inflammatory diseases.
Collapse
Affiliation(s)
- Linda M Hartkamp
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jay S Fine
- Department of Inflammation Discovery, Hoffmann-La Roche, Inc., Nutley, New Jersey, USA
| | - Inge E van Es
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Man Wai Tang
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael Smith
- Department of Inflammation Discovery, Hoffmann-La Roche, Inc., Nutley, New Jersey, USA
| | - John Woods
- Department of Inflammation Discovery, Hoffmann-La Roche, Inc., Nutley, New Jersey, USA
| | - Satwant Narula
- Department of Inflammation Discovery, Hoffmann-La Roche, Inc., Nutley, New Jersey, USA
| | - Julie DeMartino
- Department of Inflammation Discovery, Hoffmann-La Roche, Inc., Nutley, New Jersey, USA
| | - Paul P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands GlaxoSmithKline, Stevenage, and University of Cambridge, Cambridge, UK
| | - Kris A Reedquist
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
145
|
Han B, Diogo D, Eyre S, Kallberg H, Zhernakova A, Bowes J, Padyukov L, Okada Y, González-Gay M, Rantapää-Dahlqvist S, Martin J, Huizinga T, Plenge R, Worthington J, Gregersen P, Klareskog L, de Bakker P, Raychaudhuri S. Fine mapping seronegative and seropositive rheumatoid arthritis to shared and distinct HLA alleles by adjusting for the effects of heterogeneity. Am J Hum Genet 2014; 94:522-32. [PMID: 24656864 DOI: 10.1016/j.ajhg.2014.02.013] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/24/2014] [Indexed: 11/25/2022] Open
Abstract
Despite progress in defining human leukocyte antigen (HLA) alleles for anti-citrullinated-protein-autoantibody-positive (ACPA(+)) rheumatoid arthritis (RA), identifying HLA alleles for ACPA-negative (ACPA(-)) RA has been challenging because of clinical heterogeneity within clinical cohorts. We imputed 8,961 classical HLA alleles, amino acids, and SNPs from Immunochip data in a discovery set of 2,406 ACPA(-) RA case and 13,930 control individuals. We developed a statistical approach to identify and adjust for clinical heterogeneity within ACPA(-) RA and observed independent associations for serine and leucine at position 11 in HLA-DRβ1 (p = 1.4 × 10(-13), odds ratio [OR] = 1.30) and for aspartate at position 9 in HLA-B (p = 2.7 × 10(-12), OR = 1.39) within the peptide binding grooves. These amino acid positions induced associations at HLA-DRB1(∗)03 (encoding serine at 11) and HLA-B(∗)08 (encoding aspartate at 9). We validated these findings in an independent set of 427 ACPA(-) case subjects, carefully phenotyped with a highly sensitive ACPA assay, and 1,691 control subjects (HLA-DRβ1 Ser11+Leu11: p = 5.8 × 10(-4), OR = 1.28; HLA-B Asp9: p = 2.6 × 10(-3), OR = 1.34). Although both amino acid sites drove risk of ACPA(+) and ACPA(-) disease, the effects of individual residues at HLA-DRβ1 position 11 were distinct (p < 2.9 × 10(-107)). We also identified an association with ACPA(+) RA at HLA-A position 77 (p = 2.7 × 10(-8), OR = 0.85) in 7,279 ACPA(+) RA case and 15,870 control subjects. These results contribute to mounting evidence that ACPA(+) and ACPA(-) RA are genetically distinct and potentially have separate autoantigens contributing to pathogenesis. We expect that our approach might have broad applications in analyzing clinical conditions with heterogeneity at both major histocompatibility complex (MHC) and non-MHC regions.
Collapse
|
146
|
Shao S, Li XR, Cen H, Yin ZS. Association of AIRE polymorphisms with genetic susceptibility to rheumatoid arthritis in a Chinese population. Inflammation 2014; 37:495-9. [PMID: 24170308 DOI: 10.1007/s10753-013-9763-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recently, genetic polymorphisms within the autoimmune regulator (AIRE) have been implicated in the genetic susceptibility to rheumatoid arthritis (RA) in Japanese and Spanish. The aim of this case-control study involving 232 patients with RA and 313 ethnically matched control subjects was to investigate the association of AIRE rs2075876 and rs760426 polymorphisms with genetic predisposition to RA in a Chinese population. The genotypes of AIRE rs2075876 and rs760426 polymorphisms were determined by SNaPshot assay. A significant difference in the allele frequency of AIRE rs2075876 polymorphism between cases and controls was detected (A versus G, OR 1.33, 95 %CI 1.04-1.69, P = 0.02, P corrected (Bonferroni correction) Pc = 0.04). Significant evidence was found for the association between the minor allele A of AIRE rs2075876 polymorphism and the risk of RA under the recessive model (AA versus AG + GG, P = 7.15 × 10(-3), Pc = 1.43 × 10(-2)). The frequency of the minor allele G of AIRE rs760426 polymorphism was higher in patients compared with controls (47.8 % versus 42.1 %), and this deviation showed a trend towards significant level (P = 0.06, Pc = 0.12). The association between the minor allele G of AIRE rs760426 polymorphism with RA risk under the dominant model and the recessive model revealed that significant evidence was detected under the recessive model (GG versus GA + AA, P = 0.02, Pc = 0.04). Our results indicated that AIRE rs2075876 and rs760426 polymorphisms were involved in the genetic background of RA in the Chinese population.
Collapse
Affiliation(s)
- Song Shao
- Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | | | | | | |
Collapse
|
147
|
Sparks JA, Chen CY, Jiang X, Askling J, Hiraki LT, Malspeis S, Klareskog L, Alfredsson L, Costenbader KH, Karlson EW. Improved performance of epidemiologic and genetic risk models for rheumatoid arthritis serologic phenotypes using family history. Ann Rheum Dis 2014; 74:1522-9. [PMID: 24685909 DOI: 10.1136/annrheumdis-2013-205009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/16/2014] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To develop and validate rheumatoid arthritis (RA) risk models based on family history, epidemiologic factors and known genetic risk factors. METHODS We developed and validated models for RA based on known RA risk factors, among women in two cohorts: the Nurses' Health Study (NHS, 381 RA cases and 410 controls) and the Epidemiological Investigation of RA (EIRA, 1244 RA cases and 971 controls). Model discrimination was evaluated using the area under the receiver operating characteristic curve (AUC) in logistic regression models for the study population and for those with positive family history. The joint effect of family history with genetics, smoking and body mass index (BMI) was evaluated using logistic regression models to estimate ORs for RA. RESULTS The complete model including family history, epidemiologic risk factors and genetics demonstrated AUCs of 0.74 for seropositive RA in NHS and 0.77 for anti-citrullinated protein antibody (ACPA)-positive RA in EIRA. Among women with positive family history, discrimination was excellent for complete models for seropositive RA in NHS (AUC 0.82) and ACPA-positive RA in EIRA (AUC 0.83). Positive family history, high genetic susceptibility, smoking and increased BMI had an OR of 21.73 for ACPA-positive RA. CONCLUSIONS We developed models for seropositive and seronegative RA phenotypes based on family history, epidemiological and genetic factors. Among those with positive family history, models using epidemiologic and genetic factors were highly discriminatory for seropositive and seronegative RA. Assessing epidemiological and genetic factors among those with positive family history may identify individuals suitable for RA prevention strategies.
Collapse
Affiliation(s)
- Jeffrey A Sparks
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chia-Yen Chen
- Harvard School of Public Health and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xia Jiang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Askling
- Rheumatology Unit, Department of Medicine, Karolinska Institutet/Karolinska Hospital, Stockholm, Sweden
| | - Linda T Hiraki
- Research Institute, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Susan Malspeis
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet/Karolinska Hospital, Stockholm, Sweden
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Karen H Costenbader
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Elizabeth W Karlson
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
148
|
Hebbring SJ. The challenges, advantages and future of phenome-wide association studies. Immunology 2014; 141:157-65. [PMID: 24147732 PMCID: PMC3904236 DOI: 10.1111/imm.12195] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/11/2022] Open
Abstract
Over the last decade, significant technological breakthroughs have revolutionized human genomic research in the form of genome-wide association studies (GWASs). GWASs have identified thousands of statistically significant genetic variants associated with hundreds of human conditions including many with immunological aetiologies (e.g. multiple sclerosis, ankylosing spondylitis and rheumatoid arthritis). Unfortunately, most GWASs fail to identify clinically significant associations. Identifying biologically significant variants by GWAS also presents a challenge. The GWAS is a phenotype-to-genotype approach. As a complementary/alternative approach to the GWAS, investigators have begun to exploit extensive electronic medical record systems to conduct a genotype-to-phenotype approach when studying human disease – specifically, the phenome-wide association study (PheWAS). Although the PheWAS approach is in its infancy, this method has already demonstrated its capacity to rediscover important genetic associations related to immunological diseases/conditions. Furthermore, PheWAS has the advantage of identifying genetic variants with pleiotropic properties. This is particularly relevant for HLA variants. For example, PheWAS results have demonstrated that the HLA-DRB1 variant associated with multiple sclerosis may also be associated with erythematous conditions including rosacea. Likewise, PheWAS has demonstrated that the HLA-B genotype is not only associated with spondylopathies, uveitis, and variability in platelet count, but may also play an important role in other conditions, such as mastoiditis. This review will discuss and compare general PheWAS methodologies, describe both the challenges and advantages of the PheWAS, and provide insight into the potential directions in which PheWAS may lead.
Collapse
Affiliation(s)
- Scott J Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| |
Collapse
|
149
|
Medici M, Porcu E, Pistis G, Teumer A, Brown SJ, Jensen RA, Rawal R, Roef GL, Plantinga TS, Vermeulen SH, Lahti J, Simmonds MJ, Husemoen LLN, Freathy RM, Shields BM, Pietzner D, Nagy R, Broer L, Chaker L, Korevaar TIM, Plia MG, Sala C, Völker U, Richards JB, Sweep FC, Gieger C, Corre T, Kajantie E, Thuesen B, Taes YE, Visser WE, Hattersley AT, Kratzsch J, Hamilton A, Li W, Homuth G, Lobina M, Mariotti S, Soranzo N, Cocca M, Nauck M, Spielhagen C, Ross A, Arnold A, van de Bunt M, Liyanarachchi S, Heier M, Grabe HJ, Masciullo C, Galesloot TE, Lim EM, Reischl E, Leedman PJ, Lai S, Delitala A, Bremner AP, Philips DIW, Beilby JP, Mulas A, Vocale M, Abecasis G, Forsen T, James A, Widen E, Hui J, Prokisch H, Rietzschel EE, Palotie A, Feddema P, Fletcher SJ, Schramm K, Rotter JI, Kluttig A, Radke D, Traglia M, Surdulescu GL, He H, Franklyn JA, Tiller D, Vaidya B, de Meyer T, Jørgensen T, Eriksson JG, O'Leary PC, Wichmann E, Hermus AR, Psaty BM, Ittermann T, Hofman A, Bosi E, Schlessinger D, Wallaschofski H, Pirastu N, Aulchenko YS, de la Chapelle A, Netea-Maier RT, Gough SCL, Meyer zu Schwabedissen H, Frayling TM, Kaufman JM, Linneberg A, Räikkönen K, Smit JWA, Kiemeney LA, Rivadeneira F, Uitterlinden AG, Walsh JP, Meisinger C, den Heijer M, Visser TJ, Spector TD, Wilson SG, Völzke H, Cappola A, Toniolo D, Sanna S, Naitza S, Peeters RP. Identification of novel genetic Loci associated with thyroid peroxidase antibodies and clinical thyroid disease. PLoS Genet 2014; 10:e1004123. [PMID: 24586183 PMCID: PMC3937134 DOI: 10.1371/journal.pgen.1004123] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 12/03/2013] [Indexed: 12/14/2022] Open
Abstract
Autoimmune thyroid diseases (AITD) are common, affecting 2-5% of the general population. Individuals with positive thyroid peroxidase antibodies (TPOAbs) have an increased risk of autoimmune hypothyroidism (Hashimoto's thyroiditis), as well as autoimmune hyperthyroidism (Graves' disease). As the possible causative genes of TPOAbs and AITD remain largely unknown, we performed GWAS meta-analyses in 18,297 individuals for TPOAb-positivity (1769 TPOAb-positives and 16,528 TPOAb-negatives) and in 12,353 individuals for TPOAb serum levels, with replication in 8,990 individuals. Significant associations (P<5×10(-8)) were detected at TPO-rs11675434, ATXN2-rs653178, and BACH2-rs10944479 for TPOAb-positivity, and at TPO-rs11675434, MAGI3-rs1230666, and KALRN-rs2010099 for TPOAb levels. Individual and combined effects (genetic risk scores) of these variants on (subclinical) hypo- and hyperthyroidism, goiter and thyroid cancer were studied. Individuals with a high genetic risk score had, besides an increased risk of TPOAb-positivity (OR: 2.18, 95% CI 1.68-2.81, P = 8.1×10(-8)), a higher risk of increased thyroid-stimulating hormone levels (OR: 1.51, 95% CI 1.26-1.82, P = 2.9×10(-6)), as well as a decreased risk of goiter (OR: 0.77, 95% CI 0.66-0.89, P = 6.5×10(-4)). The MAGI3 and BACH2 variants were associated with an increased risk of hyperthyroidism, which was replicated in an independent cohort of patients with Graves' disease (OR: 1.37, 95% CI 1.22-1.54, P = 1.2×10(-7) and OR: 1.25, 95% CI 1.12-1.39, P = 6.2×10(-5)). The MAGI3 variant was also associated with an increased risk of hypothyroidism (OR: 1.57, 95% CI 1.18-2.10, P = 1.9×10(-3)). This first GWAS meta-analysis for TPOAbs identified five newly associated loci, three of which were also associated with clinical thyroid disease. With these markers we identified a large subgroup in the general population with a substantially increased risk of TPOAbs. The results provide insight into why individuals with thyroid autoimmunity do or do not eventually develop thyroid disease, and these markers may therefore predict which TPOAb-positives are particularly at risk of developing clinical thyroid dysfunction.
Collapse
Affiliation(s)
- Marco Medici
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- * E-mail:
| | - Eleonora Porcu
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Universita di Sassari, Sassari, Italy
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Suzanne J. Brown
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Richard A. Jensen
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington, United States of America
| | - Rajesh Rawal
- Institute for Genetic Epidemiology, Helmholtz Zentrum Munich, Munich/Neuherberg, Germany
| | - Greet L. Roef
- Department of Endocrinology and Internal Medicine, University Hospital Ghent and Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Theo S. Plantinga
- Internal Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Sita H. Vermeulen
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Matthew J. Simmonds
- Oxford Centre for Diabetes, Endocrinology and Metabolism and NIHR Oxford Biomedical Research Centre, Oxford, UK Churchill Hospital, Headington, Oxford, United Kingdom
| | - Lise Lotte N. Husemoen
- Research Centre for Prevention and Health, Glostrup University Hospital, the Capital Region of Denmark, Glostrup, Denmark
| | - Rachel M. Freathy
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Beverley M. Shields
- Peninsula NIHR Clinical Research Facility, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Diana Pietzner
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Rebecca Nagy
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Linda Broer
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Layal Chaker
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tim I. M. Korevaar
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maria Grazia Plia
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - J. Brent Richards
- Departments of Medicine, Human Genetics, Epidemiology and Biostatistics, Lady Davis Institute, McGill University, Montreal, Canada
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Fred C. Sweep
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Christian Gieger
- Institute for Genetic Epidemiology, Helmholtz Zentrum Munich, Munich/Neuherberg, Germany
| | - Tanguy Corre
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki, Finland
- Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Betina Thuesen
- Research Centre for Prevention and Health, Glostrup University Hospital, the Capital Region of Denmark, Glostrup, Denmark
| | - Youri E. Taes
- Department of Endocrinology and Internal Medicine, University Hospital Ghent and Faculty of Medicine, Ghent University, Ghent, Belgium
| | - W. Edward Visser
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrew T. Hattersley
- Peninsula NIHR Clinical Research Facility, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Alexander Hamilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism and NIHR Oxford Biomedical Research Centre, Oxford, UK Churchill Hospital, Headington, Oxford, United Kingdom
| | - Wei Li
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Monia Lobina
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Stefano Mariotti
- Dipartimento di Scienze Biomediche, Universita di Sassari, Sassari, Italy
| | | | - Massimiliano Cocca
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christin Spielhagen
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Alec Ross
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Alice Arnold
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology and Metabolism and NIHR Oxford Biomedical Research Centre, Oxford, UK Churchill Hospital, Headington, Oxford, United Kingdom
| | - Sandya Liyanarachchi
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Margit Heier
- Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Institute of Epidemiology II, Neuherberg, Germany
| | - Hans Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, HELIOS Hospital Stralsund, Greifswald, Germany
| | - Corrado Masciullo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Tessel E. Galesloot
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ee M. Lim
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia, Australia
| | - Eva Reischl
- Research Unit of Molecular Epidemiology Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peter J. Leedman
- School of Medicine and Pharmacology, the University of Western Australia, Crawley, Western Australia, Australia
- UWA Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Western Australia, Australia
| | - Sandra Lai
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | | | - Alexandra P. Bremner
- School of Population Health, University of Western Australia, Nedlands, Western Australia, Australia
| | - David I. W. Philips
- MRC Lifecourse Epidemiology Unit, Southampton General Hospital, Southampton, United Kingdom
| | - John P. Beilby
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Matteo Vocale
- High Performance Computing and Network, CRS4, Parco Tecnologico della Sardegna, Pula, Italy
| | - Goncalo Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tom Forsen
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Vaasa Health Care Centre, Diabetes Unit, Vaasa, Finland
| | - Alan James
- School of Medicine and Pharmacology, the University of Western Australia, Crawley, Western Australia, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jennie Hui
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia, Australia
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum Munich, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Ernst E. Rietzschel
- Department of Cardiology and Internal Medicine, University Hospital Ghent and Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | | | | | - Katharina Schramm
- Institute of Human Genetics, Helmholtz Zentrum Munich, Munich, Germany
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Torrance, California, United States of America
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Alexander Kluttig
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Dörte Radke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Gabriela L. Surdulescu
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Huiling He
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Jayne A. Franklyn
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, Univeristy of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Daniel Tiller
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Bijay Vaidya
- Diabetes, Endocrinology and Vascular Health Centre, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Tim de Meyer
- BIOBIX Lab. for Bioinformatics and Computational Genomics, Dept. of Mathematical Modelling, Statistics and Bioinformatics. Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup University Hospital, the Capital Region of Denmark, Glostrup, Denmark
- Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Johan G. Eriksson
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
- Vasa Central Hospital, Vasa, Finland
| | - Peter C. O'Leary
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin University of Technology, Bentley, Western Australia, Australia
| | - Eric Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum Munich, Munich, Germany
| | - Ad R. Hermus
- Internal Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emanuele Bosi
- Department of Internal Medicine, Diabetes & Endocrinology Unit, San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Henri Wallaschofski
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Nicola Pirastu
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
- University of Trieste, Trieste, Italy
| | - Yurii S. Aulchenko
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Albert de la Chapelle
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Romana T. Netea-Maier
- Internal Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Stephen C. L. Gough
- Oxford Centre for Diabetes, Endocrinology and Metabolism and NIHR Oxford Biomedical Research Centre, Oxford, UK Churchill Hospital, Headington, Oxford, United Kingdom
| | | | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Jean-Marc Kaufman
- Department of Endocrinology and Internal Medicine, University Hospital Ghent and Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup University Hospital, the Capital Region of Denmark, Glostrup, Denmark
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Johannes W. A. Smit
- Internal Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Lambertus A. Kiemeney
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Aging, Netherlands Genomics Initiative, Leiden, The Netherlands
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Aging, Netherlands Genomics Initiative, Leiden, The Netherlands
| | - John P. Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, the University of Western Australia, Crawley, Western Australia, Australia
| | - Christa Meisinger
- Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Institute of Epidemiology II, Neuherberg, Germany
| | - Martin den Heijer
- Department of Internal Medicine, VU Medical Center, Amsterdam, The Netherlands
| | - Theo J. Visser
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Scott G. Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, the University of Western Australia, Crawley, Western Australia, Australia
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anne Cappola
- Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Institute of Molecular Genetics-CNR, Pavia, Italy
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Robin P. Peeters
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
150
|
Stahelova A, Petrkova J, Petrek M, Mrazek F. Sequence variation in promoter regions of genes for CC chemokine ligands (CCL)19 and 21 in Czech patients with myocardial infarction. Mol Biol Rep 2014; 41:3163-8. [PMID: 24493450 DOI: 10.1007/s11033-014-3175-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/16/2014] [Indexed: 11/26/2022]
Abstract
Recruitment of inflammatory cells to the arterial wall is an important pathogenic mechanism of atherosclerosis and coronary artery disease (CAD). Functional variability in the genes encoding for chemokines that promote infiltration of atherosclerotic plaques by macrophages and lymphocytes may therefore contribute to the genetic susceptibility to CAD. We, therefore, investigated the association between myocardial infarction (MI) and polymorphisms in the promoter regions of the chemokine genes CCL19 and CCL21. Based on re-sequencing screening we selected and, using PCR-SSP, determined three polymorphisms of CCL19 gene (GenBank ID rs2233872) and CCL21 gene (GenBank ID rs11574914 and rs11574915) in 211 Czech patients with MI and 150 healthy control subjects. There was no difference in allelic frequencies of the investigated SNPs between patients and controls (p>0.05). However, the proportion of homozygotes for the minor G allele of the CCL21 promoter variant (rs11574915 GG) was lower among the MI patients (1%) in comparison with the control subjects (5%, nominal p=0.03). Though rare in the Czech population, CCL21 (rs11574915) GG genotype may confer protection from myocardial infarction. Our preliminary data have to be independently replicated.
Collapse
Affiliation(s)
- Anna Stahelova
- Laboratory of Immunogenomics & Immunoproteomics, Department of Immunology, Faculty of Medicine & Dentistry, Palacky University Olomouc, I. P. Pavlova Str. 6, 775 20, Olomouc, Czech Republic
| | | | | | | |
Collapse
|