Minireviews Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Orthop. Sep 18, 2014; 5(4): 544-549
Published online Sep 18, 2014. doi: 10.5312/wjo.v5.i4.544
Rheumatoid arthritis susceptibility genes: An overview
Izabela Korczowska, Department of Rheumatology and Clinical Immunology, University of Medical Sciences in Poznan, 61-701 Poznan, Poland
Author contributions: Korczowska I solely contributed to this paper.
Correspondence to: Dr. Izabela Korczowska, Department of Rheumatology and Clinical Immunology, University of Medical Sciences in Poznan, Fredry 10, 61-701 Poznan, Poland. ikorcz@post.pl
Telephone: +48-61-8547210 Fax: +48-61-8547212
Received: January 22, 2014
Revised: May 29, 2014
Accepted: June 14, 2014
Published online: September 18, 2014

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.

Key Words: Rheumatoid arthritis, Gene, Polymorphism, Human leukocyte antigen, Genome wide association study

Core tip: This is a comprehensive review concerning genetic factors in rheumatoid arthritis.



INTRODUCTION

Rheumatoid arthritis (RA) is the most common autoimmune disease, afflicting around 0.5%-2% of the human population, especially females, but the precise etiology is still unknown. RA is characterized by chronic, systemic inflammation that may affect many tissues, principally synovial tissue, leading to joint destruction, functional disability and sometimes death[1]. Environmental and genetic factors are responsible for susceptibility and the phenotype. Environmental factors include geography, climate, endemic microbes and lifestyle, such as smoking and diet[2,3]. Native Americans show a relatively higher incidence than African or Asian populations. Familial clustering is important, with the prevalence of RA ranging from 2% to 12% in first degree relatives of patients, 5%-10% in same sex dizygotic twins and almost 12%-30% in monozygotic twins[3,4].

The human leukocyte antigen (HLA) region in the human genome is the most heterogeneous and many diseases are known to be associated with this region. The first risk alleles for RA were identified within 36Mb, the major histocompatibility complex (MHC) region[3]. Several studies beginning in the 1980s explained the strong association of the HLA-DRB1 alleles with RA. The associated alleles encode five amino acids at position 70-74 of the HLA-DRβ1 chain, which is known as a shared epitope (SE). It was established that the HLA-DRB1*01, HLA-DRB1*04 and HLADRB1*10 alleles containing the SE were associated with susceptibility to RA and amino acid sequences QKRAA, QQRAA and KKRAA were known SEs conferring susceptibility, while DERAA sequences were for protective effects[5,6]. Caucasian RA patients have been tested for ACPA antibodies, RF and HLA-DR genotype, and the results showed a correlation between the presence of RF and ACPA antibodies within the HLA-DRB1 SE[3,7]. Moreover, current smoking habits and SE, especially homozygous SE, have a strong interaction[3,8]. SE is a risk factor for the development of an extra-articular manifestation and so for more severe, destructive RA. However, the non-SE alleles DRB1*1301, *1302 and *1304 seem to be linked to the DERAA motif[9-11]. The study in Hungarian RA patients recommended that HLA-DRB *1301 allele may protect against ACPA positive or ACPA negative RA[9,12-15]. Also, enhanced production of ACPA has been connected with HLA-DRB1*15 positively in RA[9,16-18]. In a Korean population, heterozygous for HLADRB1 0404 or 0901 have up to a 60-fold increased risk of developing susceptibility to RA[19].

A new taxonomy system for the risk of developing RA has been proposed[9,11]. This new classification depends on whether the RAA (motif which represents susceptibility risk of RA) sequence occupies position 71-74 of HLA-DRB1 but is modulated by amino acids at positions 70; glutamine (Q) and arginine (R) represent a higher risk than aspartic acid (D). Lysine (K) confers the highest risk, arginine (R) intermediate risk and the lowest risk is for alanine (A) and glutamic acid (E) in position 71. According to this new classification, SE alleles are divided into S1, S2, S3P and S3D groups and allele X which denotes all non-RAA motifs. The presence of S2 and S3P alleles are a positive association with RA and also correlated with ACPA production, while S1, S3D and X were found to be low risk alleles[9,11,20].

Genome wide association studies (GWAS), large scale cohorts and Wellcome Trust Case Control Consortium databases have allowed the simultaneous evaluation of thousands of genes[9,21-23] and drawn attention to association with RA susceptibility, determining the phenotype of the disease, and response to therapy. Additional variants in the MHC contribute to the heritability of RA independently of the HLA-DRB1, leading to more consequent results of genetic associations. Alleles associated with the susceptibility with RA according to the GWAS study are shown in Table 1. Loci outside the MHC have been associated in a RA population in approximately 4% to the phenotypic variance of RA risk. One of them is peptidyl arginine deiminase, type IV (PADI4) encoding peptidylarginine deiminase type IV.

Table 1 The most relevant alleles associated with susceptibility in rheumatoid arthritis according to genome wide association studies.
Gene candidateLocusSNPOR
HLA-DRB1
PTPN221Rs24766011.23-1.75
PADI41Rs 2240340a1.4
STAT4 T/C2Rs11889341.22 (0.98-1.53)
FCGR2A1Rs127466131.1
CTLA42Rs30872430.75-1.136
CCL219Rs28123781.1
TRAF19Rs37618471.1 (0.97-1.32)
IRF57Rs104886311.16 (0.72-1.87)
CCR66Rs30930230.79 (0.64-0.98)
CD4020Rs48104850.91-1.02
IL2RA10Rs21042860.92
PADI4

One of the isoenzymes carrying the post-translational conversion of arginine residues to citrulline is known as the type 4 peptidylarginine deiminase type IV. PADI4 enzyme may be connected to the production of ACPA. PADI4 is present in bone marrow and peripheral blood leukocytes and is one of the four isoforms of PADI enzyme in humans encoded by the PADI4 gene[3,24]. PADI4 gene maps on 1p36 locus have been associated with European and Japanese RA populations. A meta-analysis done by Lee et al[25] showed that in Asian patients, all 5 researched polymorphisms (PADI4_89, PADI4_90, PADI4_93, PADI4_94 and PADI4_104) were significantly associated with RA, while in Europeans only PADI4_94 was associated with RA risk, much less than in Asian patients[26,27]. The function of this gene in the European RA population is still questionable as the results of large studies from Spain, France and the UK found no association with RA[3,28,29].

Within the genes investigated for susceptibility to RA, protein tyrosine phosphatase type 22 (PTN22) is one of the most strongly associated.

Protein tyrosine phosphatase, nonreceptor type 22

Protein tyrosine phosphatase, nonreceptor type 22 (PTPN22) encodes the intracellular tyrosine phosphatase LYP, known as a powerful inhibitor of T-cell activation. The gene encoding PTPN22 shows the second strongest (just after HLA-DRB1) association with RA. The gene was first associated with type 1 diabetes, systemic sclerosis, Graves disease and lupus erythematosus. Then it was associated with RA in a Caucasian population; rs2476601, C1885T polymorphism leading to an amino acid modification from Arg to Trp at amino acid position 620. This polymorphism resides in a rather large haplotype block encompassing the entire PTPN22 gene[3,30-32]. This SNP has been associated with RF, ACPA positive and SE. ACPA status powerfully supports the early diagnosis of RA. It is worth mentioning that in contrast to SE, C1885T polymorphism may not be associated with smoking[9,33-35]. The important fact is that this polymorphism is not associated with RA in Asian populations, maybe only with Asiatic Indians with RA positive[36].

Signal transducer and activator of transcription 4

The signal transducer and activator of transcription 4 (STAT4) is a transcription factor that intercedes the intracellular signal activation by cytokines such IL-12, IL-23 and IL-27 and type I interferons. STAT4 can be induced upon activation and maturation of monocytes as well as immature dendritic cells. STAT is also overexpressed in RA synovium. Lee et al[37] and Amos et al[38] detected linkage at chromosome 2q33 in RA and then revealed that the polymorphism located at 2q33 STAT4 gene is the marker responsible for the linkage signal in 2q33. It has been found that STAT4 rs7574865 polymorphism is associated with European, Asian and Latin American RA patients[37,39-43]. Comparison between ACPA positive and negative patients showed no significant differences[37]. It seems that the intronic variant rs11893432 C/G of STAT4 gene could also predispose to RA[26,44].

Cytotoxic T lymphocyte-associated antigen 4

Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is expressed on T cells, is a member of the immunoglobulin superfamily and performs a critical role in the inhibition of T-cell activation and peripheral tolerance. Three polymorphisms have been described in the CTLA-4 gene: first, microsatellite at position 642 of the 3’ untranslated region of exon 4; second, the polymorphism 49G/A in exon 1 causes a threonine to the alanine substitution of amino acid 17; and the third, -318 of the promoter sequence C/T transition[45]. The CT60 allele has been associated with autoimmune diseases. In the end, CTLA-4 export to the membrane reduces and decreases the inhibitory function of CTLA-4. CTLA4 increased the development of ACPA positive RA in contrast to RA patients with ACPA negative. The meta-analysis showed a positive connection of 49A/G polymorphism susceptibility with RA in Asians, but only 1 in Asians and Europeans[3,46-48]. However, the exact role of this gene in RA is quite modest and still must be clarified.

TRAF1-C5

Two biological candidate genes, TNF receptor associated factor 1 (TRAF1) and complement component 5 (C5), were described by GWAS. TRAF1 is a member of the TNF receptor associated factor family, which are a class of proteins that link TNF receptor family members associated with signaling pathways that play a function in apoptosis, cell proliferation and differentiation, activation and inhibition cytokines and bone remodeling. The most strongly associated SNPs are rs3761847 and rs10818488 in the genome. It seems that the maximal genetic signal is located between the TRAF1 and C5 gene[39,49].

TNF

TNF alpha is a pleiotropic inflammatory cytokine. TNF-308A/G (rs1800629) polymorphism is associated with RA in the Latin American population[26,50] but not in any other ethnic group. Also, the TNF promoter polymorphism -609G/T and -238A/G are not associated with RA[45]. TNF-308A/G polymorphism was associated with radiological damage in a RA patient. Khanna et al[51] showed that patients with -308 TNF alpha AA+AG genotypes had considerably higher rates of progression in erosion scores and Sharp scores equal to the GG genotype patients. In contrast, Lacki et al[52] suggest that TNF-308 polymorphism cannot serve as an indicator of the disease course in RA patients.

INTERLEUKIN

Interleukins are a large part of cytokines which promote the development and differentiation of lymphocytes T, B and hematopoietic cells. In RA patients, SNPs of cytokines have been investigated regarding an association with erosive damage. One of them is IL-1. Polymorphism -511A/G (rs16944) in promoter IL-1b was positively associated with RA. +3954T allele was associated with more severe structural damage (mainly with Larsen’s score in wrist joints)[3,53,54]. IL-6 is a multifunctional cytokine implied in the inflammatory and immune response. Some studies reported that -174G/C (rs1800795) allele was associated with radiological damage in RA patients who were ACPA and RF positive[55]. The presence of two functional polymorphisms in the promoter region of IL-6, the -174G/C and -572G/C, suggests a strong susceptibility for European RA patients compared to Asians. These two polymorphisms (rs1800795 and rs1800795) may also influence the risk of osteoporosis. Another multifunctional cytokine is IL-10, produced by monocytes and lymphocytes, a protein that inhibits the synthesis of a number of cytokines and has a range of anti-inflammatory and immunoregulatory properties. Three polymorphisms placed in the promoter IL-10 were studied, including -1082G/A (rs 1800896), -892C/T (rs1800871) and -592C/A (rs1800872). The results are controversial as one showed that -1082G/A polymorphism is not associated with RA of either European or Asian populations and the other showed a positive association with RA, indicating that the carriers of the G allele could have a decreased liability of RA[26,56]. Some studies reported that the homozygosity of -592C/A was associated with higher Larsen scores in RA patients with ACPA and RF negative[55]. Polymorphisms of the IL-2 and IL-21 genes (region 4q27) have been implicated in several autoimmune diseases, including RA. One of them is intronic change A/G rs13151961[57]. Next studied were polymorphisms in RA susceptibility which may modulate gene expression of IL-2 or IL-21 located in the noncoding region, upstream of IL-21 and downstream of IL-2 is the G/T rs6822844. In a study on European Caucasian and South American populations, significant association with RA was shown[26,58,59].

With the use of GWAS, genetic studies can examine many common genetic variants across the entire human genome. There are a lot of other gene and chromosome loci revalidated as RA susceptible regions, such as CD226, CD40, CDK6, MBP, BLK, REL and more[60].

In conclusion, rheumatoid arthritis has a strong genetic influence mediated by alleles. Human genetics should be able to determine the value of RA risk alleles by providing clinical predictions. One of the most direct clinical applications is to use human genetics to lead the development of treatment for RA. It will be crucial to determine whether a combination of RA risk alleles are able to identify patients who develop certain clinical outcomes, such as myocardium infarction, severe infection or lymphoma, as well as to identify the patients who will respond to biological medication therapy.

Footnotes

P- Reviewer: Garip Y, Saviolas G S- Editor: Wen LL L- Editor: Roemmele A E- Editor: Wu HL

References
1.  Vandenbroucke JP, Hazevoet HM, Cats A. Survival and cause of death in rheumatoid arthritis: a 25-year prospective followup. J Rheumatol. 1984;11:158-161.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Youinou P, Pers JO, Gershwin ME, Shoenfeld Y. Geo-epidemiology and autoimmunity. J Autoimmun. 2010;34:J163-J167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
3.  Perricone C, Ceccarelli F, Valesini G. An overview on the genetic of rheumatoid arthritis: a never-ending story. Autoimmun Rev. 2011;10:599-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 117]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
4.  MacGregor AJ, Snieder H, Rigby AS, Koskenvuo M, Kaprio J, Aho K, Silman AJ. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000;43:30-37.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis. An approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum. 1987;30:1205-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Diogo D, Okada Y, Plenge RM. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
7.  Irigoyen P, Lee AT, Wener MH, Li W, Kern M, Batliwalla F, Lum RF, Massarotti E, Weisman M, Bombardier C. Regulation of anti-cyclic citrullinated peptide antibodies in rheumatoid arthritis: contrasting effects of HLA-DR3 and the shared epitope alleles. Arthritis Rheum. 2005;52:3813-3818.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Källberg H, Ding B, Padyukov L, Bengtsson C, Rönnelid J, Klareskog L, Alfredsson L. Smoking is a major preventable risk factor for rheumatoid arthritis: estimations of risks after various exposures to cigarette smoke. Ann Rheum Dis. 2011;70:508-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 249]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
9.  Kurkó J, Besenyei T, Laki J, Glant TT, Mikecz K, Szekanecz Z. Genetics of rheumatoid arthritis - a comprehensive review. Clin Rev Allergy Immunol. 2013;45:170-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 193]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
10.  van der Helm-van Mil AH, Wesoly JZ, Huizinga TW. Understanding the genetic contribution to rheumatoid arthritis. Curr Opin Rheumatol. 2005;17:299-304.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  du Montcel ST, Michou L, Petit-Teixeira E, Osorio J, Lemaire I, Lasbleiz S, Pierlot C, Quillet P, Bardin T, Prum B. New classification of HLA-DRB1 alleles supports the shared epitope hypothesis of rheumatoid arthritis susceptibility. Arthritis Rheum. 2005;52:1063-1068.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Besenyei T, Gyetvai A, Szabó Z, Fekete A, Kapitány A, Szodoray P, Laki J, Soós L, Sipka S, Szegedi G. Associations of HLA-shared epitope, anti-citrullinated peptide antibodies and lifestyle-related factors in Hungarian patients with rheumatoid arthritis: data from the first Central-Eastern European cohort. Joint Bone Spine. 2011;78:652-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
13.  Szodoray P, Szabó Z, Kapitány A, Gyetvai A, Lakos G, Szántó S, Szücs G, Szekanecz Z. Anti-citrullinated protein/peptide autoantibodies in association with genetic and environmental factors as indicators of disease outcome in rheumatoid arthritis. Autoimmun Rev. 2010;9:140-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 118]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
14.  van der Woude D, Houwing-Duistermaat JJ, Toes RE, Huizinga TW, Thomson W, Worthington J, van der Helm-van Mil AH, de Vries RR. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum. 2009;60:916-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 157]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
15.  van der Woude D, Lie BA, Lundström E, Balsa A, Feitsma AL, Houwing-Duistermaat JJ, Verduijn W, Nordang GB, Alfredsson L, Klareskog L. Protection against anti-citrullinated protein antibody-positive rheumatoid arthritis is predominantly associated with HLA-DRB1*1301: a meta-analysis of HLA-DRB1 associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in four European populations. Arthritis Rheum. 2010;62:1236-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 125]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
16.  van der Helm-van Mil AH, Verpoort KN, Breedveld FC, Huizinga TW, Toes RE, de Vries RR. The HLA-DRB1 shared epitope alleles are primarily a risk factor for anti-cyclic citrullinated peptide antibodies and are not an independent risk factor for development of rheumatoid arthritis. Arthritis Rheum. 2006;54:1117-1121.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Laki J, Lundström E, Snir O, Rönnelid J, Ganji I, Catrina AI, Bengtsson C, Saevarsdottir S, Wick MC, Alfredsson L. Very high levels of anti-citrullinated protein antibodies are associated with HLA-DRB1*15 non-shared epitope allele in patients with rheumatoid arthritis. Arthritis Rheum. 2012;64:2078-2084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
18.  Zsilák S, Gál J, Hodinka L, Rajczy K, Balog A, Sipka S, Baráth S, Kapitány A, Zilahi E, Szekanecz Z. HLA-DR genotypes in familial rheumatoid arthritis: increased frequency of protective and neutral alleles in a multicase family. J Rheumatol. 2005;32:2299-2302.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Oliver JE, Worthington J, Silman AJ. Genetic epidemiology of rheumatoid arthritis. Curr Opin Rheumatol. 2006;18:141-146.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Gyetvai A, Szekanecz Z, Soós L, Szabó Z, Fekete A, Kapitány A, Teodorescu M, Sipka S, Szegedi G, Lakos G. New classification of the shared epitope in rheumatoid arthritis: impact on the production of various anti-citrullinated protein antibodies. Rheumatology (Oxford). 2010;49:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
21.  Plant D, Bowes J, Potter C, Hyrich KL, Morgan AW, Wilson AG, Isaacs JD, Barton A. Genome-wide association study of genetic predictors of anti-tumor necrosis factor treatment efficacy in rheumatoid arthritis identifies associations with polymorphisms at seven loci. Arthritis Rheum. 2011;63:645-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 124]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
22.  Feng T, Zhu X. Genome-wide searching of rare genetic variants in WTCCC data. Hum Genet. 2010;128:269-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
23.  Craddock N, Hurles ME, Cardin N, Pearson RD, Plagnol V, Robson S, Vukcevic D, Barnes C, Conrad DF, Giannoulatou E. Genome-wide association study of CNVs in 16,000 cases of eight common diseases and 3,000 shared controls. Nature. 2010;464:713-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 625]  [Cited by in F6Publishing: 591]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
24.  Vossenaar ER, Zendman AJ, van Venrooij WJ, Pruijn GJ. PAD, a growing family of citrullinating enzymes: genes, features and involvement in disease. Bioessays. 2003;25:1106-1118.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Lee YH, Rho YH, Choi SJ, Ji JD, Song GG. PADI4 polymorphisms and rheumatoid arthritis susceptibility: a meta-analysis. Rheumatol Int. 2007;27:827-833.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Chatzikyriakidou A, Voulgari PV, Lambropoulos A, Drosos AA. Genetics in rheumatoid arthritis beyond HLA genes: what meta-analyses have shown? Semin Arthritis Rheum. 2013;43:29-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Lee YH, Bae SC, Choi SJ, Ji JD, Song GG. Genome-wide pathway analysis of genome-wide association studies on systemic lupus erythematosus and rheumatoid arthritis. Mol Biol Rep. 2012;39:10627-10635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 84]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
28.  Burr ML, Naseem H, Hinks A, Eyre S, Gibbons LJ, Bowes J, Wilson AG, Maxwell J, Morgan AW, Emery P. PADI4 genotype is not associated with rheumatoid arthritis in a large UK Caucasian population. Ann Rheum Dis. 2010;69:666-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
29.  Iwamoto T, Ikari K, Nakamura T, Kuwahara M, Toyama Y, Tomatsu T, Momohara S, Kamatani N. Association between PADI4 and rheumatoid arthritis: a meta-analysis. Rheumatology (Oxford). 2006;45:804-807.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Hinks A, Worthington J, Thomson W. The association of PTPN22 with rheumatoid arthritis and juvenile idiopathic arthritis. Rheumatology (Oxford). 2006;45:365-368.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Lee AT, Li W, Liew A, Bombardier C, Weisman M, Massarotti EM, Kent J, Wolfe F, Begovich AB, Gregersen PK. The PTPN22 R620W polymorphism associates with RF positive rheumatoid arthritis in a dose-dependent manner but not with HLA-SE status. Genes Immun. 2005;6:129-133.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Steer S, Lad B, Grumley JA, Kingsley GH, Fisher SA. Association of R602W in a protein tyrosine phosphatase gene with a high risk of rheumatoid arthritis in a British population: evidence for an early onset/disease severity effect. Arthritis Rheum. 2005;52:358-360.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Morgan AW, Thomson W, Martin SG, Carter AM, Erlich HA, Barton A, Hocking L, Reid DM, Harrison P, Wordsworth P. Reevaluation of the interaction between HLA-DRB1 shared epitope alleles, PTPN22, and smoking in determining susceptibility to autoantibody-positive and autoantibody-negative rheumatoid arthritis in a large UK Caucasian population. Arthritis Rheum. 2009;60:2565-2576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 77]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
34.  Farago B, Talian GC, Komlosi K, Nagy G, Berki T, Gyetvai A, Szekanecz Z, Nyarady Z, Kiss CG, Nemeth P. Protein tyrosine phosphatase gene C1858T allele confers risk for rheumatoid arthritis in Hungarian subjects. Rheumatol Int. 2009;29:793-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
35.  Goëb V, Dieudé P, Daveau R, Thomas-L’otellier M, Jouen F, Hau F, Boumier P, Tron F, Gilbert D, Fardellone P. Contribution of PTPN22 1858T, TNFRII 196R and HLA-shared epitope alleles with rheumatoid factor and anti-citrullinated protein antibodies to very early rheumatoid arthritis diagnosis. Rheumatology (Oxford). 2008;47:1208-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
36.  Mastana S, Gilmour A, Ghelani A, Smith H, Samanta A. Association of PTPN22 with rheumatoid arthritis among South Asians in the UK. J Rheumatol. 2007;34:1984-1986.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Association between the rs7574865 polymorphism of STAT4 and rheumatoid arthritis: a meta-analysis. Rheumatol Int. 2010;30:661-666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
38.  Amos CI, Chen WV, Lee A, Li W, Kern M, Lundsten R, Batliwalla F, Wener M, Remmers E, Kastner DA. High-density SNP analysis of 642 Caucasian families with rheumatoid arthritis identifies two new linkage regions on 11p12 and 2q33. Genes Immun. 2006;7:277-286.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Palomino-Morales RJ, Rojas-Villarraga A, González CI, Ramírez G, Anaya JM, Martín J. STAT4 but not TRAF1/C5 variants influence the risk of developing rheumatoid arthritis and systemic lupus erythematosus in Colombians. Genes Immun. 2008;9:379-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
40.  Martínez A, Varadé J, Márquez A, Cénit MC, Espino L, Perdigones N, Santiago JL, Fernández-Arquero M, de la Calle H, Arroyo R. Association of the STAT4 gene with increased susceptibility for some immune-mediated diseases. Arthritis Rheum. 2008;58:2598-2602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 104]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
41.  Orozco G, Alizadeh BZ, Delgado-Vega AM, González-Gay MA, Balsa A, Pascual-Salcedo D, Fernández-Gutierrez B, González-Escribano MF, Petersson IF, van Riel PL. Association of STAT4 with rheumatoid arthritis: a replication study in three European populations. Arthritis Rheum. 2008;58:1974-1980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 87]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
42.  Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, de Bakker PI, Le JM, Lee HS, Batliwalla F. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 2007;357:977-986.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Zervou MI, Sidiropoulos P, Petraki E, Vazgiourakis V, Krasoudaki E, Raptopoulou A, Kritikos H, Choustoulaki E, Boumpas DT, Goulielmos GN. Association of a TRAF1 and a STAT4 gene polymorphism with increased risk for rheumatoid arthritis in a genetically homogeneous population. Hum Immunol. 2008;69:567-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
44.  Raychaudhuri S, Remmers EF, Lee AT, Hackett R, Guiducci C, Burtt NP, Gianniny L, Korman BD, Padyukov L, Kurreeman FA. Common variants at CD40 and other loci confer risk of rheumatoid arthritis. Nat Genet. 2008;40:1216-1223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 404]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
45.  Miterski B, Drynda S, Böschow G, Klein W, Oppermann J, Kekow J, Epplen JT. Complex genetic predisposition in adult and juvenile rheumatoid arthritis. BMC Genet. 2004;5:2.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Li X, Zhang C, Zhang J, Zhang Y, Wu Z, Yang L, Xiang Z, Qi Z, Zhang X, Xiao X. Polymorphisms in the CTLA-4 gene and rheumatoid arthritis susceptibility: a meta-analysis. J Clin Immunol. 2012;32:530-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
47.  Han S, Li Y, Mao Y, Xie Y. Meta-analysis of the association of CTLA-4 exon-1 +49A/G polymorphism with rheumatoid arthritis. Hum Genet. 2005;118:123-132.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Daha NA, Toes RE. Rheumatoid arthritis: Are ACPA-positive and ACPA-negative RA the same disease? Nat Rev Rheumatol. 2011;7:202-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
49.  Plenge RM, Seielstad M, Padyukov L, Lee AT, Remmers EF, Ding B, Liew A, Khalili H, Chandrasekaran A, Davies LR. TRAF1-C5 as a risk locus for rheumatoid arthritis--a genomewide study. N Engl J Med. 2007;357:1199-1209.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Lee YH, Ji JD, Song GG. Tumor necrosis factor-alpha promoter -308 A/G polymorphism and rheumatoid arthritis susceptibility: a metaanalysis. J Rheumatol. 2007;34:43-49.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Khanna D, Wu H, Park G, Gersuk V, Gold RH, Nepom GT, Wong WK, Sharp JT, Reed EF, Paulus HE. Association of tumor necrosis factor alpha polymorphism, but not the shared epitope, with increased radiographic progression in a seropositive rheumatoid arthritis inception cohort. Arthritis Rheum. 2006;54:1105-1116.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Lacki JK, Moser R, Korczowska I, Mackiewicz S, Muller W. TNF-alpha gene polymorphism does not affect the clinical and radiological outcome of rheumatoid arthritis. Rheumatol Int. 2000;19:137-140.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Buchs N, di Giovine FS, Silvestri T, Vannier E, Duff GW, Miossec P. IL-1B and IL-1Ra gene polymorphisms and disease severity in rheumatoid arthritis: interaction with their plasma levels. Genes Immun. 2001;2:222-228.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Pawlik A, Kurzawski M, Florczak M, Gawronska Szklarz B, Herczyńska M. IL1beta+3953 exon 5 and IL-2 -330 promoter polymorphisms in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2005;23:159-164.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Marinou I, Healy J, Mewar D, Moore DJ, Dickson MC, Binks MH, Montgomery DS, Walters K, Wilson AG. Association of interleukin-6 and interleukin-10 genotypes with radiographic damage in rheumatoid arthritis is dependent on autoantibody status. Arthritis Rheum. 2007;56:2549-2556.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Zhang J, Zhang Y, Jin J, Li M, Xie K, Wen C, Cheng R, Chen C, Lu J. The -1082A/G polymorphism in the Interleukin-10 gene and the risk of rheumatoid arthritis: a meta-analysis. Cytokine. 2011;56:351-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
57.  Stahl EA, Raychaudhuri S, Remmers EF, Xie G, Eyre S, Thomson BP, Li Y, Kurreeman FA, Zhernakova A, Hinks A. Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet. 2010;42:508-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 949]  [Cited by in F6Publishing: 961]  [Article Influence: 68.6]  [Reference Citation Analysis (0)]
58.  Coenen MJ, Trynka G, Heskamp S, Franke B, van Diemen CC, Smolonska J, van Leeuwen M, Brouwer E, Boezen MH, Postma DS. Common and different genetic background for rheumatoid arthritis and coeliac disease. Hum Mol Genet. 2009;18:4195-4203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 107]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
59.  Maiti AK, Kim-Howard X, Viswanathan P, Guillén L, Rojas-Villarraga A, Deshmukh H, Direskeneli H, Saruhan-Direskeneli G, Cañas C, Tobön GJ. Confirmation of an association between rs6822844 at the Il2-Il21 region and multiple autoimmune diseases: evidence of a general susceptibility locus. Arthritis Rheum. 2010;62:323-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
60.  Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K, Kochi Y, Ohmura K, Suzuki A, Yoshida S. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506:376-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1549]  [Cited by in F6Publishing: 1545]  [Article Influence: 140.5]  [Reference Citation Analysis (0)]