ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

HLA and other susceptibility genes in rheumatoid arthritis

HLA and other susceptibility genes in rheumatoid arthritis
Literature review current through: Jan 2024.
This topic last updated: Jun 29, 2023.

INTRODUCTION — The association of particular human leukocyte antigen (HLA) alleles with rheumatoid arthritis (RA) was first noted in the late 1970s, when the frequency of individuals with the HLA-Dw4 serotype was found to be increased among RA patients compared with healthy controls [1,2]. This particular serotype represented a set of alleles at the HLA-DRB1 gene locus. Subsequently, hundreds of studies have examined and expanded that association in order to better elucidate the genetic underpinnings of RA. Both linkage and association studies of the HLA-DRB1 gene have consistently confirmed that it is the major genetic susceptibility locus for RA. As such, it provides an important clue to RA pathogenesis.

Since approximately 2005, discoveries using high-throughput genotyping in large sample collections have identified over 100 additional genetic loci outside of the HLA locus that play a relatively modest role in RA risk, but implicate critical pathways in its pathogenesis [3]. Further investigation has also begun to fine-map these RA loci, including the DRB1 gene. However, the issue of clinical utility of genotyping in patients with RA remains unresolved.

The role of HLA and other susceptibility genes in RA will be addressed in this topic review. The epidemiology of, risk factors for, causes of, and pathogenesis of RA, as well as an overview of HLA genes and their nomenclature, are discussed separately. (See "Epidemiology of, risk factors for, and possible causes of rheumatoid arthritis" and "Pathogenesis of rheumatoid arthritis" and "Human leukocyte antigens (HLA): A roadmap".)

RHEUMATOID ARTHRITIS SUSCEPTIBILITY GENES

HLA alleles and susceptibility to rheumatoid arthritis — Both linkage and association studies have established that the human leukocyte antigen (HLA) DRB1 gene is the major genetic susceptibility locus for rheumatoid arthritis (RA). Subsequently, further studies in large data sets have also identified independent associations with other HLA genes: HLA-A [4], HLA-B [5], and HLA-DPB1 [5].

Interpretation of early studies of HLA associations in RA has been complicated by evolving changes in nomenclature and methods of HLA typing [6]. Originally, HLA typing was achieved with immunologic reagents. The methods subsequently used for HLA typing in RA have involved amplification by the polymerase chain reaction of the highly polymorphic second exon of the HLA-DRB1 gene, followed by probing with a DNA sequence-specific oligonucleotide probe. Direct nucleotide sequencing is an alternative method of genotyping. A newer technique for HLA typing has since been developed for further analysis of individuals with available dense single-nucleotide polymorphism (SNP) genotyping data over the HLA region [7]. (See "Human leukocyte antigens (HLA): A roadmap" and 'Individual amino acid sites and susceptibility to rheumatoid arthritis' below.)

Individual alleles and the shared epitope — Two alleles of the HLA-DRB1 gene, DRB1*04:01 and DRB1*04:04, primarily account for the originally observed serologic association of DR4 with RA in White American and European patients. In addition, serologic associations with DRB1*01 and DRB1*10 alleles have been noted in many such RA patients who are negative for DRB1*04 alleles. (See 'Associations in different ethnic populations' below.)

Since many of these most strongly associated RA alleles shared a region of highly similar amino acid sequence, it had been postulated that this portion of the DRB1 molecule (amino acids 67 to 74) controls susceptibility to disease [8]. The most strongly associated RA alleles code for HLA proteins that share a region of highly similar amino acid sequence at positions 70 to 74, called the shared epitope (SE; 70QKRAA74, 70QRRAA74, or 70RRRAA74 – one-letter amino acid code). This hypothesis had been further bolstered by the observation that among ethnic populations in which DRB1*04 alleles are rare, the alleles associated with RA still contain the SE. As an example, some Native American populations, such as the Yakima Nation, have a high prevalence of RA. DRB1*04 alleles are rare in this population, and RA is possibly associated with the DRB1*14:02 allele. This allele has an amino acid sequence from positions 67 to 74 that is identical to the SE [9].

The strength of the association of particular HLA susceptibility alleles with RA varies in different studies. To some degree, this variability is due to differences in the criteria for inclusion of patients, in genetic backgrounds of the populations studied, and in the HLA typing methods used. However, a remarkable degree of consensus emerges when these variables are controlled. If, for example, only White American and European patients are included and if the HLA typing is DNA-based, then similar results have been obtained in different reports [10-13]:

HLA-DRB1*04:01 – Present in 50 to 61 percent of patients with RA with a relative risk of 5 to 11.

HLA-DRB1*04:04 – Present in 27 to 37 percent with a relative risk of 5 to 14. In one report, an increased risk of RA was noted when there were two copies of the HLA-DRB1*04:04 allele or when it was present along with another gene encoding the SE [13].

HLA-DRB1*01:01 – Present in 13 to 27 percent with a relative risk of 1 to 2.

HLA-DRB1*10 – Present in 1.5 percent of RA patients with a relative risk of 2.3 [13].

Protective HLA alleles — While some specific HLA-DRB1 alleles confer a high risk for the development of RA, other alleles provide protection from disease risk. As an example, certain alleles with an aspartic acid (versus glutamine or arginine) at the 70th amino acid position result in an odds ratio (OR) of 0.23 and 0.34 for RA among English and Spanish populations, respectively [14], and in an OR of 0.52 among North American RA populations. In addition to the presence of aspartic acid at position 70, the presence of isoleucine at position 67 is also associated with a protective effect [15]. A large meta-analysis across multiple European populations has shown that HLA-DRB1*13:01 confers protection [16].

Individual amino acid sites and susceptibility to rheumatoid arthritis

Seropositive rheumatoid arthritis — A small number of individual amino acid sites within HLA-DRB1 that explain seropositive (rheumatoid factor [RF] positive or anti-citrullinated peptide antibody [ACPA]-positive) RA susceptibility have been identified. It has become possible to use large SNP genotyping data sets to infer HLA-DRB1 alleles indirectly using advanced statistical methods [7,17]. Using these approaches, a study of 20,000 individuals of European descent has demonstrated that individual amino acid sites within HLA-DRB1 explain seropositive RA risk susceptibility most effectively [5]. In particular, position 11 (and equivalently position 13, which is tightly linked to it) modulates more RA risk than any position in the genome; the presence of a valine at that site (or a histidine at position 13) confers an increased risk, with an allelic OR of approximately 4, whereas the presence of a serine (at position 13 or 11) confers a decreased risk, with an allelic OR of 0.4 (table 1) [5]. Additionally, positions 71 and 74 (located within the SE consensus sequence) mediate disease risk independently of the amino acid present at position 11/13 [5]. This study has also suggested that any individual of White European descent, instead of being classified as SE-positive or SE-negative, can be classified into 16 different risk categories solely based on their four-digit HLA-DRB1 type or the amino acids carried at positions 11/13, 71, and 74 of HLA-DRB1 [5]. The highest-risk categories correspond to SE-positive individuals, while the lower-risk categories are defined by protective HLA alleles (for example, HLA-DRB1*13:01 (table 1)).

Side chains of amino acids at positions 13, 71, and 74 point into the antigen-binding groove and together fall within a specific pocket within the DR molecule known as the P4 pocket. These results suggest that the P4 pocket may play an important role in differential antigen binding, affecting disease risk. Investigators have also studied the implications of variation in amino acid residues for citrullinated peptide antigen binding [18,19]. Citrullination is a process by which positively charged arginine residues are converted to uncharged citrulline. Modeling and structural studies have shown that the shared epitope P4 pocket of HLA-DRB1 binds citrulline more effectively than arginine [18,20]. HLA-DRB1 alleles that result in an electropositive P4 pocket increase the risk of RA, while those resulting in a neutral or negative charge in the P4 pockets are protective, as they are associated with a reduced risk of seropositive RA [21].

After controlling for HLA-DRB1 associations at positions 11/13, 71, and 74, independent associations with seropositive RA are observed at HLA-DPB1, corresponding to amino acid position 9 of that molecule; in HLA-B, corresponding to HLA-B*08 and equivalently to position 9 of that molecule [5]; and at HLA-A position 77 [4]. These amino acid sites also sit at the base of the binding groove of those proteins.

Seronegative rheumatoid arthritis — Seronegative RA is genetically distinct from seropositive RA. As amino acids associated with seronegative RA are also located within the peptide-binding grooves, distinct peptide autoantigens may be involved in the etiology of the two RA serotypes. The strongest genetic associations between HLA amino acids and seronegative RA are also located at HLA-DRB1 position 11 (but in contrast to seropositive RA correspond to HLA-DRB1*03) and HLA-B position 9 (corresponding to HLA-B*08) [4,22]. However, HLA-DRB1 positions 71 and 74 were not found to be associated with seronegative RA. Although the association with HLA-DRB1 position 11 is shared between seropositive and seronegative RA, the effects of individual amino acids are different; the carriage of a serine at HLA-DRB1 position 11 increases the risk of developing seronegative RA, but decreases the risk of developing seropositive RA. The pattern of association is different for different amino acids at the same position: the carriage of a glycine at HLA-DRB1 position 11 is protective for both serotypes. These studies confirm that seropositive and seronegative RA are two genetically distinct entities.

Mechanistic implications of the HLA associations — The identification of class I and II HLA allelic associations implicates both CD8+ and CD4+ T cells in the pathogenesis of RA. Theories that explain how expression of the HLA-DRB1 alleles mediate RA risk focus on the role of the HLA molecule in binding specific peptides and in presenting them to CD4+ T lymphocytes via recognition by the T-cell receptor. The polymorphic residues in the class II HLA molecule determine which peptides are bound. Molecules containing the SE bind a different repertoire of peptides than those not containing it [23,24].

In an attempt to explain how the SE functions in its contribution to RA, different models have been advanced [25]:

One model suggests that certain pathogenic peptides bind risk alleles more efficiently and are more likely to be presented to T cells only by HLA-DRB1 risk alleles.

In a second model, this differential peptide binding alters the specific T cells that are positively selected in the thymus, thereby leading to a different T-cell repertoire that could include autoreactive, pathogenic T cells.

Still another theory suggests direct T-cell recognition of the HLA-DRB1 risk alleles, which is more dominant than recognition of the specific peptide bound by the DR molecule.

Relationship between HLA-DRB1 and ACPA — Anti-citrullinated peptide antibody (ACPA) status shows a stronger association with the presence of erosions than HLA alleles or the SE status; the presence of anti-cyclic citrullinated peptide (anti-CCP) antibodies at baseline is strongly associated with both prevalent erosions (OR = 2.5) and developing erosions at five years (OR = 10.2) [26]. Therefore, the presence of ACPA at disease onset is the strongest known predictor of later radiographic damage in patients with RA.

Since the identification of ACPA and the development of commercial tests that detect and determine the level of anti-CCP antibodies, there is emerging evidence that anti-CCP-positive and anti-CCP-negative RA may behave very differently and may actually comprise distinct diseases underpinned by distinct genetic contributions. This is particularly striking for the HLA-DRB1 gene in which, for example, the linkage to the HLA region observed in RA families is absent in anti-CCP-negative families [27]. (See "Biologic markers in the assessment of rheumatoid arthritis", section on 'Anti-citrullinated peptide antibodies'.)

Subsequent studies have confirmed that there is a significant association between SE alleles and RA in patients who are anti-CCP antibody-positive [27-30]. For example, a significantly greater prevalence of anti-CCP antibodies was found in those who carried two SE alleles than in those with one or no allele (85, 58, and 30 percent, respectively). Hence, the SE seems to predispose to anti-CCP-positive RA, and the development of anti-CCP antibodies is likely to be an intermediate stage in disease pathogenesis and to mediate the effect of the SE with RA susceptibility and/or severity. Subsequent studies in much larger cohorts of anti-CCP-negative RA patients have, however, detected significant associations between the SE, HLA alleles, specific amino acids, and anti-CCP-negative RA [4,31]. The effect of the HLA on anti-CCP-negative RA susceptibility has been localized to HLA-DRB1 position 11/13 and HLA-B position 9, but different residues at position 11/13 confer risk and protection for anti-CCP-negative RA compared with anti-CCP-positive RA [4].

A gene environment interaction between smoking and SE carriage in ACPA-positive RA has been reported in two European populations [32,33] but has not been replicated in three populations from the United States [34]. Smoking has also been reported to be associated with the production of ACPA in SE-negative individuals, so the relationship appears complex [35].

It has been speculated that smoking leads to increased citrullination of proteins. Carriage of SE alleles in this environmental background increases susceptibility to RA because they bind citrullinated peptides more strongly and induce an exaggerated T-cell response. The exaggerated T-cell response, in turn, may drive the increased autoantibody production by B cells, including anti-CCP antibodies, seen in RA [36,37].

Non-classical HLA genes and non-HLA genes within the HLA region — The HLA region on chromosome 6 contains over 200 genes of immunologic relevance, including the HLA genes. Several early studies have suggested that loci other than HLA genes may also contribute to susceptibility to RA, but these studies were challenging to interpret due to the strong effects of the HLA alleles and to the high degree of linkage disequilibrium across the HLA region. For example, early reports of an association of RA with other classical HLA genes, such as alleles of HLA-DQA1, and with other non-HLA genes, such as tumor necrosis factor (TNF), were likely to be secondary to linkage to HLA-DRB1, -DPB1, -B, or -A risk alleles in White populations. The genes for TNF lie within the HLA region and have been a focus of intense interest, given the demonstration that TNF-alpha plays a central role in the inflammatory cascade in affected joints of patients with RA and the efficacy of TNF-alpha antagonists as therapeutic agents for patients with RA. Results of such investigations, however, have been conflicting; some studies report an association of particular TNF markers with RA [38-42], while others find no differences [43-47]. The association with TNF is most likely a function of alleles in linkage with HLA-B*08 [5].

A large-scale multi-ethnic fine-mapping study of the HLA region conducted in over 60,000 cases and controls from Japanese, Eastern Asian, and European populations identified an independent association with the non-classical HLA gene, HLA-DOA, with seropositive RA [48]. The associated allele appeared to be correlated with the expression of HLA-DOA, putatively demonstrating a new pathogenic mechanism for susceptibility alleles within the HLA region.

Associations in different ethnic populations — Data have accumulated on HLA associations with RA in a diverse range of ethnic groups, many of which display a very different genetic background from populations of White European ancestry [49,50]. Nevertheless, the majority of these studies confirm a significant increase in the frequency of alleles containing the SE in RA patients:

DRB1*14:02 confers susceptibility to RA in the Yakima, Tlingit, and Pima Native American populations [9,19,51,52]. The DR4 specificity is also present and associated with RA in the Chippewa Indians [53].

Among Japanese, the most prevalent DRB1*04 allele is DRB1*04:05, which contains the SE and which is increased in Japanese patients with RA [54].

Both DRB1*04:05 and *04:04 are increased in Chinese RA patients [55].

However, a subsequent study in Han Chinese subjects, in which a deep sequencing of the HLA region was performed in ACPA-positive RA patients and healthy controls, identified HLA-DQA1 as the major genetic risk factor for seropositive RA in this population, instead of HLA-DRB1*04:05 [56].

In some population groups, such as the Spanish, the Basque, and Israeli Jews, DRB1*04 alleles are rare. In these settings, RA is associated with DRB1*01 or DRB1*10, two other alleles carrying the SE [57-60].

In other populations, such as African Americans, the majority of patients with RA do not carry the SE in their DRB1 genes [61]. Even in this study, however, DR4-positive (DR4+) susceptibility alleles were significantly increased among RF-positive patients. A similar observation was made in a study of Greek patients with RA; although 57 percent lacked the SE, its presence was still significantly increased compared with local controls (43 versus 15 percent) [62].

Indeed, further studies examining amino acid sites observed, similar to populations of European descent, that position 13 mediated much of the risk of RA in Asian populations, including patients from Japan, Korea, and China; and the same residues at that site that conferred risk in European populations also did so in Asian populations [63]. Similarly, residues that protected from risk in European populations at position 13 also protected from risk in Asian populations.

In African American populations, it was also observed that position 11 mediated risk of RA, with a similar risk profile as in Europeans [64]. A multinational Arab genome-wide association study also identified HLA-DRB1 amino acid position 11 as the strongest genetic risk factor for RA in this population [65].

Although the majority of studies seem to show that RA susceptibility genetic variations within the HLA are shared across populations, it should be noted that the HLA allele frequency remains highly population dependent: valine at HLA-DRB1 position 11 might be more frequent in RA cases compared with controls within each population, nonetheless, the prevalence of valine at position 11 is highly variable across populations. Moreover, studies of genetic variations outside the HLA region have shown that some non-HLA susceptibility polymorphisms are shared across populations, while others appear to be population-specific [3,65,66].

Rheumatoid arthritis susceptibility genes outside the HLA region — Although a role in conferring susceptibility to RA is well-accepted for HLA genes within the HLA region on chromosome 6, most investigators estimate that HLA contributes to less than 50 percent of the overall genetic risk. The availability of increasingly powerful molecular genetic techniques has ignited the search for other relevant genes in RA. This has generally taken two approaches: investigating candidate genes whose function suggests a possible role in disease pathogenesis, and screening the entire human genome using genome-wide association studies.

A number of genes outside of the HLA region also exhibit an association with RA, and some are associated with other autoimmune conditions as well. Over 150 confirmed associations with RA susceptibility have been reported [67]. Since 2007, there has been an explosion in the number of RA susceptibility genes identified and confirmed in well-powered cohorts [3,68,69]. For most of the reported associations, the actual causal variant has not been identified, and the gene thought to be responsible for the association may change as fine-mapping studies are undertaken. Most associated variants are intergenic or intronic variants that do not confer a change in the amino acid sequence of gene protein products. These studies are underway, so a better understanding of which genes are involved, whether the same variants are associated with multiple autoimmune diseases, and whether multiple variants at a locus contribute to susceptibility should become clearer as research progresses.

The R620W PTPN22 polymorphism has been consistently associated with RA in populations of northern European descent, but the variant does not exist in Japanese or Korean populations. The association with ACPA-positive RA is much stronger than with ACPA-negative RA [31]. STAT4, on the other hand, has been associated with RA in populations of both European and Asian ancestry and seems to be equally associated with ACPA-positive and -negative RA [31].

The identification of susceptibility loci for RA is a rapidly moving field, and there is an ever-expanding list of confirmed hits; six examples of this expanding list of genes include:

PTPN22 gene – The protein tyrosine phosphatase N22 (PTPN22) gene helps regulate both T and B cells. The frequency of an SNP in the gene was increased in patients versus healthy controls in studies of multiple White North American and European populations but not in Koreans [50]. The SNP encodes an amino acid substitution at position 620 (R620W), which affects binding to an intracellular signaling molecule called Csk [70].

The polymorphism is associated with multiple autoimmune diseases, including type 1 diabetes, juvenile idiopathic arthritis, Hashimoto's thyroiditis, systemic lupus erythematosus (SLE), and Addison's disease.

In mice, the corresponding gene is a negative regulator of T cells [70]. The W620 variant of PTPN22*R620W binds Csk less efficiently than the R620 allele, which may predispose to autoimmunity by resulting in a failure to switch off T cells. In other experiments, however, the polymorphism has been shown to be a "gain-of-function" variant (ie, it increases the downregulation of T cells). This is postulated to predispose to autoimmunity by failure to delete autoreactive T cells during thymic development [71]. A study has also demonstrated that levels of the 620W PTPN22 variant are decreased in human T and B cells, and its calpain binding and cleavage were increased relative to wild-type 620R. Based upon mouse experiments, this difference was attributed to calpain-mediated degradation. Reduced levels of the protein correlated with an increased number of T cells and increased T-cell activation, and with dendritic-cell hyperresponsiveness [72].

STAT4 gene – STAT4 encodes a transcription factor that transmits signals induced by several key cytokines, including interleukin (IL)-12 and type 1 interferons, as well as IL-23 [73]. STAT4-dependent signaling by IL-12 receptors plays a critical role in the development of Th1-type responses. A variant allele of STAT4 confers an increased risk of both RA and SLE [74]. Homozygosity of the risk allele is associated with a more-than-doubled risk of SLE and with a 60 percent increased risk of RA.

TRAF1-C5 gene locus – The TRAF1 gene encodes TNF receptor-associated factor 1, and the C5 gene encodes complement component 5. A genome-wide analysis of North American and Swedish patients revealed that a common genetic variant at the TRAF1-C5 locus on chromosome nine, identified by the rs3761847 SNP, appeared to increase the risk of anti-CCP antibody-positive RA [75]. This association, however, was not seen in a cohort of Korean patients with RA [76]. By contrast, the rs7021206 SNP of the TRAF1 gene was associated with RA in populations of White European descent, as well as Korean populations [77].

Chromosome 6q23 – An intergenic region between the OLIG3 and TNFAIP3 genes on chromosome 6q23 has been associated with RA susceptibility in both United States and United Kingdom populations [78,79]. The association in the region is complex with at least three variants independently contributing to overall susceptibility [80]. It is thought that the variants may be affecting the function and/or regulation of the TNFAIP3 gene as knock-out mice develop severe inflammation, which includes inflammation of joints [81]. However, although the intergenic region is certainly associated with RA susceptibility, the involvement of the TNFAIP3 gene in humans has not been confirmed. Using techniques to detect chromatin interactions, it has been shown that the DNA containing polymorphisms associated with RA interacts through chromatin looping with IL20RA, located 680 kb upstream. One of the risk alleles is correlated with an increased expression of IL20RA and is most likely located within an active enhancer in T cells [82].

PADI-4 gene – Genes for enzymes that are responsible for post-translational modification of arginine to citrulline, an antigenic side chain against which autoantibodies are found in many patients with RA, contribute to the risk of developing disease. Of the four isotypes of PADI, types 2 and 4 are considered to be the most likely to be present in human synovium in which citrullination of matrix proteins could potentially create antigenic peptides [83-85]. Furthermore, levels of PAD-2 and PAD-4 were correlated with the level of inflammation in RA synovium [85].

In two case-control studies, significant association was noted between genetic variants coding for one of four such enzymes (peptidylarginine deiminase 4 or PADI4) and the presence of RA in a Japanese population [86,87]; an association has also been identified in patients from Europe, but with different genetic variants [88]. A large trans-ethnic association study confirmed two independent associations with SNPs at the PADI4-PADI2 locus and RA [68].

CTLA-4 gene – A meta-analysis and separate well-powered validation studies have confirmed association of RA with variants within the CTLA-4 gene [89,90]. Previous work has shown that the associated variant maps within the 3' region of the CTLA-4 gene and is correlated with the ratio of soluble to full-length CTLA4 mRNA levels [91]. Interestingly, abatacept, a biologic drug in use for the treatment of RA, mimics the physiological role of CTLA-4. (See "Overview of biologic agents in the rheumatic diseases", section on 'Abatacept'.)

RHEUMATOID ARTHRITIS SEVERITY GENES

HLA alleles and severity of rheumatoid arthritis — Most studies support an association of the shared epitope (SE) with rheumatoid arthritis (RA) disease severity, based upon an association with erosive disease [92-95], but not all studies do so [96-98]. Studies have found a correlation between DRB1*04 RA-associated alleles and more severe, erosive disease, as measured by the severity of radiographic changes [93-95,99,100]; some have also noted an increase in DRB1*01 among patients with milder disease [100]. A large prospective study that was performed in over 2000 patients of White British origin concluded that the hierarchy of HLA-DRB1 alleles with regards to RA susceptibility is exactly the same for disease severity, measured as the extent of radiographic damage or by mortality [101]. Therefore, the 16 different risk categories defined by amino acids at positions 11/13, 71, and 74 of HLA-DRB1 (see 'Individual amino acid sites and susceptibility to rheumatoid arthritis' above) also represent 16 different severity categories. However, the clinical utility of this genetic stratification system remains to be evaluated. Moreover, it seems that the association of HLA with severe disease acts mainly through the presence of anti-citrullinated peptide antibody (ACPA), and it is clear that ACPA status is a better predictor of erosions than are human leukocyte antigen (HLA) alleles or the SE status. (See 'Relationship between HLA-DRB1 and ACPA' above.)

This association between HLA-DRB1*04 alleles and erosive disease has been confirmed among non-White patients as well, including Koreans [102] and Japanese [103].

The specific combination of SE-positive alleles also may be important. One report evaluated a large group of patients with severe RA, including many with Felty syndrome [104]. A hierarchy of risk was found, depending upon the particular combination of HLA-DRB1 alleles: the relative risk was 5 with the heterozygous HLA-DRB1*04/X combination (where X is any allele other than HLA-DRB1*04 or HLA-DRB1*01); 16 with HLA-DRB1*04/HLA-DRB1*01; 25 with DRB1*04/HLA-DRB1*04 combinations; and 49 with HLA-DRB1*04:01/04:04 (Dw4/Dw14).

The presence of an SE in an HLA-DRB1 allele may predispose to other extraarticular disease manifestations as well. This was illustrated in a group of rheumatoid factor (RF)-positive patients with erosive disease, almost one-half of whom had two SE-positive alleles [105]. Individuals whose SEs were contained in DR4+ alleles (HLA-DRB1*04:01, *04:04, and/or *04:08) had the highest risk of extraarticular disease, including rheumatoid vasculitis, rheumatoid lung disease, and Felty syndrome. Patients heterozygous for two SE-containing alleles, one DR4+ and one DR4-, had disease of intermediate severity.

HLA genes and prognosis — The presence of ACPA at disease onset is the strongest predictor of later radiographic damage in patients with RA [26]. Most studies also support an association of the SE with erosive disease [92]. The role of genetic predictors of disease outcome remains a focus of active research for the following reasons: (1) the relative effect of ACPA titers and HLA alleles on several measures of disease outcome is still unknown; (2) genetic markers are likely to have added value to ACPA on the prediction of severity; and (3) there is substantial interest in the prediction of disease onset (inextricably linked to future severity) before ACPA seroconversion (for example, in cohorts of first-degree relatives of patients with RA).

The presence of the SE alleles is not only associated with more severe joint injury, but also with increased risk of cardiovascular disease and mortality. In 2004, a meta-analysis of the impact of SE-positive alleles on erosive disease summarized the results of 10 studies (1027 patients) and found that carriage of one or two such alleles was associated with an odds ratio (OR) of having joint erosion of approximately 2 [106]. The retrospective association of specific HLA alleles with severe RA raised the question of whether typing may be useful to predict which patients will progress to destructive disease, as HLA markers, unlike levels of serologic markers like RF or acute phase reactants, are stable over time and are not subject to fluctuation with disease course or treatment.

Cardiovascular disease is a common comorbidity in RA and is associated with reduced life expectancy in patients with RA. Several studies have shown that carrying two copies of SE alleles is associated with premature mortality and with cardiovascular mortality, and this effect appears independent of ACPA status [107-111]. Indeed, one study reported that patients with inflammatory polyarthritis who were smokers, had ACPA, and carried two copies of SE alleles had a 7.8-fold increased risk of premature mortality from cardiovascular disease compared with patients without these risk factors [107].

Indeed, further studies reexamined RA clinical outcomes and mortality in light of position 11 residues and found that residues that drive risk at that site are connected to worse clinical outcomes, including increased mortality [101,112].

Rheumatoid arthritis severity genes outside the HLA region — Some genetic loci outside the HLA region have been associated with disease severity (reviewed in [113]). A number of non-HLA genetic loci have been investigated to assess whether they can predict severity. Since the power of severity studies is much lower than the power of susceptibility studies, the results of many severity studies are still conflicting, but the emerging picture suggests that some genetic variants associated with susceptibility are also associated with severity, while others are associated either only with susceptibility or only with severity. The number of confirmed genetic predictors located outside the HLA region is small. As examples:

Genetic variants located near the TRAF1 gene, known to be associated with susceptibility to RA, have been shown to be associated with radiographic damage in multiple independent studies in populations of different ethnicities [114-117].

An SNP located near the FOXO3 gene has been shown to be associated with prognosis in several tumor necrosis factor (TNF) alpha-driven diseases (Crohn disease, RA, malaria). This SNP leads to altered production of pro- and antiinflammatory cytokines by monocytes through a transforming growth factor (TGF) beta-1-dependent pathway. In RA, minor allele carriage of the SNP reduces TNF-alpha, interleukin (IL)-6, IL-1-beta, and IL-8 production, and increases IL-10 production. The SNP is associated with a milder course of disease (ie, less joint damage over time) [118]. Although associated with RA severity in patients with RA, FOXO3 genetic variants are not associated with an increase in susceptibility to RA in healthy individuals.

GENES DETERMINING RESPONSE TO TREATMENT IN RHEUMATOID ARTHRITIS — Studies to determine genetic predictors of treatment response [113,119-121], including genome-wide association studies for responsiveness to methotrexate [122] or to tumor necrosis factor (TNF) inhibitor therapy [123], have not yet produced clinically actionable or reproducible results. Although some data suggest that knowledge of a patient's human leukocyte antigen (HLA) status may be useful in predicting the response to specific therapies, results have been conflicting. As an example, a large study performed in 1846 patients enrolled at initiation of TNF inhibitor suggested that the 16 susceptibility/severity categories defined by HLA-DRB1 amino acids at position 11/13, 71, and 74 also represented TNF inhibitor-response categories, with carriers of high-risk/high-severity haplotypes likely to respond better to treatment [101], but these results have not been confirmed [124]. Another study performed with nonbiologic disease-modifying antirheumatic drugs (DMARDs) observed a marked difference in treatment response in patients when stratified by shared epitope (SE) status [125]. Response rates were higher in those without the SE compared with those with a single copy or two copies (also termed a single or double dose; 83, 36, and 25 percent, respectively).

These results might suggest that patients with one or especially two copies of the SE (one copy on the maternal and one copy on the paternal chromosome) may benefit from earlier, more aggressive treatment to control disease. A patient's genetic profile is likely to influence clinical decision-making in the future (precision medicine). However, limited numbers of patients have been studied, and further evidence is required [126,127]. Thus, additional prospective and longer-term studies are needed to better assess the role of testing for the SE or HLA-DRB1 amino acids or haplotypes in choosing treatment for patients with RA. Such testing is only appropriately performed as a research tool.

CLINICAL USE OF GENETIC MARKERS

For diagnosis or screening of rheumatoid arthritis — Genetic markers are not useful for diagnosis or screening of rheumatoid arthritis (RA). Although certain human leukocyte antigen (HLA) alleles are strongly associated with severe RA, these alleles are common in the normal population. The absolute risk of developing RA among White individuals carrying HLA-DRB1 alleles *0401, *0404, or *0101 is approximately 1 in 46 [128]. The highest calculated absolute risk is present in individuals expressing both HLA-DRB1*0401 and *0404, but this is still only approximately one in seven. Even knowing the genotype at the second confirmed RA susceptibility locus, the PTPN22 gene, the predictive value remains too low to be useful for either diagnosis or population screening.

Presence of anticyclic citrullinated peptide (anti-CCP) antibodies does predict the future development of RA, particularly severe RA, and one report suggested that the presence of two copies of the shared epitope (SE) with one or two copies of PTPN22 risk alleles increased the risk of developing anti-CCP-positive RA by 25-fold [129]. Separate investigations have shown an interactive relationship between smoking, HLA alleles, and anti-CCP antibody development [32].

The combination of a large number of disease-associated genetic susceptibility polymorphisms (within and outside the HLA) into genetic risk scores (GRS) might have the potential, in the future, to help clinicians with the differential diagnosis of inflammatory polyarthritis. One publication reports the development of a genetic probability tool, which helps clinicians to distinguish between RA, systemic lupus erythematosus (SLE), spondyloarthritis, psoriatic arthritis, and gout [130]. For all patients included in the study, at least one disease could be ruled out, while in 45 percent of patients, a diagnosis was suggested with a 64 percent positive predictive value [130]. However, the performance of genetic diagnostic tools remains to be assessed prospectively in a clinical setting; controlled trials comparing standard practice with the use of genetic probability tools are required before clinical utility can be claimed.

For estimating prognosis — Routine typing for HLA markers and SE status cannot be recommended, given the predictive value of other markers, including anti-CCP antibodies and rheumatoid factor (RF), and given the limited availability and potential cost of the technology required to type for SE alleles.

Once the diagnosis of RA is established, genotyping for SE alleles may help predict which patients are at highest risk of severe and erosive disease, and thus which are candidates for early and aggressive intervention. This information may be less important clinically if patients with early RA are aggressively treated with potent disease-modifying antirheumatic drug (DMARD) regimens, consistent with recommended approaches to disease management.

RF testing and anti-CCP antibody testing are the most useful tests that are widely available in predicting which patients with early inflammatory arthritis will develop erosions [26]. The presence of erosions is a predictor of poor prognosis, regardless of HLA type. The presence of HLA SE alleles is strongly correlated with the presence of anti-CCP antibodies. Whether knowledge of SE status provides additional predictive value for the development of erosive disease beyond knowledge of anti-CCP status alone remains uncertain [26,131].

It is also unclear whether genetic markers can be identified that could predict a subgroup which would not require such aggressive intervention and could be spared the risk of potential adverse events.

Role of ethnicity — Most of the information and hypotheses regarding clinical utility of genetic testing are based primarily upon data derived from White North American and northern European patients and patients of Asian ancestries. HLA typing of patients from other ethnic groups will have different implications. As an example, the lack of a disease-associated HLA-DRB1*04 allele in an African American patient does not necessarily imply that they are not at high risk of developing erosive disease [132]. On the other hand, as described above, most populations show a strong association with one or more SE-positive DR alleles; testing methods should be easily adaptable to inclusion of those alleles for appropriate ethnic groups in any RA-oriented "panel" of susceptibility alleles. However, prediction models will depend on the prevalence of the tested susceptibility alleles in the population of interest, and will therefore be population-dependent.

SUMMARY AND RECOMMENDATIONS

Rheumatoid arthritis susceptibility – Two alleles, human leukocyte antigen (HLA)-DRB1*0401 and HLA-DRB1*0404, primarily account for the originally observed serologic association of HLA-DRB1*04 alleles with rheumatoid arthritis (RA) in White individuals.

Some HLA-DRB1 alleles protect against the development of RA rather than enhance the risk. HLA-DRB1 risk is best explained by a single amino acid position at the base of the binding groove, position 13 (or equivalently position 11).

Patients from other ethnicities display a different genetic background from White individuals, but most studies confirm the role of HLA-DRB1 position 11/13 across different ethnic groups. (See 'HLA alleles and susceptibility to rheumatoid arthritis' above.)

Rheumatoid arthritis severity – HLA-DRB1 susceptibility alleles are also severity alleles: the hierarchy of HLA-DRB1 alleles or amino acids with regards to RA susceptibility, ranging from risk to protective, is the same for disease severity. Certain risk alleles may also predispose to extraarticular disease manifestations. A small number of non-HLA genetic loci also appear associated with more severe disease. (See 'HLA alleles and severity of rheumatoid arthritis' above and 'Rheumatoid arthritis severity genes outside the HLA region' above.)

Shared epitope hypothesis – Theories to explain how expression of the shared epitope (SE) contributes to RA focus on the role of the HLA molecule in binding specific peptides and in presenting them to CD4+ T lymphocytes via recognition by the T-cell receptor. Molecules containing the SE bind a different array of peptides than those not containing it. Several different models have been proposed to explain how the SE functions to contribute to RA. (See 'Mechanistic implications of the HLA associations' above.)

Seropositive versus seronegative rheumatoid arthritis – There is emerging evidence that anti-citrullinated peptide antibody (ACPA)-positive and ACPA-negative RA may represent distinct diseases underpinned by distinct genetic contributions within and outside the HLA region. The SE predisposes mainly (but not only) to ACPA-positive RA, and the development of ACPA may be an intermediate step mediating the effect of the SE with RA susceptibility and/or severity. The effect of the HLA on ACPA-negative RA susceptibility is smaller and has been localized to some HLA positions also involved in ACPA-positive RA, but different amino acids confer risk and protection for ACPA-negative versus ACPA-positive RA. (See 'Relationship between HLA-DRB1 and ACPA' above.)

Non-HLA rheumatoid arthritis susceptibility genes – HLA may contribute less than 50 percent of the overall genetic risk for RA. Loci within the HLA region other than HLA-DRB1 also contribute to susceptibility, and a number of genes outside of the HLA region also exhibit an association with RA, some of which are also associated with other autoimmune conditions. Most associated variants outside the HLA region are intergenic or intronic variants that do not confer a change in the amino acid sequence of gene protein products. (See 'Rheumatoid arthritis susceptibility genes outside the HLA region' above.)

Clinical use of genetic markers – Knowledge of a patient's HLA status and other genetic markers may be useful with further developments in diagnosis and screening, in estimating prognosis, and in predicting the response to specific therapies, but a role for HLA testing in RA outside the research setting has not been established. However, most of the information and hypotheses regarding clinical utility of genetic testing are based primarily upon data derived from White North American and northern European patients, and HLA typing of patients from other ethnic groups will have different implications. (See 'Clinical use of genetic markers' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Anne Barton, FRCP, PhD and Soumya Raychaudhuri, MD, PhD, who contributed to an earlier version of this topic review.

  1. McMichael AJ, Sasazuki T, McDevitt HO, Payne RO. Increased frequency of HLA-Cw3 and HLA-Dw4 in rheumatoid arthritis. Arthritis Rheum 1977; 20:1037.
  2. Stastny P. Association of the B-cell alloantigen DRw4 with rheumatoid arthritis. N Engl J Med 1978; 298:869.
  3. Okada Y, Wu D, Trynka G, et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014; 506:376.
  4. Han B, Diogo D, Eyre S, et al. 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.
  5. Raychaudhuri S, Sandor C, Stahl EA, et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet 2012; 44:291.
  6. Nepom BS, Nepom GT. Polyglot and polymorphism. An HLA update. Arthritis Rheum 1995; 38:1715.
  7. Jia X, Han B, Onengut-Gumuscu S, et al. Imputing amino acid polymorphisms in human leukocyte antigens. PLoS One 2013; 8:e64683.
  8. 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.
  9. Willkens RF, Nepom GT, Marks CR, et al. Association of HLA-Dw16 with rheumatoid arthritis in Yakima Indians. Further evidence for the "shared epitope" hypothesis. Arthritis Rheum 1991; 34:43.
  10. Nepom GT, Byers P, Seyfried C, et al. HLA genes associated with rheumatoid arthritis. Identification of susceptibility alleles using specific oligonucleotide probes. Arthritis Rheum 1989; 32:15.
  11. Wordsworth BP, Lanchbury JS, Sakkas LI, et al. HLA-DR4 subtype frequencies in rheumatoid arthritis indicate that DRB1 is the major susceptibility locus within the HLA class II region. Proc Natl Acad Sci U S A 1989; 86:10049.
  12. Rønningen KS, Spurkland A, Egeland T, et al. Rheumatoid arthritis may be primarily associated with HLA-DR4 molecules sharing a particular sequence at residues 67-74. Tissue Antigens 1990; 36:235.
  13. Fries JF, Wolfe F, Apple R, et al. HLA-DRB1 genotype associations in 793 white patients from a rheumatoid arthritis inception cohort: frequency, severity, and treatment bias. Arthritis Rheum 2002; 46:2320.
  14. Mattey DL, Dawes PT, Gonzalez-Gay MA, et al. HLA-DRB1 alleles encoding an aspartic acid at position 70 protect against development of rheumatoid arthritis. J Rheumatol 2001; 28:232.
  15. de Vries N, Tijssen H, van Riel PL, van de Putte LB. Reshaping the shared epitope hypothesis: HLA-associated risk for rheumatoid arthritis is encoded by amino acid substitutions at positions 67-74 of the HLA-DRB1 molecule. Arthritis Rheum 2002; 46:921.
  16. van der Woude D, Lie BA, Lundström E, et al. 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.
  17. Leslie S, Donnelly P, McVean G. A statistical method for predicting classical HLA alleles from SNP data. Am J Hum Genet 2008; 82:48.
  18. Scally SW, Petersen J, Law SC, et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. J Exp Med 2013; 210:2569.
  19. Scally SW, Law SC, Ting YT, et al. Molecular basis for increased susceptibility of Indigenous North Americans to seropositive rheumatoid arthritis. Ann Rheum Dis 2017; 76:1915.
  20. Hill JA, Southwood S, Sette A, et al. Cutting edge: the conversion of arginine to citrulline allows for a high-affinity peptide interaction with the rheumatoid arthritis-associated HLA-DRB1*0401 MHC class II molecule. J Immunol 2003; 171:538.
  21. Reviron D, Perdriger A, Toussirot E, et al. Influence of shared epitope-negative HLA-DRB1 alleles on genetic susceptibility to rheumatoid arthritis. Arthritis Rheum 2001; 44:535.
  22. Bossini-Castillo L, de Kovel C, Kallberg H, et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann Rheum Dis 2015; 74:e15.
  23. Fu XT, Bono CP, Woulfe SL, et al. Pocket 4 of the HLA-DR(alpha,beta 1*0401) molecule is a major determinant of T cells recognition of peptide. J Exp Med 1995; 181:915.
  24. Kirschmann DA, Duffin KL, Smith CE, et al. Naturally processed peptides from rheumatoid arthritis associated and non-associated HLA-DR alleles. J Immunol 1995; 155:5655.
  25. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011; 365:2205.
  26. Bukhari M, Thomson W, Naseem H, et al. The performance of anti-cyclic citrullinated peptide antibodies in predicting the severity of radiologic damage in inflammatory polyarthritis: results from the Norfolk Arthritis Register. Arthritis Rheum 2007; 56:2929.
  27. Huizinga TW, Amos CI, van der Helm-van Mil AH, et al. Refining the complex rheumatoid arthritis phenotype based on specificity of the HLA-DRB1 shared epitope for antibodies to citrullinated proteins. Arthritis Rheum 2005; 52:3433.
  28. Irigoyen P, Lee AT, Wener MH, et al. 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.
  29. Michou L, Teixeira VH, Pierlot C, et al. Associations between genetic factors, tobacco smoking and autoantibodies in familial and sporadic rheumatoid arthritis. Ann Rheum Dis 2008; 67:466.
  30. Ding B, Padyukov L, Lundström E, et al. Different patterns of associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in the extended major histocompatibility complex region. Arthritis Rheum 2009; 60:30.
  31. Viatte S, Plant D, Bowes J, et al. Genetic markers of rheumatoid arthritis susceptibility in anti-citrullinated peptide antibody negative patients. Ann Rheum Dis 2012; 71:1984.
  32. Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum 2006; 54:38.
  33. van der Helm-van Mil AH, Verpoort KN, le Cessie S, et al. The HLA-DRB1 shared epitope alleles differ in the interaction with smoking and predisposition to antibodies to cyclic citrullinated peptide. Arthritis Rheum 2007; 56:425.
  34. Lee HS, Irigoyen P, Kern M, et al. Interaction between smoking, the shared epitope, and anti-cyclic citrullinated peptide: a mixed picture in three large North American rheumatoid arthritis cohorts. Arthritis Rheum 2007; 56:1745.
  35. Verpoort KN, Papendrecht-van der Voort EA, van der Helm-van Mil AH, et al. Association of smoking with the constitution of the anti-cyclic citrullinated peptide response in the absence of HLA-DRB1 shared epitope alleles. Arthritis Rheum 2007; 56:2913.
  36. Auger I, Sebbag M, Vincent C, et al. Influence of HLA-DR genes on the production of rheumatoid arthritis-specific autoantibodies to citrullinated fibrinogen. Arthritis Rheum 2005; 52:3424.
  37. van der Helm-van Mil AH, Verpoort KN, Breedveld FC, et al. 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.
  38. Danis VA, Millington M, Hyland V, et al. Increased frequency of the uncommon allele of a tumour necrosis factor alpha gene polymorphism in rheumatoid arthritis and systemic lupus erythematosus. Dis Markers 1995; 12:127.
  39. Mulcahy B, Waldron-Lynch F, McDermott MF, et al. Genetic variability in the tumor necrosis factor-lymphotoxin region influences susceptibility to rheumatoid arthritis. Am J Hum Genet 1996; 59:676.
  40. Tuokko J, Nejentsev S, Luukkainen R, et al. HLA haplotype analysis in Finnish patients with rheumatoid arthritis. Arthritis Rheum 2001; 44:315.
  41. Waldron-Lynch F, Adams C, Amos C, et al. Tumour necrosis factor 5' promoter single nucleotide polymorphisms influence susceptibility to rheumatoid arthritis (RA) in immunogenetically defined multiplex RA families. Genes Immun 2001; 2:82.
  42. van Krugten MV, Huizinga TW, Kaijzel EL, et al. Association of the TNF +489 polymorphism with susceptibility and radiographic damage in rheumatoid arthritis. Genes Immun 1999; 1:91.
  43. Wilson AG, de Vries N, van de Putte LB, Duff GW. A tumour necrosis factor alpha polymorphism is not associated with rheumatoid arthritis. Ann Rheum Dis 1995; 54:601.
  44. Low AS, Gonzalez-Gay MA, Akil M, et al. TNF +489 polymorphism does not contribute to susceptibility to rheumatoid arthritis. Clin Exp Rheumatol 2002; 20:829.
  45. Yen JH, Chen CJ, Tsai WC, et al. Tumor necrosis factor promoter polymorphisms in patients with rheumatoid arthritis in Taiwan. J Rheumatol 2001; 28:1788.
  46. Vinasco J, Beraún Y, Nieto A, et al. Polymorphism at the TNF loci in rheumatoid arthritis. Tissue Antigens 1997; 49:74.
  47. Field M, Gallagher G, Eskdale J, et al. Tumor necrosis factor locus polymorphisms in rheumatoid arthritis. Tissue Antigens 1997; 50:303.
  48. Okada Y, Suzuki A, Ikari K, et al. Contribution of a Non-classical HLA Gene, HLA-DOA, to the Risk of Rheumatoid Arthritis. Am J Hum Genet 2016; 99:366.
  49. Yamada R, Yamamoto K. Mechanisms of disease: genetics of rheumatoid arthritis--ethnic differences in disease-associated genes. Nat Clin Pract Rheumatol 2007; 3:644.
  50. Kochi Y, Suzuki A, Yamada R, Yamamoto K. Genetics of rheumatoid arthritis: underlying evidence of ethnic differences. J Autoimmun 2009; 32:158.
  51. Nelson JL, Boyer G, Templin D, et al. HLA antigens in Tlingit Indians with rheumatoid arthritis. Tissue Antigens 1992; 40:57.
  52. Williams RC, Jacobsson LT, Knowler WC, et al. Meta-analysis reveals association between most common class II haplotype in full-heritage Native Americans and rheumatoid arthritis. Hum Immunol 1995; 42:90.
  53. Harvey J, Lotze M, Arnett FC, et al. Rheumatoid arthritis in a Chippewa band. II. Field study with clinical serologic and HLA-D correlations. J Rheumatol 1983; 10:28.
  54. Ohta N, Nishimura YK, Tanimoto K, et al. Association between HLA and Japanese patients with rheumatoid arthritis. Hum Immunol 1982; 5:123.
  55. Molkentin J, Gregersen PK, Lin X, et al. Molecular analysis of HLA-DR beta and DQ beta polymorphism in Chinese with rheumatoid arthritis. Ann Rheum Dis 1993; 52:610.
  56. Guo J, Zhang T, Cao H, et al. Sequencing of the MHC region defines HLA-DQA1 as the major genetic risk for seropositive rheumatoid arthritis in Han Chinese population. Ann Rheum Dis 2019; 78:773.
  57. Yelamos J, Garcia-Lozano JR, Moreno I, et al. Association of HLA-DR4-Dw15 (DRB1*0405) and DR10 with rheumatoid arthritis in a Spanish population. Arthritis Rheum 1993; 36:811.
  58. de Juan MD, Belmonte I, Barado J, et al. Differential associations of HLA-DR antigens with rheumatoid arthritis (RA) in Basques: high frequency of DR1 and DR10 and lack of association with HLA-DR4 or any of its subtypes. Tissue Antigens 1994; 43:320.
  59. Schiff B, Mizrachi Y, Orgad S, et al. Association of HLA-Aw31 and HLA-DR1 with adult rheumatoid arthritis. Ann Rheum Dis 1982; 41:403.
  60. de Vries N, Rønningen KS, Tilanus MG, et al. HLA-DR1 and rheumatoid arthritis in Israeli Jews: sequencing reveals that DRB1*0102 is the predominant HLA-DR1 subtype. Tissue Antigens 1993; 41:26.
  61. Hughes LB, Morrison D, Kelley JM, et al. The HLA-DRB1 shared epitope is associated with susceptibility to rheumatoid arthritis in African Americans through European genetic admixture. Arthritis Rheum 2008; 58:349.
  62. Boki KA, Panayi GS, Vaughan RW, et al. HLA class II sequence polymorphisms and susceptibility to rheumatoid arthritis in Greeks. The HLA-DR beta shared-epitope hypothesis accounts for the disease in only a minority of Greek patients. Arthritis Rheum 1992; 35:749.
  63. Okada Y, Kim K, Han B, et al. Risk for ACPA-positive rheumatoid arthritis is driven by shared HLA amino acid polymorphisms in Asian and European populations. Hum Mol Genet 2014; 23:6916.
  64. Reynolds RJ, Ahmed AF, Danila MI, et al. HLA-DRB1-associated rheumatoid arthritis risk at multiple levels in African Americans: hierarchical classification systems, amino acid positions, and residues. Arthritis Rheumatol 2014; 66:3274.
  65. Saxena R, Plenge RM, Bjonnes AC, et al. A Multinational Arab Genome-Wide Association Study Identifies New Genetic Associations for Rheumatoid Arthritis. Arthritis Rheumatol 2017; 69:976.
  66. Danila MI, Laufer VA, Reynolds RJ, et al. Dense Genotyping of Immune-Related Regions Identifies Loci for Rheumatoid Arthritis Risk and Damage in African Americans. Mol Med 2017; 23:177.
  67. Padyukov L. Genetics of rheumatoid arthritis. Semin Immunopathol 2022; 44:47.
  68. Ishigaki K, Sakaue S, Terao C, et al. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat Genet 2022; 54:1640.
  69. Saevarsdottir S, Stefansdottir L, Sulem P, et al. Multiomics analysis of rheumatoid arthritis yields sequence variants that have large effects on risk of the seropositive subset. Ann Rheum Dis 2022; 81:1085.
  70. Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet 2004; 75:330.
  71. Vang T, Congia M, Macis MD, et al. Autoimmune-associated lymphoid tyrosine phosphatase is a gain-of-function variant. Nat Genet 2005; 37:1317.
  72. Zhang J, Zahir N, Jiang Q, et al. The autoimmune disease-associated PTPN22 variant promotes calpain-mediated Lyp/Pep degradation associated with lymphocyte and dendritic cell hyperresponsiveness. Nat Genet 2011; 43:902.
  73. Watford WT, Hissong BD, Bream JH, et al. Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4. Immunol Rev 2004; 202:139.
  74. Remmers EF, Plenge RM, Lee AT, et al. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med 2007; 357:977.
  75. Plenge RM, Seielstad M, Padyukov L, et al. TRAF1-C5 as a risk locus for rheumatoid arthritis--a genomewide study. N Engl J Med 2007; 357:1199.
  76. Lee HS, Korman BD, Le JM, et al. Genetic risk factors for rheumatoid arthritis differ in Caucasian and Korean populations. Arthritis Rheum 2009; 60:364.
  77. Han TU, Bang SY, Kang C, Bae SC. TRAF1 polymorphisms associated with rheumatoid arthritis susceptibility in Asians and in Caucasians. Arthritis Rheum 2009; 60:2577.
  78. Thomson W, Barton A, Ke X, et al. Rheumatoid arthritis association at 6q23. Nat Genet 2007; 39:1431.
  79. Plenge RM, Cotsapas C, Davies L, et al. Two independent alleles at 6q23 associated with risk of rheumatoid arthritis. Nat Genet 2007; 39:1477.
  80. Orozco G, Hinks A, Eyre S, et al. Combined effects of three independent SNPs greatly increase the risk estimate for RA at 6q23. Hum Mol Genet 2009; 18:2693.
  81. Lee EG, Boone DL, Chai S, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science 2000; 289:2350.
  82. McGovern A, Schoenfelder S, Martin P, et al. Capture Hi-C identifies a novel causal gene, IL20RA, in the pan-autoimmune genetic susceptibility region 6q23. Genome Biol 2016; 17:212.
  83. Vossenaar ER, Radstake TR, van der Heijden A, et al. Expression and activity of citrullinating peptidylarginine deiminase enzymes in monocytes and macrophages. Ann Rheum Dis 2004; 63:373.
  84. Chang X, Yamada R, Suzuki A, et al. Localization of peptidylarginine deiminase 4 (PADI4) and citrullinated protein in synovial tissue of rheumatoid arthritis. Rheumatology (Oxford) 2005; 44:40.
  85. Foulquier C, Sebbag M, Clavel C, et al. Peptidyl arginine deiminase type 2 (PAD-2) and PAD-4 but not PAD-1, PAD-3, and PAD-6 are expressed in rheumatoid arthritis synovium in close association with tissue inflammation. Arthritis Rheum 2007; 56:3541.
  86. Suzuki A, Yamada R, Chang X, et al. Functional haplotypes of PADI4, encoding citrullinating enzyme peptidylarginine deiminase 4, are associated with rheumatoid arthritis. Nat Genet 2003; 34:395.
  87. Ikari K, Kuwahara M, Nakamura T, et al. Association between PADI4 and rheumatoid arthritis: a replication study. Arthritis Rheum 2005; 52:3054.
  88. Eyre S, Bowes J, Diogo D, et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet 2012; 44:1336.
  89. Raychaudhuri S, Remmers EF, Lee AT, et al. Common variants at CD40 and other loci confer risk of rheumatoid arthritis. Nat Genet 2008; 40:1216.
  90. Barton A, Eyre S, Ke X, et al. Identification of AF4/FMR2 family, member 3 (AFF3) as a novel rheumatoid arthritis susceptibility locus and confirmation of two further pan-autoimmune susceptibility genes. Hum Mol Genet 2009; 18:2518.
  91. Ueda H, Howson JM, Esposito L, et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003; 423:506.
  92. Viatte S, Barton A. The role of rheumatoid arthritis genetic susceptibility markers in the prediction of erosive disease. Eur Musculoskelet Rev 2012; 7:102.
  93. Young A, Jaraquemada D, Awad J, et al. Association of HLA-DR4/Dw4 and DR2/Dw2 with radiologic changes in a prospective study of patients with rheumatoid arthritis. Preferential relationship with HLA-Dw rather than HLA-DR specificities. Arthritis Rheum 1984; 27:20.
  94. Calin A, Elswood J, Klouda PT. Destructive arthritis, rheumatoid factor, and HLA-DR4. Susceptibility versus severity, a case-control study. Arthritis Rheum 1989; 32:1221.
  95. van Zeben D, Hazes JM, Zwinderman AH, et al. Association of HLA-DR4 with a more progressive disease course in patients with rheumatoid arthritis. Results of a followup study. Arthritis Rheum 1991; 34:822.
  96. Eberhardt K, Fex E, Johnson U, Wollheim FA. Associations of HLA-DRB and -DQB genes with two and five year outcome in rheumatoid arthritis. Ann Rheum Dis 1996; 55:34.
  97. Valenzuela-Castaño A, García-López A, Pérez-Vilches D, et al. The predictive value of the HLA shared epitope for severity of radiological joint damage in patients with rheumatoid arthritis. A 10 year observational prospective study. J Rheumatol 2000; 27:571.
  98. Rau R, Herborn G, Zueger S, Fenner H. The effect of HLA-DRB1 genes, rheumatoid factor, and treatment on radiographic disease progression in rheumatoid arthritis over 6 years. J Rheumatol 2000; 27:2566.
  99. Olsen NJ, Callahan LF, Brooks RH, et al. Associations of HLA-DR4 with rheumatoid factor and radiographic severity in rheumatoid arthritis. Am J Med 1988; 84:257.
  100. McMahon MJ, Hillarby MC, Clarkson RW, et al. Major histocompatibility complex variants and articular disease severity in rheumatoid arthritis. Br J Rheumatol 1993; 32:899.
  101. Viatte S, Plant D, Han B, et al. Association of HLA-DRB1 haplotypes with rheumatoid arthritis severity, mortality, and treatment response. JAMA 2015; 313:1645.
  102. Kim HY, Min JK, Yang HI, et al. The impact of HLA-DRB1*0405 on disease severity in Korean patients with seropositive rheumatoid arthritis. Br J Rheumatol 1997; 36:440.
  103. Wakitani S, Murata N, Toda Y, et al. The relationship between HLA-DRB1 alleles and disease subsets of rheumatoid arthritis in Japanese. Br J Rheumatol 1997; 36:630.
  104. Wordsworth P, Pile KD, Buckely JD, et al. HLA heterozygosity contributes to susceptibility to rheumatoid arthritis. Am J Hum Genet 1992; 51:585.
  105. Weyand CM, Hicok KC, Conn DL, Goronzy JJ. The influence of HLA-DRB1 genes on disease severity in rheumatoid arthritis. Ann Intern Med 1992; 117:801.
  106. Gorman JD, Lum RF, Chen JJ, et al. Impact of shared epitope genotype and ethnicity on erosive disease: a meta-analysis of 3,240 rheumatoid arthritis patients. Arthritis Rheum 2004; 50:400.
  107. Farragher TM, Goodson NJ, Naseem H, et al. Association of the HLA-DRB1 gene with premature death, particularly from cardiovascular disease, in patients with rheumatoid arthritis and inflammatory polyarthritis. Arthritis Rheum 2008; 58:359.
  108. Socioeconomic deprivation and rheumatoid disease: what lessons for the health service? ERAS Study Group. Early Rheumatoid Arthritis Study. Ann Rheum Dis 2000; 59:794.
  109. Gonzalez-Juanatey C, Testa A, Garcia-Castelo A, et al. HLA-DRB1 status affects endothelial function in treated patients with rheumatoid arthritis. Am J Med 2003; 114:647.
  110. Gonzalez-Gay MA, Gonzalez-Juanatey C, Lopez-Diaz MJ, et al. HLA-DRB1 and persistent chronic inflammation contribute to cardiovascular events and cardiovascular mortality in patients with rheumatoid arthritis. Arthritis Rheum 2007; 57:125.
  111. Mattey DL, Thomson W, Ollier WE, et al. Association of DRB1 shared epitope genotypes with early mortality in rheumatoid arthritis: results of eighteen years of followup from the early rheumatoid arthritis study. Arthritis Rheum 2007; 56:1408.
  112. van Steenbergen HW, Raychaudhuri S, Rodríguez-Rodríguez L, et al. Association of valine and leucine at HLA-DRB1 position 11 with radiographic progression in rheumatoid arthritis, independent of the shared epitope alleles but not independent of anti-citrullinated protein antibodies. Arthritis Rheumatol 2015; 67:877.
  113. Viatte S, Barton A. Genetics of rheumatoid arthritis susceptibility, severity, and treatment response. Semin Immunopathol 2017; 39:395.
  114. Kurreeman FA, Padyukov L, Marques RB, et al. A candidate gene approach identifies the TRAF1/C5 region as a risk factor for rheumatoid arthritis. PLoS Med 2007; 4:e278.
  115. Viatte S, Plant D, Lunt M, et al. Investigation of rheumatoid arthritis genetic susceptibility markers in the early rheumatoid arthritis study further replicates the TRAF1 association with radiological damage. J Rheumatol 2013; 40:144.
  116. Mohamed RH, Pasha HF, El-Shahawy EE. Influence of TRAF1/C5 and STAT4 genes polymorphisms on susceptibility and severity of rheumatoid arthritis in Egyptian population. Cell Immunol 2012; 273:67.
  117. van Steenbergen HW, Rodríguez-Rodríguez L, Berglin E, et al. A genetic study on C5-TRAF1 and progression of joint damage in rheumatoid arthritis. Arthritis Res Ther 2015; 17:1.
  118. Lee JC, Espéli M, Anderson CA, et al. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell 2013; 155:57.
  119. Liu C, Batliwalla F, Li W, et al. Genome-wide association scan identifies candidate polymorphisms associated with differential response to anti-TNF treatment in rheumatoid arthritis. Mol Med 2008; 14:575.
  120. Cui J, Stahl EA, Saevarsdottir S, et al. Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis. PLoS Genet 2013; 9:e1003394.
  121. Umiċeviċ Mirkov M, Cui J, Vermeulen SH, et al. Genome-wide association analysis of anti-TNF drug response in patients with rheumatoid arthritis. Ann Rheum Dis 2013; 72:1375.
  122. Taylor JC, Bongartz T, Massey J, et al. Genome-wide association study of response to methotrexate in early rheumatoid arthritis patients. Pharmacogenomics J 2018; 18:528.
  123. Massey J, Plant D, Hyrich K, et al. Genome-wide association study of response to tumour necrosis factor inhibitor therapy in rheumatoid arthritis. Pharmacogenomics J 2018; 18:657.
  124. Jiang X, Askling J, Saevarsdottir S, et al. A genetic risk score composed of rheumatoid arthritis risk alleles, HLA-DRB1 haplotypes, and response to TNFi therapy - results from a Swedish cohort study. Arthritis Res Ther 2016; 18:288.
  125. O'Dell JR, Nepom BS, Haire C, et al. HLA-DRB1 typing in rheumatoid arthritis: predicting response to specific treatments. Ann Rheum Dis 1998; 57:209.
  126. Plenge RM, Criswell LA. Genetic variants that predict response to anti-tumor necrosis factor therapy in rheumatoid arthritis: current challenges and future directions. Curr Opin Rheumatol 2008; 20:145.
  127. Plant D, Wilson AG, Barton A. Genetic and epigenetic predictors of responsiveness to treatment in RA. Nat Rev Rheumatol 2014; 10:329.
  128. Nepom GT, Nepom BS. Prediction of susceptibility to rheumatoid arthritis by human leukocyte antigen genotyping. Rheum Dis Clin North Am 1992; 18:785.
  129. Kallberg H, Padyukov L, Plenge RM, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet 2007; 80:867.
  130. Knevel R, le Cessie S, Terao CC, et al. Using genetics to prioritize diagnoses for rheumatology outpatients with inflammatory arthritis. Sci Transl Med 2020; 12.
  131. Rojas-Villarraga A, Diaz FJ, Calvo-Páramo E, et al. Familial disease, the HLA-DRB1 shared epitope and anti-CCP antibodies influence time at appearance of substantial joint damage in rheumatoid arthritis. J Autoimmun 2009; 32:64.
  132. McDaniel DO, Alarcón GS, Pratt PW, Reveille JD. Most African-American patients with rheumatoid arthritis do not have the rheumatoid antigenic determinant (epitope). Ann Intern Med 1995; 123:181.
Topic 7487 Version 25.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟