Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Independent association of HLA-DPB1*02:01 with rheumatoid arthritis in Japanese populations

  • Hiroshi Furukawa ,

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Resources, Writing – original draft

    furukawa-tky@umin.org

    Affiliations Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan, Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan

  • Shomi Oka,

    Roles Investigation

    Affiliations Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan, Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan

  • Kota Shimada,

    Roles Resources

    Affiliations Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan, Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan

  • Atsushi Hashimoto,

    Roles Resources

    Affiliation Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan

  • Akiko Komiya,

    Roles Resources

    Affiliation Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan

  • Shinichiro Tsunoda,

    Roles Resources

    Affiliations Division of Rheumatology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan, Department of Rheumatology, Sumitomo Hospital, Osaka, Japan

  • Akiko Suda,

    Roles Resources

    Affiliation Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Japan

  • Satoshi Ito,

    Roles Resources

    Affiliation Department of Rheumatology, Niigata Rheumatic Center, Shibata, Japan

  • Koichiro Saisho,

    Roles Resources

    Affiliation Department of Orthopedics/Rheumatology, Miyakonojo Medical Center, National Hospital Organization, Miyakonojo, Japan

  • Masao Katayama,

    Roles Resources

    Affiliation Department of Internal Medicine, Nagoya Medical Center, National Hospital Organization, Nagoya, Japan

  • Satoshi Shinohara,

    Roles Resources

    Affiliation Tochigi Rheumatology Clinic, Utsunomiya, Japan

  • Takeo Sato,

    Roles Resources

    Affiliation Division of Rheumatology and Clinical Immunology, Jichi Medical University, Shimotsuke, Japan

  • Katsuya Nagatani,

    Roles Resources

    Affiliation Division of Rheumatology and Clinical Immunology, Jichi Medical University, Shimotsuke, Japan

  • Seiji Minota,

    Roles Resources

    Affiliation Division of Rheumatology and Clinical Immunology, Jichi Medical University, Shimotsuke, Japan

  • Toshihiro Matsui,

    Roles Resources

    Affiliation Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan

  • Naoshi Fukui,

    Roles Resources

    Affiliation Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan

  • Shoji Sugii,

    Roles Resources

    Affiliation Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan

  • Hajime Sano,

    Roles Resources

    Affiliation Division of Rheumatology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan

  • Kiyoshi Migita,

    Roles Resources

    Affiliations Clinical Research Center, Nagasaki Medical Center, National Hospital Organization, Omura, Japan, Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, Japan

  • Shouhei Nagaoka,

    Roles Resources

    Affiliation Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Japan

  •  [ ... ],
  • Shigeto Tohma

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliations Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan, Tokyo National Hospital, National Hospital Organization, Kiyose, Japan

  • [ view all ]
  • [ view less ]

Abstract

Objective

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized with joint destructions; environmental and genetic factors were thought to be involved in the etiology of RA. The production of anti-citrullinated peptide antibodies (ACPA) is specifically associated with RA. DRB1 is associated with the susceptibility of RA, especially ACPA-positive RA [ACPA(+)RA]. However, a few studies reported on the independent associations of DPB1 alleles with RA susceptibility. Thus, we investigated the independent association of DPB1 alleles with RA in Japanese populations.

Methods

Association analyses of DPB1 were conducted by logistic regression analysis in 1667 RA patients and 413 controls.

Results

In unconditioned analysis, DPB1*04:02 was nominally associated with the susceptibility of ACPA(+)RA (P = 0.0021, corrected P (Pc) = 0.0275, odds ratio [OR] 1.52, 95% confidence interval [CI] 1.16–1.99). A significant association of DPB1*02:01 with the susceptibility of ACPA(+)RA was observed, when conditioned on DRB1 (Padjusted = 0.0003, Pcadjusted = 0.0040, ORadjusted 1.47, 95%CI 1.19–1.81). DPB1*05:01 was tended to be associated with the protection against ACPA(+)RA, when conditioned on DRB1 (Padjusted = 0.0091, Pcadjusted = 0.1184, ORadjusted 0.78, 95%CI 0.65–0.94). When conditioned on DRB1, the association of DPB1*04:02 with ACPA(+)RA was disappeared. No association of DPB1 alleles with ACPA-negative RA was detected.

Conclusion

The independent association of DPB1*02:01 with Japanese ACPA(+)RA was identified.

Introduction

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized with synovial joint destructions and extra-articular manifestations. The etiology of RA is still unknown, but environmental and genetic factors were thought to be involved in the pathogenesis of RA [1,2,3]. Human leukocyte antigen (HLA) is the strongest genetic factor in RA and it was confirmed in genome wide association studies based on single nucleotide polymorphisms [4]. DRB1 was believed to be the most important locus in HLA for the susceptibility of RA; some DRB1 alleles were associated with the susceptibility of RA and have common motifs of amino acid residues at position 70–74 (QKRAA, RRRAA, or QRRAA) in DRβ chain [5]. These were designated as shared epitope (SE) alleles [6]. DRB1*04:01 was mainly associated with RA in European populations [5] and DRB1*04:05 in Asian [7]. Although both of DRB1*04:01 and DRB1*04:05 are SE alleles, these differences could be explained by the different frequencies of these susceptibility alleles for RA in different ethnic groups. The production of anti-citrullinated peptide antibodies (ACPA) is specifically associated with RA. ACPA-positive RA [ACPA(+)RA] is strongly associated with SE alleles, but ACPA-negative RA [ACPA(-)RA] is weakly [7,8,9].

Some reports also suggested that B, DQB1, or DPB1 would be involved in the pathogenesis of RA [10,11,12,13,14,15,16,17,18,19,20]. Since HLA region is in strong linkage disequilibrium, it is important to eliminate the effects of DRB1 to elucidate the role of other loci in HLA. The independent associations of amino acid residues in B and DPB1 loci were recently reported [21,22,23]. However, few studies reported on the independent associations of DPB1 alleles for RA susceptibility. Since DRB1 is the strongest genetic risk factor for ACPA(+)RA, we investigated the independent association of DPB1 alleles from DRB1 in Japanese ACPA(+)RA.

Materials and methods

Patients

One thousand six hundred sixty seven Japanese RA patients were recruited at Hyogo College of Medicine, Jichi Medical University, Miyakonojo Medical Center, Nagasaki Medical Center, Nagoya Medical Center, Niigata Rheumatic Center, Sagamihara National Hospital, Tochigi Rheumatology Clinic, Tokyo Metropolitan Tama Medical Center, or Yokohama Minami Kyosai Hospital. RA patients fulfilled the 1987 American College of Rheumatology criteria for RA [24] or the 2010 Rheumatoid Arthritis Classification Criteria [25]. Four hundred thirteen Japanese healthy controls (mean age ± SD, 39.3 ± 11.0 years, vs. ACPA(+)RA: P = 6.50X10-130, vs. ACPA(-)RA, P = 5.51X10-71, 61 male [14.8%], vs. ACPA(+)RA: P = 0.0792, vs. ACPA(-)RA, P = 0.2195) were recruited at Kanazawa University, Sagamihara National Hospital, and Teikyo University [26] or by the Pharma SNP Consortium (Tokyo, Japan) [27,28]. Rheumatoid factor and ACPA were measured by N-latex RF kit (Siemens Healthcare Diagnostics, München, Germany) or Mesacup-2 test CCP (Medical & Biological Laboratories, Nagoya, Japan), respectively. This study was reviewed and approved by Hyogo College of Medicine Research Ethics Committee, Jichi Medical University Research Ethics Committee, Miyakonojo Medical Center Research Ethics Committee, Nagasaki Medical Center Research Ethics Committee, Nagoya Medical Center Research Ethics Committee, Niigata Rheumatic Center Research Ethics Committee, Sagamihara National Hospital Research Ethics Committee, Tokyo Metropolitan Tama Medical Center Research Ethics Committee, Yokohama Minami Kyosai Hospital Research Ethics Committee, and University of Tsukuba Research Ethics Committee. Written informed consent was obtained from all study participants. This study was conducted in accordance with the principles expressed in the Declaration of Helsinki.

Genotyping of DRB1 and DPB1

Genotyping of DRB1 and DPB1 was performed by polymerase chain reaction with reverse sequence-specific oligonucleotide probes (WAKFlow HLA typing kit, Wakunaga Pharmaceutical Co., Ltd., Akitakata, Japan) and Bio-Plex 200 (Bio-Rad, Hercules, CA). SE alleles contain DRB1*01:01, DRB1*04:01, DRB1*04:05, DRB1*04:10, DRB1*10:01, and DRB1*14:06 [5]. Genotyping results for some of the RA patients and the healthy controls were previously reported [7,26,29,30,31,32].

Statistical analysis

Clinical features of the RA patients were analyzed by Fisher’s exact test using 2X2 contingency tables or Student’s t-test. Unconditioned logistic regression analysis under the additive model was performed to analyze nominal associations of HLA alleles with the susceptibility of RA. On the other hand, conditioned logistic regression analysis was used to investigate the independent contribution of each DPB1 allele from DRB1 to the susceptibility of RA. Padjusted and ORadjusted were calculated for DPB1 alleles, when conditioned on DRB1. Alleles detected in both case and control groups were tested. The two-locus analysis was also conducted by logistic regression analysis under the additive model to identify the primary role of associated DRB1 or DPB1 alleles. Haplotype frequencies of DRB1-DPB1 were estimated with expectation-maximization algorithm with SNPAlyze ver.8.0.4 Pro (Dynacom, Chiba, Japan) and Permutation P values were established by 100000 permutations. Logistic regression analysis under the additive model was also performed to analyze associations of amino acid residues; conditional logistic regression analysis was used to investigate the independent contribution of each DPβ chain amino acid residue from DRβ chain amino acid residues to the susceptibility of RA. Padjusted values were calculated for amino acid residues in the DPβ chains, when conditioned on DRβ chain amino acid residues. Multiple comparisons were adjusted by Bonferroni method; corrected P (Pc) values were derived from multiplying the P values by the number of alleles or amino acid residues tested.

Results

Clinical manifestations of RA patients

Characteristics of RA patients are shown in Table 1. Steinbrocker stage and class [33] were higher in ACPA(+)RA than ACPA(-)RA. The rheumatoid factor positivity rate was also higher.

Association of DPB1 with ACPA(+)RA

Association of DRB1 with ACPA(+)RA was confirmed (S1 Table), as reported in the previous study [7]; DRB1*04:05 and *04:01 were associated with the susceptibility of ACPA(+)RA and DRB1*04:06, *08:02, *08:03, *13:02, and *14:03 were protectively associated. Next, it was analyzed whether DPB1 was also associated with ACPA(+)RA (Table 2). In unconditioned analysis, DPB1*04:02 was nominally associated with the susceptibility of ACPA(+)RA (P = 0.0021, Pc = 0.0275, odds ratio [OR] 1.52, 95% confidence interval [CI] 1.16–1.99, Table 2, left column). Since DRB1 and DPB1 are in strong linkage disequilibrium, nominal associations of DPB1 alleles were influences by the associations of DRB1 alleles with ACPA(+)RA. In order to clarify whether each DPB1 allele was independently associated with ACPA(+)RA, conditional logistic regression analysis was performed (Table 2, right column). The significant association of DPB1*02:01 with the susceptibility of ACPA(+)RA was observed, when conditioned on DRB1 (Padjusted = 0.0003, Pcadjusted = 0.0040, ORadjusted 1.47, 95%CI 1.19–1.81, Table 2, right column). DPB1*05:01 was tended to be associated with the protection against ACPA(+)RA, when conditioned on DRB1 (Padjusted = 0.0091, Pcadjusted = 0.1184, ORadjusted 0.78, 95%CI 0.65–0.94, Table 2, right column). When conditioned on DRB1, DPB1*04:02 was not associated with the susceptibility of ACPA(+)RA, suggesting the influence of DRB1 on the nominal association of DPB1*04:02. Thus, DPB1*02:01 was independently associated with the susceptibility of ACPA(+)RA.

thumbnail
Table 2. Conditional logistic regression analysis of DPB1 alleles in ACPA(+) RA and controls.

https://doi.org/10.1371/journal.pone.0204459.t002

In order to reveal whether each DRB1 allele influenced on the association of DPB1*02:01 with the susceptibility of ACPA(+)RA, conditional logistic regression analysis was conducted (Table 3). When conditioned on DRB1*04:05, the significant association of DPB1*02:01 with the susceptibility of ACPA(+)RA was observed (Padjusted = 0.0073, ORadjusted 1.29, 95%CI 1.07–1.56, Table 3). Because DRB1*04:05 is the strongest risk factor for RA in Asian [7], the influence of DRB1*04:05 on the nominal association of DPB1*02:01 would be strongest. However, the stronger association of DPB1*02:01 with the susceptibility of ACPA(+)RA was observed, when conditioned on SE alleles (Padjusted = 0.0016, ORadjusted 1.37, 95%CI 1.13–1.66, Table 3) or DRB1 (Padjusted = 0.0003, ORadjusted 1.47, 95%CI 1.19–1.81, Table 3). These data suggested that many DRB1 alleles including DRB1*04:05 had influenced on the nominal association of DPB1*02:01 with the susceptibility of ACPA(+)RA.

thumbnail
Table 3. Conditional logistic regression analysis of DPB1*02:01 between ACPA(+) RA and controls.

https://doi.org/10.1371/journal.pone.0204459.t003

The two-locus analysis was conducted to identify the primary role of DRB1*04:05 and DPB1*02:01 for the susceptibility of ACPA(+)RA (S2 Table). The OR for DPB1*02:01 in ACPA(+)RA patients with DRB1*04:05 was 1.45 (P = 0.0688, S2 Table), while the OR for DPB1*02:01 in ACPA(+)RA patients without DRB1*04:05 was 1.26 (P = 0.0356, S2 Table). On the other hand, the OR for DRB1*04:05 in ACPA(+)RA patients with DPB1*02:01 was 4.21 (P = 3.71X10-12, S2 Table), and the OR for DRB1*04:05 in ACPA(+)RA patients without DPB1*02:01 was 3.33 (P = 7.58X10-15, S2 Table). These results suggested the independent roles of DRB1*04:05 and DPB1*02:01 on the susceptibility of ACPA(+)RA.

When haplotype frequencies were compared between ACPA(+)RA patients and controls, three haplotypes including DRB1*04:05 were associated with the susceptibility of ACPA(+)RA (DRB1*04:05-DPB1*02:01; Permutation P<0.0001, DRB1*04:05-DPB1*04:02,; Permutation P = 0.0004, DRB1*04:05-DPB1*05:01; Permutation P<0.0001, S3 Table), suggesting the primary role of DRB1*04:05. On the other hand, some haplotypes including DPB1*02:01 were associated with the ACPA(+)RA susceptibility (DRB1*04:05-DPB1*02:01; Permutation P <0.0001, DRB1*09:01-DPB1*02:01; Permutation P = 0.0048, S3 Table) or the protection (DRB1*04:06-DPB1*02:01; Permutation P = 0.0149, DRB1*08:02-DPB1*02:01; Permutation P<0.0001, DRB1*13:02-DPB1*02:01; Permutation P = 0.0015, DRB1*15:01-DPB1*02:01; Permutation P = 0.0208, S3 Table), suggesting the influences of DRB1 alleles on the effects of DPB1*02:01. These data suggested the stronger effects of DRB1 alleles on the ACPA(+)RA susceptibility or the protection.

Association of DPB1 with ACPA(-)RA

It was analyzed whether DPB1 was also associated with ACPA(-)RA (Table 4). In unconditioned analysis, no DPB1 allele was associated with the susceptibility of ACPA(-)RA (Table 4, left column). In order to elucidate whether each DPB1 allele was independently associated with ACPA(-)RA, conditional logistic regression analysis was performed (Table 4, right column). No association of DPB1 alleles with the susceptibility of ACPA(-)RA was observed, when conditioned on DRB1. Association of DPB1 was also analyzed with overall RA (S4 Table). In unconditioned analysis, no DPB1 allele was associated with the overall RA (S4 Table, left column). When conditioned on DRB1, DPB1*02:01 was associated with the overall RA (S4 Table, right column). However, the association was weaker than ACPA(+)RA.

thumbnail
Table 4. Conditional logistic regression analysis of DPB1 alleles between ACPA(-)RA and controls.

https://doi.org/10.1371/journal.pone.0204459.t004

Association of DPβ chain amino acid residues with ACPA(+)RA

Association of DRβ chain amino acid residues with ACPA(+)RA was confirmed (S1 Fig), as reported in the previous study [7]; 10Y, 11S, 12T, 13H, 33N, 70D, 96Y, and 98K in the DRβ chain showed associations. Two amino acid residues, 36A 55A, in the DPβ chain were slightly associated with ACPA(+)RA in unconditioned analysis (Fig 1A). In order to clarify whether each DPβ chain amino acid residue was independently associated with ACPA(+)RA, conditional logistic regression analysis was performed. Five amino acid residues, 84G (P = 3.20X10-5, Pc = 0.0005, OR = 1.48, 95% CI 1.23–1.79), 85G (P = 3.20X10-5, Pc = 0.0005, OR = 1.48, 95% CI 1.23–1.79), 86P (P = 3.20X10-5, Pc = 0.0005, OR = 1.48, 95% CI 1.23–1.79), 87M (P = 3.20X10-5, Pc = 0.0005, OR = 1.48, 95% CI 1.23–1.79), and 96R (P = 3.94X10-5, Pc = 0.0006, OR = 1.48, 95% CI 1.23–1.78), in the DPβ chain were significantly associated with ACPA(+)RA, when conditioned on DRβ chain amino acid residues (Fig 1B). Since there are three haplotypes of these amino acid residues in the DPβ chain (84G-85G-86P-87M-96R, 84D-85E-86A-87V-96K, 84D-85E-86A-87V-96R), the results might reflect the effects of the haplotype of 84G-85G-86P-87M-96R on the susceptibility of ACPA(+)RA. The haplotype was actually associated with ACPA(+)RA in unconditioned analysis (P = 0.0078, OR = 1.24, 95% CI 1.06–1.44), or when conditioned on DRβ chain amino acid residues (Padjusted = 4.11X10-5, ORadjusted 1.47, 95%CI 1.22–1.77).

thumbnail
Fig 1. Associations of amino acid residues in the DPβ chains with ACPA(+)RA.

(A) Association was established between ACPA(+)RA and controls by logistic regression analysis. (B) Conditional logistic regression analysis was performed to clarify whether each DPβ chain amino acid residue was independently associated with ACPA(+)RA. Padjusted values were calculated for amino acid residues in the DPβ chains, when conditioned on DRβ chain amino acid residues. Corrected P (Pc) values were obtained by multiplying the P value by the number of amino acid residues tested. RA: rheumatoid arthritis, ACPA: anti-citrullinated peptide antibody, ACPA(+)RA: ACPA positive RA.

https://doi.org/10.1371/journal.pone.0204459.g001

Discussion

Although many investigations on the associations of DRB1 alleles with the susceptibility of RA were performed, relatively fewer studies on the genetic effects of DPB1 alleles for RA were conducted. There are a few direct reports on the independent association of DPB1 alleles for the susceptibility of RA, though results of some studies suggested the role of DPB1 alleles for RA [10,13,14,16,18]. DPB1*03:01 was associated with rheumatoid factor negative RA in European descent [10]. When arginine at position 71 of DRβ chain was possessed, DPB1*04:01 was associated with European RA [13]. DPB1*02:01 and DPB1*06:01 were associated with European RA without SE, whereas DPB1*04:01 was associated with European RA with SE [14]. DPB1*02:01 was associated with Japanese RA without DRB1*04:05 [16]. The association of DPB1*02:01 with the susceptibility of Japanese ACPA(+)RA and that of DPB1*04:01and DPB1*09:01 with the protection were suggested [18]. However, the results of these previous studies could not conclude the independent association of DPB1*02:01 from DRB1. In the present study, we directly revealed it in Japanese populations.

It was reported that phenylalanine at position 9 in DPβ chain was reported to be independently associated with European ACPA(+)RA [21]. Glycine at position 84 in DPβ chain was also independently associated with Japanese ACPA(+)RA [23]. However, the independently associated DPB1 alleles were not reported in these studies. In the present study, glycine at position 84 in DPβ chain was independently associated with ACPA(+)RA and 84G-85G-86P-87M-96R in DPβ chain was the ACPA(+)RA susceptible haplotype. This ACPA(+)RA associated haplotype was included in DPB1*02:01, DPB1*02:02, DPB1*04:01, DPB1*04:02, and DPB1*41:01. Since the alleles other than DPB1*02:01 were not directly associated with ACPA(+)RA susceptibility (Table 2, right column), DPB1*02:01 would be mainly contributed to the risk of ACPA(+)RA among them. The independent association of phenylalanine at position 9 in DPβ chain was not confirmed in the present study, though this amino acid residue was included in DPB1*02:01 and other alleles. This could be explained by the different distribution of HLA alleles in other ethnic populations. The amino acid residues 84, 85, 86, and 87 form the pocket 1 of DP peptide-binding groove [34]. This information suggested the involvement of peptides loaded on DP2 in the generation of ACPA or rheumatoid factor.

The association of DPB1*02:01 with ACPA(-)RA was not detected in the present study (Table 4), because of the limited sample size of ACPA(-)RA. Although the association of DPB1with ACPA(-)RA was not found in the study on European populations [22], weak association was shown around DP loci in the other study on Japanese populations [23]. Therefore, this could be detected in future large scale studies. The independent association of DPB1*02:01 with ACPA(+)RA should be replicated in future studies in Japanese populations and should be also analyzed in other populations. It was the limitation of this study that the population stratification was not excluded [35,36]. Thus, the present study revealed the independent association of DPB1*02:01 with ACPA(+)RA in Japanese populations.

Supporting information

S1 Fig. Associations of amino acid residues in the DRβ chains with ACPA(+)RA.

Association was established between ACPA(+)RA and controls by logistic regression analysis. Corrected P (Pc) values were obtained by multiplying the P value by the number of amino acid residues tested. RA: rheumatoid arthritis, ACPA: anti-citrullinated peptide antibody, ACPA(+)RA: ACPA positive RA.

https://doi.org/10.1371/journal.pone.0204459.s001

(PDF)

S1 Table. Logistic regression analysis of DRB1 alleles in ACPA(+) RA and controls.

RA: rheumatoid arthritis, ACPA: anticitrullinated peptide antibody, ACPA(+)RA: ACPA positive RA, OR: odds ratio, CI: confidence interval, P c: corrected P value, NS: not significant. Allele frequencies are shown in parenthesis (%). Association was tested by logistic regression analysis.

https://doi.org/10.1371/journal.pone.0204459.s002

(PDF)

S2 Table. Logistic regression analysis in the ACPA(+)RA patients and controls with or without DRB1*04:05 or DPB1*02:01.

RA: rheumatoid arthritis, ACPA: anti-citrullinated peptide antibody, ACPA(+)RA: ACPA positive RA, OR: odds ratio, CI: confidence interval. Association was tested between the RA patients and the controls with or without DRB1*04:05 or DPB1*02:01 by logistic regression analysis.

https://doi.org/10.1371/journal.pone.0204459.s003

(PDF)

S3 Table. DRB1-DPB1 haplotype frequency in the ACPA(+)RA patients and controls.

RA: rheumatoid arthritis, ACPA: anti-citrullinated peptide antibody, ACPA(+)RA: ACPA positive RA. Haplotypes with more than 1% frequency in controls are shown.

https://doi.org/10.1371/journal.pone.0204459.s004

(PDF)

S4 Table. Conditional logistic regression analysis of HLA-DPB1 alleles in the RA patients and controls.

RA: rheumatoid arthritis, OR: odds ratio, CI: confidence interval. Association was tested between the RA patients and the controls by Logistic regression analysis.

https://doi.org/10.1371/journal.pone.0204459.s005

(PDF)

Acknowledgments

We thank Ms. Mayumi Yokoyama and Ms. Tomomi Hanawa (Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital) for secretarial assistance.

References

  1. 1. Perricone C, Ceccarelli F, Valesini G. An overview on the genetic of rheumatoid arthritis: a never-ending story. Autoimmun Rev. 2011;10(10):599–608. Epub 2011 Apr 22. pmid:21545847
  2. 2. Scott IC, Steer S, Lewis CM, Cope AP. Precipitating and perpetuating factors of rheumatoid arthritis immunopathology: linking the triad of genetic predisposition, environmental risk factors and autoimmunity to disease pathogenesis. Best Pract Res Clin Rheumatol. 2011;25(4):447–68. pmid:22137917
  3. 3. Lewis SN, Nsoesie E, Weeks C, Qiao D, Zhang L. Prediction of disease and phenotype associations from genome-wide association studies. PLoS ONE. 2011;6(11):e27175. Epub 2011 Nov 4. pmid:22076134
  4. 4. Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K, et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506(7488):376–81. pmid:24390342.
  5. 5. Reveille JD. The genetic contribution to the pathogenesis of rheumatoid arthritis. Curr Opin Rheumatol. 1998;10(3):187–200. pmid:9608321
  6. 6. 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(11):1205–13. pmid:2446635.
  7. 7. Oka S, Furukawa H, Kawasaki A, Shimada K, Sugii S, Hashimoto A, et al. Protective effect of the HLA-DRB1*13:02 allele in Japanese rheumatoid arthritis patients. PLoS One. 2014;9(6):e99453. pmid:24911054
  8. 8. Holoshitz J. The rheumatoid arthritis HLA-DRB1 shared epitope. Curr Opin Rheumatol. 2010;22(3):293–8. pmid:20061955
  9. 9. Terao C, Ohmura K, Ikari K, Kochi Y, Maruya E, Katayama M, et al. ACPA-negative RA consists of two genetically distinct subsets based on RF positivity in Japanese. PLoS One. 2012;7(7):e40067. pmid:22792215.
  10. 10. Gao X, Fernandez-Vina M, Olsen NJ, Pincus T, Stastny P. HLA-DPB1*0301 is a major risk factor for rheumatoid factor-negative adult rheumatoid arthritis. Arthritis Rheum. 1991;34(10):1310–2. pmid:1930320.
  11. 11. Angelini G, Morozzi G, Delfino L, Pera C, Falco M, Marcolongo R, et al. Analysis of HLA DP, DQ, and DR alleles in adult Italian rheumatoid arthritis patients. Hum Immunol. 1992;34(2):135–41. pmid:1429034.
  12. 12. Ploski R, McDowell TL, Symons JA, Flato B, Duff GW, Thorsby E, et al. Interaction between HLA-DR and HLA-DP, and between HLA and interleukin 1 alpha in juvenile rheumatoid arthritis indicates heterogeneity of pathogenic mechanisms of the disease. Hum Immunol. 1995;42(4):343–7. pmid:7558921.
  13. 13. Perdriger A, Guggenbuhl P, Chales G, Le Dantec P, Yaouanq J, Genetet B, et al. The role of HLA-DR-DR and HLA-DR-DP interactions in genetic susceptibility to rheumatoid arthritis. Hum Immunol. 1996;46(1):42–8. pmid:9157088.
  14. 14. Seidl C, Koch U, Brunnler G, Buhleier T, Frank R, Moller B, et al. HLA-DR/DQ/DP interactions in rheumatoid arthritis. Eur J Immunogenet. 1997;24(5):365–76. pmid:9442804.
  15. 15. Zanelli E, Huizinga TW, Guerne PA, Vischer TL, Tiercy JM, Verduyn W, et al. An extended HLA-DQ-DR haplotype rather than DRB1 alone contributes to RA predisposition. Immunogenetics. 1998;48(6):394–401. pmid:9799335.
  16. 16. Tsuchiya K, Kimura A, Kondo M, Nishimura Y, Sasazuki T. Combination of HLA-A and HLA class II alleles controls the susceptibility to rheumatoid arthritis. Tissue Antigens. 2001;58(6):395–401. pmid:11929590.
  17. 17. Vignal C, Bansal AT, Balding DJ, Binks MH, Dickson MC, Montgomery DS, et al. Genetic association of the major histocompatibility complex with rheumatoid arthritis implicates two non-DRB1 loci. Arthritis Rheum. 2009;60(1):53–62. pmid:19116923.
  18. 18. Mitsunaga S, Suzuki Y, Kuwana M, Sato S, Kaneko Y, Homma Y, et al. Associations between six classical HLA loci and rheumatoid arthritis: a comprehensive analysis. Tissue Antigens. 2012;80(1):16–25. pmid:22471586.
  19. 19. Huang Z, Niu Q, Yang B, Zhang J, Yang M, Xu H, et al. Genetic polymorphism of rs9277535 in HLA-DP associated with rheumatoid arthritis and anti-CCP production in a Chinese population. Clin Rheumatol. 2018;2018(23):018–4030. pmid:29476350.
  20. 20. Jiang L, Jiang D, Han Y, Shi X, Ren C. Association of HLA-DPB1 polymorphisms with rheumatoid arthritis: A systemic review and meta-analysis. Int J Surg. 2018;52:98–104. pmid:29425827.
  21. 21. Raychaudhuri S, Sandor C, Stahl EA, Freudenberg J, Lee HS, Jia X, et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet. 2012;44(3):291–6. pmid:22286218.
  22. 22. Han B, Diogo D, Eyre S, Kallberg H, Zhernakova A, Bowes J, 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(4):522–32. pmid:24656864.
  23. 23. Okada Y, Suzuki A, Ikari K, Terao C, Kochi Y, Ohmura K, et al. Contribution of a Non-classical HLA Gene, HLA-DOA, to the Risk of Rheumatoid Arthritis. Am J Hum Genet. 2016;99(2):366–74. pmid:27486778.
  24. 24. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31(3):315–24. pmid:3358796
  25. 25. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–81. pmid:20872595.
  26. 26. Oka S, Furukawa H, Yasunami M, Kawasaki A, Nakamura H, Nakamura M, et al. HLA-DRB1 and DQB1 alleles in Japanese type 1 autoimmune hepatitis: The predisposing role of the DR4/DR8 heterozygous genotype. PLoS One. 2017;12(10):e0187325. pmid:29088299.
  27. 27. Kamatani N, Kawamoto M, Kitamura Y, Harigai M, Okumoto T, Sumino Y. Establishment of B-cell lines derived from 996 Japanese individuals. Tissue Culture Res Commun. 2004;23(2):71–80.
  28. 28. Kamitsuji S, Matsuda T, Nishimura K, Endo S, Wada C, Watanabe K, et al. Japan PGx Data Science Consortium Database: SNPs and HLA genotype data from 2994 Japanese healthy individuals for pharmacogenomics studies. J Hum Genet. 2015;60(6):319–26. pmid:25855068.
  29. 29. Furukawa H, Oka S, Shimada K, Sugii S, Ohashi J, Matsui T, et al. Association of human leukocyte antigen with interstitial lung disease in rheumatoid arthritis: A protective role for shared epitope. PLoS ONE. 2012;7(5):e33133. pmid:22586441
  30. 30. Furukawa H, Oka S, Shimada K, Sugii S, Hashimoto A, Komiya A, et al. Association of increased frequencies of HLA-DPB1*05:01 with the presence of anti-Ro/SS-A and anti-La/SS-B antibodies in Japanese rheumatoid arthritis and systemic lupus erythematosus patients. PLoS ONE. 2013;8(1):e53910. pmid:23320107
  31. 31. Furukawa H, Oka S, Kawasaki A, Shimada K, Sugii S, Matsushita T, et al. Human Leukocyte Antigen and Systemic Sclerosis in Japanese: The Sign of the Four Independent Protective Alleles, DRB1*13:02, DRB1*14:06, DQB1*03:01, and DPB1*02:01. PLoS One. 2016;11(4):e0154255. pmid:27116456.
  32. 32. Oka S, Furukawa H, Shimada K, Sugii S, Hashimoto A, Komiya A, et al. Association of Human Leukocyte Antigen Alleles with Chronic Lung Diseases in Rheumatoid Arthritis. Rheumatology (Oxford). 2016;55(7):1301–7.
  33. 33. Steinbrocker O, Traeger CH, Batterman RC. Therapeutic criteria in rheumatoid arthritis. J Am Med Assoc. 1949;140(8):659–62. pmid:18150288.
  34. 34. Diaz G, Amicosante M, Jaraquemada D, Butler RH, Guillen MV, Sanchez M, et al. Functional analysis of HLA-DP polymorphism: a crucial role for DPbeta residues 9, 11, 35, 55, 56, 69 and 84–87 in T cell allorecognition and peptide binding. Int Immunol. 2003;15(5):565–76. pmid:12697658.
  35. 35. Li Q, Yu K. Improved correction for population stratification in genome-wide association studies by identifying hidden population structures. Genet Epidemiol. 2008;32(3):215–26. pmid:18161052.
  36. 36. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38(8):904–9. pmid:16862161.