1
|
Tahamoli-Roudsari A, Rasouli-Saravani A, Basiri Z, Hajilooi M, Solgi G. A Potential Link Between HLA-DRB1/DQB1 Alleles and Response to Treatment in Rheumatoid Arthritis Patients. Int J Immunogenet 2025; 52:141-154. [PMID: 40195278 DOI: 10.1111/iji.12711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
The present study aimed to determine the association of HLA-DRB1/-DQB1 alleles with response to treatment and anti-citrullinated peptide antibody (ACPA) status in Iranian rheumatoid arthritis (RA) patients. A total of 167 RA patients, including 114 good responders (GRs) and 53 poor responders (PRs) as well as 330 ethnic-matched healthy controls, participated in this study. Disease activity and treatment response were assessed using the Disease Activity Score 28-joint (DAS28) scores during the 9-month post-treatment. HLA-DRB1/-DQB1 alleles were identified using polymerase chain reaction with a sequence-specific primers (PCR-SSP) method and compared between the study groups. Of the patients, 106 (63.5%) were ACPA-positive, 88 (52.7%) human leucocyte antigen (HLA)-shared epitope (SE)-positive, 64 (38.3%) ACPA+SE+ and 37 (22.2%) ACPA-SE-. HLA-SE alleles were significantly more frequent in patients (p = 3.2e - 05), in PR versus GR patients (p = 0.01) and in ACPA+ versus ACPA- patients (p = 0.009). PR patients had a higher prevalence of ACPA+SE+ compared to GR patients (p = 0.02). Higher frequencies of DRB1*04:02 (pc = 0.03), *04:04 (pc = 0.007), *04:05 (pc = 5.0e - 05), *10:01 (pc = 1.0e - 04), DQB1*03:02 (pc = 0.002) and *05:01 (pc = 0.002) alleles, and lower frequencies of DRB1*04:01 (pc = 0.007), *11:01 (pc = 0.03), *13:01 (pc = 0.03) and DQB1*06:03 (pc = 0.03) alleles were observed in patients compared to healthy controls. These findings suggest a relationship between HLA-SE alleles and ACPA development and a potential link between HLA-SE/non-SE alleles and therapeutic responses in RA patients.
Collapse
Affiliation(s)
- Ahmad Tahamoli-Roudsari
- Department of Internal Diseases Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Basiri
- Department of Internal Diseases Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdad Hajilooi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan, Iran
| |
Collapse
|
2
|
McDermott GC, Moll M, Cho MH, Hayashi K, Juge PA, Doyle TJ, Paudel ML, Kinney GL, Kronzer VL, Kim JS, O'Keeffe LA, Davis NA, Bernstein EJ, Dellaripa PF, Regan EA, Hunninghake GM, Silverman EK, Ash SY, San Jose Estepar R, Washko GR, Sparks JA. Polygenic risk scores for rheumatoid arthritis and idiopathic pulmonary fibrosis and associations with RA, interstitial lung abnormalities, and quantitative interstitial abnormalities among smokers. Semin Arthritis Rheum 2025; 72:152708. [PMID: 40090204 PMCID: PMC12048222 DOI: 10.1016/j.semarthrit.2025.152708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) facilitate construction of polygenic risk scores (PRSs) for rheumatoid arthritis (RA) and idiopathic pulmonary fibrosis (IPF). We investigated associations of RA and IPF PRSs with RA and high-resolution chest computed tomography (HRCT) parenchymal lung abnormalities. METHODS Participants in COPDGene, a prospective multicenter cohort of current/former smokers, had chest HRCT at study enrollment. Using genome-wide genotyping, RA and IPF PRSs were constructed using GWAS summary statistics. HRCT imaging underwent visual inspection for interstitial lung abnormalities (ILA) and quantitative CT (QCT) analysis using a machine-learning algorithm that quantified percentage of normal lung, interstitial abnormalities, and emphysema. RA was identified through self-report and DMARD use. We investigated associations of RA and IPF PRSs with RA, ILA, and QCT features using multivariable logistic and linear regression. RESULTS We analyzed 9,230 COPDGene participants (mean age 59.6 years, 46.4 % female, 67.2 % non-Hispanic White, 32.8 % Black/African American). In non-Hispanic White participants, RA PRS was associated with RA diagnosis (OR 1.32 per unit, 95 %CI 1.18-1.49) but not ILA or QCT features. Among non-Hispanic White participants, IPF PRS was associated with ILA (OR 1.88 per unit, 95 %CI 1.52-2.32) and quantitative interstitial abnormalities (adjusted β=+0.50 % per unit, p = 7.3 × 10-8) but not RA. There were no statistically significant associations among Black/African American participants. CONCLUSIONS RA and IPF PRSs were associated with their intended phenotypes among non-Hispanic White participants but performed poorly among Black/African American participants. PRS may have future application to risk stratify for RA diagnosis among patients with ILD or for ILD among patients with RA.
Collapse
Affiliation(s)
- Gregory C McDermott
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Matthew Moll
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Pulmonary, Allergy, Sleep and Critical Care Medicine Section, Department of Medicine, VA Boston Healthcare System, West Roxbury, MA, USA
| | - Michael H Cho
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Keigo Hayashi
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Pierre-Antoine Juge
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Université de Paris Cité, INSERM UMR 1152, F-75018 Paris, France; Service de Rhumatologie, Hôpital Bichat-Claude Bernard, AP-HP, F-75018 Paris, France
| | - Tracy J Doyle
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Misti L Paudel
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Gregory L Kinney
- Colorado School of Public Health, Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - John S Kim
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Lauren A O'Keeffe
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Natalie A Davis
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Elana J Bernstein
- Division of Rheumatology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Paul F Dellaripa
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Gary M Hunninghake
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Samuel Y Ash
- Division of Pulmonary and Critical Care Medicine, South Shore Health, South Weymouth, MA, USA
| | - Raul San Jose Estepar
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George R Washko
- Harvard Medical School, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Zou AE, Kongthong S, Mueller AA, Brenner MB. Fibroblasts in immune responses, inflammatory diseases and therapeutic implications. Nat Rev Rheumatol 2025:10.1038/s41584-025-01259-0. [PMID: 40369134 DOI: 10.1038/s41584-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Once regarded as passive bystander cells of the tissue stroma, fibroblasts have emerged as active orchestrators of tissue homeostasis and disease. From regulating immunity and controlling tissue remodelling to governing cell growth and differentiation, fibroblasts assume myriad roles in guiding normal tissue development, maintenance and repair. By comparison, in chronic inflammatory diseases such as rheumatoid arthritis, fibroblasts recruit and sustain inflammatory leukocytes, become dominant producers of pro-inflammatory factors and catalyse tissue destruction. In other disease contexts, fibroblasts promote fibrosis and impair host control of cancer. Single-cell studies have uncovered striking transcriptional and functional heterogeneity exhibited by fibroblasts in both normal tissues and diseased tissues. In particular, advances in the understanding of fibroblast pathology in rheumatoid arthritis have shed light on pathogenic fibroblast states in other chronic diseases. The differentiation and activation of these fibroblast states is driven by diverse physical and chemical cues within the tissue microenvironment and by cell-intrinsic signalling and epigenetic mechanisms. These insights into fibroblast behaviour and regulation have illuminated therapeutic opportunities for the targeted deletion or modulation of pathogenic fibroblasts across many diseases.
Collapse
Affiliation(s)
- Angela E Zou
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suppawat Kongthong
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alisa A Mueller
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA and Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Jajodia A, Mishra A, Doni Jayavelu N, Lambert K, Moss N, Yang Z, Cerosaletti K, Buckner JH, Hawkins RD. Functional dissection of noncoding variants associated with rheumatoid arthritis. Ann Rheum Dis 2025:S0003-4967(25)00890-8. [PMID: 40318978 DOI: 10.1016/j.ard.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVES Noncoding variants are critical to our understanding of the genetic basis of diseases and disorders such as rheumatoid arthritis (RA). While genome-wide association studies have identified regions of the genome associated with disease, functional studies are still lagging that can identify potentially causative variants. METHODS In order to functionally fine-map RA-associated variants, we identified variants at enhancers marked in primary activated T helper cells and conducted massively parallel reporter assay in these cells. RESULTS We found that combinations of functional variant genotypes are often exclusive to patients with RA. We leveraged 3-dimensional genome architecture and expression quantitative trait loci data to identify target genes of enhancers exhibiting allelic differences in activity. We confirmed enhancer activity and target gene interactions by Clustered Regularly Interpaced Short Palindromic Repeats Cas9 (CRISPR-Cas9) deletion in primary T cells. CONCLUSIONS The identification of functional enhancer variants suggests possible causal variants, and their target genes reveal known and novel genes as likely drivers of RA, as well as a means for therapeutic intervention.
Collapse
Affiliation(s)
- Ajay Jajodia
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Arpit Mishra
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Naresh Doni Jayavelu
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Nicholas Moss
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Zongchen Yang
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Jane H Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - R David Hawkins
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA; Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
5
|
Jha V, Freed BM, Sunderhaus ER, Lee JE, Prage EB, Miglani M, Rosloniec EF, Matsuda JL, Coulombe MG, McKee AS, Roark CL. Substitution of Glutamic Acid at Position 71 of DRβ1*04:01 and Collagen-Specific Tolerance Without Alloreactivity. Arthritis Rheumatol 2025; 77:526-535. [PMID: 39609038 PMCID: PMC12039470 DOI: 10.1002/art.43067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 10/11/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE The DRB1 locus is strongly associated with both susceptibility and resistance to rheumatoid arthritis (RA). DRB1 alleles encoding the VKA or VRA epitope in positions 11, 71, and 74 confer the highest risk of developing RA, whereas the allele encoding VEA is protective. We therefore investigated the feasibility of creating antigen-specific tolerance without inducing alloreactivity by replacing lysine with glutamic acid at position 71 in DRβ1*04:01. METHODS Individual DRB1 alleles and the DRB1*04:01K71E allele were cloned into T2 cell lines to measure binding of biotinylated peptides. Transgenic animals expressing DRB1*04:01, DRB1*01:01, or DRB1*04:01K71E were injected with collagen to measure T cell proliferation. Skin and bone marrow transplants between DRB1*04:01K71E and DRB1*04:01 mice were performed to determine if the single amino acid change at position 71 would be recognized as foreign. DRB1*04:01 mice transplanted with DRB1*04:01K71E bone marrow were injected with collagen to test if resistance to collagen sensitization could be transferred. RESULTS Replacing lysine (K) at position 71 in DRβ1*04:01 with glutamic acid (E) blocked collagen peptide binding and rendered the DRB1*04:01K71E mice resistant to collagen sensitization. Skin and bone marrow transplants from DRB1*04:01K71E mice were not rejected by DRB1*04:01 mice, suggesting the single E71 difference was not recognized as allogeneic. Bone marrow from DRB1*04:01K71E mice adoptively transferred antigen-specific tolerance to collagen to DRB1*04:01 mice. CONCLUSION These studies demonstrate that editing a single amino acid in DRβ1*04:01 blocks collagen peptide binding without inducing alloreactivity and could therefore represent a gene therapy approach to induce antigen-specific passive tolerance.
Collapse
Affiliation(s)
- Vibha Jha
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | - Brian M. Freed
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | | | - Jessica E. Lee
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | - Edward B. Prage
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | - Manjula Miglani
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | - Edward F. Rosloniec
- Lt. Col. Luke Weathers, Jr. Veterans Affairs Medical Center and University of Tennessee Health Science CenterMemphisTennessee
| | | | | | - Amy S. McKee
- University of Colorado, Anschutz Medical CampusAuroraColorado
| | | |
Collapse
|
6
|
Kaur M, Katz R, Criqui MH, Corr M, Post WS, Budoff M, Morris GP, Hughes-Austin JM. Associations of HLA-DRB1 shared epitope alleles with cardiovascular disease and events in a multi-ethnic community-living population: The multi-ethnic study of atherosclerosis (MESA). Hum Immunol 2025; 86:111265. [PMID: 40068347 DOI: 10.1016/j.humimm.2025.111265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) has been considered an independent risk factor for cardiovascular disease (CVD), where RA and CVD later manifest following genetic predisposition and an extended preclinical phase.TheHLA-DRB1Shared Epitope (SE)allelespredispose for RA andare associated withhigherrisk of CV mortality in RA patients, but have not been evaluated in a community-living population.Thus, we evaluated whetherHLA-DRB1 (SE) alleleswereassociated with subclinical CVD, CV events, and mortality in the Multi-Ethnic Study of Atherosclerosis (MESA). METHODS We performed HLA typing in 955 MESA participants and evaluated associations of SE alleles with coronary artery calcium (CAC) and abdominal aortic calcium (AAC); risk difference for CAC, AAC severity, and carotid intima media thickness (cIMT); and associations of SE with all-cause mortality, CVD and non-CVD death, and CV events. RESULTS Among 955 participants, 30 % were HLA-DRB1 SE positive. SE positivity was not significantly associated with higher risk of CAC, AAC, cIMT, CV events, or mortality. DRB1*10:01 demonstrated 2.63-fold higher risk for CAC (95 % CI 1.17-5.99); DRB1*14:02 demonstrated 42 % higher risk for AAC (95 % CI 1.17-1.74); and DRB1*04:05 demonstrated 24 % lower risk for AAC (95 % CI 0.58-0.99). CONCLUSION In a multi-ethnic community-living population, HLA-DRB1 SE alleles were not associated with subclinical CVD, CV events, or mortality. However, individual allele-associations with subclinical CVD suggested variability in risk.
Collapse
Affiliation(s)
- Manmeet Kaur
- Department of Medicine, University of California, San Diego, United States
| | - Ronit Katz
- Department of Obstetrics & Gynecology, University of Washington, United States
| | - Michael H Criqui
- Department of Family Medicine and Public Health, University of California, San Diego, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, United States
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, United States
| | - Matthew Budoff
- Department of Medicine, University of California, Los Angeles, United States
| | - Gerald P Morris
- Department of Pathology, University of California, San Diego, United States
| | - Jan M Hughes-Austin
- Department of Orthopaedic Surgery, University of California, San Diego, United States.
| |
Collapse
|
7
|
Kobayashi H, Miyazaki Y, Nakayamada S, Hanami K, Fukuyo S, Kubo S, Yamaguchi A, Inoue Y, Todoroki Y, Miyata H, Tanaka H, Fujino Y, Hirata S, Tanaka Y. Predictors of the effectiveness of first-line CTLA4-Ig in patients with RA: the FIRST registry. Rheumatology (Oxford) 2025; 64:2403-2410. [PMID: 39499185 DOI: 10.1093/rheumatology/keae598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/07/2024] Open
Abstract
OBJECTIVE This study aimed to elucidate which bio-naïve patients with RA are suitable for treatment with CTLA4-Ig. METHODS This study enrolled 953 patients with RA who were administered their first biological DMARD (CTLA4-Ig, n = 328; tumour necrosis factor inhibitor [TNFi], n = 625) from July 2013 to August 2022. The primary outcome was the Clinical Disease Activity Index (CDAI) remission rate at week 24 in each group, adjusted using propensity score (PS)-based inverse probability of treatment weighting (IPTW). RESULTS After minimizing selection bias using PS-based IPTW, the CDAI remission showed no significant difference between the CTLA4-Ig and TNFi groups (P = 0.464). Multivariable logistic regression analysis identified low baseline HAQ-Disability Index (DI) scores as a contributing factor to the CDAI remission rate at week 24 in both groups, along with high baseline ACPA levels in the CTLA4-Ig group. However, among patients with high baseline HAQ-DI scores and low baseline ACPA levels (≦57.2), the CDAI remission rate was significantly higher in the TNFi group (29.8%) compared with the CTLA4-Ig group (5.9%, P < 0.0001). Among patients with high baseline HAQ-DI scores and ACPA levels (>57.2), the CDAI remission rate was significantly higher in the CTLA4-Ig group (35.6%) compared with the TNFi group (22.1%, P = 0.0057). CONCLUSION Bio-naive RA patients with low HAQ-DI scores showed high treatment efficacy with no significant difference between CTLA4-Ig and TNFi. Among patients with high baseline HAQ-DI scores, TNFi and CTLA4-Ig were more likely to be effective in those with lower and higher baseline ACPA levels, respectively.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Japan
| | - Yusuke Miyazaki
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Kentaro Hanami
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Shunsuke Fukuyo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
- Department of Molecular Targeted Therapies (DMTT), School of Medicine, University of Occupational and Environmental Health, Japan
| | - Ayako Yamaguchi
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
- Department of Laboratory and Transfusion Medicine, University of Occupational and Environmental Health, Japan
| | - Yoshino Inoue
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
- Department of Molecular Targeted Therapies (DMTT), School of Medicine, University of Occupational and Environmental Health, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Yoshihisa Fujino
- Department of Environmental Epidemiology, University of Occupational and Environmental Health, Japan
| | - Shintaro Hirata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
8
|
Chen S, Dan L, Xiang L, He Q, Hu D, Gao Y. The role of gut flora-driven Th cell responses in preclinical rheumatoid arthritis. J Autoimmun 2025; 154:103426. [PMID: 40300482 DOI: 10.1016/j.jaut.2025.103426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder with an immune pathogenesis that evolves over decades. Preclinical RA (PreRA) represents a dynamic immune phase preceding clinical RA, marked by the loss of autoimmune tolerance, the appearance of tissue-invasive effector T cells, and the production of autoantibodies (such as antibodies against citrullinated proteins and rheumatoid factors). Extensive research has demonstrated that gut microbiota influence mucosal T-cell responses, driving the progression of PreRA through multiple mechanisms, including altered intestinal permeability, gene-environment interactions, bacterial antigenic specificity, molecular mimicry, and metabolite production. Environmental risk factors such as smoking, hormonal changes, and high-sodium (Na) diets, may contribute to RA pathogenesis via the gut microbiome. The next challenge in RA research lies in developing therapeutic strategies to intervene during the asymptomatic autoimmune phase, where dietary adjustments, natural compounds, probiotics, and other approaches could effectively modulate gut flora to prevent or delay RA onset.
Collapse
Affiliation(s)
- Shuanglan Chen
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lijuan Dan
- Department of Infection, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Li Xiang
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Qingman He
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Dongsen Hu
- Sichuan Jinxin Xi'nan Women's and Children's Hospital Co., Ltd, Chengdu, 610023, China
| | - Yongxiang Gao
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
9
|
Romero-Castillo L, Pandey RK, Xu B, Beusch CM, Oliveira-Coelho A, Zeqiraj K, Svensson C, Xu Z, Luo H, Sareila O, Sabatier P, Ge C, Cheng L, Urbonaviciute V, Krämer A, Lindgren C, Haag S, Viljanen J, Zubarev RA, Kihlberg J, Linusson A, Burkhardt H, Holmdahl R. Tolerogenic antigen-specific vaccine induces VISTA-enriched regulatory T cells and protects against arthritis in DRB1∗04:01 mice. Mol Ther 2025:S1525-0016(25)00313-2. [PMID: 40285352 DOI: 10.1016/j.ymthe.2025.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/26/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joint inflammation, cartilage damage, and bone erosion. Despite improvements with the introduction of biological disease-modifying anti-rheumatic drugs (DMARDs), RA remains an incurable life-long disease. Advancements in peptide-based vaccination may open new avenues for treating autoimmune diseases, including RA, by inducing immune tolerance while maintaining normal immune function. We have already demonstrated the efficacy of a potent vaccine against RA, consisting of the mouse major histocompatibility complex class II (Aq) protein bound to the immunodominant type II collagen peptide COL2259-273, which needed to be galactosylated at position 264. To translate the vaccine to humans and to further enhance vaccine efficacy, we modified the glycine residue at position 265 and conjugated it with the human DRB1∗04:01 molecule. Remarkably, this modified vaccine (named DR4-AL179) provided robust effectiveness in suppressing arthritis in DRB1∗04:01-expressing mice without the need for galactosylation at position 264. DR4-AL179 vaccination induces tolerance involving multiple immunoregulatory pathways, including the activation of V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA)-positive nonconventional regulatory T cells, which contribute to a potent suppressive response preventing arthritis development in mice. This modified RA vaccine offers a novel therapeutic potential for human autoimmune diseases.
Collapse
Affiliation(s)
- Laura Romero-Castillo
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden.
| | - Rajan Kumar Pandey
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Bingze Xu
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Christian M Beusch
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Ana Oliveira-Coelho
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Kejsi Zeqiraj
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Carolin Svensson
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Zhongwei Xu
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Huqiao Luo
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden; School of Medicine, Shanghai University, Shanghai 200444, China
| | - Outi Sareila
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden; Medical Inflammation Research, MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Pierre Sabatier
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Changrong Ge
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Lei Cheng
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Vilma Urbonaviciute
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Alexander Krämer
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | | | - Sabrina Haag
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden
| | - Johan Viljanen
- Department of Chemistry-BMC, Uppsala University, 75237 Uppsala, Sweden
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden; Department of Pharmacological & Technological Chemistry, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Jan Kihlberg
- Department of Chemistry-BMC, Uppsala University, 75237 Uppsala, Sweden
| | - Anna Linusson
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, & Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Division of Rheumatology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17176 Stockholm, Sweden; Medical Inflammation Research, MediCity Research Laboratory, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
10
|
González-Muñoz S, Long Y, Guzmán-Jiménez A, Cerván-Martín M, Higueras-Serrano I, Castilla JA, Clavero A, Garrido N, Luján S, Yang X, Guo X, Liu J, Bassas L, Seixas S, Gonçalves J, Lopes AM, Larriba S, Bossini-Castillo L, Palomino-Morales RJ, Wang C, Hu Z, Carmona FD. Trans-ethnic GWAS meta-analysis of idiopathic spermatogenic failure highlights the immune-mediated nature of Sertoli cell-only syndrome. Commun Biol 2025; 8:571. [PMID: 40188177 PMCID: PMC11972312 DOI: 10.1038/s42003-025-08001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Non-obstructive azoospermia, a severe form of male infertility caused by spermatogenic failure (SPGF), has a largely unknown genetic basis across ancestries. To our knowledge, this is the first trans-ethnic meta-analysis of genome-wide association studies on SPGF, involving 2255 men with idiopathic SPGF and 3608 controls from European and Asian populations. Using logistic regression and inverse variance methods, we identify two significant genetic associations with Sertoli cell-only (SCO) syndrome, the most extreme SPGF phenotype. The G allele of rs34915133, in the major histocompatibility complex class II region, significantly increases SCO risk (P = 5.25E-10, OR = 1.57), supporting a potential immune-related cause. Additionally, the rs10842262 variant in the SOX5 gene region is also a genetic marker of SCO (P = 5.29E-09, OR = 0.72), highlighting the key role of this gene in the male reproductive function. Our findings reveal shared genetic factors in male infertility across ancestries and provide insights into the molecular mechanisms underlying SCO.
Collapse
Affiliation(s)
- Sara González-Muñoz
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Yichen Long
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Andrea Guzmán-Jiménez
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miriam Cerván-Martín
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, Granada, Spain
| | - Inmaculada Higueras-Serrano
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - José A Castilla
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Departamento de Anatomía y Embriología Humana, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Ana Clavero
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Nicolás Garrido
- IVIRMA Global Research Alliance. IVI Foundation, Health Research Institute La Fe, Valencia, Spain
- Servicio de Urología. Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Saturnino Luján
- Servicio de Urología. Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Susana Seixas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - João Gonçalves
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
- ToxOmics - Centro de Toxicogenómica e Saúde Humana, Nova Medical School, Lisbon, Portugal
| | - Alexandra M Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Center for Predictive and Preventive Genetics, Institute for Cell and Molecular Biology, University of Porto, Porto, Portugal
| | - Sara Larriba
- Immune-Inflammatory Processes and Gene Therapeutics Group, Genes, Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Rogelio J Palomino-Morales
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China.
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - F David Carmona
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain.
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| |
Collapse
|
11
|
Wang WH, Xia MH, Liu XR, Lei SF, He P. A Bibliometric Analysis of GWAS on Rheumatoid Arthritis from 2002 to 2024. Hum Hered 2025; 90:18-32. [PMID: 40179854 DOI: 10.1159/000543947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/24/2025] [Indexed: 04/05/2025] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) has become a serious threat to human health and quality of life worldwide. Previous studies have demonstrated that genetic factors play a crucial role in the onset and progression of RA. Due to the rapid development of genome-wide association study (GWAS) and large-scale genetic analysis, GWAS research on RA has received widespread attention in recent years. Therefore, we conducted a comprehensive visualization and bibliometric analysis of publications to identify hotspots and future trends in GWAS research on RA. METHODS Literature on RA and GWAS published between 2002 and 2024 was extracted from the Web of Science Core Collection database by strategic screening. Collected data were further analyzed by using VOSviewer, CiteSpace, and Excel. The collaborations networks of countries, authors, institutions, and the co-citation networks of publications were visualized. Finally, research hotspots and fronts were examined. RESULTS A total of 713 publications with 45,773 citations were identified. The number of publications and citations has had a significant surge since 2007. The United States contributed the most publications globally. Okada, Yukinori, was the most influential author. The most productive institution in this field was the University of Manchester. The analysis of keywords revealed that "mendelian randomization analysis", "association", "innate", "instruments", "bias", "pathogenesis", and "genome-wide association study" are likely to be the frontiers of research in this field. CONCLUSION This study can be used to predict future research advances in the fields of GWAS on RA and helps to promote academic collaboration among scholars.
Collapse
Affiliation(s)
- Wen-Hui Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China,
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China,
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China,
| | - Ming-Hui Xia
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Xin-Ru Liu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
- Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
12
|
Kubo S, Tanaka Y. Pursuing Precision Medicine in Managing Rheumatoid Arthritis. Int J Rheum Dis 2025; 28:e70239. [PMID: 40269471 PMCID: PMC12018724 DOI: 10.1111/1756-185x.70239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Rheumatoid arthritis, characterized by immune dysregulation and joint destruction, is managed through a stepwise algorithm that combines methotrexate with biological and targeted synthetic disease-modifying antirheumatic drugs. Despite considerable advances, the lack of reliable biomarkers for selecting the most effective medication, especially in Phase II and beyond, remains a significant obstacle. As a result, achieving early clinical remission in all patients continues to be challenging. Rheumatoid arthritis demonstrates considerable clinical and molecular diversity, influenced by both genetic predispositions and environmental factors. Recent scientific and technological advances have shed light on the pathogenesis of rheumatoid arthritis, facilitating the stratification of patients into distinct phenotypic subgroups and potentially optimizing the choice of targeted therapies. However, persistent challenges include the high costs and logistical demands of these methodologies, as well as the complexities of conducting large-scale clinical trials. This review highlights the intricate pathogenesis of rheumatoid arthritis and underscores the need to address the disease's heterogeneity through precision medicine. Moving forward, a deeper investigation into rheumatoid arthritis pathogenesis, encompassing both genetic and environmental factors, is crucial. Pursuing precision medicine, grounded in accurate patient stratification, should be embraced as a "moonshot" objective in rheumatoid arthritis treatment, aiming to achieve transformative breakthroughs in management.
Collapse
Affiliation(s)
- Satoshi Kubo
- Department of Molecular Targeted TherapeuticsUniversity of Occupational and Environmental HealthKitakyushuJapan
- The First Department of Internal MedicineUniversity of Occupational and Environmental HealthKitakyushuJapan
| | - Yoshiya Tanaka
- Department of Molecular Targeted TherapeuticsUniversity of Occupational and Environmental HealthKitakyushuJapan
- The First Department of Internal MedicineUniversity of Occupational and Environmental HealthKitakyushuJapan
| |
Collapse
|
13
|
Tarjányi O, Olasz K, Rátky F, Sétáló G, Boldizsár F. Proteasome Inhibitors: Potential in Rheumatoid Arthritis Therapy? Int J Mol Sci 2025; 26:2943. [PMID: 40243560 PMCID: PMC11988683 DOI: 10.3390/ijms26072943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to the destruction of peripheral joint cartilage and bone tissue. Despite the advent of biological therapies in the past decades, the complete remission of RA patients is still out of reach. Therefore, the search for novel therapeutic approaches is still open in the field of RA. Proteasome inhibitors (PIs) were originally designed to be used in hematological malignancies like multiple myeloma. However, evidence has shown that they are potent inhibitors of the NF-κB pathway, which plays a pivotal role in inflammatory processes and RA. Furthermore, inhibition of cell activation and induction of apoptosis was also reported about PIs. In the present review, we summarize the current knowledge about the potential effects of PIs in RA based on reports from animal and human studies. We believe that there is substantial potential in the use of PIs in RA therapy either alone or in combination with the medications already used.
Collapse
Affiliation(s)
- Oktávia Tarjányi
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Katalin Olasz
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| | - Fanni Rátky
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - György Sétáló
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| |
Collapse
|
14
|
Zhu X, Lu H, Jia H, Wei X, Xue J, Li W, Zhang J, Wang Y, Yan J, Sun H, Ge Y, Zhang Z. Ferrostatin-1 reduces the inflammatory response of rheumatoid arthritis by decreasing the antigen presenting function of fibroblast-like synoviocytes. J Transl Med 2025; 23:280. [PMID: 40050869 PMCID: PMC11884008 DOI: 10.1186/s12967-025-06300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/23/2025] [Indexed: 03/10/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic chronic autoimmune disease with complex mechanism. Currently, ferroptosis is believed to play a role in it, but the specific mechanism is unknown, especially in immune response. In this study, we demonstrated that the high expression of major histocompatibility complex I (MHC-I) molecules in RA fibroblast-like synoviocytes (FLSs) is an antigen-presenting cell property and that this property is closely related to the increase in antigens after citrullination. Moreover, we detected higher levels of ferroptosis among FLSs from RA patient than among FLSs from OA patients. Ferroptosis can increase the expression of citrullinated histone H3 (cit-h3) by promoting the production of peptidyl arginine deiminase 4 (PAD4), which further promotes the expression of MHC-I molecules. We cocultured RA-FLSs treated with ferroptosis drugs with selected CD8 + T cells to assess the effect of ferroptosis on the endogenous antigen-presenting function of RA-FLSs. Ferroptosis promoted the proliferation of CD8 + T cells and the release of the inflammatory factors Tumor necrosis factor-α (TNF-α) and Interferon-gamma (IFN-γ), which enhanced the inflammatory effect. This phenomenon was also observed in a collagen-induced arthritis (CIA) mouse model. Finally, ferrostatin-1 (fer-1), a ferroptosis inhibitor, inhibited the above effects and reduced the release of inflammatory factors, indicating that ferroptosis may play a therapeutic role in RA and providing new ideas for the treatment of RA in the field of immunity.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanya Lu
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haonan Jia
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuemin Wei
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Xue
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjing Li
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Juan Zhang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanli Wang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingyao Yan
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haoyuan Sun
- Department of Osteology, Heilongjiang Provincial Hospital, Harbin, China
| | - Yanlei Ge
- Department of Respiratory Medicine, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Zhiyi Zhang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
15
|
Garrido-Mesa J, Brown MA. Antigen-driven T cell responses in rheumatic diseases: insights from T cell receptor repertoire studies. Nat Rev Rheumatol 2025; 21:157-173. [PMID: 39920282 DOI: 10.1038/s41584-025-01218-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Advances in T cell receptor (TCR) profiling techniques have substantially improved our ability to investigate T cell responses to antigens that are presented on HLA class I and class II molecules and associations between autoimmune T cells and rheumatic diseases. Early-stage studies in axial spondyloarthritis (axSpA) identified disease-associated T cell clonotypes, benefiting from the relative genetic homogeneity of the disease. However, both the genetic and the T cell immunological landscape are more complex in other rheumatic diseases. The diversity or redundancy in the TCR repertoire, epitope spreading over disease duration, genetic heterogeneity of HLA genes or other loci, and the diversity of epitopes contributing to disease pathogenesis and persistent inflammation are all likely to contribute to this complexity. TCR profiling holds promise for identifying key antigenic drivers and phenotypic T cell states that sustain autoimmunity in rheumatic diseases. Here, we review key findings from TCR repertoire studies in axSpA and other chronic inflammatory rheumatic diseases including psoriatic arthritis, rheumatoid arthritis, systemic lupus erythematosus and Sjögren syndrome. We explore how TCR profiling technologies, if applied to better controlled studies focused on early disease stages and genetically homogeneous subsets, can facilitate disease monitoring and the development of therapeutics targeting autoimmune T cells, their cognate antigens, or their underlying biology.
Collapse
Affiliation(s)
- Jose Garrido-Mesa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| | - Matthew A Brown
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK.
- Genomics England, London, UK.
| |
Collapse
|
16
|
Al-Balushi MS, Antony I, Al-Shirawi AH, Al-Riyami H, Al-Busaidi JZ, Koh CY, Al-Naamani KM, Hasson SS, Al-Jabri AA, Said EA. HLA-B*58 and HLA-C*2 Alleles Are Associated with the Occurrence of Rheumatoid Arthritis Among Omanis. J Clin Med 2025; 14:1219. [PMID: 40004750 PMCID: PMC11856611 DOI: 10.3390/jcm14041219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Rheumatoid arthritis (RA) is an autoimmune disease that is influenced by polymorphisms in the HLA molecules. Only a few studies assessed the presence of an association between HLA class I genes and RA. Moreover, ethnic background influences the association of HLA molecules and RA. HLA-I molecules are essential for the activation of CD8 T cells and natural killer (NK) cells. The implication of these cells in RA pathogenesis is controversial. Therefore, we investigated the presence of associations between HLA-I alleles and RA in Omani patients. Methods: HLA class I alleles were genotyped in a total of 206 volunteers (102 RA patients and 104 controls). The control group included volunteers who were not affected by any known disease. The Chi square test was used to investigate the significance of the associations between the HLA alleles and the occurrence of RA. A corrected p value (pc) was calculated using the Bonferroni correction. Results: The frequency of HLA-B*58 was ≈2.7-fold lower in RA patients (10.8%) compared to the control group (28.8%; pc = 0.0324). Moreover, the frequency of HLA-C*02 in RA patient was ≈8-fold higher compared to the control group (pc = 0.0104). Conclusions: This study is the first to demonstrate the presence of association between HLA-B*58 and HLA-C*02 and the occurrence of RA, which could guide future research on targeted therapies. It also suggests that these HLA alleles might influence CD8 T cells and NK cells implication in RA pathogenesis.
Collapse
Affiliation(s)
- Mohammed S. Al-Balushi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Irin Antony
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Ali H. Al-Shirawi
- Department of Medicine, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman
| | - Hamad Al-Riyami
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman
| | - Jumaa Z. Al-Busaidi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Crystal Y. Koh
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Khalid M. Al-Naamani
- Department of Medicine, The Medical City for Military and Security Services, P.O. Box 35, Muscat 123, Oman;
| | - Sidgi S. Hasson
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Ali A. Al-Jabri
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| | - Elias A. Said
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman (A.A.A.-J.)
| |
Collapse
|
17
|
Qi J, Wang T, Wang M, He P, Li Y, Shang L, Chen L, Wang X, Xu H, Ma C. Comparative study of the diversity of amino acids on human leucocyte antigen class II molecules in patients with acquired aplastic anaemia. Br J Haematol 2025; 206:735-748. [PMID: 39538961 DOI: 10.1111/bjh.19899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Human leucocyte antigen (HLA) class II molecules are critically involved in the pathology of acquired aplastic anaemia (AA) by regulating the immune response and autoreactive T cell-mediated haematopoietic cell death. In the study, amino acid residue variation and molecular structure of HLA class II have been initially investigated in 96 patients with AA. The frequencies of residues 9 and 57 in pocket 9 (P9) in DQB1, and amino acid positions 9, 11, 13, 16, 26, 38, 67 and 71 in the P4, P6 and P9 pockets in DRB1 were more prevalent among AA patients. By applying a multivariate recursive approach, the DRβ-Gln-16 (OR = 3.003, 95% CI = 1.468-6.145, pc = 0.003), DRβ-Ala-71 (OR = 1.924, 95% CI = 1.233-3.002, pc = 0.004) in P4/P7 and DQβ-Asp-57 (OR = 3.483, 95% CI = 1.079-11.242, pc = 0.037) in P9, these critical residues were significantly discovered as risk amino acid residues on the onset of AA, as well as associated with PNH-type cells and pathological somatic or cytogenetic mutations. In silico structural model analysis showed that identified DRβ-Ala-71 and DQβ-Asp-57 within the antigen-binding groove interacting with a more variable antigenic segments, may impact the repertoire of peptides presented, influence the interface HLA-antigen-T-cell receptor β (TCR β). These findings provided light on the immunogenetic pathophysiology of AA aetiology and their potential impact on upcoming immunotherapies.
Collapse
Affiliation(s)
- Jun Qi
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Tianju Wang
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Manni Wang
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Pengcheng He
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi Province, China
| | - Yuhui Li
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Lixia Shang
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Le Chen
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Xiaofang Wang
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Hua Xu
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| | - Chaofeng Ma
- HLA Laboratory, Shaanxi Province Blood Center, Institute of Xi'an Blood Bank, Xi'an, Shaanxi Province, China
| |
Collapse
|
18
|
Kronzer VL, Williamson KA, Hayashi K, Atkinson EJ, Crowson CS, Wang X, Cui J, Cerhan JR, Sletten JA, McDermott GC, Joerns EK, Vassallo R, Davis JM, Sparks JA. Uncovering specific genetic-respiratory disease endotypes for rheumatoid arthritis risk. Ann Rheum Dis 2025; 84:221-231. [PMID: 39919896 PMCID: PMC11822224 DOI: 10.1136/ard-2024-226391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVE We aimed to identify specific genetic-respiratory disease endotypes for rheumatoid arthritis (RA) risk. METHODS This case-control study used the Mass General Brigham (MGB) and Mayo Clinic (MC) Biobanks for discovery and replication, respectively. We matched criteria-confirmed incident RA cases to four non-RA controls on age, sex and health record history. Genetic exposures included the top 11 RA risk alleles, and a validated human leucocyte antigen (HLA) genetic risk score (GRS). We identified seven respiratory diseases by codes. Using logistic regression models adjusting for potential confounders, we estimated Rs with 95% CIs for the interactions between genetic and respiratory exposures for RA risk. RESULTS We identified 653 RA cases and 2607 controls in MGB, and 428 incident RA cases and 1712 non-RA controls in MC (mean age 64, 69% female). Respiratory diseases were associated with an increased risk of RA (OR 1.34, 95% CI 1.05, 1.71). Six out of 11 non-HLA RA risk alleles interacted strongly with specific respiratory diseases for RA risk, including NFKBIE and sinusitis (OR 5.49, 95% CI 1.56, 19.4 MGB; 5.26, 95% CI 2.00, 13.86 MC) and FAM167A and acute sinusitis for seronegative RA (OR 6.00, 95% CI 2.09, 17.24 MGB; 4.90, 95% CI 1.71, 14.1 MC). The RA HLA GRS interacted synergistically with interstitial lung disease for RA risk (OR 5.41, 95% CI 2.71, 10.8 in MC), with DPB1*02:01, DRB1*16:01 and DRB1*04:04 best predicting RA (positive predictive value 61%). CONCLUSION Several genetic-respiratory disease interactions strongly drive RA onset. If confirmed, these novel associations may reflect RA endotypes that can facilitate individualised prevention, diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | - Cynthia S Crowson
- Mayo Clinic, Rochester, NY, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, NY, USA
| | - Xiaosong Wang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jing Cui
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, NY, USA
| | | | - Gregory C McDermott
- Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/gregmcdermott
| | | | | | - John M Davis
- Mayo Clinic, Rochester, NY, USA. https://twitter.com/johndavis
| | - Jeffrey A Sparks
- Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/jeffsparks
| |
Collapse
|
19
|
Chin A, Small A, Wong SW, Wechalekar MD. T Cell Dysregulation in Rheumatoid Arthritis: from Genetic Susceptibility to Established Disease. Curr Rheumatol Rep 2025; 27:14. [PMID: 39862300 PMCID: PMC11762599 DOI: 10.1007/s11926-025-01180-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE OF REVIEW Rheumatoid arthritis (RA) is a complex autoimmune disease characterized by chronic inflammation of the synovial tissue, where T cells play a central role in pathogenesis. Recent research has identified T peripheral helper (Tph) cells as critical mediators of local B cell activation in inflamed tissues. This review synthesizes the latest advancements in our understanding the of the role of T cells in RA, from initiation to established disease. RECENT FINDINGS We explore recent advances regarding the genetic and epigenetic factors that predispose individuals to RA, the mechanisms of T cell activation and differentiation, and the interactions between T cells and other immune and stromal cells within the synovial microenvironment. The emergence of Tph cells as key drivers of RA pathobiology is highlighted, along with their potential as therapeutic targets. We also discuss the heterogeneity of T cell responses and their interplay with synovial cells, while addressing critical research gaps such as the drivers of T cell recruitment and the plasticity of synovial phenotypes. A deeper understanding of T cell dynamics in RA will provide valuable insights for developing targeted therapies to modulate T cell-mediated inflammation and improve patient outcomes.
Collapse
Affiliation(s)
- Athena Chin
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Annabelle Small
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Soon Wei Wong
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.
| |
Collapse
|
20
|
Rims C, Uchtenhagen H, Brooks K, Ng B, Posso SE, Carlin J, Kwok WW, Buckner JH, James EA. Antigen-specific T-cell frequency and phenotype mirrors disease activity in DRB1*04:04+ rheumatoid arthritis patients. Clin Exp Immunol 2025; 219:uxae102. [PMID: 39492692 PMCID: PMC11754868 DOI: 10.1093/cei/uxae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
Rheumatoid arthritis (RA) is associated with high-risk HLA class II alleles known as the "RA shared epitope." Among prevalent shared epitope alleles, study of DRB1*04:04 has been limited. To define relevant epitopes, we identified citrullinated peptide sequences from synovial antigens that were predicted to bind to HLA-DRB1*04:04 and utilized a systematic approach to confirm their binding and assess their recognition by CD4 T cells. After confirming the immunogenicity of 13 peptides derived from aggrecan, cartilage intermediate layer protein (CILP), α-enolase, vimentin, and fibrinogen, we assessed their recognition by T cells from a synovial tissue sample, observing measurable responses to 8 of the 13 peptides. We then implemented a multicolor tetramer panel to evaluate the frequency and phenotype of antigen-specific CD4 T cells in individuals with anti-citrullinated protein antibody-positive RA and controls. In subjects with RA, CILP-specific T-cell frequencies were significantly higher than those of other antigens. The surface phenotypes exhibited by antigen-specific T cells were heterogeneous, but Th1-like and Th2-like cells predominated. Stratifying based on disease status and activity, antigen-specific T cells were more frequent and most strongly polarized in RA subjects with high disease activity. In total, these findings identify novel citrullinated epitopes that can be used to interrogate antigen-specific CD4 T cells and show that antigen-specific T-cell frequency is elevated in subjects with high disease activity.
Collapse
Affiliation(s)
- Cliff Rims
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Hannes Uchtenhagen
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Kadin Brooks
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Bernard Ng
- VA National Rheumatology Program, Specialty Care Program Office, Washington DC, USA
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Sylvia E Posso
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jeffrey Carlin
- Department of Rheumatology, Virginia Mason Medical Center, Seattle, WA, USA
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Eddie A James
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
21
|
Kril I, Wiśniewski A, Tarnowska A, Lishchuk-Yakymovych K, Bojko Y, Kuśnierczyk P, Chopyak VV, Nowak I. Association of ERAP1 and ERAP2 gene polymorphisms and ERAP2 protein with the susceptibility and severity of rheumatoid arthritis in the Ukrainian population. Front Immunol 2025; 15:1519159. [PMID: 39906739 PMCID: PMC11790443 DOI: 10.3389/fimmu.2024.1519159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a long-term autoimmune disorder that primarily affects joints. Although RA is chiefly associated with HLA class II, nevertheless some HLA class I associations have also been observed. These molecules present antigenic peptides to CD8+ T lymphocytes and natural killer cells. HLA-I molecules bind their peptide cargo (8-10 amino acids long) in the endoplasmic reticulum. Peptides longer than 10 amino acids are trimmed by the endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 to fit the peptide binding groove of the HLA-I molecule. Here, we investigated the possible association of ERAP1 and ERAP2 polymorphisms with RA, and also any possible correlation between serum levels of the ERAP2 protein with disease severity. Methods We used Real-Time PCR to genotype ERAP1 and ERAP2 and ELISA test to detect ERAP2 protein. Results We found significant associations of ERAP1 rs30187, rs27044, and rs26618, as well as ERAP2 rs2248374, with susceptibility to RA. ERAP1 rs26653 and ERAP2 rs2248374 were also associated with the Disease Activity Score (DAS28), and some polymorphisms were also associated with anti-citrullinated protein or anti-mutated citrullinated vimentin antibodies. RA patients secreted higher concentrations of ERAP2 than controls. Patients with mild disease activity (DAS28 < 3.2) released a concentration of ERAP2 four times lower than that of patients with severe disease activity (DAS28 > 5.1). We detected a higher level of ERAP2 in rheumatoid factor (RF)-positive patients than in RF-negative patients. ERAP2 concentration above 5.85 ng/mL indicated a severe phase of RA. Conclusions Some ERAP1 and ERAP2 polymorphisms seem to be related to susceptibility to RA or the severity of the disease. The ERAP2 protein tested in serum could be a valuable biomarker of RA severity.
Collapse
Affiliation(s)
- Iryna Kril
- Department of Clinical Immunology and Allergology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Wiśniewski
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wrocław, Poland
| | - Agnieszka Tarnowska
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wrocław, Poland
| | | | - Yaryna Bojko
- Department of Clinical Immunology and Allergology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wrocław, Poland
| | - Valentyna V. Chopyak
- Department of Clinical Immunology and Allergology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Izabela Nowak
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
22
|
Sharma SD, Leung SH, Viatte S. Genetics of rheumatoid arthritis. Best Pract Res Clin Rheumatol 2024; 38:101968. [PMID: 38955657 DOI: 10.1016/j.berh.2024.101968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
In the past four decades, a plethora of genetic association studies have been carried out in cohorts of patients with rheumatoid arthritis. These studies have highlighted key aspects of disease pathogenesis and suggested causal mechanisms. In this review, we discuss major advances in our understanding of the genetic architecture of rheumatoid arthritis susceptibility, severity and treatment response and explain how genetics supports current models of disease pathogenesis and outcome. We outline future research directions, like Mendelian randomisation, and present a number of potential avenues for clinical translation, including risk and outcome prediction, patient stratification into treatment response groups and pharmacological applications.
Collapse
Affiliation(s)
- Seema D Sharma
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK; NIHR Manchester Musculoskeletal Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Shek H Leung
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| | - Sebastien Viatte
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK; NIHR Manchester Musculoskeletal Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
23
|
Hattori K, Tanaka S, Hashiba D, Tamura J, Etori K, Kageyama T, Ito T, Meguro K, Iwata A, Suto A, Suzuki K, Nakamura J, Ohtori S, Ziegler SF, Nakajima H. Synovial regulatory T cells expressing ST2 deteriorate joint inflammation through the suppression of immunoregulatory eosinophils. J Autoimmun 2024; 149:103333. [PMID: 39509740 DOI: 10.1016/j.jaut.2024.103333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic polyarthritis. It is well-established that helper T cells play crucial roles in the development and deterioration of RA. Recent studies also revealed the significant roles of regulatory T (Treg) cells in this context. Although Treg cells distributed in peripheral tissues exhibit various functions, the characteristics of synovial Treg cells remain unknown. In this study, we demonstrate that synovial Treg cells exacerbate synovial inflammation by reducing the number of immunoregulatory eosinophils through competitive consumption of IL-33. Synovial Treg cells expressed ST2 in a murine arthritis model, and surprisingly, Treg-specific ST2 knockout (ST2ΔTreg) mice exhibited attenuated arthritis. In ST2ΔTreg mice, an increase in immunoregulatory synovial eosinophils was observed. Additionally, immunoregulatory eosinophils were found to express ST2, and ST2-expressing Treg cells controlled the abundance of immunoregulatory eosinophils, possibly by consuming IL-33. Our results highlight that a subset of synovial Treg cells possesses the machinery to worsen arthritis by suppressing eosinophils. In the future landscape where Treg cell-based therapies are employed for autoimmune diseases, it is important to comprehend the characteristics of disease-related Treg cells. Understanding these aspects is crucial for ensuring safer treatment modalities that do not inadvertently worsen the diseases.
Collapse
MESH Headings
- Animals
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Eosinophils/immunology
- Eosinophils/metabolism
- Mice
- Interleukin-1 Receptor-Like 1 Protein/metabolism
- Interleukin-1 Receptor-Like 1 Protein/genetics
- Mice, Knockout
- Interleukin-33/metabolism
- Interleukin-33/immunology
- Interleukin-33/genetics
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- Synovial Membrane/metabolism
- Disease Models, Animal
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Humans
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Koto Hattori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Daisuke Hashiba
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan; Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Jun Tamura
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Keishi Etori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kazuyuki Meguro
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Junichi Nakamura
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Steven F Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101-2795, USA.
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| |
Collapse
|
24
|
Stadler M, Zhao SS, Bowes J. A review of the advances in understanding the genetic basis of spondylarthritis and emerging clinical benefit. Best Pract Res Clin Rheumatol 2024; 38:101982. [PMID: 39223061 DOI: 10.1016/j.berh.2024.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Spondyloarthropathies (SpA), including ankylosing spondylitis (AS) and psoriatic arthritis (PsA), have been shown to have a substantial genetic predisposition based on heritability estimates derived from family studies and genome-wide association studies (GWAS). GWAS have uncovered numerous genetic loci associated with susceptibility to SpA, with significant associations to human leukocyte antigen (HLA) genes, which are major genetic risk factors for both AS and PsA. Specific loci differentiating PsA from cutaneous-only psoriasis have been identified, though these remain limited. Further research with larger sample sizes is necessary to identify more PsA-specific genetic markers. Current research focuses on translating these genetic insights into clinical applications. For example, polygenic risk scores are showing promise for the classification of disease risk and diagnosis and future research should focus on refining these risk assessment tools to improve clinical outcomes for individuals with SpA. Addressing these challenges will help integrate genetic testing into patients care and impact clinical practice.
Collapse
Affiliation(s)
- Michael Stadler
- The Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Sizheng Steven Zhao
- The Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - John Bowes
- The Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| |
Collapse
|
25
|
Yao Y, Wang Q, Wei W. Association between iridocyclitis and immune-related disease: A 2-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40663. [PMID: 39612419 PMCID: PMC11608751 DOI: 10.1097/md.0000000000040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/06/2024] [Indexed: 12/01/2024] Open
Abstract
The genetic basis of iridocyclitis, an inflammatory eye disease, remains poorly understood, particularly in relation to autoimmune diseases. This study aimed to explore the causal associations between 6 immune-related diseases and iridocyclitis using Mendelian randomization (MR). A total of 230 single nucleotide polymorphisms (SNPs) significantly associated with systemic lupus erythematosus, ankylosing spondylitis (AS), rheumatoid arthritis (RA), Graves disease (GD), Crohn disease (CD), and allergic contact dermatitis were identified based on stringent MR assumptions. These SNPs served as instrumental variables to estimate the causal effect of each autoimmune disease on iridocyclitis risk. The analysis utilized the inverse variance weighted method, complemented by sensitivity analyses including MR-Egger regression and leave-one-out testing to assess pleiotropy and robustness. The MR analysis revealed significant associations between genetically predicted AS (odds ratio [OR]: 1.544, 95% confidence interval [CI]: 1.494-1.595, P = 1.99 × 10-226), RA (OR: 1.207, 95% CI: 1.052-1.385, P = .003), and CD (OR: 1.654, 95% CI: 1.263-2.166, P = 2.54 × 10⁻⁶) with an increased risk of iridocyclitis. Conversely, higher genetically predicted GD was associated with a decreased risk of iridocyclitis (OR: 0.763, 95% CI: 0.674-0.865, P = .0002). Although systemic lupus erythematosus and allergic contact dermatitis appeared to have a protective effect, these results were not statistically significant, and no causal relationship could be established. Heterogeneity was observed among the SNPs, but no significant horizontal pleiotropy was detected. This study identifies potential genetic links between AS, RA, CD, GD, and the risk of iridocyclitis, providing new insights into the genetic underpinnings of this eye disease. The results support the need for further investigation into the genetic and molecular mechanisms underlying these associations.
Collapse
Affiliation(s)
- Yao Yao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Qian Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Turcinov S, Sharma RK, De Vries C, Cîrciumaru A, Gerstner C, Mathsson-Alm L, Raposo B, Dubnovitsky A, Rönnblom L, Kwok WW, Chemin K, Malmström V, Hensvold A. Arthritis progressors have a decreased frequency of circulating autoreactive T cells during the at-risk phase of rheumatoid arthritis. RMD Open 2024; 10:e004510. [PMID: 39557489 PMCID: PMC11574433 DOI: 10.1136/rmdopen-2024-004510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024] Open
Abstract
OBJECTIVES The aim of this study was to combine deep T cell phenotyping with assessment of citrulline-reactive CD4+T cells in the pre-rheumatoid arthritis (RA) phase. METHODS 20 anti-CCP2 positive individuals (HLA-DRB1*04:01) presenting musculoskeletal complaints without clinical or ultrasound signs of synovitis; 10 arthritis progressors and 10 matched non-arthritis progressors were included. Longitudinal samples (1-3 time points) of peripheral blood mononuclear cells were assessed using HLA-class II tetramers with 12 different citrullinated candidate autoantigens combined in a >20-colour spectral flow cytometry panel. RESULTS The baseline CD4+T cell phenotype was similar between individuals who progressed to arthritis (ie, in the pre-RA phase) and the non-progressors, when studying markers associated with Th1, Th17, T-peripheral and T-regulatory cells as well as with T-cell activation. Citrulline-reactive CD4+T cells were present in both groups but at significantly lower frequency in the progressor group. CD4+T cells specific for citrullinated tenascin-C were the most frequently observed among the progressors, and their frequencies diminished during follow-up that is, closer to arthritis onset. Notably, PD-1 and CD95 expression on the memory cit-tenascin-C-specific T cells in this group indicated repeated antigen exposure. CONCLUSIONS Our data lend support to citrullinated tenascin-C as an interesting T cell antigen in RA. Moreover, lower frequency of circulating citrulline-specific cells in arthritis progressing individuals suggest an initiated homing of these cells to the joints and/or their associated lymph nodes in the pre-RA phase and a possible window of opportunity for therapeutic preventive interventions.
Collapse
Affiliation(s)
- Sara Turcinov
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Theme of Inflammation and Ageing, Medical Unit Gastro, Derma, Rheuma, Karolinska University Hospital, Solna, Sweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Charlotte De Vries
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Alexandra Cîrciumaru
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Christina Gerstner
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | | | - Bruno Raposo
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Anatoly Dubnovitsky
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Karine Chemin
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Aase Hensvold
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
27
|
He YS, Ge M, Xu YQ, Gao ZX, He T, Zhang P, Tao SS, Wang P, Chen Z, Pan HF. Associations between blue space exposure and rheumatoid arthritis: The modifying effect of genetic susceptibility and air pollutants. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117346. [PMID: 39541697 DOI: 10.1016/j.ecoenv.2024.117346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
Studies on the interaction among genetic susceptibility, blue space exposure, and rheumatoid arthritis (RA) risk have been lacking. Therefore, we examined the association between blue space exposure and RA incidence and assess the modifying effect of genetic susceptibility and air pollutants. Form the UK Biobank, 322,783 participants without RA were enrolled in this study. The association between blue space exposure and RA incidence was estimated using a cox proportional hazards model. The combined effect of blue space and genetic factors on the risk of RA was further evaluated. The polygenic risk score (PRS) for RA was calculated to represent individual genetic risk, and the potential modification effect of air pollution on the relationship between blue space, PRS, and RA were explored. During a median follow-up of 12.4 years, 3659 RA cases were identified. A 10 % increase in blue space300 m was associated with a 22.6 % reduction in RA incidence (HR=0.774, 95 % CI: 0.670, 0.895), exhibiting a consistent downward trend in the exposure-response curve. A high PRS was an independent risk factor for RA (HR=1.393, 95 % CI: 1.347, 1.439). The associations between blue space exposure, PRS, and the risk of RA were dose-dependent, with the lowest risk observed among those with high levels of blue space and lower PRS (HRbluespace300m=0.501, 95 % CI: 0.431, 0.583; HRbluespace1000m=0.476, 95 % CI: 0.408, 0.555). Interaction analysis indicated that increased concentrations of air pollutants strengthened the relationship between PRS and RA. Blue space exposure mitigated the risk of RA development, particularly in individuals with low genetic risk.
Collapse
Affiliation(s)
- Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Man Ge
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yi-Qing Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Zhao-Xing Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Peng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Peng Wang
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Zhu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
28
|
Malani K, Pradhan S, Madani MM, Roberts MB, Shadyab A, Allison M, Brasky TM, Schnatz PF, Snetselaar L, Eaton CB. Association of Diet Quality With Risk of Incident Rheumatoid Arthritis in the Women's Health Initiative. J Acad Nutr Diet 2024; 124:1451-1473. [PMID: 39025233 DOI: 10.1016/j.jand.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/03/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Various foods and nutrients are linked with higher or lower risk of rheumatoid arthritis (RA), yet these associations are inconsistent across studies. Limited research has been done evaluating the association between diet quality and RA in a larger-scale prospective study on postmenopausal women. OBJECTIVE The objective of this study was to evaluate the association between dietary quality and risk of incident RA in postmenopausal women. DESIGN This was a prospective cohort study as part of the Women's Health Initiative (WHI), with an average follow-up time of 8.1 years. Baseline diet was measured using a food frequency questionnaire (FFQ). Diet quality was evaluated by the Healthy Eating Index (HEI)-2015 total score. In addition, intake of food groups and nutrients that align with HEI-2015 components was assessed. PARTICIPANTS/SETTING Postmenopausal women (N = 109 591) were included in this study, which was conducted at various clinical centers across the United States with recruitment from 1993 to 1998. Women's Health Initiative participants who were missing outcome data, had unreliable/missing FFQ data, or had RA at baseline were excluded. MAIN OUTCOME MEASURES The primary outcome measure was incident RA. Statistical analyses performed Multivariable Cox proportional regression analysis was performed evaluating the association of diet quality with self-reported physician-diagnosed RA after adjusting for age, race, ethnicity, education status, income, and body mass index (BMI). RESULTS During 857 517 person-years of follow-up, 5823 incident RA cases were identified. After adjustment for multiple comparisons, compared with quartile 1, quartiles 2, 3, and 4 of the HEI-2015 total scores were associated with lower RA risks of 1%, 10%, and 19%, respectively (P-trend < .001). Greater consumption of total fruits (P-trend = .014), whole fruits (P-trend < .0002), total vegetables (P-trend = .008), greens and beans (P-trend < .0002), whole grains (P-trend = .008), and dairy (P-trend = .018) were significantly associated with lower rates of incident RA. Conversely, higher consumption of saturated fat (P-trend = .002) was significantly associated with higher rates of incident RA. CONCLUSION A higher-quality diet reflected by higher HEI-2015 total scores was inversely associated with incident RA in postmenopausal women.
Collapse
Affiliation(s)
- Kanika Malani
- Warren Alpert School of Medicine at Brown University, Providence, RI.
| | - Sushaili Pradhan
- Brown University Center for Primary Care and Prevention, Pawtucket, RI
| | | | - Mary B Roberts
- Brown University Center for Primary Care and Prevention, Pawtucket, RI
| | - Aladdin Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Matthew Allison
- University of California San Diego School of Medicine, San Diego, CA
| | | | - Peter F Schnatz
- Sidney Kimmel Medical College at Thomas Jefferson University, West Reading, PA
| | | | - Charles B Eaton
- Brown University Center for Primary Care and Prevention, Pawtucket, RI; Department of Epidemiology, Brown University School of Public Health, Pawtucket, RI; Department of Family Medicine, Warren Alpert Medical School of Brown University, Pawtucket, RI
| |
Collapse
|
29
|
Mori S, Kohyama M, Yasumizu Y, Tada A, Tanzawa K, Shishido T, Kishida K, Jin H, Nishide M, Kawada S, Motooka D, Okuzaki D, Naito R, Nakai W, Kanda T, Murata T, Terao C, Ohmura K, Arase N, Kurosaki T, Fujimoto M, Suenaga T, Kumanogoh A, Sakaguchi S, Ogawa Y, Arase H. Neoself-antigens are the primary target for autoreactive T cells in human lupus. Cell 2024; 187:6071-6087.e20. [PMID: 39276775 DOI: 10.1016/j.cell.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/06/2024] [Accepted: 08/12/2024] [Indexed: 09/17/2024]
Abstract
Major histocompatibility complex class II (MHC-II) is the most significant genetic risk factor for systemic lupus erythematosus (SLE), but the nature of the self-antigens that trigger autoimmunity remains unclear. Unusual self-antigens, termed neoself-antigens, are presented on MHC-II in the absence of the invariant chain essential for peptide presentation. Here, we demonstrate that neoself-antigens are the primary target for autoreactive T cells clonally expanded in SLE. When neoself-antigen presentation was induced by deleting the invariant chain in adult mice, neoself-reactive T cells were clonally expanded, leading to the development of lupus-like disease. Furthermore, we found that neoself-reactive CD4+ T cells were significantly expanded in SLE patients. A high frequency of Epstein-Barr virus reactivation is a risk factor for SLE. Neoself-reactive lupus T cells were activated by Epstein-Barr-virus-reactivated cells through downregulation of the invariant chain. Together, our findings imply that neoself-antigen presentation by MHC-II plays a crucial role in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Shunsuke Mori
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan
| | - Masako Kohyama
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yoshiaki Yasumizu
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
| | - Asa Tada
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan
| | - Kaito Tanzawa
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tatsuya Shishido
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kazuki Kishida
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Hui Jin
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Shoji Kawada
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan; Single Cell Genomics, Human Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan; Single Cell Genomics, Human Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Ryota Naito
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Wataru Nakai
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Teru Kanda
- Division of Microbiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Takayuki Murata
- Department of Virology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; Department of Virology, Fujita Health University School of Medicine, Nagoya 470-1192, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 351-0198, Japan; Clinical Research Center, Shizuoka General Hospital, Shizuoka 420-8527, Japan; The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo 650-0047, Japan
| | - Noriko Arase
- Department of Dermatology, Graduate school of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Graduate school of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Tadahiro Suenaga
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Department of Immunology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Center for advanced modalities and DDS, Osaka University, Osaka 565-0871, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Center for advanced modalities and DDS, Osaka University, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
30
|
Kim JS, Flack KF, Malik V, Manichaikul A, Sakaue S, Luo Y, McGroder CF, Salvatore M, Anderson MR, Hoffman EA, Podolanczuk AJ, Yun JH, McDermott GC, Sparks JA, Putman R, Moll M, Rich SS, Rotter JI, Noth I, Raghu G, Giles JT, Winchester R, Raychaudhuri S, Hunninghake GM, Cho MH, Garcia CK, Barr RG, Bernstein EJ. Genomic and Serological Rheumatoid Arthritis Biomarkers, MUC5B Promoter Variant, and Interstitial Lung Abnormalities. Ann Am Thorac Soc 2024; 22:64-71. [PMID: 39405163 PMCID: PMC11708761 DOI: 10.1513/annalsats.202403-238oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/27/2024] [Indexed: 11/27/2024] Open
Abstract
RATIONALE Rheumatoid arthritis (RA) has been implicated in interstitial lung disease (ILD) as majority of studies have been comprised of patients with known RA. However, it remains unclear whether an underlying risk for RA in combination with genetic risk for pulmonary fibrosis is associated with radiological markers of early lung injury and fibrosis in broader population samples. OBJECTIVE Determine whether genetic and serological biomarkers of RA risk in combination with the MUC5B (rs35705950) risk allele (T) are associated with interstitial lung abnormalities (ILA) on computed tomography (CT) scans. METHODS Associations of RA-risk HLA-DRB1 alleles (*04:01, *04:08, *04:05, *04:04, *10:01) and serum RA autoantibodies with ILA in the Multi-Ethnic Study of Atherosclerosis (MESA, n=4,018) and COPDGene (n=5,963) cohorts were modeled using logistic regression and adjusted for age, sex, self-reported race and ethnicity, smoking history, body mass index, and principal components of genetic ancestry. RESULTS The prevalence of an RA risk HLA-DRB1 allele was 16.5% and 21.9% in MESA and COPDGene, respectively. ILA was present in 3.9% and 11% in MESA and COPDGene, respectively. An RA risk HLA-DRB1 allele was not significantly associated with ILA in MESA and COPDGene. In MESA, higher serum levels of IgA rheumatoid factor (RF) and anti-cyclic citrullinated peptide were associated with an odds ratio (OR) for ILA of 1.20 (95% CI 1.07-1.35) and 1.19 (95% CI 1.04-1.37), respectively. Among smokers without baseline ILA, per doubling of IgM RF was associated with an OR for ILA 10 years later of 1.25 (95% CI 1.08-1.43). Associations were not significantly different by MUC5B risk allele status. CONCLUSIONS RA-related HLA-DRB1 alleles were not associated with ILA, whereas higher serum levels of IgM RF among smokers without baseline ILA were associated with subsequent ILA.
Collapse
Affiliation(s)
- John S Kim
- University of Virginia, Medicine, Charlottesville, Virginia, United States
- Charlottesville, Virginia, United States
| | - Kathryn F Flack
- University of Pennsylvania, Medicine, Philadelphia, Pennsylvania, United States
| | - Vidhi Malik
- Brigham and Women's Hospital, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Ani Manichaikul
- University of Virginia Center for Public Health Genomics, Charlottesville, Virginia, United States
| | - Saori Sakaue
- Broad Institute, Cambridge, Massachusetts, United States
| | - Yang Luo
- University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Claire F McGroder
- NewYork-Presbyterian Hospital, Pulmonary, Allergy and Critical Care, New York, New York, United States
| | - Mary Salvatore
- Columbia University Irving Medical Center, New York, New York, United States
| | | | - Eric A Hoffman
- University of Iowa Carver College of Medicine, Radiology, Iowa City, Iowa, United States
| | - Anna J Podolanczuk
- Weill Cornell Medical College, Department of Medicine, New York, New York, United States
| | - Jae Hee Yun
- University of Virginia, Medicine, Charlottesville, Virginia, United States
| | - Gregory C McDermott
- Brigham and Women's Hospital, Department of Rheumatology, Boston, Massachusetts, United States
| | - Jeffrey A Sparks
- Brigham and Women's Hospital, Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Boston, Massachusetts, United States
| | - Rachel Putman
- Brigham and Women's Hospital, Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Matthew Moll
- Brigham and Women's Hospital Department of Medicine, Pulmonary and Critical Care, Boston, Massachusetts, United States
| | - Stephen S Rich
- University of Virginia, Center for Public Health Genomics, Charlottesville, Virginia, United States
| | - Jerome I Rotter
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 11The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Torrance, California, United States
| | - Imre Noth
- University of Virginia, Division of Pulmonary & Critical Care & Sleep Medicine, Department of Medicine, , Charlottesville, Virginia, United States
| | - Ganesh Raghu
- University of Washington Medical Center, Division of Pulmonary and Critical Care Medicine, Seattle, Washington, United States
| | - Jon T Giles
- Columbia University, Division of Rheumatology, New York, New York, United States
| | - Robert Winchester
- Columbia University Medical Center, Medicine, NYC, New York, United States
| | - Soumya Raychaudhuri
- Broad Institute, Cambridge, Massachusetts, United States
- Brigham and Women's Hospital, Division of Rheumatology, Immunology, and Immunity , Boston, Massachusetts, United States
- Brigham and Women's Hospital, Center for Data Science and Division of Genetics, Boston, Massachusetts, United States
- The University of Manchester, Centre for Genetics and Genomics Versus Arthritis, Manchester, United Kingdom of Great Britain and Northern Ireland
| | - Gary M Hunninghake
- Brigham and Women's Hospital, Medicine, Boston, Massachusetts, United States
| | - Michael H Cho
- Harvard Medical School, Channing Division of Respiratory Medicine, Boston, Massachusetts, United States
| | - Christine Kim Garcia
- Columbia University Irving Medical Center, Medicine, New York, New York, United States
- New York, United States
| | - R Graham Barr
- Columbia University, New York, New York, United States
| | | |
Collapse
|
31
|
Ghodke-Puranik Y, Olferiev M, Crow MK. Systemic lupus erythematosus genetics: insights into pathogenesis and implications for therapy. Nat Rev Rheumatol 2024; 20:635-648. [PMID: 39232240 DOI: 10.1038/s41584-024-01152-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Systemic lupus erythematosus (SLE) is a prime example of how the interplay between genetic and environmental factors can trigger systemic autoimmunity, particularly in young women. Although clinical disease can take years to manifest, risk is established by the unique genetic makeup of an individual. Genome-wide association studies have identified almost 200 SLE-associated risk loci, yet unravelling the functional effect of these loci remains a challenge. New analytic tools have enabled researchers to delve deeper, leveraging DNA sequencing and cell-specific and immune pathway analysis to elucidate the immunopathogenic mechanisms. Both common genetic variants and rare non-synonymous mutations can interact to increase SLE risk. Notably, variants strongly associated with SLE are often located in genome super-enhancers that regulate MHC class II gene expression. Additionally, the 3D conformations of DNA and RNA contribute to genome regulation and innate immune system activation. Improved therapies for SLE are urgently needed and current and future knowledge from genetic and genomic research should provide new tools to facilitate patient diagnosis, enhance the identification of therapeutic targets and optimize testing of agents.
Collapse
Affiliation(s)
- Yogita Ghodke-Puranik
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA
| | - Mikhail Olferiev
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA
| | - Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
32
|
Shibata M, Terada A, Kawaguchi T, Kamatani Y, Okada D, Nagashima K, Ohmura K, Matsuda F, Kawaguchi S, Sese J, Yamada R. Identification of epistatic SNP combinations in rheumatoid arthritis using LAMPLINK and Japanese cohorts. J Hum Genet 2024; 69:541-547. [PMID: 39014190 DOI: 10.1038/s10038-024-01269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024]
Abstract
Genome-wide association studies have enabled the identification of important genetic factors in many trait studies. However, only a fraction of the heritability can be explained by known genetic factors, even in the most common diseases. Genetic loci combinations, or epistatic contributions expressed by combinations of single nucleotide polymorphisms (SNPs), have been argued to be one of the critical factors explaining some of the missing heritability, especially in oligogenic/polygenic diseases. Rheumatoid arthritis (RA) is a complex disease with more than 100 reported SNP associations, as well as various HLA haplotypes and amino acids; however, many associations between RA and inter-chromosomal SNP combinations are unknown. To discover novel associations of epistatic interactions with high odds ratios in RA, we applied the LAMPLINK method, a systematic enumerative procedure for identifying high-order SNP combinations, to a Japanese RA cohort (discovery cohort; 4024 patients with RA and 7731 controls). We validated the identified associations in a different Japanese cohort (validation cohort; 810 RA patients and 6303 controls). In this study, we identified 90 significant genetic associations in the discovery cohort. Among these, 74 (82.2%) associations were replicated in the validation cohort, and eight combinations were inter-chromosomal, all of which comprised rs7765379 or rs35265698 located in the HLA region. These two SNPs exhibited strong correlations with valine at amino acid position 11 in HLA-DRB1 (HLA-DRB1-11-Val). Finally, we discovered that rs9624 showed an association with RA through an epistatic interaction with HLA-DRB1-11-Val. Overall, LAMPLINK showed high reliability for identifying epistatic genetic contributions hidden in complex traits.
Collapse
Affiliation(s)
- Mio Shibata
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takahisa Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Daigo Okada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhisa Nagashima
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuji Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Jun Sese
- Humanome Lab. Inc., Tokyo, Japan.
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.
| | - Ryo Yamada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
33
|
Tanaka K, Kato K, Nonaka N, Seita J. Efficient HLA imputation from sequential SNPs data by transformer. J Hum Genet 2024; 69:533-540. [PMID: 39095607 PMCID: PMC11422163 DOI: 10.1038/s10038-024-01278-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
Human leukocyte antigen (HLA) genes are associated with a variety of diseases, yet the direct typing of HLA alleles is both time-consuming and costly. Consequently, various imputation methods leveraging sequential single nucleotide polymorphisms (SNPs) data have been proposed, employing either statistical or deep learning models, such as the convolutional neural network (CNN)-based model, DEEP*HLA. However, these methods exhibit limited imputation efficiency for infrequent alleles and necessitate a large size of reference dataset. In this context, we have developed a Transformer-based model to HLA allele imputation, named "HLA Reliable IMpuatioN by Transformer (HLARIMNT)" designed to exploit the sequential nature of SNPs data. We evaluated HLARIMNT's performance using two distinct reference panels; Pan-Asian reference panel (n = 530) and Type 1 Diabetes genetics Consortium (T1DGC) reference panel (n = 5225), alongside a combined panel (n = 1060). HLARIMNT demonstrated superior accuracy to DEEP*HLA across several indices, particularly for infrequent alleles. Furthermore, we explored the impact of varying training data sizes on imputation accuracy, finding that HLARIMNT consistently outperformed across all data size. These findings suggest that Transformer-based models can efficiently impute not only HLA types but potentially other gene types from sequential SNPs data.
Collapse
Affiliation(s)
- Kaho Tanaka
- Faculty of Engineering, Kyoto University, Kyoto, Japan
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, Japan
| | - Kosuke Kato
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, Japan
| | - Naoki Nonaka
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, Japan
| | - Jun Seita
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, Japan.
| |
Collapse
|
34
|
Steere AC, Lemieux JE. Wider recognition and greater understanding of postinfectious, antibiotic-refractory Lyme arthritis. J Clin Invest 2024; 134:e184109. [PMID: 39225104 PMCID: PMC11364397 DOI: 10.1172/jci184109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Lyme disease, caused by Borrelia burgdorferi (Bb), can progress to Lyme arthritis (LA). While most patients with LA respond successfully to antibiotic therapy, a small percentage fail to improve, a condition known as antibiotic-refractory Lyme arthritis (ARLA). While T cell responses are known to drive ARLA, molecular mechanisms for ARLA remain unknown. In this issue of the JCI, Dirks et al. isolated disease-specific Th cells from patients with ARLA residing in Germany. A distinct TCR-β motif distinguished ARLA from other rheumatic diseases. Notably, the TCR-β motif was linked predominantly to HLA-DRB1*11 or 13 alleles, which differed from alleles in patients from North America. It also mapped primarily to T peripheral helper (Tph) cells, as opposed to classical Th1 cells. These findings provide a roadmap explaining how T cell responses necessary for control of an infection can, despite antibiotic therapy, drive a disadvantageous T cell response, resulting in a postinfectious, inflammatory arthritis.
Collapse
Affiliation(s)
- Allen C. Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology and
| | - Jacob E. Lemieux
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Sysojev AÖ, Alfredsson L, Klareskog L, Silberberg GN, Saevarsdottir S, Padyukov L, Magnusson PKE, Askling J, Westerlind H. Minor Genetic Overlap Among Rheumatoid Arthritis, Myocardial Infarction, and Myocardial Infarction Risk Determinants. Arthritis Rheumatol 2024; 76:1344-1352. [PMID: 38782598 DOI: 10.1002/art.42918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE The aim of this study was to investigate whether a shared genetic susceptibility exists between individuals with rheumatoid arthritis (RA) and individuals with myocardial infarction (MI)-including major MI risk factors-and to quantify the degree of any such overlap. METHODS Genome-wide association study (GWAS) data for individuals with RA were constructed from a sample of 26,637 Swedish patients with RA and controls without RA. For patients with MI, GWAS data were obtained from a previously published meta-analysis. Genome-wide genetic correlation was estimated via linkage disequilibrium score regression. LAVA was employed to estimate local genetic correlations in ~2,500 nonoverlapping loci, including the major histocompatibility complex. The controls without RA were used for reference panel data. We also assessed stratified estimates of both genome-wide and local genetic correlation based on subsamples of individuals with seropositive RA and those with seronegative RA. Furthermore, genome-wide genetic correlation was estimated between RA and selected cardiovascular risk factors to elucidate pleiotropic relationships. RESULTS Following quality control, our GWAS of patients with RA consisted of 25,826 individuas. Genome-wide genetic correlation between patients with RA and MI was estimated to 0.13 (95% confidence interval -0.03 to 0.29). Six regions exhibited significant local genetic correlation, though none harbored any known risk single-nucleotide polymorphisms for either of the two traits. Estimates were similar in both individuals with seropositive RA and those with seronegative RA. No statistically significant genetic correlations were observed between RA risk factors and any of the MI risk factors. CONCLUSION Our findings indicate that genetic overlap between patients with RA and MI is minor. Furthermore, genetic overlap between RA and MI risk factors seem unlikely to provide a major contribution to the increased risk of MI observed in patients with RA.
Collapse
Affiliation(s)
| | | | - Lars Klareskog
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Gilad N Silberberg
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Saedis Saevarsdottir
- Karolinska Institute, Stockholm, Sweden, and deCODE genetics, Reykjavik, Iceland. Members of the Swedish Rheumatology Quality Register Biobank Group are shown in Appendix A
| | - Leonid Padyukov
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | | - Johan Askling
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
36
|
Lamba A, Taneja V. Gut microbiota as a sensor of autoimmune response and treatment for rheumatoid arthritis. Immunol Rev 2024; 325:90-106. [PMID: 38867408 PMCID: PMC11338721 DOI: 10.1111/imr.13359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Rheumatoid arthritis (RA) is considered a multifactorial condition where interaction between the genetic and environmental factors lead to immune dysregulation causing autoreactivity. While among the various genetic factors, HLA-DR4 and DQ8, have been reported to be the strongest risk factors, the role of various environmental factors has been unclear. Though events initiating autoreactivity remain unknown, a mucosal origin of RA has gained attention based on the recent observations with the gut dysbiosis in patients. However, causality of gut dysbiosis has been difficult to prove in humans. Mouse models, especially mice expressing RA-susceptible and -resistant HLA class II genes have helped unravel the complex interactions between genetic factors and gut microbiome. This review describes the interactions between HLA genes and gut dysbiosis in sex-biased preclinical autoreactivity and discusses the potential use of endogenous commensals as indicators of treatment efficacy as well as therapeutic tool to suppress pro-inflammatory response in rheumatoid arthritis.
Collapse
Affiliation(s)
| | - Veena Taneja
- Department of Immunology and Division of Rheumatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
37
|
Bashir U, Singh G, Bhatia A. Rheumatoid arthritis-recent advances in pathogenesis and the anti-inflammatory effect of plant-derived COX inhibitors. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5363-5385. [PMID: 38358467 DOI: 10.1007/s00210-024-02982-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
The majority of people with autoimmune disorders, including those with rheumatoid arthritis, osteoarthritis, and tendonitis report pain, stiffness, and inflammation as major contributors to their worse quality of life in terms of overall health. Of all the available treatment options, COX inhibitors are the ones that are utilized most frequently to ease the symptoms. Various signaling cascades have been reported to be involved in the pathogenesis of rheumatoid arthritis which includes JAK/STAT, MAPK, and NF-kB signaling pathways, and several allopathic inhibitors (tofacitinib and baricitinib) have been reported to target the components of these cascades and have received approval for RA treatment. However, the prolonged use of these COX inhibitors and other allopathic drugs can pose serious health challenges due to their significant side effects. Therefore, searching for a more effective and side effect-free treatment for rheumatoid arthritis has unveiled phytochemicals as both productive and promising. Their therapeutic ability helps develop potent and safe drugs targeting immune-inflammatory diseases including RA. Various scientific databases were used for searching articles such as NCBI, SpringerLink, BioMed Central, ResearchGate, Google Scholar, Scopus, Nature, Wiley Online Library, and ScienceDirect. This review lists various phytochemicals and discusses their potential molecular targets in RA treatment, as demonstrated by various in vitro, in vivo (pre-clinical), and clinical studies. Several pre-clinical and clinical studies suggest that various phytochemicals can be an alternative promising intervention for attenuating and managing inflammation-associated pathogenesis of rheumatoid arthritis.
Collapse
Affiliation(s)
- Ubaid Bashir
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Gurjant Singh
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Astha Bhatia
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| |
Collapse
|
38
|
Van Espen B, Prideaux EB, Wilson AR, Machado CRL, Sendo S, Parker J, Seumois G, Sacchetti C, Belongia AC, Perumal NB, Vijayanand P, Linnik MD, Benschop RJ, Wang W, Bottini N, Firestein GS, Stanford SM. Laser Capture Microscopy RNA Sequencing for Topological Mapping of Synovial Pathology During Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1243-1251. [PMID: 38556917 PMCID: PMC11949376 DOI: 10.1002/art.42853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 02/21/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is an autoimmune disease in which the joint lining or synovium becomes highly inflamed and majorly contributes to disease progression. Understanding pathogenic processes in RA synovium is critical for identifying therapeutic targets. We performed laser capture microscopy (LCM) followed by RNA sequencing (LCM-RNAseq) to study regional transcriptomes throughout RA synovium. METHODS Synovial lining, sublining, and vessel samples were captured by LCM from seven patients with RA and seven patients with osteoarthritis (OA). RNAseq was performed on RNA extracted from captured tissue. Principal component analysis was performed on the sample set by disease state. Differential expression analysis was performed between disease states based on log2 fold change and q value parameters. Pathway analysis was performed using the Reactome Pathway Database on differentially expressed genes among disease states. Significantly enriched pathways in each synovial region were selected based on the false discovery rate. RESULTS RA and OA transcriptomes were distinguishable by principal component analysis. Pairwise comparisons of synovial lining, sublining, and vessel samples between RA and OA revealed substantial differences in transcriptional patterns throughout the synovium. Hierarchical clustering of pathways based on significance revealed a pattern of association between biologic function and synovial topology. Analysis of pathways uniquely enriched in each region revealed distinct phenotypic abnormalities. As examples, RA lining samples were marked by anomalous immune cell signaling, RA sublining samples were marked by aberrant cell cycle, and RA vessel samples were marked by alterations in heme scavenging. CONCLUSION LCM-RNAseq confirms reported transcriptional differences between the RA synovium and the OA synovium and provides evidence supporting a relationship between synovial topology and molecular anomalies in RA.
Collapse
Affiliation(s)
| | | | | | | | - Sho Sendo
- University of California, San Diego, La Jolla
| | | | | | | | | | | | - Pandurangan Vijayanand
- University of California, San Diego, and La Jolla Institute for Immunology, La Jolla, California
| | | | | | - Wei Wang
- University of California, San Diego, La Jolla
| | - Nunzio Bottini
- University of California, San Diego, La Jolla, and Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | | | | |
Collapse
|
39
|
Krishna C, Chiou J, Sakaue S, Kang JB, Christensen SM, Lee I, Aksit MA, Kim HI, von Schack D, Raychaudhuri S, Ziemek D, Hu X. The influence of HLA genetic variation on plasma protein expression. Nat Commun 2024; 15:6469. [PMID: 39085222 PMCID: PMC11291675 DOI: 10.1038/s41467-024-50583-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Genetic variation in the human leukocyte antigen (HLA) loci is associated with risk of immune-mediated diseases, but the molecular effects of HLA polymorphism are unclear. Here we examined the effects of HLA genetic variation on the expression of 2940 plasma proteins across 45,330 Europeans in the UK Biobank, with replication analyses across multiple ancestry groups. We detected 504 proteins affected by HLA variants (HLA-pQTL), including widespread trans effects by autoimmune disease risk alleles. More than 80% of the HLA-pQTL fine-mapped to amino acid positions in the peptide binding groove. HLA-I and II affected proteins expressed in similar cell types but in different pathways of both adaptive and innate immunity. Finally, we investigated potential HLA-pQTL effects on disease by integrating HLA-pQTL with fine-mapped HLA-disease signals in the UK Biobank. Our data reveal the diverse effects of HLA genetic variation and aid the interpretation of associations between HLA alleles and immune-mediated diseases.
Collapse
Affiliation(s)
- Chirag Krishna
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA.
| | - Joshua Chiou
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - Saori Sakaue
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Joyce B Kang
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | | | - Isac Lee
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA
| | | | - Hye In Kim
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - David von Schack
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Daniel Ziemek
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - Xinli Hu
- Pfizer Research and Development, Pfizer Inc., Cambridge, MA, USA.
| |
Collapse
|
40
|
Loh TJ, Lim JJ, Jones CM, Dao HT, Tran MT, Baker DG, La Gruta NL, Reid HH, Rossjohn J. The molecular basis underlying T cell specificity towards citrullinated epitopes presented by HLA-DR4. Nat Commun 2024; 15:6201. [PMID: 39043656 PMCID: PMC11266596 DOI: 10.1038/s41467-024-50511-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
CD4+ T cells recognising citrullinated self-epitopes presented by HLA-DRB1 bearing the shared susceptibility epitope (SE) are implicated in rheumatoid arthritis (RA). However, the underlying T cell receptor (TCR) determinants of epitope specificity towards distinct citrullinated peptide antigens, including vimentin-64cit59-71 and α-enolase-15cit10-22 remain unclear. Using HLA-DR4-tetramers, we examine the T cell repertoire in HLA-DR4 transgenic mice and observe biased TRAV6 TCR gene usage across these two citrullinated epitopes which matches with TCR bias previously observed towards the fibrinogen β-74cit69-81 epitope. Moreover, shared TRAV26-1 gene usage is evident in four α-enolase-15cit10-22 reactive T cells in three human samples. Crystal structures of mouse TRAV6+ and human TRAV26-1+ TCR-HLA-DR4 complexes presenting vimentin-64cit59-71 and α-enolase-15cit10-22, respectively, show three-way interactions between the TCR, SE, citrulline, and the basis for the biased selection of TRAV genes. Position 2 of the citrullinated epitope is a key determinant underpinning TCR specificity. Accordingly, we provide a molecular basis of TCR specificity towards citrullinated epitopes.
Collapse
MESH Headings
- Humans
- Mice, Transgenic
- HLA-DR4 Antigen/immunology
- HLA-DR4 Antigen/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/genetics
- Mice
- Animals
- Vimentin/immunology
- Vimentin/metabolism
- Vimentin/genetics
- CD4-Positive T-Lymphocytes/immunology
- Citrullination
- Phosphopyruvate Hydratase/immunology
- Phosphopyruvate Hydratase/genetics
- Phosphopyruvate Hydratase/metabolism
- Epitopes, T-Lymphocyte/immunology
- Citrulline/metabolism
- Citrulline/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Epitopes/immunology
- Crystallography, X-Ray
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
Collapse
Affiliation(s)
- Tiing Jen Loh
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jia Jia Lim
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Claerwen M Jones
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Hien Thy Dao
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Mai T Tran
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Daniel G Baker
- Janssen Research & Development, LLC, Horsham, Philadelphia, PA, USA
| | - Nicole L La Gruta
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Hugh H Reid
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia.
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia.
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK.
| |
Collapse
|
41
|
Dirks J, Fischer J, Klaussner J, Hofmann C, Holl-Wieden A, Buck V, Klemann C, Girschick HJ, Caruana I, Erhard F, Morbach H. Disease-specific T cell receptors maintain pathogenic T helper cell responses in postinfectious Lyme arthritis. J Clin Invest 2024; 134:e179391. [PMID: 38963700 PMCID: PMC11364382 DOI: 10.1172/jci179391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUNDAntibiotic-Refractory Lyme Arthritis (ARLA) involves a complex interplay of T cell responses targeting Borrelia burgdorferi antigens progressing toward autoantigens by epitope spreading. However, the precise molecular mechanisms driving the pathogenic T cell response in ARLA remain unclear. Our aim was to elucidate the molecular program of disease-specific Th cells.METHODSUsing flow cytometry, high-throughput T cell receptor (TCR) sequencing, and scRNA-Seq of CD4+ Th cells isolated from the joints of patients with ARLA living in Europe, we aimed to infer antigen specificity through unbiased analysis of TCR repertoire patterns, identifying surrogate markers for disease-specific TCRs, and connecting TCR specificity to transcriptional patterns.RESULTSPD-1hiHLA-DR+CD4+ effector T cells were clonally expanded within the inflamed joints and persisted throughout disease course. Among these cells, we identified a distinct TCR-β motif restricted to HLA-DRB1*11 or *13 alleles. These alleles, being underrepresented in patients with ARLA living in North America, were unexpectedly prevalent in our European cohort. The identified TCR-β motif served as surrogate marker for a convergent TCR response specific to ARLA, distinguishing it from other rheumatic diseases. In the scRNA-Seq data set, the TCR-β motif particularly mapped to peripheral T helper (TPH) cells displaying signs of sustained proliferation, continuous TCR signaling, and expressing CXCL13 and IFN-γ.CONCLUSIONBy inferring disease-specific TCRs from synovial T cells we identified a convergent TCR response in the joints of patients with ARLA that continuously fueled the expansion of TPH cells expressing a pathogenic cytokine effector program. The identified TCRs will aid in uncovering the major antigen targets of the maladaptive immune response.FUNDINGSupported by the German Research Foundation (DFG) MO 2160/4-1; the Federal Ministry of Education and Research (BMBF; Advanced Clinician Scientist-Program INTERACT; 01EO2108) embedded in the Interdisciplinary Center for Clinical Research (IZKF) of the University Hospital Würzburg; the German Center for Infection Research (DZIF; Clinical Leave Program; TI07.001_007) and the Interdisciplinary Center for Clinical Research (IZKF) Würzburg (Clinician Scientist Program, Z-2/CSP-30).
Collapse
MESH Headings
- Humans
- Lyme Disease/immunology
- Lyme Disease/pathology
- Lyme Disease/genetics
- HLA-DRB1 Chains/genetics
- HLA-DRB1 Chains/immunology
- Female
- Male
- T-Lymphocytes, Helper-Inducer/immunology
- Borrelia burgdorferi/immunology
- Middle Aged
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Adult
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Johannes Dirks
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Jonas Fischer
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Julia Klaussner
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Christine Hofmann
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Annette Holl-Wieden
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Viktoria Buck
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Christian Klemann
- Department of Pediatric Immunology, Rheumatology, and Infectiology, Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | | | - Ignazio Caruana
- Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, Würzburg, Germany
| | - Florian Erhard
- Computational Systems Virology and Bioinformatics, Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Faculty for Informatics and Data Science, University of Regensburg, Regensburg, Germany
| | - Henner Morbach
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
42
|
Raposo B, Klareskog L, Robinson WH, Malmström V, Grönwall C. The peculiar features, diversity and impact of citrulline-reactive autoantibodies. Nat Rev Rheumatol 2024; 20:399-416. [PMID: 38858604 DOI: 10.1038/s41584-024-01124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Since entering the stage 25 years ago as a highly specific serological biomarker for rheumatoid arthritis, anti-citrullinated protein antibodies (ACPAs) have been a topic of extensive research. This hallmark B cell response arises years before disease onset, displays interpatient autoantigen variability, and is associated with poor clinical outcomes. Technological and scientific advances have revealed broad clonal diversity and intriguing features including high levels of somatic hypermutation, variable-domain N-linked glycosylation, hapten-like peptide interactions, and clone-specific multireactivity to citrullinated, carbamylated and acetylated epitopes. ACPAs have been found in different isotypes and subclasses, in both circulation and tissue, and are secreted by both plasmablasts and long-lived plasma cells. Notably, although some disease-promoting features have been reported, results now demonstrate that certain monoclonal ACPAs therapeutically block arthritis and inflammation in mouse models. A wealth of functional studies using patient-derived polyclonal and monoclonal antibodies have provided evidence for pathogenic and protective effects of ACPAs in the context of arthritis. To understand the roles of ACPAs, one needs to consider their immunological properties by incorporating different facets such as rheumatoid arthritis B cell biology, environmental triggers and chronic antigen exposure. The emerging picture points to a complex role of citrulline-reactive autoantibodies, in which the diversity and dynamics of antibody clones could determine clinical progression and manifestations.
Collapse
Affiliation(s)
- Bruno Raposo
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
43
|
Zou Y, Carbonetto P, Xie D, Wang G, Stephens M. Fast and flexible joint fine-mapping of multiple traits via the Sum of Single Effects model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.14.536893. [PMID: 37425935 PMCID: PMC10327118 DOI: 10.1101/2023.04.14.536893] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
We introduce mvSuSiE, a multi-trait fine-mapping method for identifying putative causal variants from genetic association data (individual-level or summary data). mvSuSiE learns patterns of shared genetic effects from data, and exploits these patterns to improve power to identify causal SNPs. Comparisons on simulated data show that mvSuSiE is competitive in speed, power and precision with existing multi-trait methods, and uniformly improves on single-trait fine-mapping (SuSiE) in each trait separately. We applied mvSuSiE to jointly fine-map 16 blood cell traits using data from the UK Biobank. By jointly analyzing the traits and modeling heterogeneous effect sharing patterns, we discovered a much larger number of causal SNPs (>3,000) compared with single-trait fine-mapping, and with narrower credible sets. mvSuSiE also more comprehensively characterized the ways in which the genetic variants affect one or more blood cell traits; 68% of causal SNPs showed significant effects in more than one blood cell type.
Collapse
Affiliation(s)
- Yuxin Zou
- Department of Statistics, University of Chicago, Chicago, IL, USA
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Dongyue Xie
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Gao Wang
- Gertrude. H. Sergievsky Center, Department of Neurology, Columbia University, New York, NY, USA
| | - Matthew Stephens
- Department of Statistics, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| |
Collapse
|
44
|
Santiago-Lamelas L, Castro-Santos P, Carracedo Á, Olloquequi J, Díaz-Peña R. Unveiling the Significance of HLA and KIR Diversity in Underrepresented Populations. Biomedicines 2024; 12:1333. [PMID: 38927540 PMCID: PMC11202227 DOI: 10.3390/biomedicines12061333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Human leukocyte antigen (HLA) molecules and their relationships with natural killer (NK) cells, specifically through their interaction with killer-cell immunoglobulin-like receptors (KIRs), exhibit robust associations with the outcomes of diverse diseases. Moreover, genetic variations in HLA and KIR immune system genes offer limitless depths of complexity. In recent years, a surge of high-powered genome-wide association studies (GWASs) utilizing single nucleotide polymorphism (SNP) arrays has occurred, significantly advancing our understanding of disease pathogenesis. Additionally, advances in HLA reference panels have enabled higher resolution and more reliable imputation, allowing for finer-grained evaluation of the association between sequence variations and disease risk. However, it is essential to note that the majority of these GWASs have focused primarily on populations of Caucasian and Asian origins, neglecting underrepresented populations in Latin America and Africa. This omission not only leads to disparities in health care access but also restricts our knowledge of novel genetic variants involved in disease pathogenesis within these overlooked populations. Since the KIR and HLA haplotypes prevalent in each population are clearly modelled by the specific environment, the aim of this review is to encourage studies investigating HLA/KIR involvement in infection and autoimmune diseases, reproduction, and transplantation in underrepresented populations.
Collapse
Affiliation(s)
- Lucía Santiago-Lamelas
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
| | - Patricia Castro-Santos
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Grupo de Medicina Xenómica, CIMUS, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jordi Olloquequi
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Roberto Díaz-Peña
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| |
Collapse
|
45
|
Dunlap G, Wagner A, Meednu N, Wang R, Zhang F, Ekabe JC, Jonsson AH, Wei K, Sakaue S, Nathan A, Bykerk VP, Donlin LT, Goodman SM, Firestein GS, Boyle DL, Holers VM, Moreland LW, Tabechian D, Pitzalis C, Filer A, Raychaudhuri S, Brenner MB, Thakar J, McDavid A, Rao DA, Anolik JH. Clonal associations between lymphocyte subsets and functional states in rheumatoid arthritis synovium. Nat Commun 2024; 15:4991. [PMID: 38862501 PMCID: PMC11167034 DOI: 10.1038/s41467-024-49186-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 05/20/2024] [Indexed: 06/13/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease involving antigen-specific T and B cells. Here, we perform single-cell RNA and repertoire sequencing on paired synovial tissue and blood samples from 12 seropositive RA patients. We identify clonally expanded CD4 + T cells, including CCL5+ cells and T peripheral helper (Tph) cells, which show a prominent transcriptomic signature of recent activation and effector function. CD8 + T cells show higher oligoclonality than CD4 + T cells, with the largest synovial clones enriched in GZMK+ cells. CD8 + T cells with possibly virus-reactive TCRs are distributed across transcriptomic clusters. In the B cell compartment, NR4A1+ activated B cells, and plasma cells are enriched in the synovium and demonstrate substantial clonal expansion. We identify synovial plasma cells that share BCRs with synovial ABC, memory, and activated B cells. Receptor-ligand analysis predicted IFNG and TNFRSF members as mediators of synovial Tph-B cell interactions. Together, these results reveal clonal relationships between functionally distinct lymphocyte populations that infiltrate the synovium of patients with RA.
Collapse
Affiliation(s)
- Garrett Dunlap
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron Wagner
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Nida Meednu
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Ruoqiao Wang
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Fan Zhang
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jabea Cyril Ekabe
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anna Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saori Sakaue
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aparna Nathan
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vivian P Bykerk
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Laura T Donlin
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Susan M Goodman
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego;, La Jolla, CA, USA
| | - David L Boyle
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego;, La Jolla, CA, USA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Larry W Moreland
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Darren Tabechian
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
- Department of Biomedical Sciences, Humanitas University and Humanitas Research Hospital, Milan, Italy
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Juilee Thakar
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Jennifer H Anolik
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
46
|
Huang F, Su Z, Huang Y, Huang Y, Zhou C, Feng S, Qin X, Xie X, Liu C, Yu C. Exploration of the combined role of immune checkpoints and immune cells in the diagnosis and treatment of ankylosing spondylitis: a preliminary study immune checkpoints in ankylosing spondylitis. Arthritis Res Ther 2024; 26:115. [PMID: 38835033 DOI: 10.1186/s13075-024-03341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/12/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE Immune checkpoints have emerged as promising therapeutic targets for autoimmune diseases. However, the specific roles of immune checkpoints in the pathophysiology of ankylosing spondylitis (AS) remain unclear. METHODS Hip ligament samples were obtained from two patient groups: those with AS and femoral head deformity, and those with femoral head necrosis but without AS, undergoing hip arthroplasty. Label-Free Quantification (LFQ) Protein Park Analysis was used to identify the protein composition of the ligaments. Peripheral blood samples of 104 AS patients from public database were used to validate the expression of key proteins. KEGG, GO, and GSVA were employed to explore potential pathways regulated by immune checkpoints in AS progression. xCell was used to calculate cell infiltration levels, LASSO regression was applied to select key cells, and the correlation between immune checkpoints and immune cells was analyzed. Drug sensitivity analysis was conducted to identify potential therapeutic drugs targeting immune checkpoints in AS. The expression of key genes was validated through immunohistochemistry (IHC). RESULTS HLA-DMB and HLA-DPA1 were downregulated in the ligaments of AS and this has been validated through peripheral blood datasets and IHC. Significant differences in expression were observed in CD8 + Tcm, CD8 + T cells, CD8 + Tem, osteoblasts, Th1 cells, and CD8 + naive T cells in AS. The infiltration levels of CD8 + Tcm and CD8 + naive T cells were significantly positively correlated with the expression levels of HLA-DMB and HLA-DPA1. Immune cell selection using LASSO regression showed good predictive ability for AS, with AUC values of 0.98, 0.81, and 0.75 for the three prediction models, respectively. Furthermore, this study found that HLA-DMB and HLA-DPA1 are involved in Th17 cell differentiation, and both Th17 cell differentiation and the NF-kappa B signaling pathway are activated in the AS group. Drug sensitivity analysis showed that AS patients are more sensitive to drugs such as doramapimod and GSK269962A. CONCLUSION Immune checkpoints and immune cells could serve as avenues for exploring diagnostic and therapeutic strategies for AS.
Collapse
Affiliation(s)
- Feihong Huang
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Zhiping Su
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yibin Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yuxiang Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chengyu Zhou
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Sitan Feng
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiong Qin
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xi Xie
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chong Liu
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| | - Chaojie Yu
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
47
|
Romero‐Castillo L, Li T, Do N, Sareila O, Xu B, Hennings V, Xu Z, Svensson C, Oliveira‐Coelho A, Sener Z, Urbonaviciute V, Ekwall O, Burkhardt H, Holmdahl R. Human MHC Class II and Invariant Chain Knock-in Mice Mimic Rheumatoid Arthritis with Allele Restriction in Immune Response and Arthritis Association. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401513. [PMID: 38602454 PMCID: PMC11187888 DOI: 10.1002/advs.202401513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Indexed: 04/12/2024]
Abstract
Transgenic mice expressing human major histocompatibility complex class II (MHCII) risk alleles are widely used in autoimmune disease research, but limitations arise due to non-physiologic expression. To address this, physiologically relevant mouse models are established via knock-in technology to explore the role of MHCII in diseases like rheumatoid arthritis. The gene sequences encoding the ectodomains are replaced with the human DRB1*04:01 and 04:02 alleles, DRA, and CD74 (invariant chain) in C57BL/6N mice. The collagen type II (Col2a1) gene is modified to mimic human COL2. Importantly, DRB1*04:01 knock-in mice display physiologic expression of human MHCII also on thymic epithelial cells, in contrast to DRB1*04:01 transgenic mice. Humanization of the invariant chain enhances MHCII expression on thymic epithelial cells, increases mature B cell numbers in spleen, and improves antigen presentation. To validate its functionality, the collagen-induced arthritis (CIA) model is used, where DRB1*04:01 expression led to a higher susceptibility to arthritis, as compared with mice expressing DRB1*04:02. In addition, the humanized T cell epitope on COL2 allows autoreactive T cell-mediated arthritis development. In conclusion, the humanized knock-in mouse faithfully expresses MHCII, confirming the DRB1*04:01 alleles role in rheumatoid arthritis and being also useful for studying MHCII-associated diseases.
Collapse
MESH Headings
- Animals
- Mice
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- Humans
- Disease Models, Animal
- Mice, Inbred C57BL
- Mice, Transgenic
- Gene Knock-In Techniques/methods
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Alleles
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- HLA-DRB1 Chains/genetics
- HLA-DRB1 Chains/immunology
- Collagen Type II/genetics
- Collagen Type II/immunology
Collapse
Affiliation(s)
- Laura Romero‐Castillo
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Taotao Li
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Nhu‐Nguyen Do
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFraunhofer Cluster of Excellence for Immune‐Mediated Diseases CIMDTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
| | - Outi Sareila
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
- Medical Inflammation ResearchMediCity Research LaboratoryUniversity of TurkuTurkuFI‐20520Finland
| | - Bingze Xu
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Viktoria Hennings
- Department of PediatricsInstitute of Clinical Sciences and Department of Rheumatology and Inflammation ResearchInstitute of MedicineThe Sahlgrenska AcademyUniversity of GothenburgGothenburg41345Sweden
| | - Zhongwei Xu
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Carolin Svensson
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Ana Oliveira‐Coelho
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Zeynep Sener
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Vilma Urbonaviciute
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Olov Ekwall
- Department of PediatricsInstitute of Clinical Sciences and Department of Rheumatology and Inflammation ResearchInstitute of MedicineThe Sahlgrenska AcademyUniversity of GothenburgGothenburg41345Sweden
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFraunhofer Cluster of Excellence for Immune‐Mediated Diseases CIMDTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
- Division of RheumatologyUniversity Hospital FrankfurtGoethe University60590Frankfurt am MainGermany
| | - Rikard Holmdahl
- Medical Inflammation ResearchDivision of ImmunologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
- Medical Inflammation ResearchMediCity Research LaboratoryUniversity of TurkuTurkuFI‐20520Finland
| |
Collapse
|
48
|
Wang T, Yang J, Yang F, Cheng Y, Huang Z, Li B, Yang L, Xing Q, Luo X. The association between HLA-B variants and amoxicillin-induced severe cutaneous adverse reactions in Chinese han population. Front Pharmacol 2024; 15:1400239. [PMID: 38863977 PMCID: PMC11165025 DOI: 10.3389/fphar.2024.1400239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
Background Amoxicillin (AMX) is among the most prescribed and the best tolerated antimicrobials worldwide. However, it can occasionally trigger severe cutaneous adverse reactions (SCAR) with a significant morbidity and mortality. The genetic factors that may be relevant to AMX-induced SCAR (AMX-SCAR) remain unclear. Identification of the genetic risk factor may prevent patients from the risk of AMX exposure and resume therapy with other falsely implicated drugs. Methodology Four patients with AMX-SCAR, 1,000 population control and 100 AMX-tolerant individuals were enrolled in this study. Both exome-wide and HLA-based association studies were conducted. Molecular docking analysis was employed to simulate the interactions between AMX and risk HLA proteins. Results Compared with AMX-tolerant controls, a significant association of HLA-B*15:01 with AMX-SCAR was validated [odds ratio (OR) = 22.9, 95% confidence interval (CI): 1.68-1275.67; p = 7.34 × 10-3]. Moreover, 75% carriers of HLA-B*15:01 in four patients with AMX-SCAR, and the carrier frequency of 10.7% in 1,000 control individuals and 11.0% in 100 AMX-tolerant controls, respectively. Within HLA-B protein, the S140 present in all cases and demonstrated the strongest association with AMX-SCAR [OR = 53.5, p = 5.18 × 10-4]. Molecular docking results also confirmed the interaction between AMX and S140 of the HLA-B protein, thus eliminating the false-positive results during in association analysis. Conclusion Our findings suggest that genetic susceptibility may be involved in the development of AMX-SCAR in Han Chinese. However, whether the HLA-B variants observed in this study can be used as an effective genetic marker of AMX-induced SCAR still needs to be further explored in larger cohort studies and other ethnic populations.
Collapse
Affiliation(s)
- Ting Wang
- Children’s Hospital of Fudan University, Institutes of Biomedical Sciences of Fudan, Shanghai, China
| | - Jin Yang
- Department of Allergy and Immunology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Research Center of Allergy and Diseases, Fudan University, Shanghai, China
| | - Fanping Yang
- Department of Allergy and Immunology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Research Center of Allergy and Diseases, Fudan University, Shanghai, China
| | - Ye Cheng
- Children’s Hospital of Fudan University, Institutes of Biomedical Sciences of Fudan, Shanghai, China
| | - Zichong Huang
- College of Chemistry, Green Catalysis Center, Zhengzhou University, Zhengzhou, China
| | - Bei Li
- Children’s Hospital of Fudan University, Institutes of Biomedical Sciences of Fudan, Shanghai, China
| | - Linlin Yang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qinghe Xing
- Children’s Hospital of Fudan University, Institutes of Biomedical Sciences of Fudan, Shanghai, China
| | - Xiaoqun Luo
- Department of Allergy and Immunology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Research Center of Allergy and Diseases, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Sugiyama N, Terry FE, Gutierrez AH, Hirano T, Hoshi M, Mizuno Y, Martin W, Yasunaga S, Niiro H, Fujio K, De Groot AS. Individual and population-level variability in HLA-DR associated immunogenicity risk of biologics used for the treatment of rheumatoid arthritis. Front Immunol 2024; 15:1377911. [PMID: 38812524 PMCID: PMC11134572 DOI: 10.3389/fimmu.2024.1377911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/24/2024] [Indexed: 05/31/2024] Open
Abstract
Hypothesis While conventional in silico immunogenicity risk assessments focus on measuring immunogenicity based on the potential of therapeutic proteins to be processed and presented by a global population-wide set of human leukocyte antigen (HLA) alleles to T cells, future refinements might adjust for HLA allele frequencies in different geographic regions or populations, as well for as individuals in those populations. Adjustment by HLA allele distribution may reveal risk patterns that are specific to population groups or individuals, which current methods that rely on global-population HLA prevalence may obscure. Key findings This analysis uses HLA frequency-weighted binding predictions to define immunogenicity risk for global and sub-global populations. A comparison of assessments tuned for North American/European versus Japanese/Asian populations suggests that the potential for anti-therapeutic responses (anti-therapeutic antibodies or ATA) for several commonly prescribed Rheumatoid Arthritis (RA) therapeutic biologics may differ, significantly, between the Caucasian and Japanese populations. This appears to align with reports of differing product-related immunogenicity that is observed in different populations. Relevance to clinical practice Further definition of population-level (regional) and individual patient-specific immunogenic risk profiles may enable prescription of the RA therapeutic with the highest probability of success to each patient, depending on their population of origin and/or their individual HLA background. Furthermore, HLA-specific immunogenicity outcomes data are limited, thus there is a need to expand HLA-association studies that examine the relationship between HLA haplotype and ATA in the clinic.
Collapse
Affiliation(s)
- Naonobu Sugiyama
- Rheumatology, Inflammation and Immunology Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | | | | | - Toshitaka Hirano
- Rheumatology, Inflammation and Immunology Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | - Masato Hoshi
- Rheumatology, Inflammation and Immunology Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | - Yasushi Mizuno
- Rheumatology, Inflammation and Immunology Medical Affairs, Pfizer Japan Inc., Tokyo, Japan
| | | | - Shin’ichiro Yasunaga
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
50
|
Hum RM, Sharma SD, Stadler M, Viatte S, Ho P, Nair N, Shi C, Yap CF, Soomro M, Plant D, Humphreys JH, MacGregor A, Yates M, Verstappen S, Barton A, Bowes J. Using Polygenic Risk Scores to Aid Diagnosis of Patients With Early Inflammatory Arthritis: Results From the Norfolk Arthritis Register. Arthritis Rheumatol 2024; 76:696-703. [PMID: 38010198 DOI: 10.1002/art.42760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE There is growing evidence that genetic data are of benefit in the rheumatology outpatient setting by aiding early diagnosis. A genetic probability tool (G-PROB) has been developed to aid diagnosis has not yet been tested in a real-world setting. Our aim was to assess whether G-PROB could aid diagnosis in the rheumatology outpatient setting using data from the Norfolk Arthritis Register (NOAR), a prospective observational cohort of patients presenting with early inflammatory arthritis. METHODS Genotypes and clinician diagnoses were obtained from patients from NOAR. Six G-probabilities (0%-100%) were created for each patient based on known disease-associated odds ratios of published genetic risk variants, each corresponding to one disease of rheumatoid arthritis, systemic lupus erythematosus, psoriatic arthritis, spondyloarthropathy, gout, or "other diseases." Performance of the G-probabilities compared with clinician diagnosis was assessed. RESULTS We tested G-PROB on 1,047 patients. Calibration of G-probabilities with clinician diagnosis was high, with regression coefficients of 1.047, where 1.00 is ideal. G-probabilities discriminated clinician diagnosis with pooled areas under the curve (95% confidence interval) of 0.85 (0.84-0.86). G-probabilities <5% corresponded to a negative predictive value of 96.0%, for which it was possible to suggest >2 unlikely diseases for 94% of patients and >3 for 53.7% of patients. G-probabilities >50% corresponded to a positive predictive value of 70.4%. In 55.7% of patients, the disease with the highest G-probability corresponded to clinician diagnosis. CONCLUSION G-PROB converts complex genetic information into meaningful and interpretable conditional probabilities, which may be especially helpful at eliminating unlikely diagnoses in the rheumatology outpatient setting.
Collapse
Affiliation(s)
- Ryan M Hum
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Seema D Sharma
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Michael Stadler
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Sebastien Viatte
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Pauline Ho
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Nisha Nair
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Chenfu Shi
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Chuan Fu Yap
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Mehreen Soomro
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Darren Plant
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Jenny H Humphreys
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | | | - Max Yates
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Suzanne Verstappen
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Anne Barton
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - John Bowes
- Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| |
Collapse
|