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.
Key points
-
Next-generation sequencing has enabled development of comprehensive and accurate T cell receptor profiling, allowing studies to search for expanded T cell clonotypes suggestive of shared antigenic drivers.
-
Using these approaches, expanded CD8 T cell clonotypes bearing highly similar T cell receptor (TCR) β-chains and α-chains have been identified in a high proportion of patients with ankylosing spondylitis, and this information has been used to identify expanded T cell clonotypes potentially involved in the pathogenesis of the disease.
-
TCR repertoire studies in other rheumatic diseases, including psoriatic arthritis, rheumatoid arthritis, systemic lupus erythematosus and Sjögren syndrome, have not found consistently expanded T cell clonotypes, although evidence of T cell expansions including at sites of inflammation have been reported.
-
Further studies focusing on early disease and more clinically, genetically and immunologically homogeneous cohorts might provide more informative to identify expanded aetiopathogenic T cell clonotypes.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Garrido-Mesa, J. & Brown, M. A. T cell repertoire profiling and the mechanism by which HLA-B27 causes ankylosing spondylitis. Curr. Rheumatol. Rep. 24, 398–410 (2022).
Oettinger, M. A., Schatz, D. G., Gorka, C. & Baltimore, D. RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science 248, 1517–1523 (1990).
Bassing, C. H., Swat, W. & Alt, F. W. The mechanism and regulation of chromosomal V(D)J recombination. Cell 109, S45–S55 (2002).
Turner, S. J., Doherty, P. C., McCluskey, J. & Rossjohn, J. Structural determinants of T-cell receptor bias in immunity. Nat. Rev. Immunol. 6, 883–894 (2006).
Wooldridge, L. et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J. Biol. Chem. 287, 1168–1177 (2012).
Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. A. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014).
Warren, R. L. et al. Exhaustive T-cell repertoire sequencing of human peripheral blood samples reveals signatures of antigen selection and a directly measured repertoire size of at least 1 million clonotypes. Genome Res. 21, 790–797 (2011).
Li, H., Ye, C., Ji, G. & Han, J. Determinants of public T cell responses. Cell Res. 22, 33–42 (2012).
Gate, D. et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 577, 399–404 (2020).
Wardemann, H. & Busse, C. E. Novel approaches to analyze immunoglobulin repertoires. Trends Immunol. 38, 471–482 (2017).
Woodsworth, D. J., Castellarin, M. & Holt, R. A. Sequence analysis of T-cell repertoires in health and disease. Genome Med. 5, 98 (2013).
Brown, S. D., Raeburn, L. A. & Holt, R. A. Profiling tissue-resident T cell repertoires by RNA sequencing. Genome Med. 7, 125 (2015).
Greiff, V., Miho, E., Menzel, U. & Reddy, S. T. Bioinformatic and statistical analysis of adaptive immune repertoires. Trends Immunol. 36, 738–749 (2015).
Miho, E. et al. Computational strategies for dissecting the high-dimensional complexity of adaptive immune repertoires. Front. Immunol. 9, 224 (2018).
Henry, V. J., Bandrowski, A. E., Pepin, A. S., Gonzalez, B. J. & Desfeux, A. OMICtools: an informative directory for multi-omic data analysis. Database 2014, bau069 (2014).
Barwell, L. J., Isaac, N. J. & Kunin, W. E. Measuring β-diversity with species abundance data. J. Anim. Ecol. 84, 1112–1122 (2015).
Miles, J. J., Douek, D. C. & Price, D. A. Bias in the αβ T-cell repertoire: implications for disease pathogenesis and vaccination. Immunol. Cell Biol. 89, 375–387 (2011).
Dash, P. et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature 547, 89–93 (2017).
Glanville, J. et al. Identifying specificity groups in the T cell receptor repertoire. Nature 547, 94–98 (2017).
Ostmeyer, J., Christley, S., Toby, I. T. & Cowell, L. G. Biophysicochemical motifs in T-cell receptor sequences distinguish repertoires from tumor-infiltrating lymphocyte and adjacent healthy tissue. Cancer Res. 79, 1671–1680 (2019).
Davis, M. M. & Boyd, S. D. Recent progress in the analysis of αβT cell and B cell receptor repertoires. Curr. Opin. Immunol. 59, 109–114 (2019).
Kim, S. M. et al. Analysis of the paired TCR α- and β-chains of single human T cells. PLoS ONE 7, e37338 (2012).
Stubbington, M. J. T., Rozenblatt-Rosen, O., Regev, A. & Teichmann, S. A. Single-cell transcriptomics to explore the immune system in health and disease. Science 358, 58–63 (2017).
Proserpio, V. & Mahata, B. Single-cell technologies to study the immune system. Immunology 147, 133–140 (2016).
Robinson, W. P. et al. HLA-Bw60 increases susceptibility to ankylosing spondylitis in HLA-B27+ patients. Arthritis Rheum. 32, 1135–1141 (1989).
Brown, M. A. et al. HLA class I associations of ankylosing spondylitis in the white population in the United Kingdom. Ann. Rheum. Dis. 55, 268–270 (1996).
Chang, S. C., Momburg, F., Bhutani, N. & Goldberg, A. L. The ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a “molecular ruler” mechanism. Proc. Natl Acad. Sci. USA 102, 17107–17112 (2005).
Evans, D. M. et al. Interaction between ERAP1 and HLA-B27 in ankylosing spondylitis implicates peptide handling in the mechanism for HLA-B27 in disease susceptibility. Nat. Genet. 43, 761–767 (2011).
Cortes, A. et al. Major histocompatibility complex associations of ankylosing spondylitis are complex and involve further epistasis with ERAP1. Nat. Commun. 6, 7146 (2015).
Colbert, R. A. The immunobiology of HLA-B27: variations on a theme. Curr. Mol. Med. 4, 21–30 (2004).
Hermann, E., Yu, D. T., Meyer zum Buschenfelde, K. H. & Fleischer, B. HLA-B27-restricted CD8 T cells derived from synovial fluids of patients with reactive arthritis and ankylosing spondylitis. Lancet 342, 646–650 (1993).
Duchmann, R. et al. HLA-B27-restricted cytotoxic T lymphocyte responses to arthritogenic enterobacteria or self-antigens are dominated by closely related TCRBV gene segments. A study in patients with reactive arthritis. Scand. J. Immunol. 43, 101–108 (1996).
Dulphy, N. et al. Common intra-articular T cell expansions in patients with reactive arthritis: identical beta-chain junctional sequences and cytotoxicity toward HLA-B27. J. Immunol. 162, 3830–3839 (1999). First report of the AS-associated TRBV9-J2S3 CDR3 motif.
May, E. et al. Conserved TCR β chain usage in reactive arthritis; evidence for selection by a putative HLA-B27-associated autoantigen. Tissue Antigens 60, 299–308 (2002).
Faham, M. et al. Discovery of T cell receptor β motifs specific to HLA-B27-positive ankylosing spondylitis by deep repertoire sequence analysis. Arthritis Rheumatol. 69, 774–784 (2017). Largest TCR profiling study in AS using NGS-based methods, HLA-B27 typing and controls including patients with non-AS rheumatic disease.
Komech, E. A. et al. CD8+ T cells with characteristic T cell receptor beta motif are detected in blood and expanded in synovial fluid of ankylosing spondylitis patients. Rheumatology 57, 1097–1104 (2018).
Zheng, M. et al. TCR repertoire and CDR3 motif analyses depict the role of αβ T cells in ankylosing spondylitis. EBioMedicine 47, 414–426 (2019).
Hanson et al. T-cell receptor immunosequencing reveals altered repertoire diversity and disease-associated clonal expansions in ankylosing spondylitis patients. Arthritis Rheumatol. 72, 1289–1302 (2020). This study provides a comprehensive description of TCR repertoire alterations in AS, including both CD8 and CD4 T cell clonotype associations, showing that this is not just a feature of HLA-B27 carriage but rather of HLA-B27-associated axSpA.
Komech, E. A. et al. TCR repertoire profiling revealed antigen-driven CD8+ T cell clonal groups shared in synovial fluid of patients with spondyloarthritis. Front. Immunol. 13, 973243 (2022). This study reports TCR associations with HLA-B38+ PsA and HLA-B27+ SpA, including the first report of AS-associated expansions among patients with PsA.
Gracey, E. et al. IL-7 primes IL-17 in mucosal-associated invariant T (MAIT) cells, which contribute to the Th17-axis in ankylosing spondylitis. Ann. Rheum. Dis. 75, 2124–2132 (2016).
Yang, X. et al. Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides. Nature 612, 771–777 (2022). This breakthrough study identifies the paired TCRαβ sequences of AS-associated TRBV9 clonotypes in AS and acute anterior uveitis and performs a yeast display peptide screening to identify potential antigenic peptides and their protein sources.
Deschler, K. et al. Antigen-specific immune reactions by expanded CD8+ T cell clones from HLA-B*27-positive patients with spondyloarthritis. J. Autoimmun. 133, 102901 (2022).
Paley, M. A. et al. Mucosal signatures of pathogenic T cells in HLA-B*27+ anterior uveitis and axial spondyloarthritis. JCI Insight 9, e174776 (2024). This study describes phenotypic signatures among AS-associated clonotypes reacting to one of the identified antigenic peptides, YeiH, using scRNA sequencing and peptide–HLA tetramer technologies for targeted TCR screening and antigenic validation.
Robinson, P. C., Wordsworth, B. P., Reveille, J. D. & Brown, M. A. Axial spondyloarthritis: a new disease entity, not necessarily early ankylosing spondylitis. Ann. Rheum. Dis. 72, 162–164 (2013).
van der Linden, S., Akkoc, N., Brown, M. A., Robinson, P. C. & Khan, M. A. The ASAS criteria for axial spondyloarthritis: strengths, weaknesses, and proposals for a way forward. Curr. Rheumatol. Rep. 17, 62 (2015).
Barkham, N., Marzo-Ortega, H., McGonagle, D. & Emery, P. How to diagnose axial spondyloarthropathy early. Ann. rheumatic Dis. 63, 471–472 (2004).
Winchester, R. & FitzGerald, O. MHC class I associations beyond HLA-B27: the peptide binding hypothesis of psoriatic arthritis and its implications for disease pathogenesis. Curr. Opin. Rheumatol. 32, 330–336 (2020).
Batko, B. Exploring the diverse immune and genetic landscape of psoriatic arthritis. J. Clin. Med. 10, 5926 (2021).
Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consirtium 2 A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat. Genet. 42, 985–990 (2010).
Costello, P., Bresnihan, B., O’Farrelly, C. & FitzGerald, O. Predominance of CD8+ T lymphocytes in psoriatic arthritis. J. Rheumatol. 26, 1117–1124 (1999).
Costello, P. J. et al. Psoriatic arthritis joint fluids are characterized by CD8 and CD4 T cell clonal expansions appear antigen driven. J. Immunol. 166, 2878–2886 (2001).
Tassiulas, I., Duncan, S. R., Centola, M., Theofilopoulos, A. N. & Boumpas, D. T. Clonal characteristics of T cell infiltrates in skin and synovium of patients with psoriatic arthritis. Hum. Immunol. 60, 479–491 (1999).
Borgato, L. et al. The T cell receptor repertoire in psoriatic synovitis is restricted and T lymphocytes expressing the same TCR are present in joint and skin lesions. J. Rheumatol. 29, 1914–1919 (2002).
Sigmundsdottir, H. et al. Circulating T cells of patients with active psoriasis respond to streptococcal M-peptides sharing sequences with human epidermal keratins. Scand. J. Immunol. 45, 688–697 (1997).
Curran, S. A. et al. Nucleotide sequencing of psoriatic arthritis tissue before and during methotrexate administration reveals a complex inflammatory T cell infiltrate with very few clones exhibiting features that suggest they drive the inflammatory process by recognizing autoantigens. J. Immunol. 172, 1935–1944 (2004).
Goldstein, I. et al. Synovial VLA-1+ T cells display an oligoclonal and partly distinct repertoire in rheumatoid and psoriatic arthritis. Clin. Immunol. 128, 75–84 (2008).
Cheuk, S. et al. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 46, 287–300 (2017).
Steel, K. J. A. et al. Polyfunctional, proinflammatory, tissue-resident memory phenotype and function of synovial interleukin-17A+CD8+ T cells in psoriatic arthritis. Arthritis Rheumatol. 72, 435–447 (2020).
Penkava, F. et al. Single-cell sequencing reveals clonal expansions of pro-inflammatory synovial CD8 T cells expressing tissue-homing receptors in psoriatic arthritis. Nat. Commun. 11, 4767 (2020).
Povoleri, G. A. M. et al. Psoriatic and rheumatoid arthritis joints differ in the composition of CD8+ tissue-resident memory T cell subsets. Cell Rep. 42, 112514 (2023).
Helliwell, P. S., Mease, P. J., FitzGerald, O., Taylor, W. J. & van der Heijde, D. Peripheral spondyloarthritis and psoriatic arthritis; overlaps and distinctions: a report from the GRAPPA 2012 annual meeting. J. Rheumatol. 40, 1446–1449 (2013).
Sokolove, J. et al. Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. PLoS ONE 7, e35296 (2012).
De Vita, S. et al. Efficacy of selective B cell blockade in the treatment of rheumatoid arthritis: evidence for a pathogenetic role of B cells. Arthritis Rheum. 46, 2029–2033 (2002).
VanderBorght, A., Geusens, P., Vandevyver, C., Raus, J. & Stinissen, P. Skewed T-cell receptor variable gene usage in the synovium of early and chronic rheumatoid arthritis patients and persistence of clonally expanded T cells in a chronic patient. Rheumatology 39, 1189–1201 (2000).
Lim, A. et al. Spread of clonal T-cell expansions in rheumatoid arthritis patients. Hum. Immunol. 48, 77–83 (1996).
Striebich, C. C., Falta, M. T., Wang, Y., Bill, J. & Kotzin, B. L. Selective accumulation of related CD4+ T cell clones in the synovial fluid of patients with rheumatoid arthritis. J. Immunol. 161, 4428–4436 (1998).
Stastny, P. Association of the B-cell alloantigen DRw4 with rheumatoid arthritis. N. Engl. J. Med. 298, 869–871 (1978).
Ikeda, Y. et al. High frequencies of identical T cell clonotypes in synovial tissues of rheumatoid arthritis patients suggest the occurrence of common antigen-driven immune responses. Arthritis Rheum. 39, 446–453 (1996).
Alam, A. et al. Persistence of dominant T cell clones in synovial tissues during rheumatoid arthritis. J. Immunol. 156, 3480–3485 (1996).
Kato, T. et al. T cell clonality in synovial fluid of a patient with rheumatoid arthritis: persistent but fluctuant oligoclonal T cell expansions. J. Immunol. 159, 5143–5149 (1997).
Cantaert, T. et al. Alterations of the synovial T cell repertoire in anti-citrullinated protein antibody-positive rheumatoid arthritis. Arthritis Rheum. 60, 1944–1956 (2009).
Klarenbeek, P. L. et al. Inflamed target tissue provides a specific niche for highly expanded T-cell clones in early human autoimmune disease. Ann. Rheum. Dis. 71, 1088–1093 (2012). This study compares patients with early and late disease RA, reporting the dynamics of TCR.
Mizushima, N. et al. HLA-dependent peripheral T cell receptor (TCR) repertoire formation and its modification by rheumatoid arthritis (RA). Clin. Exp. Immunol. 110, 428–433 (1997). Besides using low-resolution techniques, the authors used an interesting familiar study design that could facilitate the identification of repertoire findings specific to RA development.
Paliard, X. et al. Evidence for the effects of a superantigen in rheumatoid arthritis. Science 253, 325–329 (1991).
Howell, M. D. et al. Limited T-cell receptor beta-chain heterogeneity among interleukin 2 receptor-positive synovial T cells suggests a role for superantigen in rheumatoid arthritis. Proc. Natl Acad. Sci. USA 88, 10921–10925 (1991).
Sun, W. et al. Skewed T-cell receptor BV14 and BV16 expression and shared CDR3 sequence and common sequence motifs in synovial T cells of rheumatoid arthritis. Genes. Immun. 6, 248–261 (2005).
Jenkins, R. N., Nikaein, A., Zimmermann, A., Meek, K. & Lipsky, P. E. T cell receptor V beta gene bias in rheumatoid arthritis. J. Clin. Invest. 92, 2688–2701 (1993).
Waase, I., Kayser, C., Carlson, P. J., Goronzy, J. J. & Weyand, C. M. Oligoclonal T cell proliferation in patients with rheumatoid arthritis and their unaffected siblings. Arthritis Rheum. 39, 904–913 (1996).
Stamenkovic, I. et al. Clonal dominance among T-lymphocyte infiltrates in arthritis. Proc. Natl Acad. Sci. USA 85, 1179–1183 (1988).
Alam, A. et al. T-cell receptor variable region of the beta-chain gene use in peripheral blood and multiple synovial membranes during rheumatoid arthritis. Hum. Immunol. 42, 331–339 (1995).
Schmidt, D., Martens, P. B., Weyand, C. M. & Goronzy, J. J. The repertoire of CD4+ CD28− T cells in rheumatoid arthritis. Mol. Med. 2, 608–618 (1996).
Grom, A. A. et al. Dominant T-cell-receptor beta chain variable region V beta 14+ clones in juvenile rheumatoid arthritis. Proc. Natl Acad. Sci. USA 90, 11104–11108 (1993).
Davey, M. P., Burgoine, G. A. & Woody, C. N. TCRB clonotypes are present in CD4+ T cell populations prepared directly from rheumatoid synovium. Hum. Immunol. 55, 11–21 (1997).
Bröker, B. M. et al. Biased T cell receptor V gene usage in rheumatoid arthritis. Oligoclonal expansion of T cells expressing V alpha 2 genes in synovial fluid but not in peripheral blood. Arthritis Rheum. 36, 1234–1243 (1993).
Sakkas, L. I., Chen, P. F. & Platsoucas, C. D. T-cell antigen receptors in rheumatoid arthritis. Immunol. Res. 13, 117–138 (1994).
Spreafico, R. et al. A circulating reservoir of pathogenic-like CD4+ T cells shares a genetic and phenotypic signature with the inflamed synovial micro-environment. Ann. Rheum. Dis. 75, 459–465 (2016).
Chini, L. et al. Evidence of clonotypic pattern of T-cell repertoire in synovial fluid of children with juvenile rheumatoid arthritis at the onset of the disease. Scand. J. Immunol. 56, 512–517 (2002).
Liu, X. et al. T cell receptor β repertoires as novel diagnostic markers for systemic lupus erythematosus and rheumatoid arthritis. Ann. Rheum. Dis. 78, 1070–1078 (2019). Largest TCR profiling study in rheumatic conditions, including large cohorts of patients with RA and SLE and reporting specific and overlapping autoimmune signatures.
Chang, C. M. et al. Characterization of T-cell receptor repertoire in patients with rheumatoid arthritis receiving biologic therapies. Dis. Markers 2019, 2364943 (2019).
Di Sante, G. et al. Collagen specific T-cell repertoire and HLA-DR alleles: biomarkers of active refractory rheumatoid arthritis. EBioMedicine 2, 2037–2045 (2015).
Imberti, L. et al. Reduced T-cell repertoire restrictions in abatacept-treated rheumatoid arthritis patients. J. Transl. Med. 13, 12 (2015).
Sakurai, K. et al. HLA-DRB1 shared epitope alleles and disease activity are correlated with reduced T cell receptor repertoire diversity in CD4+ T cells in rheumatoid arthritis. J. Rheumatol. 45, 905–914 (2018).
Jiang, X. et al. Comprehensive TCR repertoire analysis of CD4+ T-cell subsets in rheumatoid arthritis. J. Autoimmunity 109, 102432 (2020). This study evaluates TCR repertoire targeted to defined phenotypic populations, allowing identification of the immune subtypes expanded and potentially involved in RA autoimmunity.
Zheng, Z. et al. Database of synovial T cell repertoire of rheumatoid arthritis patients identifies cross-reactive potential against pathogens including unencountered SARS-CoV-2. Ann. Rheum. Dis. 82, 438–440 (2023).
Ishigaki, K. et al. Quantitative and qualitative characterization of expanded CD4+ T cell clones in rheumatoid arthritis patients. Sci. Rep. 5, 12937 (2015).
Argyriou, A. et al. Single cell sequencing identifies clonally expanded synovial CD4+ TPH cells expressing GPR56 in rheumatoid arthritis. Nat. Commun. 13, 4046 (2022). This study identifies expansion of TPH cells in synovial fluid of patients with RA and expression of phenotypic markers of interest.
von Delwig, A., Locke, J., Robinson, J. H. & Ng, W. F. Response of Th17 cells to a citrullinated arthritogenic aggrecan peptide in patients with rheumatoid arthritis. Arthritis Rheum. 62, 143–149 (2010).
Zhou, J. et al. Skewness of TCR Vβ of peripheral blood and synovial fluid of patients with rheumatoid arthritis. J. Immunoass. Immunochem. 35, 207–219 (2014).
Wagner, U. et al. Clonally expanded CD4+CD28null T cells in rheumatoid arthritis use distinct combinations of T cell receptor BV and BJ elements. Eur. J. Immunol. 33, 79–84 (2003).
Musters, A. et al. In rheumatoid arthritis, synovitis at different inflammatory sites is dominated by shared but patient-specific T cell clones. J. Immunol. 201, 417–422 (2018). By studying paired blood and tissue samples, this study highlights the relevance of study tissue infiltrating lymphocytes in improving capture of immunological disturbances.
Lamacchia, C. et al. Detection of circulating highly expanded T-cell clones in at-risk individuals for rheumatoid arthritis before the clinical onset of the disease. Rheumatology 60, 3451–3460 (2021).
Dunlap, G. et al. Clonal associations between lymphocyte subsets and functional states in rheumatoid arthritis synovium. Nat. Commun. 15, 4991 (2024). Applying scRNA-seq to paired blood and tissue samples, this interesting study pinpoints numerous immune subsets potentially involved in antigenic responses in RA through evaluation of their clonotypic expansions and activation profile.
Hingorani, R. et al. Oligoclonality of V beta 3 TCR chains in the CD8+ T cell population of rheumatoid arthritis patients. J. Immunol. 156, 852–858 (1996).
Hall, F. C., Thomson, K., Procter, J., McMichael, A. J. & Wordsworth, B. P. TCR beta spectratyping in RA: evidence of clonal expansions in peripheral blood lymphocytes. Ann. Rheum. Dis. 57, 319–322 (1998).
Wang, E. C. et al. CD8high+ (CD57+) T cells in patients with rheumatoid arthritis. Arthritis Rheum. 40, 237–248 (1997).
Jung, J. et al. Synovial fluid CD69+CD8+ T cells with tissue-resident phenotype mediate perforin-dependent citrullination in rheumatoid arthritis. Clin. Transl. Immunol. 9, e1140 (2020).
Matulis, G. et al. Innate-like control of human iNKT cell autoreactivity via the hypervariable CDR3β loop. PLoS Biol. 8, e1000402 (2010).
Mansour, S. et al. Structural and functional changes of the invariant NKT clonal repertoire in early rheumatoid arthritis. J. Immunol. 195, 5582–5591 (2015).
Kojo, S., Adachi, Y., Keino, H., Taniguchi, M. & Sumida, T. Dysfunction of T cell receptor AV24AJ18+, BV11+ double-negative regulatory natural killer T cells in autoimmune diseases. Arthritis Rheum. 44, 1127–1138 (2001).
Tudhope, S. J. et al. Profound invariant natural killer T-cell deficiency in inflammatory arthritis. Ann. Rheum. Dis. 69, 1873–1879 (2010).
Linsen, L. et al. Peripheral blood but not synovial fluid natural killer T cells are biased towards a Th1-like phenotype in rheumatoid arthritis. Arthritis Res. Ther. 7, R493–R502 (2005).
Rao, D. A. et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114 (2017).
Sakuragi, T. et al. Autoreactivity of peripheral helper T cells in the joints of rheumatoid arthritis. J. Immunol. 206, 2045–2051 (2021).
Pitzalis, C., Jones, G. W., Bombardieri, M. & Jones, S. A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 14, 447–462 (2014).
Catrina, A. I., Svensson, C. I., Malmström, V., Schett, G. & Klareskog, L. Mechanisms leading from systemic autoimmunity to joint-specific disease in rheumatoid arthritis. Nat. Rev. Rheumatol. 13, 79–86 (2017).
Turcinov, S. et al. Diversity and clonality of T cell receptor repertoire and antigen specificities in small joints of early rheumatoid arthritis. Arthritis Rheumatol. 75, 673–684 (2023).
Fazou, C., Yang, H., McMichael, A. J. & Callan, M. F. Epitope specificity of clonally expanded populations of CD8+ T cells found within the joints of patients with inflammatory arthritis. Arthritis Rheum. 44, 2038–2045 (2001).
Bentham, J. et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat. Genet. 47, 1457–1464 (2015).
Datta, S. K., Kaliyaperumal, A., Mohan, C. & Desai-Mehta, A. T helper cells driving pathogenic anti-DNA autoantibody production in lupus: nucleosomal epitopes and CD40 ligand signals. Lupus 6, 333–336 (1997).
Desai-Mehta, A., Mao, C., Rajagopalan, S., Robinson, T. & Datta, S. K. Structure and specificity of T cell receptors expressed by potentially pathogenic anti-DNA autoantibody-inducing T cells in human lupus. J. Clin. Invest. 95, 531–541 (1995). This report highlights TCR sequences and reactivity of cell lines and CD4+ T cells with anti-double-stranded DNA antibody-inducing capacity against SLE-relevant nuclear proteins, supporting the hypothesis of cross-reactivity between humoral and cellular immunity.
Kita, Y. et al. T cell receptor clonotypes in skin lesions from patients with systemic lupus erythematosus. J. Invest. Dermatol. 110, 41–46 (1998).
Luo, W. et al. Analysis of the interindividual conservation of T cell receptor α- and β-chain variable regions gene in the peripheral blood of patients with systemic lupus erythematosus. Clin. Exp. Immunol. 154, 316–324 (2008).
Kolowos, W. et al. Detection of restricted junctional diversity of peripheral T cells in SLE patients by spectratyping. Lupus 6, 701–707 (1997).
Olive, C., Gatenby, P. A. & Serjeantson, S. W. Restricted junctional diversity of T cell receptor δ gene rearrangements expressed in systemic lupus erythematosus (SLE) patients. Clin. Exp. Immunol. 97, 430–438 (1994).
Murata, H. et al. T cell receptor repertoire of T cells in the kidneys of patients with lupus nephritis. Arthritis Rheum. 46, 2141–2147 (2002).
Winchester, R. et al. Immunologic characteristics of intrarenal T cells: trafficking of expanded CD8+ T cell β-chain clonotypes in progressive lupus nephritis. Arthritis Rheum. 64, 1589–1600 (2012). This study identified expanded CD8+ and CD4+ T cells in renal tissue of patients with SLE.
Mato, T. et al. Correlation of clonal T cell expansion with disease activity in systemic lupus erythematosus. Int. Immunol. 9, 547–554 (1997).
Holbrook, M. R., Tighe, P. J. & Powell, R. J. Restrictions of T cell receptor β chain repertoire in the peripheral blood of patients with systemic lupus erythematosus. Ann. Rheum. Dis. 55, 627–631 (1996).
Alexander, T. et al. Foxp3+ Helios+ regulatory T cells are expanded in active systemic lupus erythematosus. Ann. Rheum. Dis. 72, 1549–1558 (2013).
Costa, N. et al. Broadened T-cell repertoire diversity in ivIg-treated SLE patients is also related to the individual status of regulatory T-cells. J. Clin. Immunol. 33, 349–360 (2013).
Thapa, D. R. et al. Longitudinal analysis of peripheral blood T cell receptor diversity in patients with systemic lupus erythematosus by next-generation sequencing. Arthritis Res. Ther. 17, 132 (2015).
Yu, J. et al. Case report for recurrent and new-onset SLE patients treated by high-dose glucocorticoid therapy: characteristics of peripheral TCR beta chain CDR3 repertoires. Medicine 96, e9022 (2017).
Ye, X. et al. High-throughput sequencing-based analysis of T cell repertoire in lupus nephritis. Front. Immunol. 11, 1618 (2020).
Jakez-Ocampo, J. et al. Vβ T cell receptor (TCR) genes in circulating cells of patients with systemic lupus erythematosus and their healthy relatives [Spanish]. Gac. Med. Mex. 154, 74–79 (2018).
Sui, W. et al. Composition and variation analysis of the TCR β-chain CDR3 repertoire in systemic lupus erythematosus using high-throughput sequencing. Mol. Immunol. 67, 455–464 (2015).
Tzifi, F. et al. Flow cytometric analysis of the CD4+ TCR Vβ repertoire in the peripheral blood of children with type 1 diabetes mellitus, systemic lupus erythematosus and age-matched healthy controls. BMC Immunol. 14, 33 (2013).
Kato, T. et al. Analysis of accumulated T cell clonotypes in patients with systemic lupus erythematosus. Arthritis Rheum. 43, 2712–2721 (2000).
Massengill, S. F., Goodenow, M. M. & Sleasman, J. W. SLE nephritis is associated with an oligoclonal expansion of intrarenal T cells. Am. J. Kidney Dis. 31, 418–426 (1998).
Kolowos, W. et al. CD4 positive peripheral T cells from patients with systemic lupus erythematosus (SLE) are clonally expanded. Lupus 10, 321–331 (2001). This study suggests that the presence of acidic amino acid residues within clonally expanded CD4 T cells mediates recognition of charged epitopes such as those present in nucleosomes, relevant to SLE pathology.
Mohan, C., Adams, S., Stanik, V. & Datta, S. K. Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J. Exp. Med. 177, 1367–1381 (1993).
Perez, R. K. et al. Single-cell RNA-seq reveals cell type-specific molecular and genetic associations to lupus. Science 376, eabf1970 (2022). This scRNA-seq study in a large cohort of patients with SLE identifies expanded cytotoxic CD8 T cells with potential implication in the pathology.
Blanco, P. et al. Increase in activated CD8+ T lymphocytes expressing perforin and granzyme B correlates with disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 52, 201–211 (2005).
Bosma, A., Abdel-Gadir, A., Isenberg, D. A., Jury, E. C. & Mauri, C. Lipid-antigen presentation by CD1d+ B cells is essential for the maintenance of invariant natural killer T cells. Immunity 36, 477–490 (2012).
Rajagopalan, S., Zordan, T., Tsokos, G. C. & Datta, S. K. Pathogenic anti-DNA autoantibody-inducing T helper cell lines from patients with active lupus nephritis: isolation of CD4−8− T helper cell lines that express the γδ T-cell antigen receptor. Proc. Natl Acad. Sci. USA 87, 7020–7024 (1990).
Yin, S. et al. Hyperactivation and in situ recruitment of inflammatory Vδ2 T cells contributes to disease pathogenesis in systemic lupus erythematosus. Sci. Rep. 5, 14432 (2015).
Jonsson, M. V., Skarstein, K., Jonsson, R. & Brun, J. G. Serological implications of germinal center-like structures in primary Sjögren’s syndrome. J. Rheumatol. 34, 2044–2049 (2007).
Manoussakis, M. N., Tzioufas, A. G., Pange, P. J. & Moutsopoulos, H. M. Serological profiles in subgroups of patients with Sjögren’s syndrome. Scand. J. Rheumatol. Suppl. 61, 89–92 (1986).
Smith, M. D. et al. Selective expression of V beta families by T cells in the blood and salivary gland infiltrate of patients with primary Sjögren’s syndrome. J. Rheumatol. 21, 1832–1837 (1994).
Kay, R. A. et al. An abnormal T cell repertoire in hypergammaglobulinaemic primary Sjögren’s syndrome. Clin. Exp. Immunol. 85, 262–264 (1991).
Mizushima, N., Kohsaka, H., Tsubota, K., Saito, I. & Miyasaka, N. Diverse T cell receptor beta gene usage by infiltrating T cells in the lacrimal glands of Sjögren’s syndrome. Clin. Exp. Immunol. 101, 33–38 (1995).
Matsumoto, I. et al. Common T cell receptor clonotype in lacrimal glands and labial salivary glands from patients with Sjögren’s syndrome. J. Clin. Invest. 97, 1969–1977 (1996).
Sasaki, M. et al. Accumulation of common T cell clonotypes in the salivary glands of patients with human T lymphotropic virus type I-associated and idiopathic Sjögren’s syndrome. J. Immunol. 164, 2823–2831 (2000).
Ohyama, Y. et al. T-cell receptor Vα and Vβ gene use by infiltrating T cells in labial glands of patients with Sjögren’s syndrome. Oral. Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 79, 730–737 (1995).
Sumida, T. et al. TCR in Fas-sensitive T cells from labial salivary glands of patients with Sjögren’s syndrome. J. Immunol. 158, 1020–1025 (1997).
Ajjan, R. A. et al. Analysis of the T-cell receptor Valpha repertoire and cytokine gene expression in Sjögren’s syndrome. Br. J. Rheumatol. 37, 179–185 (1998).
Sumida, T. et al. T cell receptor repertoire of infiltrating T cells in lips of Sjögren’s syndrome patients. J. Clin. Invest. 89, 681–685 (1992).
Sumida, T. et al. T cell receptor V alpha repertoire of infiltrating T cells in labial salivary glands from patients with Sjögren’s syndrome. J. Rheumatol. 21, 1655–1661 (1994).
Murata, H. et al. Limited TCR repertoire of infiltrating T cells in the kidneys of Sjögren’s syndrome patients with interstitial nephritis. J. Immunol. 155, 4084–4089 (1995).
Matsumoto, I. et al. Single cell analysis of T cells infiltrating labial salivary glands from patients with Sjögren’s syndrome. Int. J. Mol. Med. 4, 519–527 (1999).
Lu, C. et al. Clinical significance of T cell receptor repertoire in primary Sjogren’s syndrome. EBioMedicine 84, 104252 (2022). This is a well-controlled study of a large cohort of patients with pSS evaluating the circulating TCR repertoire at three different time points throughout the disease.
Joachims, M. L. et al. Single-cell analysis of glandular T cell receptors in Sjögren’s syndrome. JCI Insight 1, e85609 (2016). This scRNA-seq study identifies tissue enrichment of potentially pathogenic CD4+ clonotypes, including some HLA-DR3/DQ2 associations, and in correlation with glandular dysfunction.
Voigt, A. et al. Unique glandular ex-vivo Th1 and Th17 receptor motifs in Sjögren’s syndrome patients using single-cell analysis. Clin. Immunol. 192, 58–67 (2018). This single-cell TCR sequencing study identifies shared CDR3 motifs among TH1 and TH17 cells from salivary gland tissue-infiltrating cells in pSS.
Rowe, J. H. et al. Abnormalities of T-cell receptor repertoire in CD4+ regulatory and conventional T cells in patients with RAG mutations: implications for autoimmunity. J. Allergy Clin. Immunol. 140, 1739–1743.e7 (2017).
Ni, P. P., Solomon, B., Hsieh, C. S., Allen, P. M. & Morris, G. P. The ability to rearrange dual TCRs enhances positive selection, leading to increased allo- and autoreactive T cell repertoires. J. Immunol. 193, 1778–1786 (2014).
Hou, X. et al. Analysis of gene expression and TCR/B cell receptor profiling of immune cells in primary Sjögren’s syndrome by single-cell sequencing. J. Immunol. 209, 238–249 (2022).
Hong, X. et al. Single-cell RNA sequencing reveals the expansion of cytotoxic CD4+ T lymphocytes and a landscape of immune cells in primary Sjögren’s syndrome. Front. Immunol. 11, 594658 (2020).
Xanthou, G. et al. CD4 cytotoxic and dendritic cells in the immunopathologic lesion of Sjögren’s syndrome. Clin. Exp. Immunol. 118, 154–163 (1999).
Tasaki, S. et al. Multiomic disease signatures converge to cytotoxic CD8 T cells in primary Sjögren’s syndrome. Ann. Rheum. Dis. 76, 1458–1466 (2017).
McHeyzer-Williams, M. G., Altman, J. D. & Davis, M. M. Enumeration and characterization of memory cells in the TH compartment. Immunol. Rev. 150, 5–21 (1996).
Altman, J. D. et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 274, 94–96 (1996).
Ornatsky, O., Baranov, V. I., Bandura, D. R., Tanner, S. D. & Dick, J. Multiple cellular antigen detection by ICP-MS. J. Immunol. Methods 308, 68–76 (2006).
Bentzen, A. K. et al. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat. Biotechnol. 34, 1037–1045 (2016).
Sumida, T., Namekawa, T., Maeda, T. & Nishioka, K. New T-cell epitope of Ro/SS-A 52 kDa protein in labial salivary glands from patients with Sjögren’s syndrome. Lancet 348, 1667 (1996).
Namekawa, T. et al. Identification of Ro(SSA) 52 kDa reactive T cells in labial salivary glands from patients with Sjögren’s syndrome. J. Rheumatol. 22, 2092–2099 (1995). This initial study identifies tissue-infiltrating T cells reactive against the Ro(SSA) protein in Sjögren syndrome with conserved CDR3 sequences.
Abe, S. et al. M3 muscarinic acetylcholine receptor-reactive Th17 cells in primary Sjögren’s syndrome. JCI Insight 5, e135982 (2020). This recent study used ELIspot technology to detect M3R-reactive TH17 cell in pSS, highlighting this as a potential autoimmune target.
Okubo, M. et al. Detection and epitope analysis of autoantigen-reactive T cells to the U1-small nuclear ribonucleoprotein A protein in autoimmune disease patients. J. Immunol. 151, 1108–1115 (1993).
Holyst, M. M., Hill, D. L., Hoch, S. O. & Hoffman, R. W. Analysis of human T cell and B cell responses against U small nuclear ribonucleoprotein 70-kd, B, and D polypeptides among patients with systemic lupus erythematosus and mixed connective tissue disease. Arthritis Rheum. 40, 1493–1503 (1997).
Song, J. et al. Shared recognition of citrullinated tenascin-C peptides by T and B cells in rheumatoid arthritis. JCI Insight 6, e145217 (2021). This study uses a combination antigen-binding or reactivity techniques for detecting citrullinated tenascin-C-specific CD4 T cells in patients with RA, reporting a TH2/TH17 phenotype.
Sharma, R. K. et al. Biased TCR gene usage in citrullinated tenascin C specific T-cells in rheumatoid arthritis. Sci. Rep. 11, 24512 (2021).
de Jong, H. et al. Cartilage proteoglycan aggrecan epitopes induce proinflammatory autoreactive T-cell responses in rheumatoid arthritis and osteoarthritis. Ann. Rheum. Dis. 69, 255–262 (2010). A large cohort of patients with RA or osteoarthritis, or heathy controls, were evaluated to validate the reactivity against cartilage proteoglycan aggrecan epitopes as an autoimmune trigger of joint inflammation, finding cross-reactivity with a bacterial protein.
Chemin, K. et al. A novel HLA-DRB1*10:01-restricted T cell epitope from citrullinated type II collagen relevant to rheumatoid arthritis. Arthritis Rheumatol. 68, 1124–1135 (2016).
Raychaudhuri, S. et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat. Genet. 44, 291–296 (2012).
Scally, S. W. et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. J. Exp. Med. 210, 2569–2582 (2013).
Loh, T. J. et al. The molecular basis underlying T cell specificity towards citrullinated epitopes presented by HLA-DR4. Nat. Commun. 15, 6201 (2024). This study uses tetramer peptide–HLA technology to investigate TCR reactivity against citrullinated peptides presented by the HLA-DR4, explaining the preferential usage of TRAV-26-1 gene segment recombination.
Scally, S. W. et al. Molecular basis for increased susceptibility of Indigenous North Americans to seropositive rheumatoid arthritis. Ann. Rheum. Dis. 76, 1915–1923 (2017).
Parkes, M., Cortes, A., van Heel, D. A. & Brown, M. A. Genetic insights into common pathways and complex relationships among immune-mediated diseases. Nat. Rev. Genet. 14, 661–673 (2013).
Gutierrez-Arcelus, M., Rich, S. S. & Raychaudhuri, S. Autoimmune diseases – connecting risk alleles with molecular traits of the immune system. Nat. Rev. Genet. 17, 160–174 (2016).
Ellinghaus, D. et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat. Genet. 48, 510–518 (2016).
Nagafuchi, Y. et al. Control of naive and effector CD4 T cell receptor repertoires by rheumatoid-arthritis-risk HLA alleles. J. Autoimmun. 133, 102907 (2022).
Rubtsova, K., Marrack, P. & Rubtsov, A. V. Sexual dimorphism in autoimmunity. J. Clin. Invest. 125, 2187–2193 (2015).
Ter Horst, R. et al. Host and environmental factors influencing individual human cytokine responses. Cell 167, 1111–1124.e3 (2016).
Hughes, T. et al. Analysis of autosomal genes reveals gene–sex interactions and higher total genetic risk in men with systemic lupus erythematosus. Ann. Rheum. Dis. 71, 694–699 (2012).
Schneider-Hohendorf, T. et al. Sex bias in MHC I-associated shaping of the adaptive immune system. Proc. Natl Acad. Sci. USA 115, 2168–2173 (2018).
Rossetti, M. et al. TCR repertoire sequencing identifies synovial Treg cell clonotypes in the bloodstream during active inflammation in human arthritis. Ann. Rheum. Dis. 76, 435–441 (2017).
Petrelli, A. et al. PD-1+CD8+ T cells are clonally expanding effectors in human chronic inflammation. J. Clin. Invest. 128, 4669–4681 (2018).
Henderson, L. A. et al. Next-generation sequencing reveals restriction and clonotypic expansion of Treg cells in juvenile idiopathic arthritis. Arthritis Rheumatol. 68, 1758–1768 (2016).
Fischer, D. C., Opalka, B., Hoffmann, A., Mayr, W. & Haubeck, H. D. Limited heterogeneity of rearranged T cell receptor Vα and Vβ transcripts in synovial fluid T cells in early stages of rheumatoid arthritis. Arthritis Rheum. 39, 454–462 (1996).
Elewaut, D., De Keyser, F., Van den Bosch, F., Verbruggen, G. & Veys, E. M. Broadening of the T cell receptor spectrum among rheumatoid arthritis synovial cell-lines in relation to disease duration. Clin. Exp. Rheumatol. 18, 201–207 (2000).
Goronzy, J. J. et al. Dominant clonotypes in the repertoire of peripheral CD4+ T cells in rheumatoid arthritis. J. Clin. Invest. 94, 2068–2076 (1994).
Ercolini, A. M. & Miller, S. D. The role of infections in autoimmune disease. Clin. Exp. Immunol. 155, 1–15 (2009).
Yin, J. et al. Shotgun metagenomics reveals an enrichment of potentially cross-reactive bacterial epitopes in ankylosing spondylitis patients, as well as the effects of TNFi therapy upon microbiome composition. Ann. Rheum. Dis. 79, 132–140 (2020).
Scher, J. U., Littman, D. R. & Abramson, S. B. Microbiome in inflammatory arthritis and human rheumatic diseases. Arthritis Rheumatol. 68, 35–45 (2016).
Prasad, S., Starck, S. R. & Shastri, N. Presentation of cryptic peptides by MHC class I is enhanced by inflammatory stimuli. J. Immunol. 197, 2981–2991 (2016).
Doyle, H. A. & Mamula, M. J. Autoantigenesis: the evolution of protein modifications in autoimmune disease. Curr. Opin. Immunol. 24, 112–118 (2012).
Pruijn, G. J., Wiik, A. & van Venrooij, W. J. The use of citrullinated peptides and proteins for the diagnosis of rheumatoid arthritis. Arthritis Res. Ther. 12, 203 (2010).
Lim, J. J. et al. The shared susceptibility epitope of HLA-DR4 binds citrullinated self-antigens and the TCR. Sci. Immunol. 6, eabe0896 (2021).
Wegner, N. et al. Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and α-enolase: implications for autoimmunity in rheumatoid arthritis. Arthritis Rheum. 62, 2662–2672 (2010).
Konig, M. F. et al. Aggregatibacter actinomycetemcomitans-induced hypercitrullination links periodontal infection to autoimmunity in rheumatoid arthritis. Sci. Transl. Med. 8, 369ra176 (2016).
Maggi, J. et al. Isolation of HLA-DR-naturally presented peptides identifies T-cell epitopes for rheumatoid arthritis. Ann. Rheum. Dis. 81, 1096–1105 (2022).
Wang, R. et al. Clonally expanded CD38hi cytotoxic CD8 T cells define the T cell infiltrate in checkpoint inhibitor-associated arthritis. Sci. Immunol. 8, eadd1591 (2023).
Kim, S. T. et al. Distinct molecular and immune hallmarks of inflammatory arthritis induced by immune checkpoint inhibitors for cancer therapy. Nat. Commun. 13, 1970 (2022).
Schmitt, H. et al. Siglec-H protects from virus-triggered severe systemic autoimmunity. J. Exp. Med. 213, 1627–1644 (2016).
Kono, D. H. et al. Endosomal TLR signaling is required for anti-nucleic acid and rheumatoid factor autoantibodies in lupus. Proc. Natl Acad. Sci. USA 106, 12061–12066 (2009).
Britanova, O. V. et al. Targeted depletion of TRBV9+ T cells as immunotherapy in a patient with ankylosing spondylitis. Nat. Med. 29, 2731–2736 (2023).
Yang, P. et al. Application of T-cell receptor repertoire as a novel monitor in dynamic tracking and assessment: a cohort-study based on RA patients. J. Cell Mol. Med. 26, 6042–6055 (2022).
Bell, G. M. et al. Autologous tolerogenic dendritic cells for rheumatoid and inflammatory arthritis. Ann. Rheum. Dis. 76, 227–234 (2017).
Sonigra, A. et al. Randomized phase I trial of antigen-specific tolerizing immunotherapy with peptide/calcitriol liposomes in ACPA+ rheumatoid arthritis. JCI Insight 7, e160964 (2022).
Kenna, T. J., Thomas, R. & Steptoe, R. J. Steady-state dendritic cells expressing cognate antigen terminate memory CD8+ T-cell responses. Blood 111, 2091–2100 (2008).
Kenna, T. J. et al. Targeting antigen to diverse APCs inactivates memory CD8+ T cells without eliciting tissue-destructive effector function. J. Immunol. 184, 598–606 (2010).
Liu, J., Zhang, X. & Cao, X. Dendritic cells in systemic lupus erythematosus: from pathogenesis to therapeutic applications. J. Autoimmunity 132, 102856 (2022).
Horwitz, D. A., Bickerton, S. & La Cava, A. Strategies to use nanoparticles to generate CD4 and CD8 regulatory T cells for the treatment of SLE and other autoimmune diseases. Front. Immunol. 12, 681062 (2021).
Wright, G. P. et al. Adoptive therapy with redirected primary regulatory T cells results in antigen-specific suppression of arthritis. Proc. Natl Acad. Sci. USA 106, 19078–19083 (2009).
Delves, P. J., Martin, S. J., Burton, D. R. & Roitt, I. M. Roitt’s Essential Immunology 13th edn (Wiley, 2017).
Bank, I. The role of gamma delta T cells in autoimmune rheumatic diseases. Cells 9, 462 (2020).
Eckle, S. B. et al. Recognition of vitamin B precursors and byproducts by mucosal associated invariant T cells. J. Biol. Chem. 290, 30204–30211 (2015).
Mortier, C., Govindarajan, S., Venken, K. & Elewaut, D. It takes “guts” to cause joint inflammation: role of innate-like T cells. Front. Immunol. 9, 1489 (2018).
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Rheumatology thanks George Tsokos, Eric Gracey, Joerg Ermann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Garrido-Mesa, J., Brown, M.A. Antigen-driven T cell responses in rheumatic diseases: insights from T cell receptor repertoire studies. Nat Rev Rheumatol 21, 157–173 (2025). https://doi.org/10.1038/s41584-025-01218-9
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41584-025-01218-9