Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides

Abstract

Human leucocyte antigen B*27 (HLA-B*27) is strongly associated with inflammatory diseases of the spine and pelvis (for example, ankylosing spondylitis (AS)) and the eye (that is, acute anterior uveitis (AAU))1. How HLA-B*27 facilitates disease remains unknown, but one possible mechanism could involve presentation of pathogenic peptides to CD8+ T cells. Here we isolated orphan T cell receptors (TCRs) expressing a disease-associated public β-chain variable region–complementary-determining region 3β (BV9–CDR3β) motif2,3,4 from blood and synovial fluid T cells from individuals with AS and from the eye in individuals with AAU. These TCRs showed consistent α-chain variable region (AV21) chain pairing and were clonally expanded in the joint and eye. We used HLA-B*27:05 yeast display peptide libraries to identify shared self-peptides and microbial peptides that activated the AS- and AAU-derived TCRs. Structural analysis revealed that TCR cross-reactivity for peptide–MHC was rooted in a shared binding motif present in both self-antigens and microbial antigens that engages the BV9–CDR3β TCRs. These findings support the hypothesis that microbial antigens and self-antigens could play a pathogenic role in HLA-B*27-associated disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of AS- and AAU-associated TRBV9–TRBJ2.3 TCRs.
Fig. 2: Screening TRBV9-TRBJ2.3 TCRs on HLA-B*27:05 yeast display libraries.
Fig. 3: Validation of TCR binding and activation by predicted human and microbial antigens.
Fig. 4: Biophysical and structural basis of AS TCR–peptide–HLA-B*27:05 complexes.

Similar content being viewed by others

Data availability

The protein sequences used for target prediction were obtained from Uniprot (http://www.uniprot.org/). The human proteome ID is UP000005640. The Chlamydia proteome ID is UP000049987. The Salmonella proteome ID is UP000000541. The Shigella proteome ID is UP000001006. The Klebsiella proteome ID is UP000019183. The Yersinia proteome ID is UP000000815. The processed and raw deep-sequencing data have been deposited in the GEO database under the accession code GSE215018. The structures have been deposited in the RCSB protein data bank with the accession codes 7N2N, 7N2O, 7N2P, 7N2Q, 7N2R, 7N2S and 8CX4. There is no restriction on data availability. Source data are provided with this paper.

Code availability

Custom Perl scripts for the deep-sequencing data processing are available from https://github.com/jlmendozabio/NGSpeptideprepandpred.

References

  1. Bowness, P. HLA-B27. Annu. Rev. Immunol. 33, 29–48 (2015).

    Article  CAS  Google Scholar 

  2. Faham, M. et al. Discovery of T cell receptor beta motifs specific to HLA-B27-positive ankylosing spondylitis by deep repertoire sequence analysis. Arthritis Rheumatol. 69, 774–784 (2017).

    Article  CAS  Google Scholar 

  3. Hanson, A. L. et al. Altered repertoire diversity and disease-associated clonal expansions revealed by T cell receptor immunosequencing in ankylosing spondylitis patients. Arthritis Rheumatol. 72, 1289–1302 (2020).

    Article  CAS  Google Scholar 

  4. 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).

    Article  CAS  Google Scholar 

  5. Schlosstein, L. et al. High association of an HL-A antigen, W27, with ankylosing spondylitis. N. Engl. J. Med. 288, 704–706 (1973).

    Article  CAS  Google Scholar 

  6. Brewerton, D. A. et al. Acute anterior uveitis and HL-A 27. Lancet 302, 994–996 (1973).

    Article  CAS  Google Scholar 

  7. Brewerton, D. A. et al. Reiter’s disease and HL-A 27. Lancet 302, 996–998 (1973).

    Article  CAS  Google Scholar 

  8. Rosenbaum, J. T. & Asquith, M. The microbiome and HLA-B27-associated acute anterior uveitis. Nat. Rev. Rheumatol. 14, 704–713 (2018).

    Article  Google Scholar 

  9. Robinson, P. C. et al. Genetic dissection of acute anterior uveitis reveals similarities and differences in associations observed with ankylosing spondylitis. Arthritis Rheumatol. 67, 149–151 (2015).

    Article  Google Scholar 

  10. Finch, M. et al. Epidemic Reiter’s syndrome following an outbreak of shigellosis. Eur. J. Epidemiol. 2, 26–30 (1986).

    Article  CAS  Google Scholar 

  11. Aho, K. et al. HL-A 27 in reactive arthritis. A study of yersinia arthritis and Reiter’s disease. Arthritis Rheumatol. 17, 521–526 (1974).

    Article  CAS  Google Scholar 

  12. Benjamin, R. & Parham, P. Guilt by association: HLA-B27 and ankylosing spondylitis. Immunol. Today 11, 137–142 (1991).

    Article  Google Scholar 

  13. Hammer, R. E. et al. Spontaneous inflammatory disease in transgenic rats expressing HLA-B27 and human beta 2m: an animal model of HLA-B27-associated human disorders. Cell 63, 1099–1112 (1990).

    Article  CAS  Google Scholar 

  14. Tran, T. M. et al. Additional human beta2-microglobulin curbs HLA-B27 misfolding and promotes arthritis and spondylitis without colitis in male HLA-B27-transgenic rats. Arthritis Rheumatol. 54, 1317–1327 (2006).

    Article  CAS  Google Scholar 

  15. Baggia, S. et al. A novel model of bacterially-induced acute anterior uveitis in rats and the lack of effect from HLA-B27 expression. J. Investig. Med. 45, 295–301 (1997).

    CAS  Google Scholar 

  16. Allen, R. L. et al. Cutting edge: HLA-B27 can form a novel beta 2-microglobulin-free heavy chain homodimer structure. J. Immunol. 1, 5045–5048 (1999).

    Google Scholar 

  17. DeLay, M. L. et al. HLA-B27 misfolding and the unfolded protein response augment interleukin-23 production and are associated with Th17 activation in transgenic rats. Arthritis Rheumatol. 60, 2633–2643 (2009).

    Article  CAS  Google Scholar 

  18. Kollnberger, S. et al. Cell-surface expression and immune receptor recognition of HLA-B27 homodimers. Arthritis Rheumatol. 46, 2972–2982 (2002).

    Article  CAS  Google Scholar 

  19. Bowness, P. et al. Th17 cells expressing KIR3DL2+ and responsive to HLA-B27 homodimers are increased in ankylosing spondylitis. J. Immunol. 186, 2672–2680 (2011).

    Article  CAS  Google Scholar 

  20. 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).

    Article  Google Scholar 

  21. 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).

    Article  CAS  Google Scholar 

  22. 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).

    CAS  Google Scholar 

  23. May, E. et al. TCRB junctional regions from HLA-B27-restricted T cells and HLA-B27 binding peptides display conserved hydropathy profiles in the absence of primary sequence homology. Int. Immunol. 8, 1815–1823 (1996).

    Article  Google Scholar 

  24. Evans, D. M. et al. Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci. Nat. Genet. 42, 123–127 (2010).

    Article  Google Scholar 

  25. York, I. A. et al. Endoplasmic reticulum aminopeptidase 1 (ERAP1) trims MHC class I-presented peptides in vivo and plays an important role in immunodominance. Proc. Natl Acad. Sci. USA 103, 9202–9207 (2006).

    Article  ADS  CAS  Google Scholar 

  26. York, I. A. et al. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by trimming epitopes to 8–9 residues. Nat. Immunol. 3, 1177–1184 (2002).

    Article  CAS  Google Scholar 

  27. Martin-Esteban, A., Gomez-Molina, P., Sanz-Bravo, A. & Lopez de Castro, J. A. Combined effects of ankylosing spondylitis-associated ERAP1 polymorphisms outside the catalytic and peptide-binding sites on the processing of natural HLA-B27 ligands. J. Biol. Chem. 289, 3978–3990 (2014).

    Article  CAS  Google Scholar 

  28. Diaz-Pena, R., Lopez-Vazquez, A. & Larrea, L. Old and new HLA associations with ankylosing spondylitis. Tissue Antigens 80, 205–213 (2012).

    Article  CAS  Google Scholar 

  29. Colmegna, I., Cuchacovich, R. & Espinoza, L. R. HLA-B27-associated reactive arthritis: pathogenetic and clinical considerations. Clin. Microbiol. Rev. 17, 348–369 (2004).

    Article  CAS  Google Scholar 

  30. Ebringer, A., Rashid, T., Tiwana, H. & Wilson, C. A possible link between Crohn’s disease and ankyloisng spondylitis via Klebsiella infections. Clin. Rheumatol. 26, 289–297 (2007).

    Article  Google Scholar 

  31. Rashid, T., Wilson, C. & Ebringer, A. The link between ankylosing spondylitis, Crohn’s disease, Klebsiella, and starch consumption. Clin. Dev. Immunol. 2013, 872632 (2013).

  32. Stewart-Jones, G. B. E. et al. Crystal structures and KIR3DL1 recognition of three immunodominant viral peptides complexed to HLA-B*2705. Eur. J. Immunol. 35, 341–351 (2005).

    Article  CAS  Google Scholar 

  33. Hulsmeyer, M. et al. HLA-B27 subtypes differentially associated with disease exhibited subtle structural alterations. J. Biol. Chem. 277, 47844–47853 (2002).

    Article  CAS  Google Scholar 

  34. Lim Kam Sian, T. C. C. et al. Allelic association with ankylosing spondylitis fails to correlate with human leukocyte antigen B27 homodimer formation. J. Biol. Chem. 294, 20185–20195 (2019).

    Article  Google Scholar 

  35. Cole, D. K. et al. Human TCR-binding affinity is governed by MHC class restriction. J. Immunol. 178, 5727–5734 (2007).

    Article  CAS  Google Scholar 

  36. Petersen, J. et al. Diverse T cell receptor gene usage in HLA-DQ8-associated celiac disease converges into a consensus binding solution. Structure 24, 1643–1657 (2016).

    Article  CAS  Google Scholar 

  37. Yang, X. et al. Structural basis for clonal diversity of the human T-cell response to a dominant influenza virus epitope. J. Biol. Chem. 292, 18618–18627 (2017).

    Article  CAS  Google Scholar 

  38. May, E. et al. Conserved TCR beta chain usage in reactive arthrits; evidence for selection by a putative HLA-B27-associated autoantigen. Tissue Antigens 60, 299–308 (2002).

    Article  CAS  Google Scholar 

  39. Broughton, S. E. et al. Biased T cell receptor usage directed against human leukocyte antigen DQ8-restricted gliadin peptides is associated with celiac disease. Immunity 37, 611–621 (2012).

    Article  CAS  Google Scholar 

  40. Venturi, V. et al. Sharing of T cell receptors in antigen-specific responses is driven by convergent recombination. Proc. Natl Acad. Sci. USA 103, 18691–18696 (2006).

    Article  ADS  CAS  Google Scholar 

  41. Roldan, E. Q. et al. Different TCRBV genes generate biased pattern of V-D-J diversity in human T cells. Immunogenetics 41, 91–100 (1995).

    Article  CAS  Google Scholar 

  42. Schittenhelm, R. B. et al. Revisiting the arthritogenic peptide theory: quantitative not qualitative changes in the peptide repertoire of HLA-B27 allotypes. Arthritis Rheumatol. 67, 702–713 (2015).

    Article  CAS  Google Scholar 

  43. Barnea, E. et al. The human leukocyte antigen (HLA)-B27 peptidome in vivo, in spondyloarthritis-susceptible HLA-B27 transgenic rats and the effect of Erap1 deletion. Mol. Cell Proteomics 16, 642–662 (2017).

    Article  CAS  Google Scholar 

  44. Garcia-Medal, N. et al. Peptide handling by HLA-B27 subtypes influences their biological behavior, association with ankylosing spondylitis and susceptibility to endoplasmic reticulum aminopeptidase 1 (ERAP1). Mol. Cell. Proteomics 13, 3367–3380 (2014).

    Article  Google Scholar 

  45. Schittenhelm, R. B. et al. Human leukocyte antigen (HLA)-B27 allotype-specific binding and candidate arthritogenic peptides revealed through heuristic clustering of data-independent acquisition mass spectrometry (DIA-MS) data. Mol. Cell. Proteomics 15, 1867–1876 (2016).

    Article  CAS  Google Scholar 

  46. Chagin, A. S. & Sävendahl, L. GPR30 estrogen receptor expression in the growth plate declines as puberty progresses. J. Clin. Endocrinol. Metab. 92, 4873–4877 (2007).

    Article  CAS  Google Scholar 

  47. Heino, T. J., Chagin, A. S. & Sävendahl, L. The novel estrogen receptor G-protein-coupled receptor 30 is expressed in human bone. J. Endocrinol. 197, R1–R6 (2008).

    Article  CAS  Google Scholar 

  48. Gautam, P. et al. Multi-species single-cell transcriptomic analysis of ocular compartment regulons. Nat. Commun. 28, 5675 (2021).

    Article  ADS  Google Scholar 

  49. Cole, D. K. et al. Hotspot autoimmune T cell receptor binding underlies pathogen and insulin peptide cross-reactivity. J. Clin. Invest. 26, 2191–2204 (2016).

    Article  Google Scholar 

  50. Petersen, J. et al. T cell receptor cross-reactivity between gliadin and bacterial peptides in celiac disease. Nat. Struct. Mol. Biol. 27, 49–61 (2020).

    Article  CAS  Google Scholar 

  51. Standardization of Uveitis Nomenclature (SUN) Working Group. Classification criteria for spondyloarthritis/HLA-B27-associated anterior uvetis. Am. J. Ophthalmol. 228, 117–125 (2021).

    Article  Google Scholar 

  52. Rudwaleit, M. et al. The development of Assessment of SpondyloArthritis International Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann. Rheum. Dis. 68, 777–783 (2009).

    Article  CAS  Google Scholar 

  53. Jabs, D. A. et al. Standardization of uveitis nonmenclature for reporting clinical data. Results of the first international workshop. Am. J. Ophthalmol. 140, 509–516 (2005).

    Article  Google Scholar 

  54. Hassman, L. M. et al. Clinicomolecular identification of conserved and individualized features of granulomatous uveitis. Ophthalmol. Sci. https://doi.org/10.1016/j.xops.2021.100010 (2021).

    Article  Google Scholar 

  55. Pogorelyy, M. V. et al. Persisting fetal clonotypes influence the structure and overlap of adult human T cell receptor repertoires. PLoS Comput. Biol. 13, e1005572 (2017).

    Article  Google Scholar 

  56. Shugay, M. et al. Towards error-free profiling of immune repertoires. Nat. Methods 11, 653–655 (2014).

    Article  CAS  Google Scholar 

  57. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  CAS  Google Scholar 

  58. Bowness, P., Allen, R. L. & McMichael, A. J. Identification of T cell receptor recognition residues for a viral peptide presented by HLA B27. Eur. J. Immunol. 24, 2357–2363 (1994).

    Article  CAS  Google Scholar 

  59. Lybarger, L. et al. Enhanced immune presentation of a single-chain major histocompatibility complex class I molecule engineered to optimize linkage of a C-terminally extended peptide. J. Biol. Chem. 278, 27105–27111 (2003).

    Article  CAS  Google Scholar 

  60. Birnbaum, M. E. et al. Deconstructing the peptide-MHC specificity of T cell recognition. Cell 157, 1073–1087 (2014).

    Article  CAS  Google Scholar 

  61. Garboczi, D. N. et al. Structure of the complex between human T-cell receptor, viral peptide and HLA-A2. Nature 384, 134–141 (1996).

    Article  ADS  CAS  Google Scholar 

  62. Aricescu, A. R., Lu, W. & Jones, E. Y. A time- and cost-efficient system for high-level protein production in mammalian cells. Acta Crystallogr. D 62, 1243–1250 (2006).

    Article  Google Scholar 

  63. Ellis, S. A., Taylor, C. & McMichael, A. Recognition of HLA-B27 and related antigen by a monoclonal antibody. Hum. Immunol. 5, 49–59 (1982).

  64. Garboczi, D. N., Hung, D. T. & Wiley, D. C. HLA-A2-peptide complexes: refolding and crystallization of molecules expressed in Escherichia coli and complexed with single antigenic peptides. Proc. Natl Acad. Sci. USA 89, 3429–3433 (1992).

    Article  ADS  CAS  Google Scholar 

  65. Emsley, P. et al. Features and development of COOT. Acta Crystallogr. D 66, 486–501 (2010).

    Article  CAS  Google Scholar 

  66. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr. D 75, 861–877 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The study received support from the National Institutes of Health (NIH; 5R01AI103867 (K.C.G.) and U19AI057229 (K.C.G.)), the Howard Hughes Medical Institute (K.C.G.), the UK Medical Research Council (MR/M019837/1; G.M.G., A.J.M. and P.B.), Oxford University McMichael Trust Fund (G.M.G and L.I.G), the Rosetrees Trust (M455; G.M.G.), Oxford University John Fell Fund and Medical Sciences Division Internal Funds (G.M.G. and L.I.G.), the UK National Institute for Health Research Oxford Biomedical Research Centre (BRC) (P.B.), the Ministry of Science and Higher Education of the Russian Federation (075-15-2019-1789; E.A.K., I.V.Z. and D.M.C.), the Rheumatology Research Foundation (M.A.P.), the Arthritis National Research Foundation (M.A.P.), the Rheumatic Diseases Research Resource-based Center at Washington University (NIH P30-AR073752; M.A.P. and W.M.Y.), the Bursky Center for Human Immunology and Immunotherapy Programs (W.M.Y.) and the Barnes-Jewish Hospital Foundation (W.M.Y.). The views expressed are those of the author(s) and not necessarily those of the NHS, the National Institute for Health Research or the Department of Health. We thank the Oxford Genomics Centre at the Wellcome Centre for Human Genetics (Wellcome Trust grant reference 203141/Z/16/Z) for the generation and initial processing of sequencing data and J. Webber (Cell Sorting Facility, Kennedy Institute of Rheumatology, University of Oxford) for conducting cell sorting. We also thank N. Ternette, W. Paes and R. Inman for data analysis advice and scientific discussions. The Berkeley Center for Structural Biology is supported in part by the Howard Hughes Medical institute. The Advanced Light Source is a Department of Energy Office of Science User Facility under Contract No. DE-AC02-05CH11231. The ALS-ENABLE beamlines are supported in part by the NIH, National institute of General Medical Sciences (grant P30 GM124169). The schematic cartoon figures in Fig. 1 and Extended Data Fig. 1 were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

X.Y., L.I.G., G.M.G., A.J.M. and K.C.G. conceived the project and wrote the manuscript. X.Y. conducted yeast display experiments, deep-sequencing data analysis, target prediction, target validation, TCR–pMHC complex purification, crystallization and structural studies. L.I.G. conducted processing of samples from patients with AS, single-cell T cell sequencing analysis and TCR tetramer staining of SCTs. M.A.P. and W.M.Y. conceived and analysed single-cell TCR sequencing from HLA-B*27+ AAU and contributed clinical perspectives and manuscript revisions. M.A.P., G.L.P. and L.M.H. identified HLA-B*27+ participants with AAU and collected aqueous and blood samples. S.B. designed the SCT staining constructs. M.N.Q. carried out thermal melt analysis. X.Z. carried out independent target validation. R.A.F., X.Y. and C.S.S. developed the algorithm for peptide prediction. K.M.J. conducted structural studies. X.Y. and L.I.G. conducted SPR experiments. E.A.K., I.V.Z. and D.M.C. identified the AS3.1 TCR αβ pair, conducted independent target validation for AS3.1 TCR and provided clinical samples. P.B. provided clinical samples and TCR analysis. W.M.Y., A.J.M., G.M.G. and K.C.G. supervised the project.

Corresponding authors

Correspondence to Wayne M. Yokoyama, Andrew J. McMichael, Geraldine M. Gillespie or K. Christopher Garcia.

Ethics declarations

Competing interests

M.A.P. has received research support from Eli Lilly and Company paid to the university and served as a consultant for AbbVie, Priovant Therapeutics and JK MarketResearch. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature thanks Robert Colbert and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Isolation of TRBV9+ TCRs from PBMC.

a. Schematic view of TRBV9+ TCRs isolation strategy from patients’ PBMC. Created with BioRender.com. b. Weblogo of CDR3β variants from HLA-B*27:05+ AS patient PBMC samples. c. Sorting strategy for scRNAseq library generation for Round 1 samples (HLA-B*27:05+ AS patient PBMC with overnight rest, post thaw). Sample AS1541 is shown. Final panel shows BV9-PerCP-Cy5.5 isotype control staining against CD8-PE. d. Representative sorting data for scRNAseq library generation. Sample AS1541 from Round 1 (HLA-B*27:05+ AS patient PBMC with overnight rest, post thaw) and sample AS1455B2 from Round 2 (HLA-B*27:05+ AS patient PBMC, 28 days expansion with αCD3 and IL-2 followed by magnetic depletion of CD4+, CD19+ and CD14+ cells) are shown. CD8+ T cell frequencies (as a % of total live cells) and BV9+ frequencies (as a % of CD8+ T cells) for each sample are as follows: Round 1 samples (CD8+ %/BV9+ %): AS1802 14.9/3.97; AS1311 16.4/10.7; AS1541 10.1/4.37. Round 2 samples: AS1455 82.9/5.60; AS1567 31.4/1.16; AS1661 66.7/7.21; AS1803 49.4/12.2. e. Enrichment of TRBV9 during first round of scRNAseq library generation by sorting. Pie charts show percentage of BV9+ cells in each sample pre- and post-mixing of sorted fractions. f. Partial amino acid sequence alignment of five AS patient PBMC-derived TCRs in this study, in RasMol colouring. The CDR1 and CDR2 sequences are shown for TRAV21 and TRBV9 in the top row. CDR3 amino acid sequences are shown in the bottom rows along with corresponding variable and joining gene usage. Structurally important bulky residues are marked with asterisks.

Extended Data Fig. 2 AS3.1 TCR alpha-beta pair identification.

a. Correlation of the top100 TCR α and β nucleotide sequence proportion in two independent samples of CD3+CD8+TRBV9+ synovial fluid T cells of patient P. Confirmation of the frequency-based paired TCR α and β with single-cell VDJ-sequencing of an independent sample of CD3+CD8+TRBV9+ synovial fluid T cells from the patient. b. Enrichment of the BV9–Y/FSTDTQ–BJ2-3 motif in the joint compared to the blood in AS. Bulk TCR sequencing was performed on CD8+ T cell cDNA from the blood and synovial fluid. The graph displays the proportion of TCR sequence reads containing the BV9–Y/FSTDTQ–BJ2-3 motif in paired blood and synovial fluid of three Ankylosing Spondylitis patients. Level of detection (LoD) of blood samples is indicated by a dashed line and is the median proportion of a single sequence read from all blood samples

Source data

Extended Data Fig. 3 Development of HLA-B*27:05 yeast library.

a. GRb TCR tetramer staining of wildtype SRY-β2M-HLA-B*27:05 single chain trimer expressed on yeast surface. Single chain trimer expression was monitored by anti-C-Myc-488. Streptavidin-647 (SA-647) was used to tetramerize and label the GRb TCR. b. Schematic of the SRY-β2m-HLA-B*27:05 single chain trimer on yeast surface. Error prone PCR introduced random mutations in SRY peptide, β2m and HLA-B*27:05 heavy chain. Sequencing of the enriched SRY-β2M-HLA-B*27:05 error prone library identified mutations located on HLA-B*27:05 heavy chain, shown as cartoon diagram and colored green. Bona fide mutations are shown as stick diagram and colored red. c. GRb TCR tetramer staining was rescued by M5L, H114Y and A153D mutation of the SRY-β2m-HLA-B*27:05 single chain trimer (HLA-B*27:053mut). Single chain trimer expression was monitored by anti-C-Myc-488. Streptavidin-647 (SA-647) was used to tetramerize and label the GRb TCR. d. Location of M5L, H114Y, and A153D mutations shown on the HLA-B*27 ribbon structure. e. Library design shows the anchor residue preference for the HLA-B*27:053mut library at the P2 and PΩ. For other positions, the NNK codon allowed all 20 amino acids and stop codons. Nucleotide abbreviations for codon usage are listed according to the IUPAC nucleotide code. Library diversities were estimated based on SDCAA plate colony numbers under serial dilution. f. GRb TCR tetramer staining of 4th round peptide-HLA-B*27:053mut yeast-display library, indicating successful enrichment. g. Deep sequencing counts on the most enriched peptides after 4 rounds of selection with the GRb TCR. h. WebLogo plots of the top 100 peptides after 4th round of selection with the GRb TCR, the two predominant clusters within the selection, and the sequence of the cognate peptide recognized by the GRb TCR in the abundance at the given position among the unique peptides.

Source data

Extended Data Fig. 4 Selection of AS TCRs on HLA-B*27:05 libraries.

a. HLA-B*27:053mut 10mer library enrichment per round of selection by the five AS TCRs as measured by flow cytometry. b. AS4.1, AS4.2, AS4.3 and AS4.4 TCR tetramer staining on the 4th round HLA-B*27:05 9-AA libraries. HLA-B*27:053mut expression was monitored by anti-C-Myc-488. Streptavidin-647 (SA-647) was used to tetramerize and fluorescently label the AS TCRs. c. AS3.1, AS4.1, AS4.2, AS4.3 and AS4.4 TCR tetramer staining on the 4th round HLA-B*27:053mut 10-AA libraries. HLA-B*27:05 expression was monitored by anti-HA-488. Streptavidin-647 (SA-647) was used to tetramerize and fluorescently label the AS TCRs. d. WebLogos representing the unique 4th round selected peptides for each AS TCR based on deep sequencing reads. The size of each amino acid letter represents its abundance at the given position among the unique peptides. e. Heatmap plots showing the amino acid composition per position of the peptides enriched after the 4th round of selection. Darker color represents greater abundance of a given amino acid at specific position.

Source data

Extended Data Fig. 5 Synthetic peptide activation by AS TCR expressing T cell line.

(a-j) CD8+ SKW-3 cells were transduced with AS TCRs via lentivirus and sorted for stable TCR (IP26) and CD3 (UTCH1) co-expression. K562 cells were transduced with wild type HLA-B*27:05 via lentivirus and sorted for stable HLA molecule expression. The antigen-presenting cell line was pulsed for 2 h with 100 µM peptides and co-incubated with the T cell lines for 18 h then analyzed for CD69 expression by flow cytometry. (a-e) AS3.1, AS4.1, AS4.2, AS4.3 and AS4.4 TCR were tested for CD69 activation by yeast-selected mimotopes from 9-AA libraries. (f-j) AS3.1, AS4.1, AS4.2, AS4.3 and AS4.4 TCR were tested for CD69 activation by yeast-selected mimotopes from 10-AA libraries. Red dots show activation, as defined by CD69% at least 2-fold greater than negative controls (DMSO and null peptide, n = 1).

Source data

Extended Data Fig. 6 AS TCR tetramer staining on SCT transfectants.

a. Representative flow cytometry showing AS8.4 cells alone, AS8.4 T cells + K562 (HLA-B*27:05+), AS8.4 T cells + K562 (HLA-B*27:05+ GPER1+), AS8.4 T cells + K562 (HLA-B*27:05+ YEIH+) and AS8.4 T cells + K562 (HLA-B*27:05+ 1uM YEIH peptide). b. Plots of percentage of CD69% positive AS8.4 T cells alone, AS8.4 T cells + K562 (HLA-B*27:05+), AS8.4 T cells + K562 (HLA-B*27:05+ GPER1+), AS8.4 T cells + K562 (HLA-B*27:05+ YEIH+) and AS8.4 T cells + K562 (HLA-B*27:05+ 1uM YEIH peptide). Data are shown as mean±s.d., n = 3 biological replicates. NS, not significant, P = 0.5453; ****P<0.0001 (one-way ANOVA). c. Representative flow cytometry histograms showing AS TCR tetramer staining for SCT transfected b2m KO 293T cells. Red histograms: negative controls (irrelevant p-HLA-B*27:05 construct); Blue histograms: potential arthritogenic peptide identified in this study.

Source data

Extended Data Fig. 7 Differential activation of AS TCRs by potential arthritogenic peptides on HLA-B*27:05 and HLA-B*27:09 backgrounds.

a. AS8.4 T cells incubated with GPER1-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. b. AS8.2 T cells incubated with GPER1-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. c. Structure models showing how the GPER1 P9 Arg interacts with HLA-B*27:05 Asp at 116 and HLA-B*27:09 His at 116. The peptide Arg 9 is colored magenta, Asp 116 is colored green, and His 116 is colored cyan. d. AU2 T cells incubated with RNASEH2B-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. e. AS4.3 T cells incubated with RNASEH2B-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. f. Structure models showing how the RNASEH2B P9 Arg interacts with HLA-B*27:05 Asp at 116 and HLA-B*27:09 His at 116. The peptide Arg 9 is colored magenta, Asp 116 is colored green, and His 116 is colored cyan. g. AU2 T cells incubated with PRPF3-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. h. AS4.3 T cells incubated with PRPF3-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. i. Structure models showing how the PRPF3 P9 Lys interacts with HLA-B*27:05 Asp at 116 and HLA-B*27:09 His at 116. The peptide Lys 9 is colored magenta, Asp 116 is colored green, and His 116 is colored cyan. j. AS8.4 T cells incubated with YEIH-pulsed K562 cells, express HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. k. AS8.2 T cells incubate with YEIH-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. l. Structure models showing how the YEIH P9 Phe interacts with HLA-B*27:05 Asp at 116 and HLA-B*27:09 His at 116. The peptide Phe 9 is colored magenta, Asp 116 is colored green, and His 116 is colored cyan. m. AU2 T cells incubate with MPP4-pulsed K562 cells, expressing HLA-B*27:05 or HLA-B*27:09, respectively, were tested for CD69 up-regulation. Data are shown as mean±s.d., n = 3 biological replicates. n. Structure models showing how the MPP4 P9 His interacts with HLA-B*27:05 Asp at 116 and HLA-B*27:09 His at 116. o. Thermal stability of refolded peptide/HLA-B*27 complex as determined by differential scanning fluorimetry. The melting temperature, Tm, of each complex is plotted as a bar graph with mean±s.d. (n = 3 technical triplicates).

Source data

Extended Data Fig. 8 SPR sensorgram of AS TCRs and peptide-HLA-B*27:05.

(a-p) Binding analysis of AS TCRs AS3.1, AS4.1, AS4.2, AS4.3 and AS8.4 to GPER1/PRPF3/RNASEH2B/YEIH-HLA-B*27:05 molecules. Increasing concentrations of each TCR was injected over immobilized GPER1/PRPF3/RNASEH2B/YEIH-HLA-B*27:05 complexes. The response unit vs concentration plots were fitted to a steady state affinity model. The SPR measurements were performed once for each TCR-pMHC interaction.

Extended Data Fig. 9 Omit maps of AS TCR-peptide-HLA-B*27:05 complex contoured at 1 σ.

a. Simulated annealing composite omit map of the entire AS4.3 TCR-RNASEH2B-HLA-B*27:05 complex contoured at 1 σ. The TCRα chain is colored red; the TCRβ chain is colored yellow; the peptide is colored magenta; the β2M is colored cyan; the HLA-B*27:05 heavy chain is colored green. (b-h) Simulated annealing composite omit maps of the AS4.3 TCR-RNASEH2B-HLA-B*27:05, AS3.1 TCR-PRPF3-HLA-B*27:05, AS4.2 TCR-PRPF3-HLA-B*27:05, AS4.2 TCR-YEIH-HLA-B*27:05, AS4.3 TCR-PRPF3-HLA-B*27:05, AS4.3 TCR-YEIH-HLA-B*27:05 and AS8.4 TCR-YEIH-HLA-B*27:05 complex interfaces contoured at 1 σ. The TCR CDR1α is colored red; the TCR CDR3β is colored yellow; the peptide is colored magenta; the HLA-B*27:05 heavy chain is colored green.

Extended Data Fig. 10 Detailed interactions between AS TCR and peptide-HLA-B*27:05.

a. Side view of superimposed AS TCR-peptide-HLA-B*27:05 complexes. b. Top view of superimposed interface between AS TCR CDR loops and peptide-HLA-B*27:05. c. Superimposed complex structures reveal CDR3β interaction with peptides and HLA-B*27:05. d. Superimposed complex structures reveal CDR2β interaction with HLA-B*27:05. (e-k) Peptide recognition by CDR1α and CDR3β in AS TCR-peptide-HLA-B*27:05 complexes. The upper panels are ‘Sticks’ indicating TCR residues and peptide residues making contacts. CDR1α is colored red or orange and depicted in cartoon. CDR3β is colored yellow or wheat and depicted in cartoon. Peptide is colored magenta and HLA-B*27:05 is colored green and depicted in cartoon. The lower panels show CDR1α, CDR3β and peptide sequences and atomic interactions. The black lines indicate van der Waals contacts; red dashed lines indicate hydrogen bonds.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1–6.

Reporting Summary

Peer Review File

Source data

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, X., Garner, L.I., Zvyagin, I.V. et al. Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides. Nature 612, 771–777 (2022). https://doi.org/10.1038/s41586-022-05501-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05501-7

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing