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:

Genetic variation in MHC proteins is associated with T cell receptor expression biases

Abstract

In each individual, a highly diverse T cell receptor (TCR) repertoire interacts with peptides presented by major histocompatibility complex (MHC) molecules. Despite extensive research, it remains controversial whether germline-encoded TCR–MHC contacts promote TCR–MHC specificity and, if so, whether differences exist in TCR V gene compatibilities with different MHC alleles. We applied expression quantitative trait locus (eQTL) mapping to test for associations between genetic variation and TCR V gene usage in a large human cohort. We report strong trans associations between variation in the MHC locus and TCR V gene usage. Fine-mapping of the association signals identifies specific amino acids from MHC genes that bias V gene usage, many of which contact or are spatially proximal to the TCR or peptide in the TCR–peptide–MHC complex. Hence, these MHC variants, several of which are linked to autoimmune diseases, can directly affect TCR–MHC interaction. These results provide the first examples of trans-QTL effects mediated by protein–protein interactions and are consistent with intrinsic TCR–MHC specificity.

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

Figure 1: Illustration of our approach.
Figure 2: Expression of TCR Vα and Vβ genes is significantly associated with genetic variation in the MHC locus.
Figure 3: Expression of TCR Vα and Vβ genes is associated with amino acid variation in MHC proteins.
Figure 4: Bayesian inference of amino acid residues encoded in MHC genes that influence expression of TCR Vα genes.
Figure 5: MHC residues that are functionally important for TCR recognition are also associated with TCR expression.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Neefjes, J., Jongsma, M.L.M., Paul, P. & Bakke, O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat. Rev. Immunol. 11, 823–836 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. McDevitt, H.O. & Bodmer, W.F. HL-A, immune-response genes, and disease. Lancet 1, 1269–1275 (1974).

    Article  CAS  PubMed  Google Scholar 

  3. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Miyadera, H. & Tokunaga, K. Associations of human leukocyte antigens with autoimmune diseases: challenges in identifying the mechanism. J. Hum. Genet. 60, 697–702 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Vantourout, P. & Hayday, A. Six-of-the-best: unique contributions of γδ T cells to immunology. Nat. Rev. Immunol. 13, 88–100 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rossjohn, J. et al. T cell antigen receptor recognition of antigen-presenting molecules. Annu. Rev. Immunol. 33, 169–200 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Rudolph, M.G., Stanfield, R.L. & Wilson, I.A. How TCRs bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 24, 419–466 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. 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).

    Article  CAS  PubMed  Google Scholar 

  9. Housset, D. & Malissen, B. What do TCR–pMHC crystal structures teach us about MHC restriction and alloreactivity? Trends Immunol. 24, 429–437 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Garcia, K.C. et al. A closer look at TCR germline recognition. Immunity 36, 887–888 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Roudier, J. Association of MHC and rheumatoid arthritis. Association of RA with HLA-DR4: the role of repertoire selection. Arthritis Res. 2, 217–220 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Robins, H.S. et al. Overlap and effective size of the human CD8+ T cell receptor repertoire. Sci. Transl. Med. 2, 47ra64 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Zvyagin, I.V. et al. Distinctive properties of identical twins' TCR repertoires revealed by high-throughput sequencing. Proc. Natl. Acad. Sci. USA 111, 5980–5985 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gulwani-Akolkar, B. et al. Do HLA genes play a prominent role in determining T cell receptor Vα segment usage in humans? J. Immunol. 154, 3843–3851 (1995).

    CAS  PubMed  Google Scholar 

  16. Miles, J.J. et al. TCRα genes direct MHC restriction in the potent human T cell response to a class I–bound viral epitope. J. Immunol. 177, 6804–6814 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Garcia, K.C. Reconciling views on T cell receptor germline bias for MHC. Trends Immunol. 33, 429–436 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Garcia, K.C., Adams, J.J., Feng, D. & Ely, L.K. The molecular basis of TCR germline bias for MHC is surprisingly simple. Nat. Immunol. 10, 143–147 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Castro, C.D., Luoma, A.M. & Adams, E.J. Coevolution of T-cell receptors with MHC and non-MHC ligands. Immunol. Rev. 267, 30–55 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Marrack, P., Scott-Browne, J.P., Dai, S., Gapin, L. & Kappler, J.W. Evolutionarily conserved amino acids that control TCR–MHC interaction. Annu. Rev. Immunol. 26, 171–203 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Scott-Browne, J.P., White, J., Kappler, J.W., Gapin, L. & Marrack, P. Germline-encoded amino acids in the αβ T-cell receptor control thymic selection. Nature 458, 1043–1046 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Van Laethem, F. et al. Lck availability during thymic selection determines the recognition specificity of the T cell repertoire. Cell 154, 1326–1341 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Van Laethem, F. et al. Deletion of CD4 and CD8 coreceptors permits generation of αβ T cells that recognize antigens independently of the MHC. Immunity 27, 735–750 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Holland, S.J. et al. The T-cell receptor is not hardwired to engage MHC ligands. Proc. Natl. Acad. Sci. USA 109, E3111–E3118 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Burrows, S.R. et al. Hard wiring of T cell receptor specificity for the major histocompatibility complex is underpinned by TCR adaptability. Proc. Natl. Acad. Sci. USA 107, 10608–10613 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Beringer, D.X. et al. T cell receptor reversed polarity recognition of a self-antigen major histocompatibility complex. Nat. Immunol. 16, 1153–1161 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Parrish, H.L., Deshpande, N.R., Vasic, J. & Kuhns, M.S. Functional evidence for TCR-intrinsic specificity for MHCII. Proc. Natl. Acad. Sci. USA 113, 3000–3005 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rockman, M.V. & Kruglyak, L. Genetics of global gene expression. Nat. Rev. Genet. 7, 862–872 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Battle, A. et al. Characterizing the genetic basis of transcriptome diversity through RNA-sequencing of 922 individuals. Genome Res. 24, 14–24 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sottini, A., Imberti, L., Fiordalisi, G. & Primi, D. Use of variable human Vδ genes to create functional T cell receptor α chain transcripts. Eur. J. Immunol. 21, 2455–2459 (1991).

    Article  CAS  PubMed  Google Scholar 

  31. Jia, X. et al. Imputing amino acid polymorphisms in human leukocyte antigens. PLoS One 8, e64683 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sinclair, C., Bains, I., Yates, A.J. & Seddon, B. Asymmetric thymocyte death underlies the CD4:CD8 T-cell ratio in the adaptive immune system. Proc. Natl. Acad. Sci. USA 110, E2905–E2914 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yang, J., Lee, S.H., Goddard, M.E. & Visscher, P.M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kichaev, G. et al. Integrating functional data to prioritize causal variants in statistical fine-mapping studies. PLoS Genet. 10, e1004722 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Wallace, C. et al. Dissection of a complex disease susceptibility region using a Bayesian stochastic search approach to fine mapping. PLoS Genet. 11, e1005272 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Wellcome Trust Case Control Consortium. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat. Genet. 44, 1294–1301 (2012).

  37. Servin, B. & Stephens, M. Imputation-based analysis of association studies: candidate regions and quantitative traits. PLoS Genet. 3, e114 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hu, X. et al. Additive and interaction effects at three amino acid positions in HLA-DQ and HLA-DR molecules drive type 1 diabetes risk. Nat. Genet. 47, 898–905 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Patsopoulos, N.A. et al. Fine-mapping the genetic association of the major histocompatibility complex in multiple sclerosis: HLA and non-HLA effects. PLoS Genet. 9, e1003926 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Messaoudi, I., Guevara Patiño, J.A., Dyall, R., LeMaoult, J. & Nikolich-Zugich, J. Direct link between MHC polymorphism, T cell avidity, and diversity in immune defense. Science 298, 1797–1800 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Price, D.A. et al. Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection. J. Exp. Med. 206, 923–936 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Luz, J.G. et al. Structural comparison of allogeneic and syngeneic T cell receptor–peptide–major histocompatibility complex complexes: a buried alloreactive mutation subtly alters peptide presentation substantially increasing Vβ interactions. J. Exp. Med. 195, 1175–1186 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Murray, J.S. An old Twist in HLA-A: CDR3α hook up at an R65-joint. Front. Immunol. 6, 268 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Levin, A.M. et al. Exploiting a natural conformational switch to engineer an interleukin-2 'superkine'. Nature 484, 529–533 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jerne, N.K. The somatic generation of immune recognition. Eur. J. Immunol. 1, 1–9 (1971).

    Article  CAS  PubMed  Google Scholar 

  47. Dai, S. et al. Crossreactive T cells spotlight the germline rules for αβ T cell-receptor interactions with MHC molecules. Immunity 28, 324–334 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Adams, J.J. et al. Structural interplay between germline interactions and adaptive recognition determines the bandwidth of TCR–peptide–MHC cross-reactivity. Nat. Immunol. 17, 87–94 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Feng, D., Bond, C.J., Ely, L.K., Maynard, J. & Garcia, K.C. Structural evidence for a germline-encoded T cell receptor–major histocompatibility complex interaction 'codon'. Nat. Immunol. 8, 975–983 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Blevins, S.J. et al. How structural adaptability exists alongside HLA-A2 bias in the human αβ TCR repertoire. Proc. Natl. Acad. Sci. USA 113, E1276–E1285 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sim, B.C., Zerva, L., Greene, M.I. & Gascoigne, N.R. Control of MHC restriction by TCR Vα CDR1 and CDR2. Science 273, 963–966 (1996).

    Article  CAS  PubMed  Google Scholar 

  52. Ferreira, M.A.R. et al. Quantitative trait loci for CD4:CD8 lymphocyte ratio are associated with risk of type 1 diabetes and HIV-1 immune control. Am. J. Hum. Genet. 86, 88–92 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Klarenbeek, P.L. et al. Somatic variation of T-cell receptor genes strongly associate with HLA class restriction. PLoS One 10, e0140815 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Anders, S., Pyl, P.T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. de Bakker, P.I.W. et al. A high-resolution HLA and SNP haplotype map for disease association studies in the extended human MHC. Nat. Genet. 38, 1166–1172 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Delaneau, O. & Marchini, J. Integrating sequence and array data to create an improved 1000 Genomes Project haplotype reference panel. Nat. Commun. 5, 3934 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Howie, B.N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009).

    PubMed  PubMed Central  Google Scholar 

  60. 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  61. Visscher, P.M., Yang, J. & Goddard, M.E. A commentary on 'common SNPs explain a large proportion of the heritability for human height' by Yang et al. (2010). Twin Res. Hum. Genet. 13, 517–524 (2010).

    Article  PubMed  Google Scholar 

  62. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    Google Scholar 

  63. Mitchell, T.J. & Beauchamp, J.J. Bayesian variable selection in linear regression. J. Am. Stat. 83, 1023–1032 (1988).

    Article  Google Scholar 

  64. Ishwaran, H. & Rao, J.S. Spike and slab gene selection for multigroup microarray data. J. Am. Stat. 100, 764–780 (2005).

    Article  CAS  Google Scholar 

  65. Geman, S. & Geman, D. Stochastic relaxation, gibbs distributions, and the Bayesian restoration of images. IEEE Trans. Pattern Anal. Mach. Intell. 6, 721–741 (1984).

    Article  CAS  PubMed  Google Scholar 

  66. Lefranc, M.-P. et al. IMGT®, the international ImMunoGeneTics information system® 25 years on. Nucleic Acids Res. 43, D413–D422 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Berman, H.M. et al. The Protein Data Bank. Nucleic Acids Res. 28, 235–242 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. UniProt Consortium. UniProt: a hub for protein information. Nucleic Acids Res. 43, D204–D212 (2015).

Download references

Acknowledgements

We thank D. Golan, D. Knowles, A. Fu, M. Birnbaum, M. Gee, J. Mendoza, A. Bhaskar and T. Raj for helpful discussions and the anonymous referees for valuable comments. This work was supported by NIH grants HG0070736, 1R01GM097171-01A1, RO1AI03867 and U19AI057229, the Howard Hughes Medical Institute, the EMBO Long-Term Fellowship and a National Science Foundation Graduate Research Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

E.S., J.K.P. and K.C.G. conceived the project. E.S. performed genetic analyses with input from A.B., H.B.F. and J.K.P. E.S. and L.V.S. performed structural analyses with input from K.C.G. E.S., L.V.S., K.C.G. and J.K.P. wrote the manuscript. The work was supervised by K.C.G. and J.K.P. All authors reviewed, revised and provided feedback on the manuscript.

Corresponding authors

Correspondence to K Christopher Garcia or Jonathan K Pritchard.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–32 and Supplementary Table 1 (PDF 17162 kb)

Supplementary Table 2

TCR V genes expression. (XLSX 930 kb)

Supplementary Table 3

Ig V-genes expression. (XLSX 1070 kb)

Supplementary Table 4

TCR and Ig V genes association with short-range genetic variation and genetic variation in the MHC locus. (XLSX 88 kb)

Supplementary Table 5

Most significance association between expression of each TCR and Ig V gene and genotyped SNPs in the MHC locus. (XLSX 76 kb)

Supplementary Table 6

Nucleotide and amino acid variants in the MHC locus that are independently associated with single TCR Vα or Vβ gene expression. (XLSX 70 kb)

Supplementary Table 7

Expression variation of TCR Vα and Vβ genes explained by indepedent associations with SNP and amino acid variation in the MHC locus. (XLSX 54 kb)

Supplementary Table 8

Expression variation of TCR Vα and Vβ genes explained by indepedent associations with four-digit haplotypes for classical MHC genes. (XLSX 12 kb)

Supplementary Table 9

Probabilities that classical MHC gene amino acid positions influences expression of any TCR Vα gene. (XLSX 62 kb)

Supplementary Table 10

Probabilities that classical MHC gene amino acid positions influences expression of any TCR Vβ gene. (XLSX 61 kb)

Supplementary Table 11

A list of PDB accession codes, MHC alleles and TCR Vα gene used in the analysis of TCR–pMHC complexes. (XLSX 58 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sharon, E., Sibener, L., Battle, A. et al. Genetic variation in MHC proteins is associated with T cell receptor expression biases. Nat Genet 48, 995–1002 (2016). https://doi.org/10.1038/ng.3625

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3625

This article is cited by

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