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:

HLA class I signal peptide polymorphism determines the level of CD94/NKG2–HLA-E-mediated regulation of effector cell responses

Abstract

Human leukocyte antigen (HLA)-E binds epitopes derived from HLA-A, HLA-B, HLA-C and HLA-G signal peptides (SPs) and serves as a ligand for CD94/NKG2A and CD94/NKG2C receptors expressed on natural killer and T cell subsets. We show that among 16 common classical HLA class I SP variants, only 6 can be efficiently processed to generate epitopes that enable CD94/NKG2 engagement, which we term ‘functional SPs’. The single functional HLA-B SP, known as HLA-B/−21M, induced high HLA-E expression, but conferred the lowest receptor recognition. Consequently, HLA-B/−21M SP competes with other SPs for providing epitope to HLA-E and reduces overall recognition of target cells by CD94/NKG2A, calling for reassessment of previous disease models involving HLA-B/−21M. Genetic population data indicate a positive correlation between frequencies of functional SPs in humans and corresponding cytomegalovirus mimics, suggesting a means for viral escape from host responses. The systematic, quantitative approach described herein will facilitate development of prediction algorithms for accurately measuring the impact of CD94/NKG2–HLA-E interactions in disease resistance/susceptibility.

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: HLA class I signal peptide polymorphism influences HLA-E expression.
Fig. 2: JurkatNKG2 reporter cells respond differentially to VL9-pulsed .221 and .221-SPE cells.
Fig. 3: 221-SPE*01:03 cells differentially stimulate primary NK and NKL cells.
Fig. 4: Frequencies of combined functional signal peptides at each HLA locus across NMDP populations.
Fig. 5: HLA-A, HLA-B and HLA-C haplotypes and genotypes stratified by the presence of alleles encoding functional signal peptides.
Fig. 6: HLA-E expression levels on BLCLs and recognition of BLCLs by reporter cells suggest competition between SP variants in providing VL9 epitopes to HLA-E.
Fig. 7: Increasing copy number of alleles encoding SP-1C versus SP-6B associate with opposing effects on JurkatNKG2A reporter cell activity.

Similar content being viewed by others

Data availability

Publicly available HLA genetic datasets used in this study include Allele Frequency Net database (http://www.allelefrequencies.net/), IPD-IMGT/HLA database (https://www.ebi.ac.uk/ipd/imgt/hla/), NMDP Registry Haplotype Frequencies (https://frequency.nmdp.org/) and the 2014 1000 Genomes Project HLA data (https://www.internationalgenome.org/category/hla/). The structure of the human CD94/NKG2A complex with HLA-E (3CDG) was downloaded from the Protein Data Bank (https://www.rcsb.org/). Source data are provided with this paper. All other data are available within the article and Supplementary Information.

Code availability

PyMOL script is provided in the Supplementary Information.

References

  1. Quatrini, L. et al. Human NK cells, their receptors and function. Eur. J. Immunol. 51, 1566–1579 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. McMahon, C. W. & Raulet, D. H. Expression and function of NK cell receptors in CD8+ T cells. Curr. Opin. Immunol. 13, 465–470 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Borrego, F., Ulbrecht, M., Weiss, E. H., Coligan, J. E. & Brooks, A. G. Recognition of human histocompatibility leukocyte antigen (HLA)-E complexed with HLA class I signal sequence-derived peptides by CD94/NKG2 confers protection from natural killer cell-mediated lysis. J. Exp. Med. 187, 813–818 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Braud, V. M. et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 391, 795–799 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Lee, N. et al. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proc. Natl Acad. Sci. USA 95, 5199–5204 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Andre, P. et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175, 1731–1743 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Vales-Gomez, M., Reyburn, H. T., Erskine, R. A., Lopez-Botet, M. & Strominger, J. L. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J. 18, 4250–4260 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tomasec, P. et al. Surface expression of HLA-E, an inhibitor of natural killer cells, enhanced by human cytomegalovirus gpUL40. Science 287, 1031 (2000).

    Article  CAS  PubMed  Google Scholar 

  9. Orr, M. T. & Lanier, L. L. Natural killer cell education and tolerance. Cell 142, 847–856 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Strong, R. K. et al. HLA-E allelic variants. Correlating differential expression, peptide affinities, crystal structures, and thermal stabilities. J. Biol. Chem. 278, 5082–5090 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Lee, N., Goodlett, D. R., Ishitani, A., Marquardt, H. & Geraghty, D. E. HLA-E surface expression depends on binding of TAP-dependent peptides derived from certain HLA class I signal sequences. J. Immunol. 160, 4951–4960 (1998).

    Article  CAS  PubMed  Google Scholar 

  12. Bland, F. A., Lemberg, M. K., McMichael, A. J., Martoglio, B. & Braud, V. M. Requirement of the proteasome for the trimming of signal peptide-derived epitopes presented by the nonclassical major histocompatibility complex class I molecule HLA-E. J. Biol. Chem. 278, 33747–33752 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Llano, M. et al. HLA-E-bound peptides influence recognition by inhibitory and triggering CD94/NKG2 receptors: preferential response to an HLA-G-derived nonamer. Eur. J. Immunol. 28, 2854–2863 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Miller, J. D. et al. Analysis of HLA-E peptide-binding specificity and contact residues in bound peptide required for recognition by CD94/NKG2. J. Immunol. 171, 1369–1375 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Kaiser, B. K. et al. Interactions between NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J. Immunol. 174, 2878–2884 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Sullivan, L. C. et al. The heterodimeric assembly of the CD94-NKG2 receptor family and implications for human leukocyte antigen-E recognition. Immunity 27, 900–911 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Hammer, Q. et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 19, 453–463 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Walters, L. C., McMichael, A. J. & Gillespie, G. M. Detailed and atypical HLA-E peptide binding motifs revealed by a novel peptide exchange binding assay. Eur. J. Immunol. 50, 2075–2091 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Walters, L. C. et al. Primary and secondary functions of HLA-E are determined by stability and conformation of the peptide-bound complexes. Cell Rep. 39, 110959 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gustafson, K. S. & Ginder, G. D. Interferon-gamma induction of the human leukocyte antigen-E gene is mediated through binding of a complex containing STAT1α to a distinct interferon-γ-responsive element. J. Biol. Chem. 271, 20035–20046 (1996).

    Article  CAS  PubMed  Google Scholar 

  21. Garcia-Beltran, W. F. et al. Open conformers of HLA-F are high-affinity ligands of the activating NK cell receptor KIR3DS1. Nat. Immunol. 17, 1067–1074 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Petrie, E. J. et al. CD94-NKG2A recognition of human leukocyte antigen (HLA)-E bound to an HLA class I leader sequence. J. Exp. Med. 205, 725–735 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lopez-Botet, M., De Maria, A., Muntasell, A., Della Chiesa, M. & Vilches, C. Adaptive NK cell response to human cytomegalovirus: facts and open issues. Semin. Immunol. 65, 101706 (2023).

    Article  CAS  PubMed  Google Scholar 

  24. Robertson, M. J. et al. Characterization of a cell line, NKL, derived from an aggressive human natural killer cell leukemia. Exp. Hematol. 24, 406–415 (1996).

    CAS  PubMed  Google Scholar 

  25. Gragert, L., Madbouly, A., Freeman, J. & Maiers, M. Six-locus high resolution HLA haplotype frequencies derived from mixed-resolution DNA typing for the entire US donor registry. Hum. Immunol. 74, 1313–1320 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Gourraud, P. A. et al. HLA diversity in the 1000 Genomes dataset. PLoS ONE 9, e97282 (2014).

  27. Pietra, G. et al. HLA-E-restricted recognition of cytomegalovirus-derived peptides by human CD8+ cytolytic T lymphocytes. Proc. Natl Acad. Sci. USA 100, 10896–10901 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bjorkstrom, N. K., Strunz, B. & Ljunggren, H. G. Natural killer cells in antiviral immunity. Nat. Rev. Immunol. 22, 112–123 (2022).

    Article  PubMed  Google Scholar 

  29. Barber, C. et al. Structure-guided stabilization of pathogen-derived peptide-HLA-E complexes using non-natural amino acids conserves native TCR recognition. Eur. J. Immunol. 52, 618–632 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Horowitz, A. et al. Class I HLA haplotypes form two schools that educate NK cells in different ways. Sci. Immunol. 1, eaag1672 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Hallner, A. et al. The HLA-B -21 dimorphism impacts on NK cell education and clinical outcome of immunotherapy in acute myeloid leukemia. Blood 133, 1479–1488 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Merino, A. M. et al. HLA-B signal peptide polymorphism influences the rate of HIV-1 acquisition but not viral load. J. Infect. Dis. 205, 1797–1805 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ramsuran, V. et al. Elevated HLA-A expression impairs HIV control through inhibition of NKG2A-expressing cells. Science 359, 86–90 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Petersdorf, E. W. et al. Role of HLA-B exon 1 in graft-versus-host disease after unrelated haemopoietic cell transplantation: a retrospective cohort study. Lancet Haematol. 7, e50–e60 (2020).

    Article  PubMed  Google Scholar 

  35. van Hall, T. et al. Monalizumab: inhibiting the novel immune checkpoint NKG2A. J. Immunother. Cancer 7, 263 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Martin-Villa, J. M. et al. HLA-G: too much or too little? Role in cancer and autoimmune disease. Front. Immunol. 13, 796054 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Battin, C. et al. NKG2A-checkpoint inhibition and its blockade critically depends on peptides presented by its ligand HLA-E. Immunology 166, 507–521 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Bansal, A. et al. HLA-E-restricted HIV-1-specific CD8+ T cell responses in natural infection. J. Clin. Investig. 131, e148979 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Verweij, M. C. et al. Modulation of MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine efficacy. Science 372, eabe9233 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hansen, S. G. et al. Broadly targeted CD8+ T cell responses restricted by major histocompatibility complex E. Science 351, 714–720 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Partridge, T. et al. Discrimination between human leukocyte antigen class I-bound and co-purified HIV-derived peptides in immunopeptidomics workflows. Front. Immunol. 9, 912 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Shimizu, Y. & DeMars, R. Production of human cells expressing individual transferred HLA-A,-B,-C genes using an HLA-A,-B,-C null human cell line. J. Immunol. 142, 3320–3328 (1989).

    Article  CAS  PubMed  Google Scholar 

  43. Kaiser, B. K., Pizarro, J. C., Kerns, J. & Strong, R. K. Structural basis for NKG2A/CD94 recognition of HLA-E. Proc. Natl Acad. Sci. USA 105, 6696–6701 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Case, D. A. et al. Amber 2022 (University of California, 2022).

  45. Tian, C. et al. ff19SB: amino-acid-specific protein backbone parameters trained against quantum mechanics energy surfaces in solution. J. Chem. Theory Comput. 16, 528–552 (2020).

    Article  PubMed  Google Scholar 

  46. Izadi, S., Anandakrishnan, R. & Onufriev, A. V. Building water models: a different approach. J. Phys. Chem. Lett. 5, 3863–3871 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Q. Hammer, G. Nelson and M. P. Martin for discussions and M. Thompson for technical assistance. We also thank the NIH Blood Bank, the University Medical Center Hamburg-Eppendorf, and donors for providing blood samples. This project has been funded in whole or in part with federal funds from the Frederick National Laboratory for Cancer Research, under contract no. 75N91019D00024. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the US Government. This research was supported in part by the Intramural Research Program of the NIH, Frederick National Laboratory, Center for Cancer Research. A. Hoelzemer and M.B. are supported by the Federal Ministry of Education and Research (01KI2110) and DZIF (German Center for Infection Research).

Author information

Authors and Affiliations

Authors

Contributions

Z.L. and A.A.B. designed and performed experiments, analyzed data and wrote the manuscript; M.V. performed bioinformatic analysis; L.G., M.Q. and G.M.G. performed SPR and peptide binding analyses; M.B. and A. Hoelzemer performed macrophage experiments; W.K.K. performed molecular dynamics simulation analysis; M.A. assisted Z.L. and A.A.B. in cloning and flow cytometry experiments; Y.Y. performed HLA typing; P.O. and W.F.G.-B. designed and made the JurkatNKG2 reporter cells; S.D. and T.A. performed mass spectrometry analysis; V.N., A. Horowitz, A.J.M., A. Hoelzemer, G.M.G. and W.F.G.-B. contributed to the design of the work; M.C. designed the study, supervised all work and wrote the manuscript. All authors reviewed and approved the manuscript.

Corresponding author

Correspondence to Mary Carrington.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Peter Parham and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary handling editor: S. Houston, in collaboration with the Nature Immunology team.

Additional information

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

Extended data

Extended Data Fig. 1 HLA class I SP frequencies in human populations.

Allelic frequencies of SP variants were determined based on HLA class I allele frequencies obtained from www.allelefrequencies.net for four USA NMDP populations, including African (n = 28,557), European (n = 1,242,890), Chinese (n = 99,672), and Southeast Asian (n = 27,978). SP sequences and corresponding HLA class I alleles are shown in Table 1.

Source data

Extended Data Fig. 2 Differential binding of VL9 peptides to HLA-E.

a, HLA-E surface expression level on VL9-pulsed .221 cells measured by flow cytometry using 3D12 antibody. Cells were pulsed with peptides at 100, 30, and 10 μM concentrations. Peptide sequence alignments labeled with the corresponding SP variants are shown on the left. The expression index was calculated using 3D12 MFI as follows: ((sample – neg_ctrl) ÷ (pos_ctrl – neg_ctrl)) × 100, where neg_ctrl represents unpulsed .221 cells mixed with DMSO and pos_ctrl represents unpulsed .221 cells incubated constantly at 26 °C. Data represent triplicate experiments and reflect endogenous HLA-E*01:01 expression. b, VL9 binding to HLA-E*01:03 estimated using ELISA-based peptide binding and thermal stability assays. Bar charts represent absorbance signals at 450 nm reflecting the degree of VL9HLA-E complex recovery in the sandwich ELISA assay and thermal melting temperatures of VL9HLA-E determined using differential scanning fluorimetry. Data represent six experiments. a, b, Light gray bars depict the three peptides that showed the lowest binding levels consistently across experiments. Error bars represent the mean ± SD.

Source data

Extended Data Fig. 3 Differential HLA-E expression on the surface of .221-SPE cells and correlations of HLA-E expression levels between pairs of distinct measurements.

a, Cell surface expression levels of HLA-E on .221-SPE*01:01 and .221-SPE*01:03 cells measured by flow cytometry using anti-FLAG antibody. Data represent measurements on different days (n = 3). Error bars represent the mean ± SD. Amino acid sequence alignments of corresponding SP variants are shown on the left. VL9 peptide is shown in red. b, Correlations between HLA-E expression levels measured using anti-FLAG antibody and those measured using 3D12 antibody on the surface of .221-SPE*01:01 or .221-SPE*01:03 cells. c, Correlations between HLA-E expression levels on .221-SPE*01:01 cells and those on .221-SPE*01:03 cells measured using 3D12 or anti-FLAG antibodies. d, Correlations of HLA-E expression levels on .221-SPB*57:01 and those on .221-SPE*01:01 or .221-SPE*01:03. b-d, R2 was determined by Spearman correlation analysis and is shown with a two-tailed P value.

Source data

Extended Data Fig. 4 HLA-E expression levels on various cell types.

a, HLA-E expression levels on .221-SPE*01:03 cells, .221 cells pulsed with 100 μM VL9 peptides, PBMCs, and BLCLs. Error bars for PBMCs reflect variation across four donors (mean ± SD). The Y axis represents MFI obtained by 3D12 antibody staining minus MFI obtained by isotype control staining for each cell type. b, HLA-E expression levels on PBMCs and cell type subsets after 48 hours in culture with or without IFN-γ treatment. Data for two healthy donors (HD) are shown (HD75 and HD77) and represent triplicate experiments. Error bars represent the mean ± SD. P values for comparison between IFN-γ-treated and untreated cells were determined by a two-sided unpaired t-test: * - P < 0.05, ** - P < 0.01, *** - P < 0.001, **** - P < 0.0001.

Source data

Extended Data Fig. 5 JurkatNKG2A and JurkatNKG2C reporter activity against VL9-pulsed .221 cells.

a, Correlations between JurkatNKG2A and JurkatNKG2C reporter activity against VL9-pulsed (100uM) .221 expressing endogenous HLA-E*01:01 (shown in Fig. 2b). b, Absence of a correlation between HLA-E expression level on VL9-pulsed (100uM) .221 cells (shown in Fig. 1a) and Jurkat reporter cell activity against these cells (shown in Fig. 2b). Peptides are labeled with representative SPs. c, Correlations between Jurkat reporter cell activity against VL9-pulsed (100uM) .221 cells and KD values for HLA-E*01:03/VL9 binding to CD94/NKG2 determined by SPR analysis (Supplementary Table 1). a-c, R2 was determined by Spearman correlation analysis and is shown with a two-tailed P value.

Source data

Extended Data Fig. 6 Increase of HLA-E surface expression on monocyte-derived macrophages and enhanced JurkatNKG2A activity against these cells after IFN-γ treatment.

Monocytes were isolated from nine healthy donors and differentiated to monocyte-derived macrophages that were exposed to IFN-γ overnight (both unpulsed and VL9G-pulsed). Peptide pulsing was performed at 37°C for one hour prior to co-culture with reporter cells. P values for comparison between IFN-γ-treated and untreated cells were determined by a two-sided paired t-test.

Source data

Extended Data Fig. 7 Influence of valine at P7 of VL96B on CD94/NKG2A recognition.

a, Mutation analysis of P7 using JurkatNKG2A reporter cell response to .221-SPE*01:03 cells. Schematic representation of hybrid swap constructs transduced into .221 cells is shown on top. HLA-E expression level on .221-SPE*01:03 cells expressing wild-type (1A, 6B) and mutant (1AV, 6BL) SPs, and JurkatNKG2A activity against these cells are shown below. Data represent triplicate experiments. Error bars represent the mean ± SD. P values for comparisons between wild type and mutant SPs were determined by a two-sided unpaired t-test. b, Molecular dynamics simulation analysis of CD94/NKG2A–HLA-E*01:03 complexes in the presence of VL91A and VL96B peptides. Box plots show the root-mean-square deviations (RMSDs) of the full receptor, CD94, or NKG2A depending on the presence of the two distinct VL9 peptides in the binding groove during a 5 μs simulation (n = 500). Box boundaries span the 25–75 percentiles with the median marked in the middle; the whiskers extend to 10 and 90 percentiles, and the remaining data points are shown as gray circles. The RMSD box plots demonstrate higher receptor motions in VL96B-loaded complexes P values for comparisons between VL91A and VL96B were determined by a two-sided Mann-Whitney test.

Source data

Extended Data Fig. 8 NKG2A surface expression levels on different effector cells.

Jurkat reporter cells, primary NK cells from four healthy donors (HD), and NKL cells were stained with anti-NKG2A antibody (clone REA110). Flow cytometry histograms are shown on top, and graphical representation of the corresponding MFI data is shown below. The vertical line on the REA110 histogram separates NKG2A from NKG2A+ populations. NKG2A expression levels are determined as MFI values of NKG2A+ cells obtained by REA110 staining minus MFI values obtained by isotype control staining for each cell type. (-) represents untransduced parental Jurkat cells.

Source data

Extended Data Fig. 9 Distribution of specific functional SP haplotype groups present in NMDP samples.

Each SP haplotype group presented in Fig. 5a (main text) was further stratified by specific SP variants encoded by HLA-A, -B, -C haplotypes. Haplotype groupings are labeled according to the encoded functional SP variants.

Source data

Extended Data Fig. 10 HLA-E surface expression on BLCLs and reporter cell recognition of BLCLs stratified by copy number of alleles encoding SP-1A, SP-2A, or SP-2C.

HLA-E surface expression levels on BLCLs measured by flow cytometry using 3D12 antibody is shown on top and the corresponding JurkatNKG2A reporter cell activity (% of CD69+ cells) is shown below (n = 360). Lines in each group represent the median. P values for multi-group comparisons were determined by the Kruskal-Wallis test.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Peer Review File

Supplementary Table 1

Equilibrium Kd values for HLA-E*01:03/VL9 binding to CD94/NKG2.

Supplementary Code 1

PyMOL script for root-mean-square deviation calculations in the molecular dynamics simulation analysis.

Supplementary Data 1

Antibody and reagents.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical 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

Lin, Z., Bashirova, A.A., Viard, M. et al. HLA class I signal peptide polymorphism determines the level of CD94/NKG2–HLA-E-mediated regulation of effector cell responses. Nat Immunol 24, 1087–1097 (2023). https://doi.org/10.1038/s41590-023-01523-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01523-z

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