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:

A dynamic CD2-rich compartment at the outer edge of the immunological synapse boosts and integrates signals

Subjects

An Author Correction to this article was published on 29 October 2020

This article has been updated

Abstract

The CD2–CD58 recognition system promotes adhesion and signaling and counters exhaustion in human T cells. We found that CD2 localized to the outer edge of the mature immunological synapse, with cellular or artificial APC, in a pattern we refer to as a ‘CD2 corolla’. The corolla captured engaged CD28, ICOS, CD226 and SLAM-F1 co-stimulators. The corolla amplified active phosphorylated Src-family kinases (pSFK), LAT and PLC-γ over T cell receptor (TCR) alone. CD2–CD58 interactions in the corolla boosted signaling by 77% as compared with central CD2–CD58 interactions. Engaged PD-1 invaded the CD2 corolla and buffered CD2-mediated amplification of TCR signaling. CD2 numbers and motifs in its cytoplasmic tail controlled corolla formation. CD8+ tumor-infiltrating lymphocytes displayed low expression of CD2 in the majority of people with colorectal, endometrial or ovarian cancer. CD2 downregulation may attenuate antitumor T cell responses, with implications for checkpoint immunotherapies.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: A unique ring pattern, called corolla, formed by CD2–CD58 interactions in the IS.
Fig. 2: The corolla organizes multiple co-stimulatory receptor interactions.
Fig. 3: CD2 corolla boosts CD2-dependent TCR-signal amplification.
Fig. 4: Regulation of signaling in the corolla by PD-1 engagement.
Fig. 5: Number of CD2 per human T cell predicts CD58 engagement and corolla formation.
Fig. 6: CD2 expression determines corolla formation independent of signaling.
Fig. 7: CD8+ TILs from people with cancer can express considerably low levels of CD2.

Similar content being viewed by others

Data availability

scRNA-seq data are available in Supplementary Table 7 in the NCBI Gene Expression Omnibus single-cell-sequencing data section. Additional data and information that support the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

No custom code or mathematical algorithm was used when acquiring or analyzing data included in this study.

Change history

  • 29 October 2020

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Sanchez-Madrid, F. et al. Three distinct antigens associated with human T-lymphocyte-mediated cytolysis: LFA-1, LFA-2, and LFA-3. Proc. Natl Acad. Sci. USA 79, 7489–7493 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Sanders, M. E. et al. Human memory T lymphocytes express increased levels of three cell adhesion molecules (LFA-3, CD2, and LFA-1) and three other molecules (UCHL1, CDw29, and Pgp-1) and have enhanced IFN-γ production. J. Immunol. 140, 1401–1407 (1988).

    CAS  PubMed  Google Scholar 

  3. Davis, S. J. & van der Merwe, P. A. The kinetic-segregation model: TCR triggering and beyond. Nat. Immunol. 7, 803–809 (2006).

    CAS  PubMed  Google Scholar 

  4. Carmo, A. M., Mason, D. W. & Beyers, A. D. Physical association of the cytoplasmic domain of CD2 with the tyrosine kinases p56lck and p59fyn. Eur. J. Immunol. 23, 2196–2201 (1993).

    CAS  PubMed  Google Scholar 

  5. Dustin, M. L. et al. A novel adapter protein orchestrates receptor patterning and cytoskeletal polarity in T cell contacts. Cell 94, 667–677 (1998).

    CAS  PubMed  Google Scholar 

  6. Espagnolle, N. et al. CD2 and TCR synergize for the activation of phospholipase Cγ1/calcium pathway at the immunological synapse. Int. Immunol. 19, 239–248 (2007).

    CAS  PubMed  Google Scholar 

  7. Freiberg, B. A. et al. Staging and resetting T cell activation in SMACs. Nat. Immunol. 3, 911–917 (2002).

    CAS  PubMed  Google Scholar 

  8. Saliba, D. G. et al. Composition and structure of synaptic ectosomes exporting antigen receptor linked to functional CD40 ligand from helper T-cells. Elife 8, 600551 (2019).

    Google Scholar 

  9. Dustin, M. L., Ferguson, L. M., Chan, P. Y., Springer, T. A. & Golan, D. E. Visualization of CD2 interaction with LFA-3 and determination of the two-dimensional dissociation constant for adhesion receptors in a contact area. J. Cell Biol. 132, 465–474 (1996).

    CAS  PubMed  Google Scholar 

  10. Kaizuka, Y., Douglass, A. D., Vardhana, S., Dustin, M. L. & Vale, R. D. The coreceptor CD2 uses plasma membrane microdomains to transduce signals in T cells. J. Cell Biol. 185, 521–534 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Zaru, R., Cameron, T. O., Stern, L. J., Müller, S. & Valitutti, S. Cutting edge: TCR engagement and triggering in the absence of large-scale molecular segregation at the T cell-APC contact site. J. Immunol. 168, 4287–4291 (2002).

    CAS  PubMed  Google Scholar 

  12. De Jager, P. L. et al. The role of the CD58 locus in multiple sclerosis. Proc. Natl Acad. Sci. USA 106, 5264–5269 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Raychaudhuri, S. et al. Genetic variants at CD28, PRDM1 and CD2/CD58 are associated with rheumatoid arthritis risk. Nat. Genet. 41, 1313–1318 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. McKinney, E. F., Lee, J. C., Jayne, D. R., Lyons, P. A. & Smith, K. G. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Abdul Razak, F. R., Diepstra, A., Visser, L. & van den Berg, A. CD58 mutations are common in Hodgkin lymphoma cell lines and loss of CD58 expression in tumor cells occurs in Hodgkin lymphoma patients who relapse. Genes Immun. 17, 363–366 (2016).

    CAS  PubMed  Google Scholar 

  16. Koneru, M., Monu, N., Schaer, D., Barletta, J. & Frey, A. B. Defective adhesion in tumor infiltrating CD8+ T cells. J. Immunol. 176, 6103–6111 (2006).

    CAS  PubMed  Google Scholar 

  17. Abu-Shah, E. et al. A tissue-like platform for studying engineered quiescent human T-cells’ interactions with dendritic cells. Elife 8, e48221 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Vendrame, E. et al. TIGIT is upregulated by HIV-1 infection and marks a highly functional adaptive and mature subset of natural killer cells. AIDS 34, 801–813 (2020).

  19. Ramsbottom, K. M. et al. Cutting edge: DNAX accessory molecule 1-deficient CD8+ T cells display immunological synapse defects that impair antitumor immunity. J. Immunol. 192, 553–557 (2014).

    CAS  PubMed  Google Scholar 

  20. Aversa, G., Chang, C. C., Carballido, J. M., Cocks, B. G. & de Vries, J. E. Engagement of the signaling lymphocytic activation molecule (SLAM) on activated T cells results in IL-2-independent, cyclosporin A-sensitive T cell proliferation and IFN-gamma production. J. Immunol. 158, 4036–4044 (1997).

    CAS  PubMed  Google Scholar 

  21. Colin-York, H., Kumari, S., Barbieri, L., Cords, L. & Fritzsche, M. Distinct actin cytoskeleton behaviour in primary and immortalised T-cells. J. Cell Sci. 133, jcs232322 (2019).

  22. Hui, E. et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Clarkson, N. G. & Brown, M. H. Inhibition and activation by CD244 depends on CD2 and phospholipase C-γ1. J. Biol. Chem. 284, 24725–24734 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kaye, J. et al. Selective development of CD4+ T cells in transgenic mice expressing a class II MHC-restricted antigen receptor. Nature 341, 746–749 (1989).

    CAS  PubMed  Google Scholar 

  25. Pages, F. et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).

    PubMed  Google Scholar 

  26. Huyghe, N., Baldin, P. & Van den Eynde, M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol. Rep. (Oxf.) 8, 11–24 (2020).

    Google Scholar 

  27. Chirica, M. et al. Phenotypic analysis of T cells infiltrating colon cancers: correlations with oncogenetic status. Oncoimmunology 4, e1016698 (2015).

    PubMed  PubMed Central  Google Scholar 

  28. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    CAS  PubMed  Google Scholar 

  29. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    CAS  PubMed  Google Scholar 

  30. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356.e16 (2017).

    CAS  PubMed  Google Scholar 

  31. Grakoui, A. et al. The immunological synapse: a molecular machine controlling T cell activation. Science 285, 221–227 (1999).

    CAS  PubMed  Google Scholar 

  32. Valitutti, S., Dessing, M., Aktories, K., Gallati, H. & Lanzavecchia, A. Sustained signaling leading to T cell activation results from prolonged T cell receptor occupancy. Role of T cell actin cytoskeleton. J. Exp. Med. 181, 577–584 (1995).

    CAS  PubMed  Google Scholar 

  33. Kumari, S. et al. Actin foci facilitate activation of the phospholipase C-γ in primary T lymphocytes via the WASP pathway. Elife 4, e04953 (2015).

    PubMed Central  Google Scholar 

  34. Shen, K., Thomas, V. K., Dustin, M. L. & Kam, L. C. Micropatterning of costimulatory ligands enhances CD4+ T cell function. Proc. Natl Acad. Sci. USA 105, 7791–7796 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Cai, E. et al. Visualizing dynamic microvillar search and stabilization during ligand detection by T cells. Science 356, eaal3118 (2017).

  36. Dustin, M. L. Adhesive bond dynamics in contacts between T lymphocytes and glass-supported planar bilayers reconstituted with the immunoglobulin-related adhesion molecule CD58. J. Biol. Chem. 272, 15782–15788 (1997).

    CAS  PubMed  Google Scholar 

  37. Siokis, A. et al. In silico characterization of mechanisms positioning costimulatory and checkpoint complexes in immune synapses. Preprint at bioRxiv https://doi.org/10.1101/2020.01.16.908723 (2020).

  38. Douglass, A. D. & Vale, R. D. Single-molecule microscopy reveals plasma membrane microdomains created by protein–protein networks that exclude or trap signaling molecules in T cells. Cell 121, 937–950 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hutchings, N. J., Clarkson, N., Chalkley, R., Barclay, A. N. & Brown, M. H. Linking the T cell surface protein CD2 to the actin-capping protein CAPZ via CMS and CIN85. J. Biol. Chem. 278, 22396–22403 (2003).

    CAS  PubMed  Google Scholar 

  40. Wei, F. et al. Strength of PD-1 signaling differentially affects T-cell effector functions. Proc. Natl Acad. Sci. USA 110, E2480–2489 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Lo, D. J. et al. Selective targeting of human alloresponsive CD8+ effector memory T cells based on CD2 expression. Am. J. Transplant. 11, 22–33 (2011).

    CAS  PubMed  Google Scholar 

  42. Leitner, J., Herndler-Brandstetter, D., Zlabinger, G. J., Grubeck-Loebenstein, B. & Steinberger, P. CD58/CD2 is the primary costimulatory pathway in human CD28-CD8+ T Cells. J. Immunol. 195, 477–487 (2015).

    CAS  PubMed  Google Scholar 

  43. Choudhuri, K. et al. Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse. Nature 507, 118–123 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Yao, X. et al. Isolation and characterization of an HLA-DPB1*04:01-restricted MAGE-A3 T-cell receptor for cancer immunotherapy. J. Immunother. 39, 191–201 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, Y. et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat. Biotechnol. 23, 349–354 (2005).

    CAS  PubMed  Google Scholar 

  46. Dustin, M. L., Starr, T., Varma, R. & Thomas, V. K. Supported planar bilayers for study of the immunological synapse. Curr. Protoc. Immunol. 76, 18.13.1–18.13.35 (2007).

    Google Scholar 

  47. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    PubMed  Google Scholar 

  49. Zar, J. H. in Biostatistical Analysis 2nd edn, Ch. 18 (Prentice-Hall, 1984).

Download references

Acknowledgements

We thank S. Davis, P.A. Van Der Merwe, O. Dushek and R. Owens (University of Oxford) for kindly providing plasmids and HLA-A2 pMHC monomers. We also thank M. Dumoux (The Rosalind Franklin Institute) for kindly providing plasmids. We thank M. H. Brown and S. Sivakumar for helpful discussion and feedback on experiments and writing of the manuscript, and all the Dustin lab members for their kind support. We thank S. Balint for his help on some experiments and for maintaining the TIRF microscope, C. Laggerholm for access to and assistance with the Airyscan confocal microscope and P. Cespedes for his contribution in methods development for quantification of surface molecules on T cells. The results published here are in part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga. A Kennedy Trust for Rheumatology (KTRR) Prize Studentship supported P.D. An UCB-Oxford Post-doctoral Fellowship supported E.A.S. A grant from The Research Council of Norway in conjunction with Marie Sklodowska-Curie Actions (275466) supported A.K. Wellcome Trust Principal Research Fellowship 100262Z/12/Z, and a grant from KTRR supported M.L.D. KTRR supports the Kennedy Institute Microscopy Facility and the Wolfson Foundation supports the Weatherall Institute Microscopy Facility. A collaborative grant from the Human Frontiers Science Program supported E.A.-S., A.S. and M.M.-H. A.S. was supported by the German Research Foundation (DFG) and Collaborative Research Center (SFB 854) ‘Molecular Organization of Cellular Communication within the Immune System’. A fellowship from Philippe Foundation partially supported D.D. A Wellcome Trust Senior Research Fellowship 207537/Z/17/Z supported E.A.-S. and M.A.K. NIHR Biomedical Research Centre, Oxford, supports the Oxford Gastro-Intestinal Biobank and the Oxford Inflammatory Bowel Disease Cohort study. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. We acknowledge the contribution to this study made by the Oxford Centre for Histopathology Research and the Oxford Radcliffe Biobank, which are supported by the NIHR Oxford Biomedical Research Centre.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

P.D. conceptualized the project, designed and performed experiments, analyzed the data and co-wrote the manuscript. E.A.-S. designed, performed and analyzed experiments and co-wrote the manuscript. S.V. prepared and performed experiments and maintained critical infrastructure. K.K. assisted in confocal micrsocpy experiments and acquiring confocal microscopy images. A.K. performed and analyzed experiments. S.M., M.F. and E.M. prepared single-cell suspensions from CRC tissue. E.A.-S. prepared single-cell suspensions from EndoC and OC samples. S.M. performed transcriptional analysis. P.D. and E.A.S. performed the staining and acquisition experiments of CRC tissues. E.A.S. performed the staining and acquisition experiments of EndoC and OC tissues. J.A., H.R., S.Y., S.V., V.M. and M.A.K. prepared essential reagents for experiments. V.M. designed image-analysis software and trained P.D. in its use. L.Y.W.L. and T.S. performed transcriptional analysis of CRC, HCC, NSCLC and melanoma cohorts. The Oxford IBD Cohort Investigators provided access to CRC tissue and clinical data. M.M. provided EndoC and OC samples and clinical data and contributed to discussion of patient data. N.W. and A.A.A. provided access to Endo and OC samples. P.D., E.A.S., V.M., D.D., A.S., M.M.-H. and M.L.D. made intellectual contributions to the project through regular discussions. D.D. provided training to P.D. and conceptualized and designed experiments. M.L.D. supervised the research and facilitated collaboration. P.D. drafted the manuscript, and E.A.-S., V.M., S.M., D.D. and M.L.D. contributed to writing and editing.

Corresponding author

Correspondence to Michael L. Dustin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Supplementary information

Supplementary Information

Suppplementary Figs. 1–6.

Reporting Summary

Supplementary Video 1

Tracking of IS formation by a human T cell incubated on SLBs reconstituted with ICAM-1 (200 molecules per μm2), anti-CD3 Fab (30 molecules per μm2) or CD58 (200 molecules per μm2). Cells were imaged at 4-s intervals with TIRFM. Scale bar, 5 μm.

Supplementary Video 2

Tracking of IS formation by a human T cell incubated on SLBs reconstituted with ICAM-1 (200 molecules per μm2), anti-CD3 Fab (30 molecules per μm2) or CD58 (200 molecules per μm2). Cells were imaged at 4-s intervals with TIRFM. Scale bar, 5 μm.

Supplementary Video 3

Tracking of IS formation by a human T cell incubated on SLBs reconstituted with ICAM-1 (200 molecules per μm2), anti-CD3 Fab (30 molecules per μm2), CD58 (200 molecules per μm2) or CD80 (100 molecules per μm2). Cells were imaged at 4-s intervals with TIRFM.

Supplementary Tables

Tables supporting main data presented in the manuscript.

Supplementary Data

Numerical data used for plotting Supplementary Figs. 3–6.

Source data

Source Data Fig. 3

Numerical data used for plotting various plots

Source Data Fig. 4

Numerical data used for plotting various plots

Source Data Fig. 5

Numerical data used for plotting various plots

Source Data Fig. 6

Numerical data used for plotting various plots

Source Data Fig. 7

Numerical data used for plotting various plots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Demetriou, P., Abu-Shah, E., Valvo, S. et al. A dynamic CD2-rich compartment at the outer edge of the immunological synapse boosts and integrates signals. Nat Immunol 21, 1232–1243 (2020). https://doi.org/10.1038/s41590-020-0770-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0770-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer