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:

N-glycosylation bidirectionally extends the boundaries of thymocyte positive selection by decoupling Lck from Ca2+ signaling

Abstract

Positive selection of diverse yet self-tolerant thymocytes is vital to immunity and requires a limited degree of T cell antigen receptor (TCR) signaling in response to self peptide–major histocompatibility complexes (self peptide–MHCs). Affinity of newly generated TCR for peptide-MHC primarily sets the boundaries for positive selection. We report that N-glycan branching of TCR and the CD4 and CD8 coreceptors separately altered the upper and lower affinity boundaries from which interactions between peptide-MHC and TCR positively select T cells. During thymocyte development, N-glycan branching varied approximately 15-fold. N-glycan branching was required for positive selection and decoupled Lck signaling from TCR-driven Ca2+ flux to simultaneously promote low-affinity peptide-MHC responses while inhibiting high-affinity ones. Therefore, N-glycan branching imposes a sliding scale on interactions between peptide-MHC and TCR that bidirectionally expands the affinity range for positive selection.

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: N-glycan branching is required for T cell development.
Figure 2: Branching deficiency enhances death by neglect.
Figure 3: N-glycan branching prevents death by neglect by promoting Lck activation and coreceptor surface retention.
Figure 4: Branching deficiency sensitizes thymocytes to TCR-induced death.
Figure 5: N-glycan branching inhibits death by negative selection.
Figure 6: Branching deficiency enhances TCR-triggered Ca2+ flux.

Similar content being viewed by others

References

  1. Starr, T.K., Jameson, S.C. & Hogquist, K.A. Positive and negative selection of T cells. Annu. Rev. Immunol. 21, 139–176 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Morris, G.P. & Allen, P.M. How the TCR balances sensitivity and specificity for the recognition of self and pathogens. Nat. Immunol. 13, 121–128 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Gallo, E.M. et al. Calcineurin sets the bandwidth for discrimination of signals during thymocyte development. Nature 450, 731–735 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Parsons, S.A. et al. Genetic loss of calcineurin blocks mechanical overload-induced skeletal muscle fiber type switching but not hypertrophy. J. Biol. Chem. 279, 26192–26200 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Li, Q.J. et al. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129, 147–161 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Staton, T.L. et al. Dampening of death pathways by schnurri-2 is essential for T-cell development. Nature 472, 105–109 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fischer, A.M., Katayama, C.D., Pages, G., Pouyssegur, J. & Hedrick, S.M. The role of erk1 and erk2 in multiple stages of T cell development. Immunity 23, 431–443 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Alberola-Lla, J., Forbush, K.A., Seger, R., Krebs, E.G. & Perlmutter, R.M. Selective requirement for MAP kinase activation in thymocyte differentiation. Nature 373, 620–623 (1995).

    Article  Google Scholar 

  9. Wang, D. et al. Tespa1 is involved in late thymocyte development through the regulation of TCR-mediated signaling. Nat. Immunol. 13, 560–568 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Daniels, M.A. et al. Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling. Nature 444, 724–729 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Fu, G. et al. Themis sets the signal threshold for positive and negative selection in T-cell development. Nature 504, 441–445 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Artyomov, M.N., Lis, M., Devadas, S., Davis, M.M. & Chakraborty, A.K. CD4 and CD8 binding to MHC molecules primarily acts to enhance Lck delivery. Proc. Natl. Acad. Sci. USA 107, 16916–16921 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Trobridge, P.A., Forbush, K.A. & Levin, S.D. Positive and negative selection of thymocytes depends on Lck interaction with the CD4 and CD8 coreceptors. J. Immunol. 166, 809–818 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Demetriou, M., Granovsky, M., Quaggin, S. & Dennis, J.W. Negative regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation. Nature 409, 733–739 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Grigorian, A., Mkhikian, H. & Demetriou, M. Interleukin-2, interleukin-7, T cell-mediated autoimmunity, and N-glycosylation. Ann. NY Acad. Sci. 1253, 49–57 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Mkhikian, H. et al. Genetics and the environment converge to dysregulate N-glycosylation in multiple sclerosis. Nat. Commun. 2, 334 (2011).

    Article  PubMed  CAS  Google Scholar 

  18. Lee, S.U. et al. N-glycan processing deficiency promotes spontaneous inflammatory demyelination and neurodegeneration. J. Biol. Chem. 282, 33725–33734 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Chen, I.J., Chen, H.L. & Demetriou, M. Lateral compartmentalization of T cell receptor versus CD45 by galectin-N-glycan binding and microfilaments coordinate basal and activation signaling. J. Biol. Chem. 282, 35361–35372 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Lau, K.S. et al. Complex N-glycan number and degree of branching cooperate to regulate cell proliferation and differentiation. Cell 129, 123–134 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Dennis, J.W., Nabi, I.R. & Demetriou, M. Metabolism, cell surface organization, and disease. Cell 139, 1229–1241 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Cummings, R.D. & Kornfeld, S. Characterization of the structural determinants required for the high affinity interaction of asparagine-linked oligosaccharides with immobilized Phaseolus vulgaris leukoagglutinating and erythroagglutinating lectins. J. Biol. Chem. 257, 11230–11234 (1982).

    Article  CAS  PubMed  Google Scholar 

  23. Grigorian, A. et al. Control of T cell-mediated autoimmunity by metabolite flux to N-glycan biosynthesis. J. Biol. Chem. 282, 20027–20035 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Metzler, M. et al. Complex asparagine-linked oligosaccharides are required for morphogenic events during post-implantation development. EMBO J. 13, 2056–2065 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ioffe, E. & Stanley, P. Mice lacking N-acetylglucosaminyltransferase I activity die at mid-gestation, revealing an essential role for complex or hybrid N-linked carbohydrates. Proc. Natl. Acad. Sci. USA 91, 728–732 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen, W. & Stanley, P. Five Lec1 CHO cell mutants have distinct Mgat1 gene mutations that encode truncated N-acetylglucosaminyltransferase I. Glycobiology 13, 43–50 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Rathmell, J.C., Lindsten, T., Zong, W.X., Cinalli, R.M. & Thompson, C.B. Deficiency in Bak and Bax perturbs thymic selection and lymphoid homeostasis. Nat. Immunol. 3, 932–939 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Bouillet, P. et al. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 286, 1735–1738 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Grillot, D.A., Merino, R. & Nunez, G. Bcl-XL displays restricted distribution during T cell development and inhibits multiple forms of apoptosis but not clonal deletion in transgenic mice. J. Exp. Med. 182, 1973–1983 (1995).

    Article  CAS  PubMed  Google Scholar 

  30. Nika, K. et al. Constitutively active Lck kinase in T cells drives antigen receptor signal transduction. Immunity 32, 766–777 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Robertson, J.M., Jensen, P.E. & Evavold, B.D. DO11.10 and OT-II T cells recognize a C-terminal ovalbumin 323–339 epitope. J. Immunol. 164, 4706–4712 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Vidal, K., Daniel, C., Hill, M., Littman, D.R. & Allen, P.M. Differential requirements for CD4 in TCR-ligand interactions. J. Immunol. 163, 4811–4818 (1999).

    CAS  PubMed  Google Scholar 

  33. Hampl, J., Chien, Y.H. & Davis, M.M. CD4 augments the response of a T cell to agonist but not to antagonist ligands. Immunity 7, 379–385 (1997).

    Article  CAS  PubMed  Google Scholar 

  34. Demotte, N. et al. Restoring the association of the T cell receptor with CD8 reverses anergy in human tumor-infiltrating lymphocytes. Immunity 28, 414–424 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Partridge, E.A. et al. Regulation of cytokine receptors by Golgi N-glycan processing and endocytosis. Science 306, 120–124 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Cainan, B.J., Szychowski, S., Chan, F.K., Cado, D. & Winoto, A. A role for the orphan steroid receptor Nur77 in apoptosis accompanying antigen-induced negative selection. Immunity 3, 273–282 (1995).

    Article  Google Scholar 

  37. Bouillet, P. et al. BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes. Nature 415, 922–926 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Villunger, A. et al. Negative selection of semimature CD4+8–HSA+ thymocytes requires the BH3-only protein Bim but is independent of death receptor signaling. Proc. Natl. Acad. Sci. USA 101, 7052–7057 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Taylor-Fishwick, D.A. & Siegel, J.N. Raf-1 provides a dominant but not exclusive signal for the induction of CD69 expression on T cells. Eur. J. Immunol. 25, 3215–3221 (1995).

    Article  CAS  PubMed  Google Scholar 

  40. Li, C.F. et al. Hypomorphic MGAT5 polymorphisms promote multiple sclerosis cooperatively with MGAT1 and interleukin-2 and -7 receptor variants. J. Neuroimmunol. 256, 71–76 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Brynedal, B. et al. MGAT5 alters the severity of multiple sclerosis. J. Neuroimmunol. 220, 120–124 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Grigorian, A. et al. Pathogenesis of multiple sclerosis via environmental and genetic dysregulation of N-glycosylation. Semin. Immunopathol. 34, 415–424 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yu, Z. et al. Family studies of type 1 diabetes reveal additive and epistatic effects between MGAT1 and three other polymorphisms. Genes Immun. 15, 218–223 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Huse, M. et al. Spatial and temporal dynamics of T cell receptor signaling with a photoactivatable agonist. Immunity 27, 76–88 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Shi, X. et al. Ca2+ regulates T-cell receptor activation by modulating the charge property of lipids. Nature 493, 111–115 (2013).

    Article  PubMed  CAS  Google Scholar 

  46. Gwack, Y. et al. Hair loss and defective T- and B-cell function in mice lacking ORAI1. Mol. Cell. Biol. 28, 5209–5222 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Feske, S., Picard, C. & Fischer, A. Immunodeficiency due to mutations in ORAI1 and STIM1. Clin. Immunol. 135, 169–182 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, P.P. et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Andersen for K14-Cre mice and K.H. Khachikyan for helping with experiments. We thank the members of M.D.'s lab for proofreading the manuscript. Supported by the US National Institutes of Health through the National Institute of Allergy and Infectious Disease (R01AI053331 to M.D.) and the National Heart, Lung, and Blood Institute (F30HL108451 to H.M.).

Author information

Authors and Affiliations

Authors

Contributions

R.W.Z. performed all experiments with the assistance of H.M., A.H., D.C., A.G. and A.A. M.D. wrote the paper with assistance from R.W.Z.

Corresponding author

Correspondence to Michael Demetriou.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 N-glycan branching regulates T cell development.

(a) The Golgi N-glycan GlcNAc branching pathway. The N-acetylglucosaminyltransferases Mgat1, 2, 4, and 5 act sequentially to generate N-acetylglucosamine (GlcNAc) branched N-glycans on glycoproteins transiting the Golgi. These are variably extended with galactose to generate N-acetyllactosamine units. Galectins bind N-acetyllactosamine, with avidity increasing with the number of N-acetyllactosamine units (i.e., branching). Loss of Mgat1 blocks all branching and results in only high-mannose type N-glycans. The plant lectin L-PHA binds tri- and tetra-antennary GlcNAc branched N-glycans and serves as a marker for Mgat1 deleted cells. GalT3, galactosyltransferase3; iGnT, β1,3 N-acetylglucosaminyl transferase; MII/MIIx, mannosidase II/IIx. (b) Flow cytometric analysis of ConA binding on wildtype thymic subpopulations, normalized to CD4SP thymocytes. NS, not significant, *P<0.05. P-values by unpaired t-test (2-tailed) with Welch’s and Bonferroni corrections. Data are one experiment representative of three (b; mean ± s.e.m. of three technical replicates) independent experiments.

Supplementary Figure 2 Characterization of Mgat1-deficient mice.

(a) Histology of the spleen. (b) Quantification of DN, DP, CD4, CD8 SP thymocyte and splenic CD4 and CD8 cells. Each symbol represents one mouse; small horizontal lines indicate the mean (n=10 mice). (c) Flow cytometric analysis of surface CD24 expression on thymocytes. (d) Flow cytometric analysis of 7-AAD staining of total thymocytes after one day culture at rest. Error bars indicate s.e.m. of three technical replicates. (e) Pictures of mice (left) revealing lack of gross skin defects and flow cytometric analysis of skin epithelial cells (right) from Mgat1f/f and Mgat1f/fK14-Cre+ mouse confirming deletion of Mgat1 in Mgat1f/fK14-Cre+ mouse. Numbers indicate the mean percent cells of indicated areas. (f) Percentage of L-PHA (left) and Annexin V+ (right) ex vivo splenic T cells after 56 days doxycycline treatment provided in the drinking water; error bars indicate s.e.m. of three technical replicates. NS, not significant, *P<0.05, ***P<0.001. P-values by unpaired t-test (1-tailed) with Welch’s corrections. Data are one experiment representative of three (a, c), or two (d, e) independent experiment, or pooled from 10 (b) independent experiments.

Supplementary Figure 3 Characterization of Mgat2-deficient mice.

(a) Total number (left) of thymocytes (n=15 mice) and splenocytes (n=7 mice) from 4-8 weeks old mice; quantification (right) of thymic and splenic subpopulations (n=6 mice). Each symbol represents one mouse; small horizontal lines indicate the mean. (b) Flow cytometric analysis of CD4 and CD8 expression (left) and L-PHA binding (right) of thymocytes and splenocytes. Numbers indicate the mean percent cells of indicated areas. (c) Flow cytometric analysis of Annexin V binding on ex vivo thymocytes; DP thymocytes were gated for analysis. NS, not significant, *P<0.05, **P<0.01, ***P<0.001. P-values by unpaired t-test (1-tailed) with Welch’s corrections. Data are pooled from 15 (a) independent experiments, and one experiment representative of six (b; mean of three technical replicates) or three (c; mean ± s.e.m of three technical replicates) experiments.

Supplementary Figure 4 Characterization of Mgat1-deficient mice and non-TCR induced death.

(a) Flow cytometric analysis of CD4 and CD8 expression (left) and L-PHA binding of thymocytes and splenocytes (right). Numbers indicate the mean percent cells of indicated areas. Note that although Mgat1 is expected to be deleted at the DP stage in Mgat1f/f CD4-Cre+ mice, loss of cell surface branching is delayed until the SP stage due to the time required for membrane turnover of cell surface glycoproteins. (b) Flow cytometric analysis of cell surface expression of IL-7R on thymic subpopulations. (c-e) Annexin V+ DP thymocytes following one day of culture at rest in the presence of dexamethasone (c), TNF (d), and α-Fas (e); gated on DP thymocytes for analysis. Data for each genotype are normalized to its own non-treated control. Relative Annexin V+ cells (%) = % Annexin V+treated – % Annexin+non-treated. Data are one representative experiment of four (a, mean of three technical replicates) or two (b-e; mean ± s.e.m. of three technical replicates) independent experiments.

Supplementary Figure 5 Characterization of cell death.

(a-e) Flow cytometric analysis of Annexin V and L-PHA binding on gated DP thymocytes cultured at rest (a, c), with and without z-VAD-FMK for one day (b), or with and without 40 mM GlcNAc, 10 mM Uridine, and 10 μM Kifunensine for two days (d, e). (f, g) Flow cytometric analysis of L-PHA and Annexin V binding on thymocyte without (f) and with ConA binding (g); DP thymocytes were gated for analysis. (h) Immunoblot of total Bcl-xL, Bax, Bim(EL), and Mcl-1 in ex vivo thymocytes. (i) Flow cytometric analysis of cell surface expression of CD69 and TCRβ in total thymocytes. Numbers indicate the mean percent cells of indicated areas. (j) Annexin V+ DP thymocytes after one day of culture with and without PMA and Ionomycin. NS, not significant, *P<0.05, **P<0.01, ***P<0.001. P-values by unpaired (a-c, j) or paired (d, e, g) t-test (1-tailed) with Welch’s and Bonferroni (d, e) corrections. Data are one experiment representative of three (a, b, f-h, j; mean (± s.e.m.) of three technical replicates), two (c-e; mean ± s.e.m. of three technical replicates) or four (i) independent experiments.

Supplementary Figure 6 Characterization of CD4, CD8 expression and cell death in Mgat1- and Mgat2-deficient thymocytes.

(a) Real-time RT-PCR analysis of Cd4 and Cd8α mRNA expression in total thymocytes; results are presented relative to Actb expression. (b) Immunoblot of total CD4 and CD8α of ex vivo thymocytes. (c) Flow cytometric analysis of CD4 and CD8α on gated Annexin V+ and Annexin V- DP thymocytes from Mgat1f/fLck-Cre+ mouse after one day culture at rest. (d, e) Flow cytometric analysis of surface CD4 and CD8α expression on ex vivo Mgat2f/fLck-Cre+ and Mgat5/ DP (d, e) and SP (d) thymocytes; gated on L-PHAlo cells for Mgat2f/fLck-Cre+ mice for analysis; data were normalized to control and each symbol represents one mouse, small horizontal lines indicate the mean (d). (f) Nur77+ DP thymocytes following one day stimulation by plate-bound α-CD3ɛ plus α-CD28. (g, i, j) Flow cytometric analysis of Annexin V binding on gated DP thymocytes after one day culture in the presence of PMA and Ionomycin (g), Ionomycin (i), or stimulated by α-CD3ɛ (10 μg/ml) plus α-CD28 (50 μg/ml) with cyclosporine A (j). (h) Annexin V+ ex vivo thymocytes, gated on T3.70+CD8+ cells for analysis. NS, not significant, *P<0.05, **P<0.01, ***P<0.001. P-values by unpaired t-test (1-tailed) with Welch’s corrections. Data are one experiment representative of three (a-c, g, h), or two (e, f, i, j) independent experiments (mean ± s.e.m. of three technical replicates).

Supplementary Figure 7 A model for N-glycan branching control of positive selection.

(a) A model for the interaction of branching with pMHC-TCR affinity in controlling positive selection. N-glycosylation provides a novel sliding scale that dynamically expands the range for positive selection in two directions by differentially controlling both the lower and upper limits of affinity from which pMHC-TCR interactions positively select T cells. (b) In response to low affinity pMHC, branching promotes positive selection by enhancing cell surface retention of the CD4 and CD8 co-receptors, which stabilize binding of low-affinity pMHC to TCR and enhance recruitment of Lck to TCR for low level signaling. Reduced branching disrupts binding of glycoproteins to galectins, leading to CD4/CD8 endocytosis, non-responsiveness to low affinity pMHC and death by neglect. In response to high affinity pMHC, high branching limits TCR clustering and downstream signaling to promote positive selection. Reduced branching disrupts galectin-TCR interactions, thereby promoting TCR clustering/signaling, and Ca2+ flux in response to high affinity pMHC, therefore enhancing negative selection.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 3657 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, R., Mkhikian, H., Grigorian, A. et al. N-glycosylation bidirectionally extends the boundaries of thymocyte positive selection by decoupling Lck from Ca2+ signaling. Nat Immunol 15, 1038–1045 (2014). https://doi.org/10.1038/ni.3007

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3007

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