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:

TAP dysfunction in dendritic cells enables noncanonical cross-presentation for T cell priming

Abstract

Classic major histocompatibility complex class I (MHC-I) presentation relies on shuttling cytosolic peptides into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). Viruses disable TAP to block MHC-I presentation and evade cytotoxic CD8+ T cells. Priming CD8+ T cells against these viruses is thought to rely solely on cross-presentation by uninfected TAP-functional dendritic cells. We found that protective CD8+ T cells could be mobilized during viral infection even when TAP was absent in all hematopoietic cells. TAP blockade depleted the endosomal recycling compartment of MHC-I molecules and, as such, impaired Toll-like receptor–regulated cross-presentation. Instead, MHC-I molecules accumulated in the ER–Golgi intermediate compartment (ERGIC), sequestered away from Toll-like receptor control, and coopted ER-SNARE Sec22b-mediated vesicular traffic to intersect with internalized antigen and rescue cross-presentation. Thus, when classic MHC-I presentation and endosomal recycling compartment–dependent cross-presentation are impaired in dendritic cells, cell-autonomous noncanonical cross-presentation relying on ERGIC-derived MHC-I counters TAP dysfunction to nevertheless mediate CD8+ T cell priming.

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: MHC-I molecules are retained in the ERGIC of murine DCs upon TAP deficiency.
Fig. 2: MHC-I molecules are retained in the ERGIC of human DCs upon TAP blockade by virus or TAP inhibitor.
Fig. 3: Intact cross-presentation occurs independently of TAP function and despite ERC depletion of MHC-I.
Fig. 4: Altered subcellular MHC-I localization in the absence of TAP bypasses TLR regulation of phagocytic antigen cross-presentation.
Fig. 5: Noncanonical cross-presentation in the absence of TAP is independent of TLR signal-dependent phosphorylation of phagosomal SNAP23.
Fig. 6: Noncanonical cross-presentation is dependent on Sec22b-mediated trafficking of MHC-I to phagosomes.
Fig. 7: Noncanonical cross-presentation cell-autonomously counters TAP deficiency by presenting viral- but also cellular-derived antigens.
Fig. 8: TAP deficiency in hematopoietic cells does not preclude the generation of a protective CD8+ T cell response to viral infection.

Similar content being viewed by others

Data availability

Source data and uncropped immunoblot images are provided with this paper. All other data supporting the findings of the paper are available from the corresponding author upon reasonable request.

References

  1. Blum, J. S., Wearsch, P. A. & Cresswell, P. Pathways of antigen processing. Annu. Rev. Immunol. 31, 443–473 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Neefjes, J., Jongsma, M. L., 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  Google Scholar 

  3. Blees, A. et al. Structure of the human MHC-I peptide-loading complex. Nature 551, 525–528 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Mayerhofer, P. U. & Tampe, R. Antigen translocation machineries in adaptive immunity and viral immune evasion. J. Mol. Biol. 427, 1102–1118 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Praest, P., Liaci, A. M., Forster, F. & Wiertz, E. New insights into the structure of the MHC class I peptide-loading complex and mechanisms of TAP inhibition by viral immune evasion proteins. Mol. Immunol. 113, 103–114 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Blander, J. M. Regulation of the cell biology of antigen cross-presentation. Annu. Rev. Immunol. 36, 717–753 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ackerman, A. L., Kyritsis, C., Tampe, R. & Cresswell, P. Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens. Proc. Natl Acad. Sci. USA 100, 12889–12894 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Delamarre, L., Holcombe, H. & Mellman, I. Presentation of exogenous antigens on major histocompatibility complex (MHC) class I and MHC class II molecules is differentially regulated during dendritic cell maturation. J. Exp. Med. 198, 111–122 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nair-Gupta, P. et al. TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation. Cell 158, 506–521 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Montealegre, S., Abramova, A., Manceau, V., de Kanter, A.F. & van Endert, P. The role of MHC class I recycling and Arf6 in cross-presentation by murine dendritic cells. Life Sci. Alliance 2, e201900464 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Oliveira, C. C. & van Hall, T. Alternative antigen processing for MHC class I: multiple roads lead to Rome. Front. Immunol. 6, 298 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Rock, K. L. & Shen, L. Cross-presentation: underlying mechanisms and role in immune surveillance. Immunol. Rev. 207, 166–183 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Sengupta, D., Graham, M., Liu, X. & Cresswell, P. Proteasomal degradation within endocytic organelles mediates antigen cross-presentation. EMBO J. 38, e99266 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Cruz, F. M., Colbert, J. D., Merino, E., Kriegsman, B. A. & Rock, K. L. The biology and underlying mechanisms of cross-presentation of exogenous antigens on MHC-I molecules. Annu. Rev. Immunol. 35, 149–176 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Heath, W. R. & Carbone, F. R. Cross-presentation in viral immunity and self-tolerance. Nat. Rev. Immunol. 1, 126–134 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Joffre, O. P., Segura, E., Savina, A. & Amigorena, S. Cross-presentation by dendritic cells. Nat. Rev. Immunol. 12, 557–569 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Gutiérrez-Martínez, E. et al. Cross-presentation of cell-associated antigens by MHC class I in dendritic cell subsets. Front. Immunol. 6, 363 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Helft, J. et al. Cross-presenting CD103+ dendritic cells are protected from influenza virus infection. J. Clin. Investig. 122, 4037–4047 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Silvin, A. et al. Constitutive resistance to viral infection in human CD141+ dendritic cells. Sci. Immunol. 2, eaai8071 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sigal, L. J., Crotty, S., Andino, R. & Rock, K. L. Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen. Nature 398, 77–80 (1999).

    Article  CAS  PubMed  Google Scholar 

  21. Whitney, P. G. et al. Effective priming of herpes simplex virus-specific CD8+ T cells in vivo does not require infected dendritic cells. J. Virol. 92, e01508–e01517 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Donaldson, J. G. & Williams, D. B. Intracellular assembly and trafficking of MHC class I molecules. Traffic 10, 1745–1752 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nebenfuhr, A., Ritzenthaler, C. & Robinson, D. G. Brefeldin A: deciphering an enigmatic inhibitor of secretion. Plant Physiol. 130, 1102–1108 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cebrian, I. et al. Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells. Cell 147, 1355–1368 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Hewitt, E. W. The MHC class I antigen presentation pathway: strategies for viral immune evasion. Immunology 110, 163–169 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Noriega, V., Redmann, V., Gardner, T. & Tortorella, D. Diverse immune evasion strategies by human cytomegalovirus. Immunol. Res. 54, 140–151 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Shen, L., Sigal, L. J., Boes, M. & Rock, K. L. Important role of cathepsin S in generating peptides for TAP-independent MHC class I crosspresentation in vivo. Immunity 21, 155–165 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Tiwari, N. et al. A transporter associated with antigen-processing independent vacuolar pathway for the MHC class I-mediated presentation of endogenous transmembrane proteins. J. Immunol. 178, 7932–7942 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Bertholet, S. et al. Leishmania antigens are presented to CD8+ T cells by a transporter associated with antigen processing-independent pathway in vitro and in vivo. J. Immunol. 177, 3525–3533 (2006).

  30. Merzougui, N., Kratzer, R., Saveanu, L. & van Endert, P. A proteasome-dependent, TAP-independent pathway for cross-presentation of phagocytosed antigen. EMBO Rep. 12, 1257–1264 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lawand, M., Abramova, A., Manceau, V., Springer, S. & van Endert, P. TAP-dependent and -independent peptide import into dendritic cell phagosomes. J. Immunol. 197, 3454–3463 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Blander, J. M. & Medzhitov, R. Toll-dependent selection of microbial antigens for presentation by dendritic cells. Nature 440, 808–812 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Huang, A. Y., Bruce, A. T., Pardoll, D. M. & Levitsky, H. I. In vivo cross-priming of MHC class I-restricted antigens requires the TAP transporter. Immunity 4, 349–355 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Lankat-Buttgereit, B. & Tampe, R. The transporter associated with antigen processing: function and implications in human diseases. Physiol. Rev. 82, 187–204 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Halenius, A. & Hengel, H. Human cytomegalovirus and autoimmune disease. Biomed. Res. Int. 2014, 472978 (2014).

  36. Fu, Y., Yan, G., Shi, L. & Faustman, D. Antigen processing and autoimmunity. Evaluation of mRNA abundance and function of HLA-linked genes. Ann. N. Y. Acad. Sci. 842, 138–155 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. Aldrich, C. J. et al. Positive selection of self- and alloreactive CD8+ T cells in Tap-1 mutant mice. Proc. Natl Acad. Sci. USA 91, 6525–6528 (1994).

  38. Van Kaer, L., Ashton-Rickardt, P. G., Ploegh, H. L. & Tonegawa, S. TAP1 mutant mice are deficient in antigen presentation, surface class I molecules, and CD48+ T cells. Cell 71, 1205–1214 (1992).

  39. Behar, S. M., Dascher, C. C., Grusby, M. J., Wang, C. R. & Brenner, M. B. Susceptibility of mice deficient in CD1D or TAP1 to infection with Mycobacterium tuberculosis. J. Exp. Med. 189, 1973–1980 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Alloatti, A. et al. Critical role for Sec22b-dependent antigen cross-presentation in antitumor immunity. J. Exp. Med. 214, 2231–2241 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wu, S. J. et al. A critical analysis of the role of SNARE protein SEC22B in antigen cross-presentation. Cell Rep. 19, 2645–2656 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sigal, L. J. & Rock, K. L. Bone marrow–derived antigen-presenting cells are required for the generation of cytotoxic T lymphocyte responses to viruses and use transporter associated with antigen presentation (Tap)-dependent and -independent pathways of antigen presentation. J. Exp. Med. 192, 1143–1150 (2000).

  43. Geng, J., Zaitouna, A. J. & Raghavan, M. Selected HLA-B allotypes are resistant to inhibition or deficiency of the transporter associated with antigen processing (TAP). PLoS Pathog. 14, e1007171 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Cerundolo, V. & de la Salle, H. Description of HLA class I- and CD8-deficient patients: insights into the function of cytotoxic T lymphocytes and NK cells in host defense. Semin. Immunol. 18, 330–336 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. de la Salle, H. et al. HLA class I deficiencies due to mutations in subunit 1 of the peptide transporter TAP1. J. Clin. Investig. 103, R9–R13 (1999).

    Article  PubMed  Google Scholar 

  46. Garrido, G. et al. Tumor-targeted silencing of the peptide transporter TAP induces potent antitumor immunity. Nat. Commun. 10, 3773 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Marijt, K. A., Van Der Burg, S. H. & van Hall, T. TEIPP peptides: exploration of unTAPped cancer antigens. Oncoimmunology 8, 1599639 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Marijt, K. A. & van Hall, T. To TAP or not to TAP: alternative peptides for immunotherapy of cancer. Curr. Opin. Immunol. 64, 15–19 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Uger, R. A. & Barber, B. H. Presentation of an influenza nucleoprotein epitope incorporated into the H-2Db signal sequence requires the transporter-associated with antigen presentation. J. Immunol. 158, 685–692 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Zhong, W., Reche, P. A., Lai, C. C., Reinhold, B. & Reinherz, E. L. Genome-wide characterization of a viral cytotoxic T lymphocyte epitope repertoire. J. Biol. Chem. 278, 45135–45144 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Perez, O. A. et al. CD169+ macrophages orchestrate innate immune responses by regulating bacterial localization in the spleen. Sci. Immunol. 2, eaah5520 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Suchin, E. J. et al. Quantifying the frequency of alloreactive T cells in vivo: new answers to an old question. J. Immunol. 166, 973–981 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Drutman, S. B. & Trombetta, E. S. Dendritic cells continue to capture and present antigens after maturation in vivo. J. Immunol. 185, 2140–2146 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Moltedo, B., Li, W., Yount, J. S. & Moran, T. M. Unique type I interferon responses determine the functional fate of migratory lung dendritic cells during influenza virus infection. PLoS Pathog. 7, e1002345 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Rodriguez-Madoz, J. R. et al. Inhibition of the type I interferon response in human dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex. J. Virol. 84, 9760–9774 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Noriega, V. M. et al. Human cytomegalovirus modulates monocyte-mediated innate immune responses during short-term experimental latency in vitro. J. Virol. 88, 9391–9405 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Parham, P., Barnstable, C. J. & Bodmer, W. F. Use of a monoclonal antibody (W6/32) in structural studies of HLA-A,B,C, antigens. J. Immunol. 123, 342–349 (1979).

    Article  CAS  PubMed  Google Scholar 

  58. Bolte, S. & Cordelieres, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Husebye, H. et al. The Rab11a GTPase controls Toll-like receptor 4-induced activation of interferon regulatory factor-3 on phagosomes. Immunity 33, 583–596 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Gillespie for expert pathology on mouse lung tissues. We thank S. Trombetta (Boehringer Ingelheim); K. Rock (University of Massachusetts); P. Cresswell (Yale University); and W. Li, T. M. Moran, D. B. Rubiov and A. Fernandez-Sesma (Icahn School of Medicine at Mount Sinai) for reagents and technical advice. We are grateful to current Blander laboratory members and to H. Gupta, M. A. Blander and S. J. Blander for discussions and support. The ISMMS-Microscopy Shared Resource Facility was supported by grants no. NIH 5R24 CA095823-04, no. S10 RR0 9145-01 and no. NSF DBI-9724504. This work was supported by NIH grants no. AI073899 and no. AI123284 to J.M.B.; an NIH/NIAID Center for Research on Influenza Pathogenesis contract as part of the CEIRS Network no. HHSN266200700010C to A.G.-S., grant nos. AI101820 and AI112318 to D.T., and grant no. AI143861 to K.M.K.; German Research Foundation grants no. SFB 807–Membrane Transport and Communication and no. TA157/7; and by the European Research Council (ERC Advanced Grant no. 789121) to R.T. Support to J.M.B. was also provided by NIH grants no. DK111862 and no. AI127658, the Burroughs Wellcome Fund, and the Leukemia and Lymphoma Society. G.B. has been supported by a fellowship and is currently supported by a career development award from the Crohn’s and Colitis Foundation. T.G. was supported by an American Heart Association pre-doctoral fellowship and NIH grant no. F32CA224438.

Author information

Authors and Affiliations

Authors

Contributions

G.B., P.N.-G. and J.M.B. designed experiments, directed the study and wrote the manuscript. P.N.-G. and G.B. performed most in vitro and in vivo experiments, respectively, and curated data. M.S., G.M., A.C. and A.G.-S. provided influenza A virus animal model expertise and management and conducted animal weight loss and lung viral titer measurements. F.S. and R.T. prepared and provided recombinant soluble TAP inhibitor US6. J.M. and G.B. performed immunoblots for knockdown validation. T.G. and D.T. provided reagents and methodology for HCMV infections. S.T.Y. and K.M.K. sectioned, stained and imaged lung tissues by confocal microscopy. J.M.B. supervised and conceived of the study.

Corresponding author

Correspondence to J. Magarian Blander.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Malini Raghavan, Scheherazade Sadegh-Nasseri and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey 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.

Extended data

Extended Data Fig. 1 Steady state residual expression of MHC-I at the plasma membrane of Tap–/– DC.

a, Confocal micrographs of WT or Tap−⁄− resting DCs stained for cholera toxin B subunit (CTB), H2-Kb and ERGIC-53. b, FACS analyses of WT or Tap−⁄− DCs for surface H2-Kb. Scale bars represent 10 µm. Data represent at least three independent experiments.

Extended Data Fig. 2 HCMV protein expression is detected only in DC treated with live and not UV-inactivated virus.

Confocal micrographs of human DCs stained for immediate early (IE) viral protein along with CTB, 48h post infection with either HCMVTB40/E or its UV-irradiated counterpart at a MOI=10. Scale bars represent 10 µm. Data represent at least three independent experiments.

Extended Data Fig. 3 Impaired TAP function blocks classic MHC-I presentation but not cross-presentation.

a, Classic MHC-I presentation by WT DCs of SIINFEKL derived from influenza PR/8-OTI virus to OVA-specific CD8+ T cells. DCs were infected with influenza PR/8-OT-I virus for 5h and incubated in the presence of active or inactive US6. b, c, WT and Tap−⁄− DC cross-presentation of SIINFEKL from E. coli-OVA at several time points (b), or at 4.5h post-phagocytosis of either E. coli-OVA or 5h post-infection with influenza PR/8-OTI virus (c). Data represent at least three independent experiments except for (b) depicting two independent experiments. The mean ± SEM are presented and each symbol represents biological replicate. **P < 0.01; N.S.= non statistically significant (P>0.05) using an unpaired two-tailed t-test.

Source data

Extended Data Fig. 4 Classic MHC-I presentation of viral antigen by Wt DC is not affected by Sec22b or Rab11a inhibition.

a, Immunoblots for the expression of β-actin, Sec22b and Rab11a on whole cell extracts prepared from DCs seven days post-differentiation of bone marrow progenitors that had been transduced with lentiviruses encoding for control shRNA or shRNA targeting either Rab11a or Sec22b. Immunoblots were cropped to show indicated proteins. b, WT DC progenitors were transduced with recombinant lentiviruses expressing scrambled, Sec22b or Rab11a specific shRNA. Classic MHC-I presentation of SIINFEKL at 5h after infection with recombinant SIINFEKL-expressing Influenza-OTI (PR/8-OTI) (left panels, no secondary treatment) or cross-presentation of SIINFEKL from heat-inactivated Influenza-OTI given to DCs at 3h following infection by Influenza-OTI virus. Cross-presentation was assessed 2h later. Data represent at least three independent experiments. The mean ± SEM are presented and each symbol represents biological replicate. N.S.= non statistically significant (P>0.05) using an unpaired two-tailed t-test.

Source data

Extended Data Fig. 5 Lung CD8 T cells during lethal challenge with influenza A virus.

a, Percent of influenza A specific CD8+ T cells in the lungs of WTWT and Tap/WT mice at days 3 and 5 post lethal challenge with 75 p.f.u. influenza A PR8 virus. Data show two different H-2Dd tetramers loaded with either a polymerase acidic protein epitope (SSLENFRAYV) or a nucleoprotein epitope (ASNENMETM). b, Representative FACS plot and % CD8+CD44+ cells in the lung of naïve chimeric mice. c, Flow cytometry plots showing CD8+ T cells in the lungs or spleens of WT mice on days 1 and 2 following injection with anti-CD8α antibody (clone 2.43) intranasally (i.n., 200µg), intraperitoneally (i.p., 100µg), or via both routes (i.p.+i.n.). Percent of CD3+CD8β+ T cells remaining are indicated in the blue gates. The mean ± SEM are presented and each symbol represents a mouse. N.S.= non statistically significant (P>0.05) using an unpaired two-tailed t-test.

Source data

Extended Data Fig. 6 CD8 T cells mediate protection of Tap/ chimeric mice against lethal challenge with influenza A virus.

a, Confocal micrographs at 20X magnification of lung sections from indicated anti-CD8α-treated mice stained for CD8α, CD11c, EpCAM and influenza A virus. Legend to the right shows the color code for each antibody. Scale bar represents 50 µm. b, Lung PR8 viral titers at days 3 and 5 in WTWT and Tap/WT chimeric mice. Each symbol is one mouse. c, Pathology scores of infected mice at different time points post lethal PR8 challenge in WTWT and Tap/WT mice. Slides were scored by a blinded pathologist for perivascular, bronchiolar or alveolar inflammation, epithelial degeneration or necrosis, and intraluminal debris or hemorrhage. Each symbol is one mouse. d, Hematoxylin-and-eosin staining of lung sections from WTWT and Tap/WT mice on days 3 or 5 post PR8 challenge. Scale bar represents 100 μm. The mean ± SEM are presented and each symbol represents a mouse. Data represent 2 experiments (for a total n of 140 mice).

Source data

Supplementary information

Supplementary Information

Supplementary gating strategy.

Reporting Summary

Supplementary Video 1

A schematic illustrating the cell biology of noncanonical cross-presentation based on the data presented in this study. The model phagocytic cargo shown here is a bacterium that engages TLR signaling. Based on previous work, two pathways of vesicular traffic contribute to the cross-presenting phagosome: TLR-regulated traffic from the ERC to the phagosome mediated by TLR–MyD88–IKK2-dependent phosphorylation of SNAP23 (not shown, refer to ref. 9) which delivers MHC-I molecules, and TLR-independent traffic from the ERGIC to phagosomes requiring the ER-SNARE Sec22b, which delivers the components of the peptide-loading complex, including TAP and Sec61 (refs. 9,24). Accumulation of MHC-I molecules in the ERGIC upon TAP dysfunction enables their Sec22b-dependent recruitment to phagosomes to prime CD8+ T cells through noncanonical cross-presentation.

Supplementary Video 2

3D reconstruction of confocal stacks showing CD11c+ cells with infected epithelial cell inclusions (white merge) in the lungs of WT  WT at day 3 post influenza A/PR8 challenge. Staining for CD11c is in red, influenza in green, CD8 in cyan and EpCAM in magenta.

Supplementary Video 3

3D reconstruction of confocal stacks showing CD11c+ cells with infected epithelial cell inclusions (white merge) in the lungs of Tap/ → WT at day 3 post influenza A/PR8 challenge. Staining for CD11c is in red, influenza in green, CD8 in cyan and EpCAM in magenta.

Source data

Source Data Fig. 1

Numerical data used for plots.

Source Data Fig. 2

Numerical data used for plots.

Source Data Fig. 3

Numerical data used for plots.

Source Data Fig. 4

Numerical data used for plots.

Source Data Fig. 4

Unprocessed immunoblots.

Source Data Fig. 5

Numerical data used for plots.

Source Data Fig. 6

Numerical data used for plots.

Source Data Fig. 7

Numerical data used for plots.

Source Data Fig. 8

Numerical data used for plots.

Source Data Extended Data Fig. 3

Numerical data used for plots.

Source Data Extended Data Fig. 4

Numerical data used for plots.

Source Data Extended Data Fig. 4

Unprocessed immunoblots.

Source Data Extended Data Fig. 5

Numerical data used for plots.

Source Data Extended Data Fig. 6

Numerical data used for plots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barbet, G., Nair-Gupta, P., Schotsaert, M. et al. TAP dysfunction in dendritic cells enables noncanonical cross-presentation for T cell priming. Nat Immunol 22, 497–509 (2021). https://doi.org/10.1038/s41590-021-00903-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00903-7

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