Skip to main content

Advertisement

Log in

Extracellular vesicle-mediated MHC cross-dressing in immune homeostasis, transplantation, infectious diseases, and cancer

  • Review
  • Published:
Seminars in Immunopathology Aims and scope Submit manuscript

Abstract

Eukaryotic cells employ different types of extracellular vesicles (EVs) to exchange proteins, mRNAs, non-coding regulatory RNAs, carbohydrates, and lipids. Cells of the immune system, in particular antigen (Ag)-presenting cells (APCs), acquire major histocompatibility complex (MHC) class I and II molecules loaded with antigenic peptides from leukocytes and tissue parenchymal and stromal cells, through a mechanism known as MHC cross-dressing. Increasing evidence indicates that cross-dressing of APCs with pre-formed Ag-peptide/MHC complexes (pMHCs) is mediated via passage of clusters of EVs with characteristics of exosomes. A percentage of the transferred EVs remain attached to the acceptor APCs, with the appropriate orientation, at sufficient concentration within localized areas of the plasma membrane, and for sufficient time, so the preformed pMHCs carried by the EVs are presented without further processing, to cognate T cells. Although its biological relevance is not fully understood, numerous studies have demonstrated that MHC cross-dressing of APCs represents a pathway of Ag presentation of acquired pre-formed pMHCs to T cells—alternative to direct and cross-presentation—participate in immune homeostasis and T cell tolerance, cross-regulate alloreactive T cells with different MHC restricted specificities, and is a mechanism of Ag spreading for autologous, allogeneic, microbial, tumor, or vaccine-delivered Ags. Here, we compare MHC cross-dressing with other mechanisms and terminologies used for pMHC transfer, including trogocytosis. We discuss the experimental evidence, mostly from in vitro and ex vivo studies, of the role of MHC cross-dressing of APCs via EVs in positive or negative regulation of T cell immunity in the steady state, transplantation, microbial diseases, and cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Thery C, Ostrowski M, Segura E (2009) Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9:581–593

    CAS  Google Scholar 

  2. Colombo M, Raposo G, Thery C (2014) Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 30:255–289

    CAS  Google Scholar 

  3. Robbins PD, Morelli AE (2014) Regulation of immune responses by extracellular vesicles. Nat Rev Immunol 14:195–208

    CAS  PubMed Central  Google Scholar 

  4. Nakayama M (2014) Antigen presentation by MHC-dressed cells. Front Immunol 5:672

    Google Scholar 

  5. Harshyne LA, Watkins SC, Gambotto A, Barratt-Boyes SM (2001) Dendritic cells acquire antigens from live cells for cross-presentation to CTL. J Immunol 166:3717–3723

    CAS  Google Scholar 

  6. Herrera OB, Golshayan D, Tibbott R, Salcido Ochoa F, James MJ, Marelli-Berg FM, Lechler RI (2004) A novel pathway of alloantigen presentation by dendritic cells. J Immunol 173:4828–4837

    CAS  Google Scholar 

  7. Zhang QJ, Li XL, Wang D, Huang XC, Mathis JM, Duan WM, Knight D, Shi R, Glass J, Zhang DQ, Eisenbach L, Jefferies WA (2008) Trogocytosis of MHC-I/peptide complexes derived from tumors and infected cells enhances dendritic cell cross-priming and promotes adaptive T cell responses. PLoS One 3:e3097

    PubMed Central  Google Scholar 

  8. Huang JF, Yang Y, Sepulveda H, Shi W, Hwang I, Peterson PA, Jackson MR, Sprent J, Cai Z (1999) TCR-mediated internalization of peptide-MHC complexes acquired by T cells. Science 286:952–954

    CAS  Google Scholar 

  9. Hudrisier D, Riond J, Mazarguil H, Gairin JE, Joly E (2001) Cutting edge: CTLs rapidly capture membrane fragments from target cells in a TCR signaling-dependent manner. J Immunol 166:3645–3649

    CAS  Google Scholar 

  10. Batista FD, Iber D, Neuberger MS (2001) B cells acquire antigen from target cells after synapse formation. Nature 411:489–494

    CAS  Google Scholar 

  11. Patel DM, Arnold PY, White GA, Nardella JP, Mannie MD (1999) Class II MHC/peptide complexes are released from APC and are acquired by T cell responders during specific antigen recognition. J Immunol 163:5201–5210

    CAS  Google Scholar 

  12. Wetzel SA, McKeithan TW, Parker DC (2005) Peptide-specific intercellular transfer of MHC class II to CD4+ T cells directly from the immunological synapse upon cellular dissociation. J Immunol 174:80–89

    CAS  Google Scholar 

  13. Arnold PY, Mannie MD (1999) Vesicles bearing MHC class II molecules mediate transfer of antigen from antigen-presenting cells to CD4+ T cells. Eur J Immunol 29:1363–1373

    CAS  Google Scholar 

  14. Patel DM, Mannie MD (2001) Intercellular exchange of class II major histocompatibility complex/peptide complexes is a conserved process that requires activation of T cells but is constitutive in other types of antigen presenting cell. Cell Immunol 214:165–172

    CAS  Google Scholar 

  15. Hwang I, Shen X, Sprent J (2003) Direct stimulation of naive T cells by membrane vesicles from antigen-presenting cells: distinct roles for CD54 and B7 molecules. Proc Natl Acad Sci USA 100:6670–6675

    CAS  Google Scholar 

  16. Nolte-‘t Hoen EN, Buschow SI, Anderton SM, Stoorvogel W, Wauben MH (2009) Activated T cells recruit exosomes secreted by dendritic cells via LFA-1. Blood 113:1977–1981

    Google Scholar 

  17. Muntasell A, Berger AC, Roche PA (2007) T cell-induced secretion of MHC class II-peptide complexes on B cell exosomes. EMBO J 26:4263–4272

    CAS  PubMed Central  Google Scholar 

  18. Choudhuri K, Llodra J, Roth EW, Tsai J, Gordo S, Wucherpfennig KW, Kam LC, Stokes DL, Dustin ML (2014) Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse. Nature 507:118–123

    CAS  PubMed Central  Google Scholar 

  19. Game DS, Rogers NJ, Lechler RI (2005) Acquisition of HLA-DR and costimulatory molecules by T cells from allogeneic antigen presenting cells. Am J Transplant 5:1614–1625

    CAS  Google Scholar 

  20. Hwang I, Huang JF, Kishimoto H, Brunmark A, Peterson PA, Jackson MR, Surh CD, Cai Z, Sprent J (2000) T cells can use either T cell receptor or CD28 receptors to absorb and internalize cell surface molecules derived from antigen-presenting cells. J Exp Med 191:1137–1148

    CAS  PubMed Central  Google Scholar 

  21. Tsang JY, Chai JG, Lechler R (2003) Antigen presentation by mouse CD4+ T cells involving acquired MHC class II: peptide complexes: another mechanism to limit clonal expansion? Blood 101:2704–2710

    CAS  Google Scholar 

  22. Romagnoli PA, Premenko-Lanier MF, Loria GD, Altman JD (2013) CD8 T cell memory recall is enhanced by novel direct interactions with CD4 T cells enabled by MHC class II transferred from APCs. PLoS One 8:e56999

    CAS  PubMed Central  Google Scholar 

  23. Riond J, Elhmouzi J, Hudrisier D, Gairin JE (2007) Capture of membrane components via trogocytosis occurs in vivo during both dendritic cells and target cells encounter by CD8(+) T cells. Scand J Immunol 66:441–450

    CAS  Google Scholar 

  24. Hudrisier D, Aucher A, Puaux AL, Bordier C, Joly E (2007) Capture of target cell membrane components via trogocytosis is triggered by a selected set of surface molecules on T or B cells. J Immunol 178:3637–3647

    CAS  Google Scholar 

  25. Daubeuf S, Puaux AL, Joly E, Hudrisier D (2006) A simple trogocytosis-based method to detect, quantify, characterize and purify antigen-specific live lymphocytes by flow cytometry, via their capture of membrane fragments from antigen-presenting cells. Nat Protoc 1:2536–2542

    CAS  Google Scholar 

  26. Machlenkin A, Uzana R, Frankenburg S, Eisenberg G, Eisenbach L, Pitcovski J, Gorodetsky R, Nissan A, Peretz T, Lotem M (2008) Capture of tumor cell membranes by trogocytosis facilitates detection and isolation of tumor-specific functional CTLs. Cancer Res 68:2006–2013

    CAS  Google Scholar 

  27. Uzana R, Eisenberg G, Sagi Y, Frankenburg S, Merims S, Amariglio N, Yefenof E, Peretz T, Machlenkin A, Lotem M (2012) Trogocytosis is a gateway to characterize functional diversity in melanoma-specific CD8+ T cell clones. J Immunol 188:632–640

    CAS  Google Scholar 

  28. Eisenberg G, Uzana R, Pato A, Frankenburg S, Merims S, Yefenof E, Ferrone S, Peretz T, Machlenkin A, Lotem M (2013) Imprinting of lymphocytes with melanoma antigens acquired by trogocytosis facilitates identification of tumor-reactive T cells. J Immunol 190:5856–5865

    CAS  PubMed Central  Google Scholar 

  29. Uzana R, Eisenberg G, Merims S, Frankenburg S, Pato A, Yefenof E, Engelstein R, Peretz T, Machlenkin A, Lotem M (2015) Human T cell crosstalk is induced by tumor membrane transfer. PLoS One 10:e0118244

    PubMed Central  Google Scholar 

  30. Harshyne LA, Zimmer MI, Watkins SC, Barratt-Boyes SM (2003) A role for class a scavenger receptor in dendritic cell nibbling from live cells. J Immunol 170:2302–2309

    CAS  Google Scholar 

  31. Yewdell JW, Haeryfar SM (2005) Understanding presentation of viral antigens to CD8+ T cells in vivo: the key to rational vaccine design. Annu Rev Immunol 23:651–682

    CAS  Google Scholar 

  32. Russo V, Zhou D, Sartirana C, Rovere P, Villa A, Rossini S, Traversari C, Bordignon C (2000) Acquisition of intact allogeneic human leukocyte antigen molecules by human dendritic cells. Blood 95:3473–3477

    CAS  Google Scholar 

  33. Dolan BP, Gibbs KD Jr, Ostrand-Rosenberg S (2006) Dendritic cells cross-dressed with peptide MHC class I complexes prime CD8+ T cells. J Immunol 177:6018–6024

    CAS  Google Scholar 

  34. Thacker RI, Janssen EM (2012) Cross-presentation of cell-associated antigens by mouse splenic dendritic cell populations. Front Immunol 3:41

    PubMed Central  Google Scholar 

  35. Stinchcombe JC, Bossi G, Booth S, Griffiths GM (2001) The immunological synapse of CTL contains a secretory domain and membrane bridges. Immunity 15:751–761

    CAS  Google Scholar 

  36. Onfelt B, Nedvetzki S, Yanagi K, Davis DM (2004) Cutting edge: membrane nanotubes connect immune cells. J Immunol 173:1511–1513

    Google Scholar 

  37. Davis DM (2007) Intercellular transfer of cell-surface proteins is common and can affect many stages of an immune response. Nat Rev Immunol 7:238–243

    CAS  Google Scholar 

  38. Liu Q, Rojas-Canales DM, Divito SJ, Shufesky WJ, Stolz DB, Erdos G, Sullivan ML, Gibson GA, Watkins SC, Larregina AT, Morelli AE (2016) Donor dendritic cell-derived exosomes promote allograft-targeting immune response. J Clin Invest 126:2805–2820

    PubMed Central  Google Scholar 

  39. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183:1161–1172

    CAS  Google Scholar 

  40. Segura E, Nicco C, Lombard B, Veron P, Raposo G, Batteux F, Amigorena S, Thery C (2005) ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood 106:216–223

    CAS  Google Scholar 

  41. Admyre C, Bohle B, Johansson SM, Focke-Tejkl M, Valenta R, Scheynius A, Gabrielsson S (2007) B cell-derived exosomes can present allergen peptides and activate allergen-specific T cells to proliferate and produce TH2-like cytokines. J Allergy Clin Immunol 120:1418–1424

    CAS  Google Scholar 

  42. Montecalvo A, Shufesky WJ, Stolz DB, Sullivan MG, Wang Z, Divito SJ, Papworth GD, Watkins SC, Robbins PD, Larregina AT, Morelli AE (2008) Exosomes as a short-range mechanism to spread alloantigen between dendritic cells during T cell allorecognition. J Immunol 180:3081–3090

    CAS  Google Scholar 

  43. Qazi KR, Gehrmann U, Domange Jordo E, Karlsson MC, Gabrielsson S (2009) Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood 113:2673–2683

    CAS  Google Scholar 

  44. Vincent-Schneider H, Stumptner-Cuvelette P, Lankar D, Pain S, Raposo G, Benaroch P, Bonnerot C (2002) Exosomes bearing HLA-DR1 molecules need dendritic cells to efficiently stimulate specific T cells. Int Immunol 14:713–722

    CAS  Google Scholar 

  45. Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S (2002) Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol 3:1156–1162

    CAS  Google Scholar 

  46. Hsu DH, Paz P, Villaflor G, Rivas A, Mehta-Damani A, Angevin E, Zitvogel L, Le Pecq JB (2003) Exosomes as a tumor vaccine: enhancing potency through direct loading of antigenic peptides. J Immunother 26:440–450

    CAS  Google Scholar 

  47. Kovar M, Boyman O, Shen X, Hwang I, Kohler R, Sprent J (2006) Direct stimulation of T cells by membrane vesicles from antigen-presenting cells. Proc Natl Acad Sci USA 103:11671–11676

    CAS  Google Scholar 

  48. Andre F, Chaput N, Schartz NE, Flament C, Aubert N, Bernard J, Lemonnier F, Raposo G, Escudier B, Hsu DH, Tursz T, Amigorena S, Angevin E, Zitvogel L (2004) Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol 172:2126–2136

    CAS  Google Scholar 

  49. Hao S, Bai O, Li F, Yuan J, Laferte S, Xiang J (2007) Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology 120:90–102

    CAS  PubMed Central  Google Scholar 

  50. Mallegol J, Van Niel G, Lebreton C, Lepelletier Y, Candalh C, Dugave C, Heath JK, Raposo G, Cerf-Bensussan N, Heyman M (2007) T84-intestinal epithelial exosomes bear MHC class II/peptide complexes potentiating antigen presentation by dendritic cells. Gastroenterology 132:1866–1876

    CAS  Google Scholar 

  51. Giri PK, Schorey JS (2008) Exosomes derived from M. bovis BCG infected macrophages activate antigen-specific CD4+ and CD8+ T cells in vitro and in vivo. PLoS One 3:e2461

    PubMed Central  Google Scholar 

  52. Wakim LM, Bevan MJ (2011) Cross-dressed dendritic cells drive memory CD8+ T-cell activation after viral infection. Nature 471:629–632

    CAS  PubMed Central  Google Scholar 

  53. Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Papworth GD, Zahorchak AF, Logar AJ, Wang Z, Watkins SC, Falo LD Jr, Thomson AW (2004) Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 104:3257–3266

    CAS  Google Scholar 

  54. Segura E, Guerin C, Hogg N, Amigorena S, Thery C (2007) CD8+ dendritic cells use LFA-1 to capture MHC-peptide complexes from exosomes in vivo. J Immunol 179:1489–1496

    CAS  Google Scholar 

  55. Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, Zhou Q, Sui SF (2010) Cellular internalization of exosomes occurs through phagocytosis. Traffic 11:675–687

    CAS  Google Scholar 

  56. Fitzner D, Schnaars M, van Rossum D, Krishnamoorthy G, Dibaj P, Bakhti M, Regen T, Hanisch UK, Simons M (2011) Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J Cell Sci 124:447–458

    CAS  Google Scholar 

  57. Gallegos AM, Bevan MJ (2004) Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med 200:1039–1049

    CAS  PubMed Central  Google Scholar 

  58. Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci USA 105:657–662

    CAS  Google Scholar 

  59. Millet V, Naquet P, Guinamard RR (2008) Intercellular MHC transfer between thymic epithelial and dendritic cells. Eur J Immunol 38:1257–1263

    CAS  Google Scholar 

  60. Kroger CJ, Spidale NA, Wang B, Tisch R (2017) Thymic dendritic cell subsets display distinct efficiencies and mechanisms of intercellular MHC transfer. J Immunol 198:249–256

    CAS  Google Scholar 

  61. Koble C, Kyewski B (2009) The thymic medulla: a unique microenvironment for intercellular self-antigen transfer. J Exp Med 206:1505–1513

    CAS  PubMed Central  Google Scholar 

  62. Hubert FX, Kinkel SA, Davey GM, Phipson B, Mueller SN, Liston A, Proietto AI, Cannon PZ, Forehan S, Smyth GK, Wu L, Goodnow CC, Carbone FR, Scott HS, Heath WR (2011) Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood 118:2462–2472

    CAS  Google Scholar 

  63. Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH (2006) Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 7:1092–1100

    CAS  Google Scholar 

  64. Atibalentja DF, Byersdorfer CA, Unanue ER (2009) Thymus-blood protein interactions are highly effective in negative selection and regulatory T cell induction. J Immunol 183:7909–7918

    CAS  PubMed Central  Google Scholar 

  65. Li J, Park J, Foss D, Goldschneider I (2009) Thymus-homing peripheral dendritic cells constitute two of the three major subsets of dendritic cells in the steady-state thymus. J Exp Med 206:607–622

    CAS  PubMed Central  Google Scholar 

  66. Atibalentja DF, Murphy KM, Unanue ER (2011) Functional redundancy between thymic CD8alpha+ and Sirpalpha+ conventional dendritic cells in presentation of blood-derived lysozyme by MHC class II proteins. J Immunol 186:1421–1431

    CAS  Google Scholar 

  67. Hadeiba H, Lahl K, Edalati A, Oderup C, Habtezion A, Pachynski R, Nguyen L, Ghodsi A, Adler S, Butcher EC (2012) Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity 36:438–450

    CAS  PubMed Central  Google Scholar 

  68. Dubrot J, Duraes FV, Potin L, Capotosti F, Brighouse D, Suter T, LeibundGut-Landmann S, Garbi N, Reith W, Swartz MA, Hugues S (2014) Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4(+) T cell tolerance. J Exp Med 211:1153–1166

    CAS  PubMed Central  Google Scholar 

  69. Lee RS, Grusby MJ, Glimcher LH, Winn HJ, Auchincloss H Jr (1994) Indirect recognition by helper cells can induce donor-specific cytotoxic T lymphocytes in vivo. J Exp Med 179:865–872

    CAS  Google Scholar 

  70. Wise MP, Bemelman F, Cobbold SP, Waldmann H (1998) Linked suppression of skin graft rejection can operate through indirect recognition. J Immunol 161:5813–5816

    CAS  Google Scholar 

  71. Brown K, Sacks SH, Wong W (2008) Extensive and bidirectional transfer of major histocompatibility complex class II molecules between donor and recipient cells in vivo following solid organ transplantation. FASEB J 22:3776–3784

    CAS  Google Scholar 

  72. Marino J, Babiker-Mohamed MH, Crosby-Bertorini P, Paster JT, LeGuern C, Germana S, Abdi R, Uehara M, Kin JI, Markmann JF, Tocco G, Benichou G (2016) Donor exosomes rather than passenger leukocytes initiate alloreactive T cell responses after transplantation. Sci Immunol 1:aaf8759

    PubMed Central  Google Scholar 

  73. Brown K, Sacks SH, Wong W (2011) Coexpression of donor peptide/recipient MHC complex and intact donor MHC: evidence for a link between the direct and indirect pathways. Am J Transplant 11:826–831

    CAS  Google Scholar 

  74. Sivaganesh S, Harper SJ, Conlon TM, Callaghan CJ, Saeb-Parsy K, Negus MC, Motallebzadeh R, Bolton EM, Bradley JA, Pettigrew GJ (2013) Copresentation of intact and processed MHC alloantigen by recipient dendritic cells enables delivery of linked help to alloreactive CD8 T cells by indirect-pathway CD4 T cells. J Immunol 190:5829–5838

    CAS  PubMed Central  Google Scholar 

  75. Harper SJ, Ali JM, Wlodek E, Negus MC, Harper IG, Chhabra M, Qureshi MS, Mallik M, Bolton E, Bradley JA, Pettigrew GJ (2015) CD8 T-cell recognition of acquired alloantigen promotes acute allograft rejection. Proc Natl Acad Sci USA 112:12788–12793

    CAS  Google Scholar 

  76. Smyth LA, Harker N, Turnbull W, El-Doueik H, Klavinskis L, Kioussis D, Lombardi G, Lechler R (2008) The relative efficiency of acquisition of MHC:peptide complexes and cross-presentation depends on dendritic cell type. J Immunol 181:3212–3220

    CAS  Google Scholar 

  77. Markey KA, Koyama M, Gartlan KH, Leveque L, Kuns RD, Lineburg KE, Teal BE, MacDonald KP, Hill GR (2014) Cross-dressing by donor dendritic cells after allogeneic bone marrow transplantation contributes to formation of the immunological synapse and maximizes responses to indirectly presented antigen. J Immunol 192:5426–5433

    CAS  Google Scholar 

  78. Wang X, Li H, Matte-Martone C, Cui W, Li N, Tan HS, Roopenian D, Shlomchik WD (2011) Mechanisms of antigen presentation to T cells in murine graft-versus-host disease: cross-presentation and the appearance of cross-presentation. Blood 118:6426–6437

    CAS  PubMed Central  Google Scholar 

  79. Naslund TI, Gehrmann U, Qazi KR, Karlsson MC, Gabrielsson S (2013) Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 190:2712–2719

    Google Scholar 

  80. Bedford P, Garner K, Knight SC (1999) MHC class II molecules transferred between allogeneic dendritic cells stimulate primary mixed leukocyte reactions. Int Immunol 11:1739–1744

    CAS  Google Scholar 

  81. Bedford PA, Burke F, Stagg AJ, Knight SC (2008) Dendritic cells derived from bone marrow cells fail to acquire and present major histocompatibility complex antigens from other dendritic cells. Immunology 124:542–552

    CAS  PubMed Central  Google Scholar 

  82. Snell GD (1957) The homograft reaction. Annu Rev Microbiol 11:439–458

    CAS  Google Scholar 

  83. Celli S, Albert ML, Bousso P (2011) Visualizing the innate and adaptive immune responses underlying allograft rejection by two-photon microscopy. Nat Med 17:744–749

    CAS  Google Scholar 

  84. Barker CF, Billingham RE (1968) The role of afferent lymphatics in the rejection of skin homografts. J Exp Med 128:197–221

    CAS  PubMed Central  Google Scholar 

  85. Steinmuller D (1980) Passenger leukocytes and the immunogenicity of skin allografts. J Invest Dermatol 75:107–115

    CAS  Google Scholar 

  86. McMaster PD, Hudack SS (1934) The participation of skin lymphatics in repair of the lesions due to incisions and burns. J Exp Med 60:479–501

    CAS  PubMed Central  Google Scholar 

  87. Saunderson SC, Dunn AC, Crocker PR, McLellan AD (2014) CD169 mediates the capture of exosomes in spleen and lymph node. Blood 123:208–216

    CAS  PubMed Central  Google Scholar 

  88. Smyth LA, Lechler RI, Lombardi G (2017) Continuous acquisition of MHC:peptide complexes by recipient cells contributes to the generation of anti-graft CD8(+) T cell immunity. Am J Transplant 17:60–68

    CAS  Google Scholar 

  89. Zhuang Q, Liu Q, Divito SJ, Zeng Q, Yatim KM, Hughes AD, Rojas-Canales DM, Nakao A, Shufesky WJ, Williams AL, Humar R, Hoffman RA, Shlomchik WD, Oberbarnscheidt MH, Lakkis FG, Morelli AE (2016) Graft-infiltrating host dendritic cells play a key role in organ transplant rejection. Nat Commun 7:12623

    CAS  PubMed Central  Google Scholar 

  90. Ono Y, Perez-Gutierrez A, Nakao T, Dai H, Camirand G, Yoshida O, Yokota S, Stolz DB, Ross MA, Morelli AE, Geller DA, Thomson AW (2017) Graft-infiltrating PD-L1(hi) cross-dressed dendritic cells regulate anti-donor T cell responses in mouse liver transplant tolerance. Hepatology. https://doi.org/10.1002/hep.29529

    CAS  Google Scholar 

  91. Bracamonte-Baran W, Florentin J, Zhou Y, Jankowska-Gan E, Haynes WJ, Zhong W, Brennan TV, Dutta P, Claas FH, van Rood JJ, Burlingham WJ (2017) Modification of host dendritic cells by microchimerism-derived extracellular vesicles generates split tolerance. Proc Natl Acad Sci USA 114:1099–1104

    CAS  Google Scholar 

  92. Qu C, Nguyen VA, Merad M, Randolph GJ (2009) MHC class I/peptide transfer between dendritic cells overcomes poor cross-presentation by monocyte-derived APCs that engulf dying cells. J Immunol 182:3650–3659

    CAS  PubMed Central  Google Scholar 

  93. Smyth LA, Hervouet C, Hayday T, Becker PD, Ellis R, Lechler RI, Lombardi G, Klavinskis LS (2012) Acquisition of MHC:peptide complexes by dendritic cells contributes to the generation of antiviral CD8+ T cell immunity in vivo. J Immunol 189:2274–2282

    CAS  Google Scholar 

  94. Li L, Kim S, Herndon JM, Goedegebuure P, Belt BA, Satpathy AT, Fleming TP, Hansen TH, Murphy KM, Gillanders WE (2012) Cross-dressed CD8alpha+/CD103+ dendritic cells prime CD8+ T cells following vaccination. Proc Natl Acad Sci USA 109:12716–12721

    CAS  Google Scholar 

  95. Tian T, Zhu YL, Hu FH, Wang YY, Huang NP, Xiao ZD (2013) Dynamics of exosome internalization and trafficking. J Cell Physiol 228:1487–1495

    CAS  Google Scholar 

  96. Coppieters K, Barral AM, Juedes A, Wolfe T, Rodrigo E, Thery C, Amigorena S, von Herrath MG (2009) No significant CTL cross-priming by dendritic cell-derived exosomes during murine lymphocytic choriomeningitis virus infection. J Immunol 182:2213–2220

    CAS  Google Scholar 

  97. Allan RS, Smith CM, Belz GT, van Lint AL, Wakim LM, Heath WR, Carbone FR (2003) Epidermal viral immunity induced by CD8alpha+ dendritic cells but not by Langerhans cells. Science 301:1925–1928

    CAS  Google Scholar 

  98. Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, Zhan Y, Lew AM, Shortman K, Heath WR, Carbone FR (2006) Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 25:153–162

    CAS  Google Scholar 

  99. Oliphant CJ, Hwang YY, Walker JA, Salimi M, Wong SH, Brewer JM, Englezakis A, Barlow JL, Hams E, Scanlon ST, Ogg GS, Fallon PG, McKenzie AN (2014) MHCII-mediated dialog between group 2 innate lymphoid cells and CD4(+) T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity 41:283–295

    CAS  PubMed Central  Google Scholar 

  100. Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, Angevin E, Amigorena S, Zitvogel L (2001) Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med 7:297–303

    CAS  Google Scholar 

  101. Andre F, Schartz NE, Movassagh M, Flament C, Pautier P, Morice P, Pomel C, Lhomme C, Escudier B, Le Chevalier T, Tursz T, Amigorena S, Raposo G, Angevin E, Zitvogel L (2002) Malignant effusions and immunogenic tumour-derived exosomes. Lancet 360:295–305

    CAS  PubMed Central  Google Scholar 

  102. Chaput N, Schartz NE, Andre F, Taieb J, Novault S, Bonnaventure P, Aubert N, Bernard J, Lemonnier F, Merad M, Adema G, Adams M, Ferrantini M, Carpentier AF, Escudier B, Tursz T, Angevin E, Zitvogel L (2004) Exosomes as potent cell-free peptide-based vaccine. II. Exosomes in CpG adjuvants efficiently prime naive Tc1 lymphocytes leading to tumor rejection. J Immunol 172:2137–2146

    CAS  Google Scholar 

  103. Hao S, Bai O, Yuan J, Qureshi M, Xiang J (2006) Dendritic cell-derived exosomes stimulate stronger CD8+ CTL responses and antitumor immunity than tumor cell-derived exosomes. Cell Mol Immunol 3:205–211

    CAS  Google Scholar 

  104. Bonaccorsi I, Morandi B, Antsiferova O, Costa G, Oliveri D, Conte R, Pezzino G, Vermiglio G, Anastasi GP, Navarra G, Munz C, Di Carlo E, Mingari MC, Ferlazzo G (2014) Membrane transfer from tumor cells overcomes deficient phagocytic ability of plasmacytoid dendritic cells for the acquisition and presentation of tumor antigens. J Immunol 192:824–832

    CAS  Google Scholar 

  105. Josien R, Li HL, Ingulli E, Sarma S, Wong BR, Vologodskaia M, Steinman RM, Choi Y (2000) TRANCE, a tumor necrosis factor family member, enhances the longevity and adjuvant properties of dendritic cells in vivo. J Exp Med 191:495–502

    CAS  PubMed Central  Google Scholar 

  106. Barratt-Boyes SM, Zimmer MI, Harshyne LA, Meyer EM, Watkins SC, Capuano S 3rd, Murphey-Corb M, Falo LD Jr, Donnenberg AD (2000) Maturation and trafficking of monocyte-derived dendritic cells in monkeys: implications for dendritic cell-based vaccines. J Immunol 164:2487–2495

    CAS  Google Scholar 

  107. Smith AL, Fazekas de St Groth B (1999) Antigen-pulsed CD8alpha+ dendritic cells generate an immune response after subcutaneous injection without homing to the draining lymph node. J Exp Med 189:593–598

    CAS  PubMed Central  Google Scholar 

  108. Kleindienst P, Brocker T (2003) Endogenous dendritic cells are required for amplification of T cell responses induced by dendritic cell vaccines in vivo. J Immunol 170:2817–2823

    CAS  Google Scholar 

Download references

Acknowledgments

Supported by the National Institutes of Health grant R01 HL130191 (to A.E.M.). Furong Zeng is a Research Fellow of the Third Xiangya Hospital of Central South University (Changsha, Hunan, China).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Adrian E. Morelli.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Additional information

This article is a contribution to the special issue on Extracellular Vesicles -- Guest Editor: Esther Nolte-‘t Hoen

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zeng, F., Morelli, A.E. Extracellular vesicle-mediated MHC cross-dressing in immune homeostasis, transplantation, infectious diseases, and cancer. Semin Immunopathol 40, 477–490 (2018). https://doi.org/10.1007/s00281-018-0679-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00281-018-0679-8

Keywords

Navigation