T Lymphocyte–Antigen Presenting Cell Immune Synapse and exosome secretion: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Immunology

Exosomes are extracellular vesicles (EV) of endosomal origin (multivesicular bodies, MVB) constitutively released by many different eukaryotic cells by fusion of MVB to the plasma membrane. However, inducible exosome secretion controlled by cell surface receptors is restricted to very few cell types and a limited number of cell surface receptors. Among these, exosome secretion is induced in T lymphocytes and B lymphocytes when stimulated at the immune synapse (IS) via T-cell receptor (TCR) and B-cell receptor (BCR), respectively. IS formation by T and B lymphocytes constitutes a crucial event involved in antigen-specific, cellular and humoral immune responses. Upon IS formation by T and B lymphocytes with antigen-presenting cells (APC) the convergence of MVB towards the microtubule organization center (MTOC), and MTOC polarization to the IS, are involved in polarized exosome secretion at the synaptic cleft. This specialized mechanism provides the immune system with a finely-tuned strategy to increase the specificity and efficiency of crucial secretory effector functions of B and T lymphocytes. Since inducible exosome secretion by antigen-receptors is a critical and unique feature of the immune system this review considers the study of the traffic events leading to polarized exosome secretion at the IS and some of their biological consequences.

  • exosomes
  • T lymphocytes
  • B lymphocytes
  • polarized secretion
  • immune synapse
  • T-cell receptor
  • B-cell receptor
  • multivesicular bodies
  • diacylglycerol
  • MHC-class II compartment

T Lymphocyte–Antigen Presenting Cell Immune Synapse: Exosome Secretion by T Lymphocytes

To constitute an IS, T lymphocytes must recognize processed antigenic peptides loaded onto MHC molecules present on the cell surface of professional antigen-presenting cells (APC) or pathogen-infected cells. TCR interaction with peptide-MHC-I complexes (pMHC-I) induces naïve CD8+ cytotoxic T lymphocytes (CTL) activation (priming), whereas TCR interaction with peptide-MHC-II complexes (pMHC-II) leads to CD4+ Th lymphocyte activation (Figure 2) [61]. Primed CTL form IS with target cells resulting in a specific killing. In addition, mature IS formation can induce T lymphocyte anergy or activation-induced apoptosis (AICD)[62].

CTL-Target Cell Immune Synapse

CTL-target cell IS induces the rapid polarization (from seconds to few minutes) of CTL MTOC and lytic granules (secretory granules or secretory lysosomes-SL-with MVB structure) towards the central supramolecular activation cluster (cSMAC) at the IS (Figure 2). Lytic granules fusion with the plasma membrane (degranulation) induces the secretion of certain cytotoxic factors such as perforin and granzymes to the synaptic cleft, triggering target cell apoptosis [63]. Upon degranulation, FasL located at the secretory granule limiting membrane becomes exposed to the plasma membrane at the IS and induces target cell Fas crosslinking leading to target cell apoptosis [58,64,65,66].
 
Another consequence of degranulation is ILV secretion as nanosize EV at the CTL-target cell synaptic cleft, first described by Peters et al. [17]. Although the vesicles secreted by CTL were not referred at that time as exosomes, their formation and mode of exocytosis justifies such a nomenclature [6,17].
Subsequent publications demonstrated that T lymphocyte stimulation of T lymphoblasts (including CD4+ and CD8+ lymphocytes) with activation agonists produced non-directional secretion of nanosize EV (quoted as microvesicles) carrying pro-apoptotic FasL and Apo2L [11] via MVB-mediated degranulation [12], providing an alternative mechanism of TCR-controlled AICD that does not necessarily imply cell-to-cell contact [12,67]. Moreover, it was shown that upon TCR triggering T lymphoblasts secrete exosomes [67,68] containing TCR/CD3 [68], extending the early observations obtained in CTL forming synapses [17]. CTL MTOC reorientation is initially guided by a diacylglycerol (DAG) gradient centered at the IS [69], generated by TCR-stimulated phospholipase C (PLC). DAG phosphorylation by diacylglycerol kinase α(DGKα) is involved in the spatiotemporal control of the DAG gradient [70,71] and MTOC polarization to the IS in CTL [69]. DAG activates, among others, several members of the PKC and PKD families [72], such as PKCδ, which is necessary for the polarization of lytic granules and cytotoxicity in mouse CTL [73,74].

Th Immune Synapse

Polarized secretion upon Th IS formation has been less studied than polarized CTL secretion [75]. Th IS are more stable and longer (from minutes up to several hours) than CTL IS (few minutes) [30,53]. Th IS are required for both directional and continuous cytokine secretion [30,53]. These cytokines are contained in secretory vesicles and IL-2, IFN-γ-containing secretory vesicles undergo polarized traffic to the F-actin poor area at cSMAC [49,53,54,76] as CTL lytic granules. Although the identity of the cytokine-containing secretory vesicles has not been characterized yet [75,77] they, most probably, are not MVB [77] (Figure 2).
 
Early reports in CD4+ Jurkat cells and T lymphoblasts demonstrated that stimulation with activation agonists [11] or anti-TCR [67,68] induced exosome secretion. Stimulation with a heterologous receptor agonist, that mimics TCR-derived signals leading to full T cell activation [78] and AICD [79], also induced exosome secretion in CD4+ Jurkat cells [67], suggesting that exosome secretion is a general consequence of T lymphocyte activation. IS formation by CD4+ Jurkat cells and superantigen-coated Raji B cells acting as an APC, which constitutes a well-established IS model [61,80,81], induces polarized MVB traffic towards the IS, MVB degranulation and exosome release [15,18] (Figure 2, right side panel). In this Th-APC IS model, a positive role of TCR-triggered DAG and its regulator DGKα [70], in polarized MVB traffic towards the IS was demonstrated [15,67,82]. As DGKα also controls late endosomes polarized traffic during invasive migration [83], and MTOC and lytic granules polarized traffic in CTL (described above), DAG and DGKα can be considered as general regulators of polarized traffic [1]. DAG-activated PKCδ is needed for cortical actin reorganization at the IS, MTOC and MVB polarization to the IS and exosome secretion in this IS model [84]. Overall, this leads us to hypothesize that an altered actin reorganization at the IS may underlie the deficient MVB polarization occurring in PKCδ-interfered T cell clones [84]. In this model, DAG-activated PKD1/2 regulates MVB maturation and polarization leading to exosome secretion [59], suggesting that several regulatory points in exosome secretion are controlled by DAG.
 
Microvesicles or ectosomes budding from the Th cell plasma membrane and accumulating at the IS have been described [85] (Figure 2, right side panel). These shedding vesicles were enriched in TCR and capable to trigger B-lymphocyte signaling via pMHC-II stimulation [85,86]. Thus, it appears that distinct types of EV from Th lymphocytes are secreted at the IS. Further research will be necessary to establish whether these subtypes of EV trigger different Th effector responses or, on the contrary, redundantly or synergistically trigger the same responses.
 
Fig. 2
  • Calvo, V.; Izquierdo, M. Imaging Polarized Secretory Traffic at the Immune Synapse in Living T Lymphocytes. Front. Immunol. 2018, 9, 684. [Google Scholar] [CrossRef] [PubMed]
  • Friedl, P.; den Boer, A.T.; Gunzer, M. Tuning immune responses: diversity and adaptation of the immunological synapse. Nat. Rev. Immunol. 2005, 5, 532–545. [Google Scholar] [CrossRef] [PubMed]
  • Peters, P.J.; Borst, J.; Oorschot, V.; Fukuda, M.; Krahenbuhl, O.; Tschopp, J.; Slot, J.W.; Geuze, H.J. Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J. Exp. Med. 1991, 173, 1099–1109. [Google Scholar] [CrossRef] [PubMed]
  • Vignaux, F.; Vivier, E.; Malissen, B.; Depraetere, V.; Nagata, S.; Golstein, P. TCR/CD3 coupling to Fas-based cytotoxicity. J. Exp. Med. 1995, 181, 781–786. [Google Scholar] [CrossRef] [PubMed]
  • Bossi, G.; Griffiths, G.M. Degranulation plays an essential part in regulating cell surface expression of Fas ligand in T cells and natural killer cells. Nat. Med. 1999, 5, 90–96. [Google Scholar] [CrossRef]
  • Nagata, S. Apoptosis by death factor. Cell 1997, 88, 355–365. [Google Scholar] [CrossRef]
  • Alonso, R.; Rodriguez, M.C.; Pindado, J.; Merino, E.; Merida, I.; Izquierdo, M. Diacylglycerol kinase alpha regulates the secretion of lethal exosomes bearing Fas ligand during activation-induced cell death of T lymphocytes. J. Biol. Chem. 2005, 280, 28439–28450. [Google Scholar] [CrossRef]
  • Blanchard, N.; Lankar, D.; Faure, F.; Regnault, A.; Dumont, C.; Raposo, G.; Hivroz, C. TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/zeta complex. J. Immunol. 2002, 168, 3235–3241. [Google Scholar] [CrossRef]
  • Quann, E.J.; Merino, E.; Furuta, T.; Huse, M. Localized diacylglycerol drives the polarization of the microtubule-organizing center in T cells. Nat. Immunol. 2009, 10, 627–635. [Google Scholar] [CrossRef]
  • Sanjuan, M.A.; Jones, D.R.; Izquierdo, M.; Merida, I. Role of diacylglycerol kinase alpha in the attenuation of receptor signaling. J. Cell Biol. 2001, 153, 207–220. [Google Scholar] [CrossRef]
  • Chauveau, A.; Le Floc’h, A.; Bantilan, N.S.; Koretzky, G.A.; Huse, M. Diacylglycerol kinase alpha establishes T cell polarity by shaping diacylglycerol accumulation at the immunological synapse. Sci. Signal. 2014, 7, ra82. [Google Scholar] [CrossRef] [PubMed]
  • Carrasco, S.; Merida, I. Diacylglycerol, when simplicity becomes complex. Trends Biochem. Sci. 2007, 32, 27–36. [Google Scholar] [CrossRef] [PubMed]
  • Ma, J.S.; Monu, N.; Shen, D.T.; Mecklenbrauker, I.; Radoja, N.; Haydar, T.F.; Leitges, M.; Frey, A.B.; Vukmanovic, S.; Radoja, S. Protein kinase Cdelta regulates antigen receptor-induced lytic granule polarization in mouse CD8+ CTL. J. Immunol. 2007, 178, 7814–7821. [Google Scholar] [CrossRef] [PubMed]
  • Ma, J.S.; Haydar, T.F.; Radoja, S. Protein kinase C delta localizes to secretory lysosomes in CD8+ CTL and directly mediates TCR signals leading to granule exocytosis-mediated cytotoxicity. J. Immunol. 2008, 181, 4716–4722. [Google Scholar] [CrossRef] [PubMed]
  • Jolly, C.; Sattentau, Q.J. Regulated secretion from CD4+ T cells. Trends Immunol. 2007, 28, 474–481. [Google Scholar] [CrossRef]
  • Huse, M.; Lillemeier, B.F.; Kuhns, M.S.; Chen, D.S.; Davis, M.M. T cells use two directionally distinct pathways for cytokine secretion. Nat. Immunol. 2006, 7, 247–255. [Google Scholar] [CrossRef]
  • Huse, M.; Quann, E.J.; Davis, M.M. Shouts, whispers and the kiss of death: directional secretion in T cells. Nat. Immunol. 2008, 9, 1105–1111. [Google Scholar] [CrossRef]
  • Desai, D.M.; Newton, M.E.; Kadlecek, T.; Weiss, A. Stimulation of the phosphatidylinositol pathway can induce T-cell activation. Nature 1990, 348, 66–69. [Google Scholar] [CrossRef]
  • Izquierdo, M.; Ruiz-Ruiz, M.C.; Lopez-Rivas, A. Stimulation of phosphatidylinositol turnover is a key event for Fas-dependent, activation-induced apoptosis in human T lymphocytes. J. Immunol. 1996, 157, 21–28. [Google Scholar]
  • Montoya, M.C.; Sancho, D.; Bonello, G.; Collette, Y.; Langlet, C.; He, H.T.; Aparicio, P.; Alcover, A.; Olive, D.; Sanchez-Madrid, F. Role of ICAM-3 in the initial interaction of T lymphocytes and APCs. Nat. Immunol. 2002, 3, 159–168. [Google Scholar] [CrossRef]
  • Bello-Gamboa, A.; Izquierdo, J.M.; Velasco, M.; Moreno, S.; Garrido, A.; Meyers, L.; Palomino, J.C.; Calvo, V.; Izquierdo, M. Imaging the Human Immunological Synapse. J. Vis. Exp. 2019. [Google Scholar] [CrossRef] [PubMed]
  • Alonso, R.; Mazzeo, C.; Merida, I.; Izquierdo, M. A new role of diacylglycerol kinase alpha on the secretion of lethal exosomes bearing Fas ligand during activation-induced cell death of T lymphocytes. Biochimie 2007, 89, 213–221. [Google Scholar] [CrossRef] [PubMed]
  • Rainero, E.; Caswell, P.T.; Muller, P.A.; Grindlay, J.; McCaffrey, M.W.; Zhang, Q.; Wakelam, M.J.; Vousden, K.H.; Graziani, A.; Norman, J.C. Diacylglycerol kinase alpha controls RCP-dependent integrin trafficking to promote invasive migration. J. Cell Biol. 2012, 196, 277–295. [Google Scholar] [CrossRef] [PubMed]
  • Herranz, G.; Aguilera, P.; Davila, S.; Sanchez, A.; Stancu, B.; Gomez, J.; Fernandez-Moreno, D.; de Martin, R.; Quintanilla, M.; Fernandez, T.; et al. Protein Kinase C delta Regulates the Depletion of Actin at the Immunological Synapse Required for Polarized Exosome Secretion by T Cells. Front. Immunol. 2019, 10, 851. [Google Scholar] [CrossRef]
  • Choudhuri, K.; Llodra, J.; Roth, E.W.; Tsai, J.; Gordo, S.; Wucherpfennig, K.W.; Kam, L.C.; Stokes, D.L.; Dustin, M.L. Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse. Nature 2014, 507, 118–123. [Google Scholar] [CrossRef]
  • Saliba, D.G.; Cespedes-Donoso, P.F.; Balint, S.; Compeer, E.B.; Korobchevskaya, K.; Valvo, S.; Mayya, V.; Kvalvaag, A.; Peng, Y.; Dong, T.; et al. Composition and structure of synaptic ectosomes exporting antigen receptor linked to functional CD40 ligand from helper T cells. Elife 2019. [Google Scholar] [CrossRef]

This entry is adapted from the peer-reviewed paper 10.3390/ijms21072631

References

  1. R Alonso; Carla Mazzeo; M C Rodriguez; Mark Marsh; Alberto Fraile-Ramos; V Calvo; Antonia Avila-Flores; Isabel Mérida; Manuel Izquierdo; Diacylglycerol kinase α regulates the formation and polarisation of mature multivesicular bodies involved in the secretion of Fas ligand-containing exosomes in T lymphocytes. Cell Death & Differentiation 2011, 18, 1161-1173, 10.1038/cdd.2010.184.
More
This entry is offline, you can click here to edit this entry!