Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Cell Rep ; 36(2): 109375, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260912

ABSTRACT

The mechanism of T cell antigen receptor (TCR-CD3) signaling remains elusive. Here, we identify mutations in the transmembrane region of TCRß or CD3ζ that augment peptide T cell antigen receptor (pMHC)-induced signaling not explicable by enhanced ligand binding, lateral diffusion, clustering, or co-receptor function. Using a biochemical assay and molecular dynamics simulation, we demonstrate that the gain-of-function mutations loosen the interaction between TCRαß and CD3ζ. Similar to the activating mutations, pMHC binding reduces TCRαß cohesion with CD3ζ. This event occurs prior to CD3ζ phosphorylation and at 0°C. Moreover, we demonstrate that soluble monovalent pMHC alone induces signaling and reduces TCRαß cohesion with CD3ζ in membrane-bound or solubilised TCR-CD3. Our data provide compelling evidence that pMHC binding suffices to activate allosteric changes propagating from TCRαß to the CD3 subunits, reconfiguring interchain transmembrane region interactions. These dynamic modifications could change the arrangement of TCR-CD3 boundary lipids to license CD3ζ phosphorylation and initiate signal propagation.


Subject(s)
Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Allosteric Regulation , Gain of Function Mutation/genetics , HEK293 Cells , Humans , Ligands , Major Histocompatibility Complex , Phosphorylation , Protein Multimerization , Protein Structure, Quaternary , Solubility
3.
Blood ; 137(15): 2033-2045, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33513601

ABSTRACT

Exocytosis of cytotoxic granules (CG) by lymphocytes is required for the elimination of infected and malignant cells. Impairments in this process underly a group of diseases with dramatic hyperferritinemic inflammation termed hemophagocytic lymphohistiocytosis (HLH). Although genetic and functional studies of HLH have identified proteins controlling distinct steps of CG exocytosis, the molecular mechanisms that spatiotemporally coordinate CG release remain partially elusive. We studied a patient exhibiting characteristic clinical features of HLH associated with markedly impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell exocytosis functions, who beared biallelic deleterious mutations in the gene encoding the small GTPase RhoG. Experimental ablation of RHOG in a model cell line and primary CTLs from healthy individuals uncovered a hitherto unappreciated role of RhoG in retaining CGs in the vicinity of the plasma membrane (PM), a fundamental prerequisite for CG exocytotic release. We discovered that RhoG engages in a protein-protein interaction with Munc13-4, an exocytosis protein essential for CG fusion with the PM. We show that this interaction is critical for docking of Munc13-4+ CGs to the PM and subsequent membrane fusion and release of CG content. Thus, our study illuminates RhoG as a novel essential regulator of human lymphocyte cytotoxicity and provides the molecular pathomechanism behind the identified here and previously unreported genetically determined form of HLH.


Subject(s)
Killer Cells, Natural/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , T-Lymphocytes, Cytotoxic/pathology , rho GTP-Binding Proteins/genetics , Cell Line , Cells, Cultured , Gene Deletion , Germ-Line Mutation , Humans , Infant , Killer Cells, Natural/metabolism , Lymphohistiocytosis, Hemophagocytic/pathology , Male , Models, Molecular , T-Lymphocytes, Cytotoxic/metabolism , rho GTP-Binding Proteins/chemistry
4.
Front Immunol ; 11: 1154, 2020.
Article in English | MEDLINE | ID: mdl-32582217

ABSTRACT

Autosomal recessive mutations in genes required for cytotoxicity are causative of a life-threatening, early-onset hyperinflammatory syndrome termed familial hemophagocytic lymphohistiocytosis (FHL). Mutations in UNC13D cause FHL type 3. UNC13D encodes Munc13-4, a member of the Unc13 protein family which control SNARE complex formation and vesicle fusion. We have previously identified FHL3-associated mutations in the first intron of UNC13D which control transcription from an alternative transcriptional start site. Using isoform specific antibodies, we demonstrate that this alternative Munc13-4 isoform with a unique N-terminus is preferentially expressed in human lymphocytes and platelets, as compared to the conventional isoform that was mostly expressed in monocytes and neutrophils. The distinct N-terminal of the two isoforms did not impact on Munc13-4 localization or trafficking to the immunological synapse of cytotoxic T cells. Moreover, ectopic expression of both isoforms efficiently restored exocytosis by FHL3 patient-derived Munc13-4 deficient T cells. Thus, we demonstrate that the conventional and alternative Munc13-4 isoforms have different expression pattern in hematopoietic cell subsets, but display similar localization and contribution to T cell exocytosis. The use of an alternative transcriptional starting site (TSS) in lymphocytes and platelets could be selected for increasing the overall levels of Munc13-4 expression for efficient secretory granule release.


Subject(s)
Blood Platelets/metabolism , Lymphocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Blood Platelets/immunology , Cells, Cultured , Humans , Lymphocytes/immunology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/metabolism , Mutation , Promoter Regions, Genetic , Protein Isoforms/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
5.
Front Immunol ; 10: 1855, 2019.
Article in English | MEDLINE | ID: mdl-31447853

ABSTRACT

Cytotoxic T lymphocytes kill infected or malignant cells through the directed release of cytotoxic substances at the site of target cell contact, the immunological synapse. While genetic association studies of genes predisposing to early-onset life-threatening hemophagocytic lymphohistiocytosis has identified components of the plasma membrane fusion machinery, the identity of the vesicular components remain enigmatic. Here, we identify VAMP7 as an essential component of the vesicular fusion machinery of primary, human T cells. VAMP7 co-localizes with granule markers throughout all stages of T cell maturation and simultaneously fuses with granule markers at the IS. Knock-down of VAMP7 expression significantly decreased the killing efficiency of T cells, without diminishing early T cell receptor signaling. VAMP7 exerts its function in a SNARE complex with Syntaxin11 and SNAP-23 on the plasma membrane. The identification of the minimal fusion machinery in T cells provides a starting point for the development of potential drugs in immunotherapy.


Subject(s)
Cell Degranulation/immunology , Cytoplasmic Granules/immunology , R-SNARE Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Cells, Cultured , Cytoplasmic Granules/metabolism , Humans , Immunological Synapses/immunology , Immunological Synapses/metabolism , R-SNARE Proteins/metabolism , Secretory Vesicles/immunology , Secretory Vesicles/metabolism , T-Lymphocytes, Cytotoxic/metabolism
6.
J Cell Sci ; 130(6): 1110-1121, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28154159

ABSTRACT

Sustained signalling at the immune synapse (IS) requires the synaptic delivery of recycling endosome-associated T cell antigen receptors (TCRs). IFT20, a component of the intraflagellar transport system, controls TCR recycling to the IS as a complex with IFT57 and IFT88. Here, we used quantitative mass spectrometry to identify additional interaction partners of IFT20 in Jurkat T cells. In addition to IFT57 and IFT88, the analysis revealed new binding partners, including IFT54 (also known as TRAF3IP1), GMAP-210 (also known as TRIP11), Arp2/3 complex subunit-3 (ARPC3), COP9 signalosome subunit-1 (CSN1, also known as GPS1) and ERGIC-53 (also known as LMAN1). A direct interaction between IFT20 and both IFT54 and GMAP-210 was confirmed in pulldown assays. Confocal imaging of antigen-specific conjugates using T cells depleted of these proteins by RNA interference showed that TCR accumulation and phosphotyrosine signalling at the IS were impaired in the absence of IFT54, ARPC3 or ERGIC-53. Similar to in IFT20-deficient T cells, this defect resulted from a reduced ability of endosomal TCRs to polarize to the IS despite a correct translocation of the centrosome towards the antigen-presenting cell contact. Our data underscore the traffic-related role of an IFT20 complex that includes components of the intracellular trafficking machinery in IS assembly.


Subject(s)
Carrier Proteins/metabolism , Immunological Synapses/metabolism , Protein Interaction Maps , T-Lymphocytes/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Cytoskeletal Proteins , Endocytosis , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Mannose-Binding Lectins/metabolism , Mass Spectrometry , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Nuclear Proteins/metabolism , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Receptors, Transferrin/metabolism
7.
Proc Natl Acad Sci U S A ; 113(2): 386-91, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26715756

ABSTRACT

Biogenesis of the immune synapse at the interface between antigen-presenting cells and T cells assembles and organizes a large number of membrane proteins required for effective signaling through the T-cell receptor. We showed previously that the intraflagellar transport protein 20 (IFT20), a component of the intraflagellar transport system, controls polarized traffic during immune synapse assembly. To investigate the role of IFT20 in primary CD4(+) T cells in vitro and in vivo, we generated mice bearing a conditional defect of IFT20 expression in T cells. We show that in the absence of IFT20, although cell spreading and the polarization of the centrosome were unaffected, T-cell receptor (TCR)-mediated signaling and recruitment of the signaling adaptor LAT (linker for activation of T cells) at the immune synapse were reduced. As a consequence, CD4(+) T-cell activation and proliferation were also defective. In vivo, conditional IFT20-deficient mice failed to mount effective antigen-specific T-cell responses, and their T cells failed to induce colitis after adoptive transfer to Rag(-/-) mice. IFT20 is therefore required for the delivery of the intracellular pool of LAT to the immune synapse in naive primary T lymphocytes and for effective T-cell responses in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Immunological Synapses/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Gene Knockdown Techniques , Humans , Immunoblotting , Jurkat Cells , Mice , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymocytes/metabolism
8.
J Cell Sci ; 128(14): 2541-52, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26034069

ABSTRACT

IFT20, a component of the intraflagellar transport (IFT) system that controls ciliogenesis, regulates immune synapse assembly in the non-ciliated T-cell by promoting T-cell receptor (TCR) recycling. Here, we have addressed the role of Rab8 (for which there are two isoforms Rab8a and Rab8b), a small GTPase implicated in ciliogenesis, in TCR traffic to the immune synapse. We show that Rab8, which colocalizes with IFT20 in Rab11(+) endosomes, is required for TCR recycling. Interestingly, as opposed to in IFT20-deficient T-cells, TCR(+) endosomes polarized normally beneath the immune synapse membrane in the presence of dominant-negative Rab8, but were unable to undergo the final docking or fusion step. This could be accounted for by the inability of the vesicular (v)-SNARE VAMP-3 to cluster at the immune synapse in the absence of functional Rab8, which is responsible for its recruitment. Of note, and similar to in T-cells, VAMP-3 interacts with Rab8 at the base of the cilium in NIH-3T3 cells, where it regulates ciliary growth and targeting of the protein smoothened. The results identify Rab8 as a new player in vesicular traffic to the immune synapse and provide insight into the pathways co-opted by different cell types for immune synapse assembly and ciliogenesis.


Subject(s)
Immunological Synapses/metabolism , Receptors, Antigen, T-Cell/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Endosomes/genetics , Endosomes/metabolism , Humans , Immunological Synapses/genetics , Jurkat Cells , Mice , NIH 3T3 Cells , Receptors, Antigen, T-Cell/genetics , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/genetics , rab GTP-Binding Proteins/genetics
9.
J Cell Sci ; 127(Pt 9): 1924-37, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24554435

ABSTRACT

T cell activation requires sustained signaling at the immune synapse, a specialized interface with the antigen-presenting cell (APC) that assembles following T cell antigen receptor (TCR) engagement by major histocompatibility complex (MHC)-bound peptide. Central to sustained signaling is the continuous recruitment of TCRs to the immune synapse. These TCRs are partly mobilized from an endosomal pool by polarized recycling. We have identified IFT20, a component of the intraflagellar transport (IFT) system that controls ciliogenesis, as a central regulator of TCR recycling to the immune synapse. Here, we have investigated the interplay of IFT20 with the Rab GTPase network that controls recycling. We found that IFT20 forms a complex with Rab5 and the TCR on early endosomes. IFT20 knockdown (IFT20KD) resulted in a block in the recycling pathway, leading to a build-up of recycling TCRs in Rab5(+) endosomes. Recycling of the transferrin receptor (TfR), but not of CXCR4, was disrupted by IFT20 deficiency. The IFT components IFT52 and IFT57 were found to act together with IFT20 to regulate TCR and TfR recycling. The results provide novel insights into the mechanisms that control TCR recycling and immune synapse assembly, and underscore the trafficking-related function of the IFT system beyond ciliogenesis.


Subject(s)
Synapses/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antigen-Presenting Cells/metabolism , Biological Transport/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Jurkat Cells , Microscopy, Fluorescence , Protein Binding/genetics , Protein Transport/genetics , Protein Transport/physiology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...