Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nat Immunol ; 18(5): 509-518, 2017 05.
Article in English | MEDLINE | ID: mdl-28319098

ABSTRACT

The retention of intracellular Toll-like receptors (TLRs) in the endoplasmic reticulum prevents their activation under basal conditions. TLR9 is activated by sensing ligands in specific endosomal-lysosomal compartments. Here we identified IRAP+ endosomes as major cellular compartments for the early steps of TLR9 activation in dendritic cells (DCs). Both TLR9 and its ligand, the dinucleotide CpG, were present as cargo in IRAP+ endosomes. In the absence of the aminopeptidase IRAP, the trafficking of CpG and TLR9 to lysosomes and signaling via TLR9 were enhanced in DCs and in mice following bacterial infection. IRAP stabilized CpG-containing endosomes by interacting with the actin-nucleation factor FHOD4, which slowed the trafficking of TLR9 toward lysosomes. Thus, endosomal retention of TLR9 via the interaction of IRAP with the actin cytoskeleton is a mechanism that prevents hyper-activation of TLR9 in DCs.


Subject(s)
Cystinyl Aminopeptidase/metabolism , Cytoskeleton/metabolism , Dendritic Cells/physiology , Endosomes/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Toll-Like Receptor 9/metabolism , Animals , Cells, Cultured , CpG Islands/genetics , Cystinyl Aminopeptidase/genetics , Dendritic Cells/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Oligodeoxyribonucleotides/immunology , Protein Binding , Signal Transduction
2.
Immunity ; 43(2): 304-17, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26253786

ABSTRACT

Antimicrobial peptides (AMPs) expressed by epithelial and immune cells are largely described for the defense against invading microorganisms. Recently, their immunomodulatory functions have been highlighted in various contexts. However how AMPs expressed by non-immune cells might influence autoimmune responses in peripheral tissues, such as the pancreas, is unknown. Here, we found that insulin-secreting ß-cells produced the cathelicidin related antimicrobial peptide (CRAMP) and that this production was defective in non-obese diabetic (NOD) mice. CRAMP administrated to prediabetic NOD mice induced regulatory immune cells in the pancreatic islets, dampening the incidence of autoimmune diabetes. Additional investigation revealed that the production of CRAMP by ß-cells was controlled by short-chain fatty acids produced by the gut microbiota. Accordingly, gut microbiota manipulations in NOD mice modulated CRAMP production and inflammation in the pancreatic islets, revealing that the gut microbiota directly shape the pancreatic immune environment and autoimmune diabetes development.


Subject(s)
Cathelicidins/metabolism , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Intestines/immunology , Microbiota/physiology , Pancreas/immunology , Animals , Antimicrobial Cationic Peptides , Cathelicidins/genetics , Diabetes Mellitus, Type 1/microbiology , Fatty Acids, Volatile/immunology , Female , Intestines/microbiology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Pancreas/microbiology
3.
Immunol Rev ; 291(1): 57-74, 2019 09.
Article in English | MEDLINE | ID: mdl-31402505

ABSTRACT

T-lymphocyte activation relies on the cognate recognition by the TCR of the MHC-associated peptide ligand (pMHC) presented at the surface of an antigen-presenting cell (APC). This leads to the dynamic formation of a cognate contact between the T lymphocyte and the APC: the immune synapse (IS). Engagement of the TCR by the pMHC in the synaptic zone induces a cascade of signaling events leading to phosphorylation and dephosphorylation of proteins and lipids, which ultimately shapes the response of T lymphocytes. Although the engagement of the T-cell receptor (TCR) takes place at the plasma membrane, the TCR/CD3 complexes and the signaling molecules involved in transduction of the TCR signal are also present in intracellular membrane pools. These pools, which are both endocytic and exocytic, have tentatively been characterized by several groups including ours. We will herein summarize what is known on the intracellular pools of TCR signaling components. We will discuss their origin and the mechanisms involved in their mobility at the IS. Finally, we will propose several hypotheses concerning the functional role(s) that these intracellular pools might play in T-cell activation. We will also discuss the tools that could be used to test these hypotheses.


Subject(s)
Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Endocytosis , Endosomes/metabolism , Humans , Intracellular Space/metabolism , Ligands , Lipid Metabolism , Phosphorylation , Protein Transport
4.
Eur J Immunol ; 45(12): 3257-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26399368

ABSTRACT

The immunodominant MART-1(26(27)-35) epitope, liberated from the differentiation antigen melanoma antigen recognized by T cells/melanoma antigen A (MART-1/Melan-A), has been frequently targeted in melanoma immunotherapy, but with limited clinical success. Previous studies suggested that this is in part due to an insufficient peptide supply and epitope presentation, since proteasomes containing the immunosubunits ß5i/LMP7 (LMP, low molecular weight protein) or ß1i/LMP2 and ß5i/LMP7 interfere with MART-1(26-35) epitope generation in tumor cells. Here, we demonstrate that in addition the IFN-γ-inducible proteasome subunit ß2i/MECL-1 (multicatalytic endopeptidase complex-like 1), proteasome activator 28 (PA28), and ER-resident aminopeptidase 1 (ERAP1) impair MART-1(26-35) epitope generation. ß2i/MECL-1 and PA28 negatively affect C- and N-terminal cleavage and therefore epitope liberation from the proteasome, whereas ERAP1 destroys the MART-1(26-35) epitope by overtrimming activity. Constitutive expression of PA28 and ERAP1 in melanoma cells indicate that both interfere with MART-1(26-35) epitope generation even in the absence of IFN-γ. In summary, our results provide first evidence that activities of different antigen-processing components contribute to an inefficient MART-1(26-35) epitope presentation, suggesting the tumor cell's proteolytic machinery might have an important impact on the outcome of epitope-specific immunotherapies.


Subject(s)
Aminopeptidases/physiology , Epitopes/immunology , Melanoma/immunology , Muscle Proteins/physiology , Neoplasm Proteins/immunology , Proteasome Endopeptidase Complex/physiology , T-Lymphocytes/immunology , Cell Line, Tumor , Cysteine Endopeptidases/physiology , Humans , Minor Histocompatibility Antigens
5.
J Immunol ; 193(2): 901-8, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24928998

ABSTRACT

The endoplasmic reticulum aminopeptidases (ERAP)1 and ERAP2 play a critical role in the production of final epitopes presented by MHC class I molecules. Formation of heterodimers by ERAP1 and ERAP2 has been proposed to facilitate trimming of epitope precursor peptides, but the effects of dimerization on ERAP function remain unknown. In this study, we produced stabilized ERAP1-ERAP2 heterodimers and found that they produced several mature epitopes more efficiently than a mix of the two enzymes unable to dimerize. Physical interaction with ERAP2 changes basic enzymatic parameters of ERAP1 and improves its substrate-binding affinity. Thus, by bringing the two enzymes in proximity and by producing allosteric effects on ERAP1, dimerization of ERAP1/2 creates complexes with superior peptide-trimming efficacy. Such complexes are likely to enhance Ag presentation by cells displaying coordinated expression of the two enzymes.


Subject(s)
Aminopeptidases/immunology , Epitopes/immunology , Peptides/immunology , Protein Multimerization , Amino Acid Sequence , Aminopeptidases/genetics , Aminopeptidases/metabolism , Animals , Antigen Presentation/immunology , Cells, Cultured , Chromatography, High Pressure Liquid , Epitopes/metabolism , HeLa Cells , Humans , Immunoblotting , Minor Histocompatibility Antigens , Molecular Sequence Data , Peptides/metabolism , Protein Binding/immunology , Sf9 Cells , Substrate Specificity
6.
J Immunol ; 188(4): 1840-6, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22238454

ABSTRACT

Dendritic cells (DCs) use cellular pathways collectively referred to as cross-presentation to stimulate CD8(+) T cells with peptide Ags derived from internalized, exogenous Ags. We have recently reported that DCs rely on aminoterminal trimming of cross-presented peptides by insulin-responsive aminopeptidase (IRAP), an enzyme localized in a regulated endosomal storage compartment. Considering a report contending that this role is limited to inflammatory DCs (Segura et al. 2009. Proc. Natl. Acad. Sci. USA 106: 20377-20381), in this study, we examined the role of IRAP in steady-state DC subpopulations. Steady-state conventional DCs (cDCs) and plasmacytoid DCs expressed similar amounts of IRAP. IRAP colocalized with the endosomal markers Rab14 and syntaxin 6, both known to be associated with regulated endosomal storage compartments, in CD8(+) and CD8(-) cDCs-however, to a greater extent in the former population. Likewise, IRAP recruitment to phagosomes was significantly stronger in CD8(+) DCs. IRAP deficiency compromised cross-presentation of soluble and particulate Ag by both CD8(+) and CD8(-) cDCs, again with a stronger effect in the former population. Thus, the requirement of IRAP in cross-presentation extends to steady-state cDCs. Moreover, these data suggest that increased recruitment of an IRAP(+)/Rab14(+) compartment to Ag-containing vesicles contributes to the superior cross-presentation efficacy of CD8(+) cDCs.


Subject(s)
Cross-Priming , Cystinyl Aminopeptidase/metabolism , Dendritic Cells/immunology , Endosomes/immunology , rab GTP-Binding Proteins/metabolism , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cystinyl Aminopeptidase/biosynthesis , Dendritic Cells/metabolism , Endosomes/metabolism , Mice , Mice, Knockout , Phagosomes/immunology , Phagosomes/metabolism , Qa-SNARE Proteins/metabolism
7.
Sci Transl Med ; 16(740): eadl6149, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38536935

ABSTRACT

Mechanisms underlying the disruption of self-tolerance in acquired autoimmunity remain unclear. Immunoglobulin A (IgA) nephropathy is an acquired autoimmune disease where deglycosylated IgA1 (IgA subclass 1) auto-antigens are recognized by IgG auto-antibodies, forming immune complexes that are deposited in the kidneys, leading to glomerulonephritis. In the intestinal microbiota of patients with IgA nephropathy, there was increased relative abundance of mucin-degrading bacteria, including Akkermansia muciniphila. IgA1 was deglycosylated by A. muciniphila both in vitro and in the intestinal lumen of mice. This generated neo-epitopes that were recognized by autoreactive IgG from the sera of patients with IgA nephropathy. Mice expressing human IgA1 and the human Fc α receptor I (α1KI-CD89tg) that underwent intestinal colonization by A. muciniphila developed an aggravated IgA nephropathy phenotype. After deglycosylation of IgA1 by A. muciniphila in the mouse gut lumen, IgA1 crossed the intestinal epithelium into the circulation by retrotranscytosis and became deposited in the glomeruli of mouse kidneys. Human α-defensins-a risk locus for IgA nephropathy-inhibited growth of A. muciniphila in vitro. A negative correlation observed between stool concentration of α-defensin 6 and quantity of A. muciniphila in the guts of control participants was lost in patients with IgA nephropathy. This study demonstrates that gut microbiota dysbiosis contributes to generation of auto-antigens in patients with IgA nephropathy and in a mouse model of this disease.


Subject(s)
Gastrointestinal Microbiome , Glomerulonephritis, IGA , Humans , Mice , Animals , Immunoglobulin A , Glomerulonephritis, IGA/genetics , Kidney , Immunoglobulin G
8.
Nat Commun ; 15(1): 3389, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649353

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by anti-nuclear autoantibodies whose production is promoted by autoreactive T follicular helper (TFH) cells. During SLE pathogenesis, basophils accumulate in secondary lymphoid organs (SLO), amplify autoantibody production and disease progression through mechanisms that remain to be defined. Here, we provide evidence for a direct functional relationship between TFH cells and basophils during lupus pathogenesis, both in humans and mice. PD-L1 upregulation on basophils and IL-4 production are associated with TFH and TFH2 cell expansions and with disease activity. Pathogenic TFH cell accumulation, maintenance, and function in SLO were dependent on PD-L1 and IL-4 in basophils, which induced a transcriptional program allowing TFH2 cell differentiation and function. Our study establishes a direct mechanistic link between basophils and TFH cells in SLE that promotes autoantibody production and lupus nephritis.


Subject(s)
B7-H1 Antigen , Basophils , Interleukin-4 , Lupus Erythematosus, Systemic , T Follicular Helper Cells , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Autoantibodies/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Basophils/immunology , Basophils/metabolism , Cell Differentiation/immunology , Interleukin-4/metabolism , Interleukin-4/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lupus Nephritis/metabolism , Mice, Inbred C57BL , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
9.
EMBO Rep ; 12(12): 1257-64, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22037009

ABSTRACT

Major histocompatibility complex (MHC) class I cross-presentation is thought to involve two pathways, one of which depends on both the TAP transporters and the proteasome and the other on neither. We found that preincubation of TAP-deficient dendritic cells at low temperature increases the density of MHC class I at the surface and fully restores cross-presentation of phagocytosed antigen, but not of soluble antigen internalized through receptors. Restoration of cross-presentation by TAP-deficient cells requires antigen degradation by the proteasome. Thus, TAP might mainly be required for recycling cell surface class I molecules during cross-presentation of phagocytosed antigens. Furthermore, phagosomes-but not endosomes-seem to have a TAP-independent mechanism to import peptides generated by cytosolic proteasome complexes.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Cross-Priming/immunology , Phagocytosis/immunology , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/immunology , ATP-Binding Cassette Transporters/metabolism , Animals , Antigen Presentation/drug effects , Cross-Priming/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Endocytosis/drug effects , Endocytosis/immunology , Histocompatibility Antigens Class I/immunology , Mice , Models, Immunological , Phagocytosis/drug effects , Protease Inhibitors/pharmacology , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Solubility/drug effects , Temperature
10.
Bioorg Med Chem Lett ; 23(17): 4832-6, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23916253

ABSTRACT

Endoplasmic reticulum aminopeptidases, ERAP1 and ERAP2, as well as Insulin regulated aminopeptidase (IRAP) play key roles in antigen processing, and have recently emerged as biologically important targets for manipulation of antigen presentation. Taking advantage of the available structural and substrate-selectivity data for these enzymes, we have rationally designed a new series of inhibitors that display low micromolar activity. The selectivity profile for these three highly homologous aminopeptidases provides a promising avenue for modulating intracellular antigen processing.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Cystinyl Aminopeptidase/antagonists & inhibitors , Endoplasmic Reticulum/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Drug Design , Humans , Models, Molecular
11.
Curr Opin Immunol ; 83: 102337, 2023 08.
Article in English | MEDLINE | ID: mdl-37216842

ABSTRACT

Antigen (Ag)-trimming aminopeptidases belong to the oxytocinase subfamily of M1 metallopeptidases. In humans, this subfamily contains the endoplasmic reticulum aminopeptidases 1 and 2 (ERAP1 and 2) and the insulin-responsive aminopeptidase (IRAP, synonym oxytocinase), an endosomal enzyme. The ability of these enzymes to trim antigenic precursors and to generate major histocompatibility class-I ligands has been demonstrated extensively for ERAP1, less for ERAP2, which is absent in rodents, and exclusively in the context of cross-presentation for IRAP. During 20 years of research on these aminopeptidases, their enzymatic function has been very well characterized and their genetic association with autoimmune diseases, cancers, and infections is well established. The mechanisms by which these proteins are associated to human diseases are not always clear. This review discusses the Ag-trimming-independent functions of the oxytocinase subfamily of M1 aminopeptidases and the new questions raised by recent publications on IRAP and ERAP2.


Subject(s)
Aminopeptidases , Cystinyl Aminopeptidase , Humans , Aminopeptidases/genetics , Aminopeptidases/metabolism , Cystinyl Aminopeptidase/genetics , Antigens , Minor Histocompatibility Antigens/genetics
12.
iScience ; 26(7): 107055, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37360697

ABSTRACT

Cell surface receptor internalization can either terminate signaling or activate alternative endosomal signaling pathways. We investigated here whether endosomal signaling is involved in the function of the human receptors for Fc immunoglobulin fragments (FcRs): FcαRI, FcγRIIA, and FcγRI. All these receptors were internalized after their cross-linking with receptor-specific antibodies, but their intracellular trafficking was different. FcαRI was targeted directly to lysosomes, while FcγRIIA and FcγRI were internalized in particular endosomal compartments described by the insulin esponsive minoeptidase (IRAP), where they recruited signaling molecules, such as the active form of the kinase Syk, PLCγ and the adaptor LAT. Destabilization of FcγR endosomal signaling in the absence of IRAP compromised cytokine secretion downstream FcγR activation and macrophage ability to kill tumor cells by antibody-dependent cell-mediated cytotoxicity (ADCC). Our results indicate that FcγR endosomal signaling is required for the FcγR-driven inflammatory reaction and possibly for the therapeutic action of monoclonal antibodies.

13.
Biomed J ; 45(2): 310-320, 2022 04.
Article in English | MEDLINE | ID: mdl-34592497

ABSTRACT

Antigen T cell receptors (TCR) recognize antigenic peptides displayed by the major histocompatibility complex (pMHC) and play a critical role in T cell activation. The levels of TCR complexes at the cell surface, where signaling is initiated, depend on the balance between TCR synthesis, recycling and degradation. Cell surface TCR interaction with pMHC leads to receptor clustering and formation of a tight T cell-APC contact, the immune synapse, from which the activated TCR is internalized. While TCR internalization from the immune synapse has been initially considered to arrest TCR signaling, recent evidence support the hypothesis that the internalized receptor continues to signal from specialized endosomes. Here, we review the molecular mechanisms of TCR endocytosis and recycling, both in steady state and after T cell activation. We then discuss the experimental evidence in favor of endosomal TCR signaling and its possible consequences on T cell activation.


Subject(s)
Lymphocyte Activation , Receptors, Antigen, T-Cell , Antigens , Endocytosis , Humans , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes
14.
Front Immunol ; 13: 1029759, 2022.
Article in English | MEDLINE | ID: mdl-36389775

ABSTRACT

The function of intracellular trafficking in immune-complex triggered inflammation remains poorly understood. Here, we investigated the role of Insulin-Regulated Amino Peptidase (IRAP)-positive endosomal compartments in Fc receptor (FcR)-induced inflammation. Less severe FcγR-triggered arthritis, active systemic anaphylaxis and FcεRI-triggered passive systemic anaphylaxis were observed in IRAP-deficient versus wild-type mice. In mast cells FcεRI stimulation induced rapid plasma membrane recruitment of IRAP-positive endosomes. IRAP-deficient cells exhibited reduced secretory responses, calcium signaling and activating SykY519/520 phosphorylation albeit receptor tyrosine phosphorylation on ß and γ subunits was not different. By contrast, in the absence of IRAP, SHP1-inactivating phosphorylation on Ser591 that controls Syk activity was decreased. Ex-vivo cell profiling after FcγR-triggered anaphylaxis confirmed decreased phosphorylation of both SykY519/520 and SHP-1S591 in IRAP-deficient neutrophils and monocytes. Thus, IRAP-positive endosomal compartments, in promoting inhibition of SHP-1 during FcR signaling, control the extent of phosphorylation events at the plasma membrane and contribute to setting the intensity of immune-complex triggered inflammatory diseases.


Subject(s)
Anaphylaxis , Insulin , Animals , Mice , Insulin/pharmacology , Aminopeptidases/metabolism , Cystinyl Aminopeptidase , Receptors, Fc , Receptors, IgG/genetics , Receptors, IgE , Antigen-Antibody Complex , Inflammation
16.
Mucosal Immunol ; 14(4): 949-962, 2021 07.
Article in English | MEDLINE | ID: mdl-33846534

ABSTRACT

Respiratory Syncytial Virus (RSV) is the major cause of lower respiratory tract infection in infants, in whom, the sensing of RSV by innate immune receptors and its regulation are still poorly described. However, the severe bronchiolitis following RSV infection in neonates has been associated with a defect in type I interferons (IFN-I) production, a cytokine produced mainly by alveolar macrophages (AMs) upon RSV infection in adults. In the present study, neonatal C57BL/6 AMs mobilized very weakly the IFN-I pathway upon RSV infection in vitro and failed to restrain virus replication. However, IFN-I productions by neonatal AMs were substantially increased by the deletion of Insulin-Responsive AminoPeptidase (IRAP), a protein previously involved in the regulation of IFN-I production by dendritic cells. Moreover, neonatal IRAPKO AMs showed a higher expression of IFN-stimulated genes than their wild-type C57BL/6 counterpart. Interestingly, depletion of IRAP did not affect adult AM responses. Finally, we demonstrated that newborn IRAPKO mice infected with RSV had more IFN-I in their lungs and eliminated the virus more efficiently than WT neonates. Taken together, early-life susceptibility to RSV infection may be related to an original age-dependent suppressive function of IRAP on the IFN-I driven-antiviral responses in neonatal AMs.


Subject(s)
Cystinyl Aminopeptidase/metabolism , Interferon Type I/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses , Animals , Animals, Newborn , Disease Models, Animal , Host-Pathogen Interactions/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Syncytial Virus Infections/virology , Signal Transduction , Toll-Like Receptors/metabolism , Virus Replication
17.
Nature ; 425(6956): 397-402, 2003 Sep 25.
Article in English | MEDLINE | ID: mdl-14508489

ABSTRACT

Induction of cytotoxic T-cell immunity requires the phagocytosis of pathogens, virus-infected or dead tumour cells by dendritic cells. Peptides derived from phagocytosed antigens are then presented to CD8+ T lymphocytes on major histocompatibility complex (MHC) class I molecules, a process called "cross-presentation". After phagocytosis, antigens are exported into the cytosol and degraded by the proteasome. The resulting peptides are thought to be translocated into the lumen of the endoplasmic reticulum (ER) by specific transporters associated with antigen presentation (TAP), and loaded onto MHC class I molecules by a complex "loading machinery" (which includes tapasin, calreticulin and Erp57). Here we show that soon after or during formation, phagosomes fuse with the ER. After antigen export to the cytosol and degradation by the proteasome, peptides are translocated by TAP into the lumen of the same phagosomes, before loading on phagosomal MHC class I molecules. Therefore, cross-presentation in dendritic cells occurs in a specialized, self-sufficient, ER-phagosome mix compartment.


Subject(s)
Antigen Presentation , Dendritic Cells/cytology , Dendritic Cells/immunology , Endoplasmic Reticulum/metabolism , Histocompatibility Antigens Class I/immunology , Membrane Fusion , Phagosomes/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antigens/immunology , Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cysteine Endopeptidases/metabolism , Cytosol/metabolism , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Endoplasmic Reticulum/ultrastructure , Histocompatibility Antigens Class I/metabolism , Mice , Multienzyme Complexes/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , Phagocytosis , Phagosomes/immunology , Phagosomes/ultrastructure , Proteasome Endopeptidase Complex , Protein Transport
18.
Front Cell Dev Biol ; 8: 585713, 2020.
Article in English | MEDLINE | ID: mdl-33425891

ABSTRACT

Dendritic cells (DCs) contribute to the immune surveillance by sampling their environment through phagocytosis and endocytosis. We have previously reported that, rapidly following uptake of extracellular antigen into phagosomes or endosomes in DCs, a specialized population of storage endosomes marked by Rab14 and insulin-regulated aminopeptidase (IRAP) is recruited to the nascent antigen-containing compartment, thereby regulating its maturation and ultimately antigen cross-presentation to CD8+ T lymphocytes. Here, using IRAP-/- DCs, we explored how IRAP modulates phagosome maturation dynamics and cross-presentation. We find that in the absence of IRAP, phagosomes acquire more rapidly late endosomal markers, are more degradative, and show increased microbicidal activity. We also report evidence for a role of vesicle trafficking from the endoplasmic reticulum (ER)-Golgi intermediate compartment to endosomes for the formation or stability of the IRAP compartment. Moreover, we dissect the dual role of IRAP as a trimming peptidase and a critical constituent of endosome stability. Experiments using a protease-dead IRAP mutant and pharmacological IRAP inhibition suggest that IRAP expression but not proteolytic activity is required for the formation of storage endosomes and for DC-typical phagosome maturation, whereas proteolysis is required for fully efficient cross-presentation. These findings identify IRAP as a key factor in cross-presentation, trimming peptides to fit the major histocompatibility complex class-I binding site while preventing their destruction through premature phagosome maturation.

19.
Front Mol Biosci ; 7: 583556, 2020.
Article in English | MEDLINE | ID: mdl-33195428

ABSTRACT

Insulin regulated aminopeptidase (IRAP) is a type II transmembrane protein with broad tissue distribution initially identified as a major component of Glut4 storage vesicles (GSV) in adipocytes. Despite its almost ubiquitous expression, IRAP had been extensively studied mainly in insulin responsive cells, such as adipocytes and muscle cells. In these cells, the enzyme displays a complex intracellular trafficking pattern regulated by insulin. Early studies using fusion proteins joining the IRAP cytosolic domain to various reporter proteins, such as GFP or the transferrin receptor (TfR), showed that the complex and regulated trafficking of the protein depends on its cytosolic domain. This domain contains several motifs involved in IRAP trafficking, as demonstrated by mutagenesis studies. Also, proteomic studies and yeast two-hybrid experiments showed that the IRAP cytosolic domain engages in multiple protein interactions with cytoskeleton components and vesicular trafficking adaptors. These findings led to the hypothesis that IRAP is not only a cargo of GSV but might be a part of the sorting machinery that controls GSV dynamics. Recent work in adipocytes, immune cells, and neurons confirmed this hypothesis and demonstrated that IRAP has a dual function. Its carboxy-terminal domain located inside endosomes is responsible for the aminopeptidase activity of the enzyme, while its amino-terminal domain located in the cytosol functions as an endosomal trafficking adaptor. In this review, we recapitulate the published protein interactions of IRAP and summarize the increasing body of evidence indicating that IRAP plays a role in intracellular trafficking of several proteins. We describe the impact of IRAP deletion or depletion on endocytic trafficking and the consequences on immune cell functions. These include the ability of dendritic cells to cross-present antigens and prime adaptive immune responses, as well as the control of innate and adaptive immune receptor signaling and modulation of inflammatory responses.

20.
Nat Commun ; 11(1): 2779, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32487999

ABSTRACT

T cell receptor (TCR) activation is modulated by mechanisms such as TCR endocytosis, which is thought to terminate TCR signalling. Here we show that, upon internalization, TCR continues to signal from a set of specialized endosomes that are crucial for T cell functions. Mechanistically, TCR ligation leads to clathrin-mediated internalization of the TCR-CD3ζ complex, while maintaining CD3ζ signalling, in endosomal vesicles that contain the insulin responsive aminopeptidase (IRAP) and the SNARE protein Syntaxin 6. Destabilization of this compartment through IRAP deletion enhances plasma membrane expression of the TCR-CD3ζ complex, yet compromises overall CD3ζ signalling; moreover, the integrity of this compartment is also crucial for T cell activation and survival after suboptimal TCR activation, as mice engineered with a T cell-specific deletion of IRAP fail to develop efficient polyclonal anti-tumour responses. Our results thus reveal a previously unappreciated function of IRAP-dependent endosomal TCR signalling in T cell activation.


Subject(s)
Cystinyl Aminopeptidase/metabolism , Endosomes/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism , Animals , Cell Membrane/metabolism , Cell Proliferation , Clathrin/metabolism , Cystinyl Aminopeptidase/genetics , Disease Models, Animal , Endocytosis/physiology , HEK293 Cells , Humans , Interleukin-2/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Qa-SNARE Proteins/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL