Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 592
Filter
1.
Annu Rev Immunol ; 31: 675-704, 2013.
Article in English | MEDLINE | ID: mdl-23330955

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) control many important aspects of immune cell development, differentiation, and function. Mammals have eight PI3K catalytic subunits that are divided into three classes based on similarities in structure and function. Specific roles for the class I PI3Ks have been broadly investigated and are relatively well understood, as is the function of their corresponding phosphatases. More recently, specific roles for the class II and class III PI3Ks have emerged. Through vertebrate evolution and in parallel with the evolution of adaptive immunity, there has been a dramatic increase not only in the genes for PI3K subunits but also in genes for phosphatases that act on 3-phosphoinositides and in 3-phosphoinositide-binding proteins. Our understanding of the PI3Ks in immunity is guided by fundamental discoveries made in simpler model organisms as well as by appreciating new adaptations of this signaling module in mammals in general and in immune cells in particular.


Subject(s)
Multigene Family/immunology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology , Animals , Catalytic Domain/immunology , Endocytosis/immunology , Humans , Multiprotein Complexes/immunology , Phagocytosis/immunology , Phosphatidylinositol 3-Kinases/classification , Protein Transport/immunology
2.
Nat Immunol ; 17(5): 531-7, 2016 May.
Article in English | MEDLINE | ID: mdl-27043408

ABSTRACT

The antigen-presenting molecule MR1 presents vitamin B-related antigens (VitB antigens) to mucosal-associated invariant T (MAIT) cells through an uncharacterized pathway. We show that MR1, unlike other antigen-presenting molecules, does not constitutively present self-ligands. In the steady state it accumulates in a ligand-receptive conformation within the endoplasmic reticulum. VitB antigens reach this location and form a Schiff base with MR1, triggering a 'molecular switch' that allows MR1-VitB antigen complexes to traffic to the plasma membrane. These complexes are endocytosed with kinetics independent of the affinity of the MR1-ligand interaction and are degraded intracellularly, although some MR1 molecules acquire new ligands during passage through endosomes and recycle back to the surface. MR1 antigen presentation is characterized by a rapid 'off-on-off' mechanism that is strictly dependent on antigen availability.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Histocompatibility Antigens Class I/immunology , Signal Transduction/immunology , Antigens/metabolism , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Endocytosis/immunology , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endosomes/immunology , Endosomes/metabolism , HeLa Cells , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immunoblotting , Intracellular Space/immunology , Intracellular Space/metabolism , Microscopy, Confocal , Minor Histocompatibility Antigens , Protein Binding/immunology , Protein Transport/immunology , Vitamin B Complex/immunology
3.
Annu Rev Cell Dev Biol ; 30: 169-206, 2014.
Article in English | MEDLINE | ID: mdl-25150009

ABSTRACT

The trans-Golgi network (TGN) is an important cargo sorting station within the cell where newly synthesized proteins are packaged into distinct transport carriers that are targeted to various destinations. To maintain the fidelity of protein transport, elaborate protein sorting machinery is employed to mediate sorting of specific cargo proteins into distinct transport carriers. Protein sorting requires assembly of the cytosolic sorting machinery onto the TGN membrane and capture of cargo proteins. We review the cytosolic and transmembrane sorting machinery that function at the TGN and describe molecular interactions and regulatory mechanisms that enable accurate protein sorting. In addition, we highlight the importance of TGN sorting in physiology and disease.


Subject(s)
Protein Transport/physiology , trans-Golgi Network/physiology , ADP-Ribosylation Factor 1/physiology , Adaptor Proteins, Vesicular Transport/physiology , Amino Acid Motifs , Animals , Carrier Proteins/physiology , Cell Polarity , Cytosol/physiology , Humans , Membrane Lipids/physiology , Membrane Transport Proteins/physiology , Models, Biological , Models, Molecular , Phospholipids/physiology , Protein Conformation , Protein Sorting Signals/physiology , Protein Transport/immunology , Structure-Activity Relationship , Transport Vesicles/physiology , Vesicular Transport Proteins/physiology , trans-Golgi Network/immunology
4.
Immunity ; 48(5): 911-922.e7, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768176

ABSTRACT

Unc-93 homolog B1 (UNC93B1) is a key regulator of nucleic acid (NA)-sensing Toll-like receptors (TLRs). Loss of NA-sensing TLR responses in UNC93B1-deficient patients facilitates Herpes simplex virus type 1 (HSV-1) encephalitis. UNC93B1 is thought to guide NA-sensing TLRs from the endoplasmic reticulum (ER) to their respective endosomal signaling compartments and to guide the flagellin receptor TLR5 to the cell surface, raising the question of how UNC93B1 mediates differential TLR trafficking. Here, we report that UNC93B1 regulates a step upstream of the differential TLR trafficking process. We discovered that UNC93B1 deficiency resulted in near-complete loss of TLR3 and TLR7 proteins in primary splenic mouse dendritic cells and macrophages, showing that UNC93B1 is critical for maintaining TLR expression. Notably, expression of an ER-retained UNC93B1 version was sufficient to stabilize TLRs and largely restore endosomal TLR trafficking and activity. These data are critical for an understanding of how UNC93B1 can regulate the function of a broad subset of TLRs.


Subject(s)
Endosomes/immunology , Membrane Transport Proteins/immunology , Molecular Chaperones/immunology , Toll-Like Receptors/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Stability , Protein Transport/immunology , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
5.
Nat Immunol ; 14(1): 19-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23238760

ABSTRACT

Cytosolic detection of pathogen-derived nucleic acids is critical for the initiation of innate immune defense against diverse bacterial, viral and eukaryotic pathogens. Conversely, inappropriate responses to cytosolic nucleic acids can produce severe autoimmune pathology. The host protein STING has been identified as a central signaling molecule in the innate immune response to cytosolic nucleic acids. STING seems to be especially critical for responses to cytosolic DNA and the unique bacterial nucleic acids called 'cyclic dinucleotides'. Here we discuss advances in the understanding of STING and highlight the many unresolved issues in the field.


Subject(s)
Autoimmune Diseases/immunology , Bacterial Infections/immunology , Cytosol/immunology , DNA, Bacterial/immunology , Membrane Proteins/immunology , Nucleotides, Cyclic/immunology , Animals , Autoimmune Diseases/etiology , Bacterial Infections/complications , Host-Pathogen Interactions , Humans , Immunity, Innate , Protein Transport/immunology , Signal Transduction
6.
Immunity ; 43(5): 909-22, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26546281

ABSTRACT

Microbe-induced receptor trafficking has emerged as an essential means to promote innate immune signal transduction. Upon detection of bacterial lipopolysaccharides (LPS), CD14 induces an inflammatory endocytosis pathway that delivers Toll-like receptor 4 (TLR4) to endosomes. Although several regulators of CD14-dependent TLR4 endocytosis have been identified, the cargo-selection mechanism during this process remains unknown. We reveal that, in contrast to classic cytosolic interactions that promoted the endocytosis of transmembrane receptors, TLR4 was selected as cargo for inflammatory endocytosis entirely through extracellular interactions. Mechanistically, the extracellular protein MD-2 bound to and dimerized TLR4 in order to promote this endocytic event. Our analysis of LPS variants from human pathogens and gut commensals revealed a common mechanism by which bacteria prevent inflammatory endocytosis. We suggest that evasion of CD14-dependent endocytosis is an attribute that transcends the concept of pathogenesis and might be a fundamental feature of bacteria that inhabit eukaryotic hosts.


Subject(s)
Bacteria/immunology , Endocytosis/immunology , Immune Evasion/immunology , Lipopolysaccharide Receptors/metabolism , Toll-Like Receptor 4/metabolism , Cells, Cultured , Humans , Inflammation/immunology , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/immunology , Protein Transport/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology
7.
Nat Immunol ; 12(11): 1119-26, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964606

ABSTRACT

Binding of antigen to the B cell antigen receptor (BCR) triggers both BCR signaling and endocytosis. How endocytosis regulates BCR signaling remains unknown. Here we report that BCR signaling was not extinguished by endocytosis of BCRs; instead, BCR signaling initiated at the plasma membrane continued as the BCR trafficked intracellularly with the sequential phosphorylation of kinases. Blocking the endocytosis of BCRs resulted in the recruitment of both proximal and downstream kinases to the plasma membrane, where mitogen-activated protein kinases (MAPKs) were hyperphosphorylated and the kinase Akt and its downstream target Foxo were hypophosphorylated, which led to the dysregulation of gene transcription controlled by these pathways. Thus, the cellular location of the BCR serves to compartmentalize kinase activation to regulate the outcome of signaling.


Subject(s)
B-Lymphocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Compartmentation/immunology , Cells, Cultured , Endocytosis/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/immunology , Protein Transport/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Transcriptional Activation/immunology
8.
Nat Immunol ; 12(11): 1105-12, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964608

ABSTRACT

Protein kinase C-θ (PKC-θ) translocates to the center of the immunological synapse, but the underlying mechanism and its importance in T cell activation are unknown. Here we found that the V3 domain of PKC-θ was necessary and sufficient for localization to the immunological synapse mediated by association with the coreceptor CD28 and dependent on the kinase Lck. We identified a conserved proline-rich motif in V3 required for association with CD28 and immunological synapse localization. We found association with CD28 to be essential for PKC-θ-mediated downstream signaling and the differentiation of T helper type 2 cells (T(H)2 cells) and interleukin 17-producing helper T cells (T(H)17 cells) but not of T helper type 1 cells (T(H)1 cells). Ectopic expression of V3 sequestered PKC-θ from the immunological synapse and interfered with its functions. Our results identify a unique mode of CD28 signaling, establish a molecular basis for the immunological synapse localization of PKC-θ and indicate V3-based 'decoys' may be therapeutic modalities for T cell-mediated inflammatory diseases.


Subject(s)
CD28 Antigens/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Amino Acid Motifs/genetics , Animals , CD28 Antigens/immunology , Cell Differentiation/immunology , Cells, Cultured , Immunological Synapses , Immunomodulation , Isoenzymes/genetics , Isoenzymes/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Proline-Rich Protein Domains/genetics , Protein Binding/immunology , Protein Kinase C/genetics , Protein Kinase C/immunology , Protein Kinase C-theta , Protein Transport/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
9.
Proc Natl Acad Sci U S A ; 117(38): 23707-23716, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32878999

ABSTRACT

Trafficking of toll-like receptor 3 (TLR3) from the endoplasmic reticulum (ER) to endolysosomes and its subsequent proteolytic cleavage are required for it to sense viral double-stranded RNA (dsRNA) and trigger antiviral response, yet the underlying mechanisms remain enigmatic. We show that the E3 ubiquitin ligase TRIM3 is mainly located in the Golgi apparatus and transported to the early endosomes upon stimulation with the dsRNA analog poly(I:C). TRIM3 mediates K63-linked polyubiquitination of TLR3 at K831, which is enhanced following poly(I:C) stimulation. The polyubiquitinated TLR3 is recognized and sorted by the ESCRT (endosomal sorting complex required for transport) complexes to endolysosomes. Deficiency of TRIM3 impairs TLR3 trafficking from the Golgi apparatus to endosomes and its subsequent activation. Trim3-/- cells and mice express lower levels of antiviral genes and show lower levels of inflammatory response following poly(I:C) but not lipopolysaccharide (LPS) stimulation. These findings suggest that TRIM3-mediated polyubiquitination of TLR3 represents a feedback-positive regulatory mechanism for TLR3-mediated innate immune and inflammatory responses.


Subject(s)
Carrier Proteins/immunology , Endosomal Sorting Complexes Required for Transport/immunology , Immunity, Innate/immunology , Toll-Like Receptor 3/immunology , Ubiquitination/immunology , Animals , Antiviral Agents/immunology , HEK293 Cells , Humans , Lysosomes/immunology , Mice , Protein Transport/immunology , RNA, Viral/immunology , Signal Transduction/immunology
10.
Nat Immunol ; 10(5): 504-13, 2009 May.
Article in English | MEDLINE | ID: mdl-19363483

ABSTRACT

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Subject(s)
Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Arenaviridae Infections/immunology , Blotting, Western , CTLA-4 Antigen , Dendritic Cells/metabolism , Flow Cytometry , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Congenic , Mice, Transgenic , Protein Transport/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
J Immunol ; 203(6): 1560-1570, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31391232

ABSTRACT

STING plays central roles in the innate immune response to pathogens that contain DNA. Sensing cytoplasmic DNA by cyclic GMP-AMP synthase produces cyclic GMP-AMP, which binds to and activates STING and induces STING translocation from the endoplasmic reticulum to the perinuclear microsome. However, this trafficking process has not been fully elucidated yet. In this study, we identified YIPF5 as a positive regulator of STING trafficking. YIPF5 is essential for DNA virus- or intracellular DNA-triggered production of type I IFNs. Consistently, knockdown of YIPF5 impairs cellular antiviral responses to DNA virus. Mechanistically, YIPF5 interacts with both STING and components of COPII, facilitating STING recruitment to COPII in the presence of cytoplasmic dsDNA. Furthermore, knockdown of components of COPII inhibits DNA virus-triggered production of type I IFNs, suggesting that COPII is involved in innate immune responses to DNA viruses. Collectively, our findings demonstrate that YIPF5 positively regulates STING-mediated innate immune responses by recruiting STING to COPII-coated vesicles and facilitating STING trafficking from the endoplasmic reticulum to Golgi, providing important insights into the molecular mechanisms of intracellular DNA-stimulated STING trafficking and activation.


Subject(s)
COP-Coated Vesicles/immunology , DNA Viruses/immunology , Immunity, Innate/immunology , Membrane Proteins/immunology , Protein Transport/immunology , Signal Transduction/immunology , Vesicular Transport Proteins/immunology , Animals , DNA, Viral/immunology , Endoplasmic Reticulum/immunology , Golgi Apparatus/immunology , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL
12.
J Immunol ; 202(8): 2360-2371, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30814309

ABSTRACT

Mast cells (MCs) contribute to the control of local inflammatory reactions and become hyporesponsive after prolonged TLR4 activation by bacterial LPS. The molecular mechanisms involved in endotoxin tolerance (ET) induction in MCs are not fully understood. In this study, we demonstrate that the endocannabinoid 2-arachidonoylglycerol (2-AG) and its receptor, cannabinoid receptor 2 (CB2), play a role in the establishment of ET in bone marrow-derived MCs from C57BL/6J mice. We found that CB2 antagonism prevented the development of ET and that bone marrow-derived MCs produce 2-AG in a TLR4-dependent fashion. Exogenous 2-AG induced ET similarly to LPS, blocking the phosphorylation of IKK and the p65 subunit of NF-κB and inducing the synthesis of molecular markers of ET. LPS caused CB2 receptor trafficking in Rab11-, Rab7-, and Lamp2-positive vesicles, indicating recycling and degradation of the receptor. 2-AG also prevented LPS-induced TNF secretion in vivo, in a MC-dependent model of endotoxemia, demonstrating that TLR4 engagement leads to 2-AG secretion, which contributes to the negative control of MCs activation. Our study uncovers a functional role for the endocannabinoid system in the inhibition of MC-dependent innate immune responses in vivo.


Subject(s)
Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Glycerides/pharmacology , Immune Tolerance/drug effects , Lipopolysaccharides/toxicity , Mast Cells/immunology , Receptor, Cannabinoid, CB2/immunology , Toll-Like Receptor 4/immunology , Animals , Immune Tolerance/immunology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/immunology , Mice , Mice, Knockout , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/immunology , Receptor, Cannabinoid, CB2/genetics , Toll-Like Receptor 4/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology , rab7 GTP-Binding Proteins
13.
J Biol Chem ; 294(21): 8412-8423, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30944173

ABSTRACT

The innate immune system plays an essential role in initial recognition of pathogen infection by producing inflammatory cytokines and type I interferons. cGAS is a cytoplasmic sensor for DNA derived from DNA viruses. cGAS binding with DNA induces the production of cGAMP, a second messenger that associates with STING in endoplasmic reticulum (ER). STING changes its cellular distribution from ER to perinuclear Golgi, where it activates the protein kinase TBK1 that catalyzes the phosphorylation of IRF3. Here we found that STING trafficking is regulated by myotubularin-related protein (MTMR) 3 and MTMR4, members of protein tyrosine phosphatases that dephosphorylate 3' position in phosphatidylinositol (PtdIns) and generate PtdIns5P from PtdIns3,5P2 and PtdIns from PtdIns3P. We established MTMR3 and MTMR4 double knockout (DKO) RAW264.7 macrophage cells and found that they exhibited increased type I interferon production after interferon-stimulatory DNA (ISD) stimulation and herpes simplex virus 1 infection concomitant with enhanced IRF3 phosphorylation. In DKO cells, STING rapidly trafficked from ER to Golgi after ISD stimulation. Notably, DKO cells exhibited enlarged cytosolic puncta positive for PtdIns3P and STING was aberrantly accumulated in this puncta. Taken together, these results suggest that MTMR3 and MTMR4 regulate the production of PtdIns3P, which plays a critical role in suppressing DNA-mediated innate immune responses via modulating STING trafficking.


Subject(s)
DNA, Viral/immunology , Herpesvirus 1, Human/immunology , Immunity, Innate , Macrophages/immunology , Membrane Proteins/immunology , Phosphatidylinositol Phosphates/immunology , Protein Tyrosine Phosphatases, Non-Receptor/immunology , Animals , DNA, Viral/genetics , Herpesvirus 1, Human/genetics , Membrane Proteins/genetics , Mice , Phosphatidylinositol Phosphates/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Transport/genetics , Protein Transport/immunology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RAW 264.7 Cells
14.
Nat Immunol ; 9(12): 1407-14, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18931679

ABSTRACT

Toll-like receptors (TLRs) activate the innate immune system in response to pathogens. Here we show that TLR9 proteolytic cleavage is a prerequisite for TLR9 signaling. Inhibition of lysosomal proteolysis rendered TLR9 inactive. The carboxy-terminal fragment of TLR9 thus generated included a portion of the TLR9 ectodomain, as well as the transmembrane and cytoplasmic domains. This cleavage fragment bound to the TLR9 ligand CpG DNA and, when expressed in Tlr9(-/-) dendritic cells, restored CpG DNA-induced cytokine production. Although cathepsin L generated the requisite TLR9 cleavage products in a cell-free in vitro system, several proteases influenced TLR9 cleavage in intact cells. Lysosomal proteolysis thus contributes to innate immunity by facilitating specific cleavage of TLR9.


Subject(s)
Enzyme Activation/immunology , Immunity, Innate/physiology , Lysosomes/metabolism , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , Blotting, Western , Cathepsins/immunology , Cathepsins/metabolism , Cell Line , CpG Islands , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoprecipitation , Lysosomes/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Protein Transport/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
15.
Cytokine ; 129: 154944, 2020 05.
Article in English | MEDLINE | ID: mdl-32146280

ABSTRACT

Effector CD4+ T cells can be classified by the cytokines they secrete, with T helper 1 (Th1) cells generating interferon (IFN)γ and Th17 cells secreting interleukin (IL)-17. Both Th1 and Th17 cells are strongly implicated in the initiation and chronicity of autoimmune diseases such as multiple sclerosis. The endoplasmic reticulum (ER) has been implicated as a potentially crucial site in regulating CD4+ T cell function. Secretory and transmembrane proteins are shuttled into the ER via the Sec61 translocon, where they undergo appropriate folding; misfolded proteins are retro-translocated from the ER in a p97-dependent manner. Here, we provide evidence that both processes are crucial to the secretion of inflammatory cytokines from effector CD4+ T cells. The pan-ER inhibitor eeeyarestatin-1 (ESI), which interferes with both Sec61 translocation and p97 retro-translocation, inhibited secretion of interferon (IFN)γ, interleukin (IL)-2 and tumor necrosis factor (TNF)α from Th1 cells in a dose-dependent manner. Selective inhibition of Sec61 by Apratoxin A (ApraA) revealed that ER translocation is crucial for Th1 cytokine secretion, while inhibition of p97 by NMS-873 also inhibited Th1 function, albeit to a lesser degree. By contrast, none of ESI, ApraA or NMS-873 could significantly reduce IL-17 secretion from Th17 cells. ApraA, but not NMS-873, reduced phosphorylation of Stat1 in Th1 cells, indicating the involvement of ER translocation in Th1 differentiation pathways. ApraA had modest effects on activation of the Th17 transcription factor Stat3, while NMS-873 had no effect. Interestingly, NMS-873 was able to reduce disease severity in CD4+ T cell-driven experimental autoimmune encephalomyelitis (EAE). Together, our data indicate that CD4+ T cell function, and Th1 cell function in particular, is dependent on protein translocation and dislocation across the ER.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endoplasmic Reticulum/immunology , Inflammation/immunology , Protein Transport/immunology , Animals , Cell Differentiation/immunology , Central Nervous System/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology
16.
Scand J Immunol ; 92(5): e12978, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32969499

ABSTRACT

MHC class I molecules on the cellular surface display peptides that either derive from endogenous proteins (self or viral), or from endocytosis of molecules, dying cells or pathogens. The conventional antigen-processing pathway for MHC class I presentation depends on proteasome-mediated degradation of the protein followed by transporter associated with antigen-processing (TAP)-mediated transport of the generated peptides into the endoplasmic reticulum (ER). Here, peptides are loaded onto MHC I molecules before transportation to the cell surface. However, several alternative mechanisms have emerged. These include TAP-independent mechanisms, the vacuolar pathway and involvement of autophagy. Autophagy is a cell intrinsic recycling system. It also functions as a defence mechanism that removes pathogens and damaged endocytic compartments from the cytosol. Therefore, it appears likely that autophagy would intersect with the MHC class I presentation pathway to alarm CD8+ T cells of an ongoing intracellular infection. However, the importance of autophagy as a source of antigen for presentation on MHC I molecules remains to be defined. Here, original research papers which suggest involvement of autophagy in MHC I antigen presentation are reviewed. The antigens are from herpesvirus, cytomegalovirus and chlamydia. The studies point towards autophagy as important in MHC class I presentation of endogenous proteins during conditions of immune evasion. Because autophagy is a regulated process which is induced upon activation of, for example, pattern recognition receptors (PRRs), it will be crucial to use relevant stimulatory conditions together with primary cells when aiming to confirm the importance of autophagy in MHC class I antigen presentation in future studies.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Transport/immunology
17.
Immunity ; 34(1): 24-38, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21194982

ABSTRACT

RAPL (an alternative spliced form of Rassf5) is a critical Ras-related protein1 (Rap1) effector that regulates lymphocyte adhesion. Here, we have shown that in addition to this previously described function, RAPL also negatively controls lymphocyte proliferation and prevents autoimmunity and lymphoma. RAPL-deficient mice experienced age-related lupus-like glomerulonephritis and developed B cell lymphomas. RAPL-deficient lymphocytes showed hyperproliferation by enhanced S phase entry after antigen receptor ligation. Compared to wild-type cells, RAPL-deficient naive lymphocytes had a 2- to 3-fold increase in Cdk2 kinase activity with a cytoplasmic mislocalization of the cyclin-dependent kinase inhibitor p27(kip1). RAPL was found to suppress the phosphorylation of p27(kip1) on serine 10 (S10) and promoted p27(kip1) nuclear translocation. An S10A mutation in p27(kip1) corrected its cytoplasmic accumulation, reduced hyperproliferation in RAPL-deficient lymphocytes, and suppressed glomerulonephritis and development of B cell lymphoma. Thus, RAPL serves as a checkpoint for S phase entry to prevent lymphoproliferative disorders through the spatial regulation of p27(kip1).


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , rap1 GTP-Binding Proteins/genetics , Animals , Autoimmunity/genetics , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lymphocytes/immunology , Mice , Mice, Knockout , Mutation/genetics , Phosphorylation/genetics , Protein Transport/genetics , Protein Transport/immunology , rap1 GTP-Binding Proteins/immunology , rap1 GTP-Binding Proteins/metabolism
18.
J Immunol ; 200(8): 2798-2808, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29540576

ABSTRACT

TLRs recognize pathogen components and drive innate immune responses. They localize at either the plasma membrane or intracellular vesicles such as endosomes and lysosomes, and proper cellular localization is important for their ligand recognition and initiation of signaling. In this study, we disrupted ATP6V0D2, a component of vacuolar-type H+ adenosine triphosphatase (V-ATPase) that plays a central role in acidification of intracellular vesicles, in a macrophage cell line. ATP6V0D2-deficient cells exhibited reduced cytokine production in response to endosome-localized, nucleic acid-sensing TLR3, TLR7, and TLR9, but enhanced inflammatory cytokine production and NF-κB activation following stimulation with LPS, a TLR4 agonist. Moreover, they had defects in internalization of cell surface TLR4 and exhibited enhanced inflammatory cytokine production after repeated LPS stimulation, thereby failing to induce LPS tolerance. A component of the V-ATPase complex interacted with ARF6, the small GTPase known to regulate TLR4 internalization, and ARF6 deficiency resulted in prolonged TLR4 expression on the cell surface. Taken together, these findings suggest that ATP6V0D2-dependent intravesicular acidification is required for TLR4 internalization, which is associated with prevention from excessive LPS-triggered inflammation and induction of tolerance.


Subject(s)
Immune Tolerance/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Toll-Like Receptor 4/metabolism , Animals , Cytoplasmic Vesicles/immunology , Cytoplasmic Vesicles/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Protein Transport/immunology , RAW 264.7 Cells , Toll-Like Receptor 4/immunology
19.
J Immunol ; 200(11): 3790-3800, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29686050

ABSTRACT

Amino acid metabolism plays important roles in innate immune cells, including macrophages. Recently, we reported that a lysosomal adaptor protein, Lamtor1, which serves as the scaffold for amino acid-activated mechanistic target of rapamycin complex 1 (mTORC1), is critical for the polarization of M2 macrophages. However, little is known about how Lamtor1 affects the inflammatory responses that are triggered by the stimuli for TLRs. In this article, we show that Lamtor1 controls innate immune responses by regulating the phosphorylation and nuclear translocation of transcription factor EB (TFEB), which has been known as the master regulator for lysosome and autophagosome biogenesis. Furthermore, we show that nuclear translocation of TFEB occurs in alveolar macrophages of myeloid-specific Lamtor1 conditional knockout mice and that these mice are hypersensitive to intratracheal administration of LPS and bleomycin. Our observation clarified that the amino acid-sensing pathway consisting of Lamtor1, mTORC1, and TFEB is involved in the regulation of innate immune responses.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Immunity, Innate/immunology , Lysosomes/immunology , Proteins/immunology , Amino Acids/immunology , Animals , Autophagy/immunology , Cell Line , Cell Nucleus/immunology , Macrophages/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/immunology , Protein Transport/immunology , RAW 264.7 Cells , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology
20.
J Immunol ; 201(9): 2624-2640, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30282749

ABSTRACT

Endosomal traffic of TCR and signaling molecules regulates immunological synapse formation and T cell activation. We recently showed that Rab11 endosomes regulate the subcellular localization of the tyrosine kinase Lck and of the GTPase Rac1 and control their functions in TCR signaling and actin cytoskeleton remodeling. HIV-1 infection of T cells alters their endosomal traffic, activation capacity, and actin cytoskeleton organization. The viral protein Nef is pivotal for these modifications. We hypothesized that HIV-1 Nef could jointly alter Lck and Rac1 endosomal traffic and concomitantly modulate their functions. In this study, we show that HIV-1 infection of human T cells sequesters both Lck and Rac1 in a pericentrosomal compartment in an Nef-dependent manner. Strikingly, the Nef-induced Lck compartment contains signaling-competent forms (phosphorylated on key Tyr residues) of Lck and some of its downstream effectors, TCRζ, ZAP70, SLP76, and Vav1, avoiding the proximal LAT adaptor. Importantly, Nef-induced concentration of signaling molecules was concomitant with the upregulation of several early and late T cell activation genes. Moreover, preventing the concentration of the Nef-induced Lck compartment by depleting the Rab11 effector FIP3 counteracted Nef-induced gene expression upregulation. In addition, Nef extensively sequesters Rac1 and downregulates Rac1-dependent actin cytoskeleton remodeling, thus reducing T cell spreading. Therefore, by modifying their endosomal traffic, Nef hijacks signaling and actin cytoskeleton regulators to dually modulate their functional outputs. Our data shed new light into the molecular mechanisms that modify T cell physiology during HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , nef Gene Products, Human Immunodeficiency Virus/immunology , rac1 GTP-Binding Protein/metabolism , Actin Cytoskeleton/immunology , Actin Cytoskeleton/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Endosomes/immunology , Endosomes/metabolism , Endosomes/virology , HIV Infections/metabolism , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Protein Transport/immunology , Signal Transduction/immunology , rac1 GTP-Binding Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL