Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nat Immunol ; 23(5): 757-767, 2022 05.
Article in English | MEDLINE | ID: mdl-35437325

ABSTRACT

LAG3 is an inhibitory receptor that is highly expressed on exhausted T cells. Although LAG3-targeting immunotherapeutics are currently in clinical trials, how LAG3 inhibits T cell function remains unclear. Here, we show that LAG3 moved to the immunological synapse and associated with the T cell receptor (TCR)-CD3 complex in CD4+ and CD8+ T cells, in the absence of binding to major histocompatibility complex class II-its canonical ligand. Mechanistically, a phylogenetically conserved, acidic, tandem glutamic acid-proline repeat in the LAG3 cytoplasmic tail lowered the pH at the immune synapse and caused dissociation of the tyrosine kinase Lck from the CD4 or CD8 co-receptor, which resulted in a loss of co-receptor-TCR signaling and limited T cell activation. These observations indicated that LAG3 functioned as a signal disruptor in a major histocompatibility complex class II-independent manner, and provide insight into the mechanism of action of LAG3-targeting immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Antigens, CD/immunology , CD3 Complex/immunology , CD8 Antigens/metabolism , Histocompatibility Antigens Class II , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Lymphocyte Activation Gene 3 Protein
2.
Cell ; 178(3): 536-551.e14, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31257024

ABSTRACT

The expression of some proteins in the autophagy pathway declines with age, which may impact neurodegeneration in diseases, including Alzheimer's Disease. We have identified a novel non-canonical function of several autophagy proteins in the conjugation of LC3 to Rab5+, clathrin+ endosomes containing ß-amyloid in a process of LC3-associated endocytosis (LANDO). We found that LANDO in microglia is a critical regulator of immune-mediated aggregate removal and microglial activation in a murine model of AD. Mice lacking LANDO but not canonical autophagy in the myeloid compartment or specifically in microglia have a robust increase in pro-inflammatory cytokine production in the hippocampus and increased levels of neurotoxic ß-amyloid. This inflammation and ß-amyloid deposition were associated with reactive microgliosis and tau hyperphosphorylation. LANDO-deficient AD mice displayed accelerated neurodegeneration, impaired neuronal signaling, and memory deficits. Our data support a protective role for LANDO in microglia in neurodegenerative pathologies resulting from ß-amyloid deposition.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Endocytosis , Microtubule-Associated Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , CD36 Antigens/metabolism , Cytokines/metabolism , Disease Models, Animal , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/cytology , Microglia/metabolism , RAW 264.7 Cells , Receptors, Immunologic/metabolism , Toll-Like Receptor 4/metabolism
3.
Cell ; 175(2): 429-441.e16, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30245008

ABSTRACT

Targeting autophagy in cancer cells and in the tumor microenvironment are current goals of cancer therapy. However, components of canonical autophagy play roles in other biological processes, adding complexity to this goal. One such alternative function of autophagy proteins is LC3-associated phagocytosis (LAP), which functions in phagosome maturation and subsequent signaling events. Here, we show that impairment of LAP in the myeloid compartment, rather than canonical autophagy, induces control of tumor growth by tumor-associated macrophages (TAM) upon phagocytosis of dying tumor cells. Single-cell RNA sequencing (RNA-seq) analysis revealed that defects in LAP induce pro-inflammatory gene expression and trigger STING-mediated type I interferon responses in TAM. We found that the anti-tumor effects of LAP impairment require tumor-infiltrating T cells, dependent upon STING and the type I interferon response. Therefore, autophagy proteins in the myeloid cells of the tumor microenvironment contribute to immune suppression of T lymphocytes by effecting LAP.


Subject(s)
Immune Tolerance/physiology , Microtubule-Associated Proteins/physiology , Phagocytosis/physiology , Animals , Autophagy/immunology , Cell Line , Host-Pathogen Interactions , Humans , Immune Tolerance/immunology , Macrophages , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Myeloid Cells/metabolism , Phagosomes/physiology , T-Lymphocytes/metabolism , Tumor Microenvironment/physiology
5.
Mol Cell ; 82(13): 2401-2414.e9, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35597236

ABSTRACT

Activated CD8+ T lymphocytes differentiate into heterogeneous subsets. Using super-resolution imaging, we found that prior to the first division, dynein-dependent vesicular transport polarized active TORC1 toward the microtubule-organizing center (MTOC) at the proximal pole. This active TORC1 was physically associated with active eIF4F, required for the translation of c-myc mRNA. As a consequence, c-myc-translating polysomes polarized toward the cellular pole proximal to the immune synapse, resulting in localized c-myc translation. Upon division, the TORC1-eIF4A complex preferentially sorted to the proximal daughter cell, facilitating asymmetric c-Myc synthesis. Transient disruption of eIF4A activity at first division skewed long-term cell fate trajectories to memory-like function. Using a genetic barcoding approach, we found that first-division sister cells often displayed differences in transcriptional profiles that largely correlated with c-Myc and TORC1 target genes. Our findings provide mechanistic insights as to how distinct T cell fate trajectories can be established during the first division.


Subject(s)
CD8-Positive T-Lymphocytes , Eukaryotic Initiation Factor-4F , Cell Differentiation , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/genetics
6.
Immunity ; 52(6): 994-1006.e8, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32428502

ABSTRACT

Cell death pathways regulate various homeostatic processes. Autoimmune lymphoproliferative syndrome (ALPS) in humans and lymphoproliferative (LPR) disease in mice result from abrogated CD95-induced apoptosis. Because caspase-8 mediates CD95 signaling, we applied genetic approaches to dissect the roles of caspase-8 in cell death and inflammation. Here, we describe oligomerization-deficient Caspase-8F122GL123G/F122GL123G and non-cleavable Caspase-8D387A/D387A mutant mice with defective caspase-8-mediated apoptosis. Although neither mouse developed LPR disease, removal of the necroptosis effector Mlkl from Caspase-8D387A/D387A mice revealed an inflammatory role of caspase-8. Ablation of one allele of Fasl, Fadd, or Ripk1 prevented the pathology of Casp8D387A/D387AMlkl-/- animals. Removing both Fadd alleles from these mice resulted in early lethality prior to post-natal day 15 (P15), which was prevented by co-ablation of either Ripk1 or Caspase-1. Our results suggest an in vivo role of the inflammatory RIPK1-caspase-8-FADD (FADDosome) complex and reveal a FADD-independent inflammatory role of caspase-8 that involves activation of an inflammasome.


Subject(s)
Caspase 8/genetics , Disease Susceptibility , Fas-Associated Death Domain Protein/metabolism , Inflammation/etiology , Inflammation/metabolism , Necroptosis/genetics , Animals , Apoptosis/genetics , Biomarkers , Caspase 8/chemistry , Caspase 8/metabolism , Disease Models, Animal , Disease Progression , Fluorescent Antibody Technique , Gene Expression Regulation , Inflammasomes/metabolism , Inflammation/mortality , Inflammation/pathology , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Mortality , Phenotype , Protein Multimerization
7.
Immunity ; 49(3): 515-530.e5, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231985

ABSTRACT

Fungi represent a significant proportion of the gut microbiota. Aberrant immune responses to fungi are frequently observed in inflammatory bowel diseases (IBD) and colorectal cancer (CRC), and mutations in the fungal-sensing pathways are associated with the pathogenesis of IBD. Fungal recognition receptors trigger downstream signaling via the common adaptor protein CARD9 and the kinase SYK. Here we found that commensal gut fungi promoted inflammasome activation during AOM-DSS-induced colitis. Myeloid cell-specific deletion of Card9 or Syk reduced inflammasome activation and interleukin (IL)-18 maturation and increased susceptibility to colitis and CRC. IL-18 promoted epithelial barrier restitution and interferon-γ production by intestinal CD8+ T cells. Supplementation of IL-18 or transfer of wild-type myeloid cells reduced tumor burden in AOM-DSS-treated Card9-/- and Sykfl/flLysMCre/+ mice, whereas treatment with anti-fungal agents exacerbated colitis and CRC. CARD9 deletion changes the gut microbial landscape, suggesting that SYK-CARD9 signaling maintains a microbial ecology that promotes inflammasome activation and thereby restrains colitis and colon tumorigenesis.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Colitis/immunology , Colonic Neoplasms/immunology , Fungi/immunology , Gastrointestinal Microbiome/immunology , Inflammasomes/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/physiology , Myeloid Cells/physiology , Syk Kinase/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Colitis/chemically induced , Disease Models, Animal , Humans , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Sodium Dodecyl Sulfate , Syk Kinase/genetics
8.
Nature ; 595(7869): 724-729, 2021 07.
Article in English | MEDLINE | ID: mdl-34234346

ABSTRACT

T follicular helper (TFH) cells are crucial for B cell-mediated humoral immunity1. Although transcription factors such as BCL6 drive the differentiation of TFH cells2,3, it is unclear whether and how post-transcriptional and metabolic programs enforce TFH cell programming. Here we show that the cytidine diphosphate (CDP)-ethanolamine pathway co-ordinates the expression and localization of CXCR5 with the responses of TFH cells and humoral immunity. Using in vivo CRISPR-Cas9 screening and functional validation in mice, we identify ETNK1, PCYT2, and SELENOI-enzymes in the CDP-ethanolamine pathway for de novo synthesis of phosphatidylethanolamine (PE)-as selective post-transcriptional regulators of TFH cell differentiation that act by promoting the surface expression and functional effects of CXCR5. TFH cells exhibit unique lipid metabolic programs and PE is distributed to the outer layer of the plasma membrane, where it colocalizes with CXCR5. De novo synthesis of PE through the CDP-ethanolamine pathway co-ordinates these events to prevent the internalization and degradation of CXCR5. Genetic deletion of Pcyt2, but not of Pcyt1a (which mediates the CDP-choline pathway), in activated T cells impairs the differentiation of TFH cells, and this is associated with reduced humoral immune responses. Surface levels of PE and CXCR5 expression on B cells also depend on Pcyt2. Our results reveal that phospholipid metabolism orchestrates post-transcriptional mechanisms for TFH cell differentiation and humoral immunity, highlighting the metabolic control of context-dependent immune signalling and effector programs.


Subject(s)
Immunity, Humoral , Phosphatidylethanolamines/metabolism , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Differentiation , Cytidine Diphosphate , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphotransferases (Alcohol Group Acceptor) , RNA Nucleotidyltransferases , Signal Transduction
10.
Nature ; 587(7834): 466-471, 2020 11.
Article in English | MEDLINE | ID: mdl-33116313

ABSTRACT

Severe respiratory infections can result in acute respiratory distress syndrome (ARDS)1. There are no effective pharmacological therapies that have been shown to improve outcomes for patients with ARDS. Although the host inflammatory response limits spread of and eventually clears the pathogen, immunopathology is a major contributor to tissue damage and ARDS1,2. Here we demonstrate that respiratory viral infection induces distinct fibroblast activation states, which we term extracellular matrix (ECM)-synthesizing, damage-responsive and interferon-responsive states. We provide evidence that excess activity of damage-responsive lung fibroblasts drives lethal immunopathology during severe influenza virus infection. By producing ECM-remodelling enzymes-in particular the ECM protease ADAMTS4-and inflammatory cytokines, damage-responsive fibroblasts modify the lung microenvironment to promote robust immune cell infiltration at the expense of lung function. In three cohorts of human participants, the levels of ADAMTS4 in the lower respiratory tract were associated with the severity of infection with seasonal or avian influenza virus. A therapeutic agent that targets the ECM protease activity of damage-responsive lung fibroblasts could provide a promising approach to preserving lung function and improving clinical outcomes following severe respiratory infections.


Subject(s)
ADAMTS4 Protein/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Influenza A virus/pathogenicity , Lung/pathology , Lung/physiopathology , ADAMTS4 Protein/antagonists & inhibitors , Animals , Birds/virology , Extracellular Matrix/enzymology , Gene Expression Profiling , Humans , Influenza in Birds/virology , Influenza, Human/pathology , Influenza, Human/therapy , Influenza, Human/virology , Interferons/immunology , Interferons/metabolism , Leukocyte Common Antigens/metabolism , Lung/enzymology , Lung/virology , Mice , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/virology , Seasons , Single-Cell Analysis , Stromal Cells/metabolism
11.
Nat Immunol ; 14(3): 262-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377202

ABSTRACT

The physiological basis and mechanistic requirements for a large number of functional immunoreceptor tyrosine-based activation motifs (ITAMs; high ITAM multiplicity) in the complex of the T cell antigen receptor (TCR) and the invariant signaling protein CD3 remain obscure. Here we found that whereas a low multiplicity of TCR-CD3 ITAMs was sufficient to engage canonical TCR-induced signaling events that led to cytokine secretion, a high multiplicity of TCR-CD3 ITAMs was required for TCR-driven proliferation. This was dependent on the formation of compact immunological synapses, interaction of the adaptor Vav1 with phosphorylated CD3 ITAMs to mediate the recruitment and activation of the oncogenic transcription factor Notch1 and, ultimately, proliferation induced by the cell-cycle regulator c-Myc. Analogous mechanistic events were also needed to drive proliferation in response to weak peptide agonists. Thus, the TCR-driven pathways that initiate cytokine secretion and proliferation are separable and are coordinated by the multiplicity of phosphorylated ITAMs in TCR-CD3.


Subject(s)
CD3 Complex/immunology , Cytokines/biosynthesis , Immunoreceptor Tyrosine-Based Activation Motif/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , CD3 Complex/metabolism , Cell Line , Cell Proliferation , HEK293 Cells , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Receptor, Notch1/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism
12.
Nature ; 573(7775): 590-594, 2019 09.
Article in English | MEDLINE | ID: mdl-31511697

ABSTRACT

The cellular stress response has a vital role in regulating homeostasis by modulating cell survival and death. Stress granules are cytoplasmic compartments that enable cells to survive various stressors. Defects in the assembly and disassembly of stress granules are linked to neurodegenerative diseases, aberrant antiviral responses and cancer1-5. Inflammasomes are multi-protein heteromeric complexes that sense molecular patterns that are associated with damage or intracellular pathogens, and assemble into cytosolic compartments known as ASC specks to facilitate the activation of caspase-1. Activation of inflammasomes induces the secretion of interleukin (IL)-1ß and IL-18 and drives cell fate towards pyroptosis-a form of programmed inflammatory cell death that has major roles in health and disease6-12. Although both stress granules and inflammasomes can be triggered by the sensing of cellular stress, they drive contrasting cell-fate decisions. The crosstalk between stress granules and inflammasomes and how this informs cell fate has not been well-studied. Here we show that the induction of stress granules specifically inhibits NLRP3 inflammasome activation, ASC speck formation and pyroptosis. The stress granule protein DDX3X interacts with NLRP3 to drive inflammasome activation. Assembly of stress granules leads to the sequestration of DDX3X, and thereby the inhibition of NLRP3 inflammasome activation. Stress granules and the NLRP3 inflammasome compete for DDX3X molecules to coordinate the activation of innate responses and subsequent cell-fate decisions under stress conditions. Induction of stress granules or loss of DDX3X in the myeloid compartment leads to a decrease in the production of inflammasome-dependent cytokines in vivo. Our findings suggest that macrophages use the availability of DDX3X to interpret stress signals and choose between pro-survival stress granules and pyroptotic ASC specks. Together, our data demonstrate the role of DDX3X in driving NLRP3 inflammasome and stress granule assembly, and suggest a rheostat-like mechanistic paradigm for regulating live-or-die cell-fate decisions under stress conditions.


Subject(s)
Cell Death/genetics , DEAD-box RNA Helicases/metabolism , Inflammasomes/genetics , Macrophages/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Stress, Physiological/genetics , Animals , Cell Line , Cell Survival/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Humans , Inflammasomes/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
13.
Nat Immunol ; 13(3): 290-9, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-22306691

ABSTRACT

Interleukin 35 (IL-35) belongs to the IL-12 family of heterodimeric cytokines but has a distinct functional profile. IL-35 suppresses T cell proliferation and converts naive T cells into IL-35-producing induced regulatory T cells (iTr35 cells). Here we found that IL-35 signaled through a unique heterodimer of receptor chains IL-12Rß2 and gp130 or homodimers of each chain. Conventional T cells were sensitive to IL-35-mediated suppression in the absence of one receptor chain but not both receptor chains, whereas signaling through both chains was required for IL-35 expression and conversion into iTr35 cells. Signaling through the IL-35 receptor required the transcription factors STAT1 and STAT4, which formed a unique heterodimer that bound to distinct sites in the promoters of the genes encoding the IL-12 subunits p35 and Ebi3. This unconventional mode of signaling, distinct from that of other members of the IL-12 family, may broaden the spectrum and specificity of IL-35-mediated suppression.


Subject(s)
Receptors, Interleukin-1/immunology , Receptors, Interleukin/immunology , Signal Transduction , Animals , Cytokine Receptor gp130/immunology , Interleukins/immunology , Mice , Mice, Knockout , Models, Molecular , Protein Multimerization , Protein Structure, Quaternary , Receptors, Interleukin/chemistry , Receptors, Interleukin/deficiency , Receptors, Interleukin/metabolism , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-12/immunology , STAT1 Transcription Factor/immunology , STAT4 Transcription Factor/immunology
14.
Cell Mol Life Sci ; 79(10): 531, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36169732

ABSTRACT

In response to infection or sterile insults, inflammatory programmed cell death is an essential component of the innate immune response to remove infected or damaged cells. PANoptosis is a unique innate immune inflammatory cell death pathway regulated by multifaceted macromolecular complexes called PANoptosomes, which integrate components from other cell death pathways. Growing evidence shows that PANoptosis can be triggered in many physiological conditions, including viral and bacterial infections, cytokine storms, and cancers. However, PANoptosomes at the single cell level have not yet been fully characterized. Initial investigations have suggested that key pyroptotic, apoptotic, and necroptotic molecules including the inflammasome adaptor protein ASC, apoptotic caspase-8 (CASP8), and necroptotic RIPK3 are conserved components of PANoptosomes. Here, we optimized an immunofluorescence procedure to probe the highly dynamic multiprotein PANoptosome complexes across various innate immune cell death-inducing conditions. We first identified and validated antibodies to stain endogenous mouse ASC, CASP8, and RIPK3, without residual staining in the respective knockout cells. We then assessed the formation of PANoptosomes across innate immune cell death-inducing conditions by monitoring the colocalization of ASC with CASP8 and/or RIPK3. Finally, we established an expansion microscopy procedure using these validated antibodies to image the organization of ASC, CASP8, and RIPK3 within the PANoptosome. This optimized protocol, which can be easily adapted to study other multiprotein complexes and other cell death triggers, provides confirmation of PANoptosome assembly in individual cells and forms the foundation for a deeper molecular understanding of the PANoptosome complex and PANoptosis to facilitate therapeutic targeting.


Subject(s)
Inflammasomes , Single-Cell Analysis , Animals , Apoptosis , Caspase 8/metabolism , Inflammasomes/metabolism , Mice , Microscopy , Pyroptosis
15.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Article in English | MEDLINE | ID: mdl-32150572

ABSTRACT

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Subject(s)
Burkholderia Infections/immunology , Burkholderia/immunology , GTP-Binding Proteins/immunology , Giant Cells/immunology , Macrophages/immunology , Nose Diseases/immunology , Protein Prenylation/immunology , Animals , Burkholderia Infections/genetics , Burkholderia Infections/pathology , Cell Fusion , GTP-Binding Proteins/genetics , Giant Cells/microbiology , Giant Cells/pathology , Interferon Type I/genetics , Interferon Type I/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Nose Diseases/genetics , Nose Diseases/microbiology , Nose Diseases/pathology
16.
Proc Natl Acad Sci U S A ; 114(1): E67-E74, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27994150

ABSTRACT

Double-positive (DP) thymocytes respond to intrathymic T-cell receptor (TCR) signals by undergoing positive selection and lineage differentiation into single-positive (SP) mature cells. Concomitant with these well-characterized events is the acquisition of a mature T-cell gene expression program characterized by the induction of the effector molecules IL-7Rα, S1P1, and CCR7, but the underlying mechanism remains elusive. We report here that transcription repressor Growth factor independent 1 (Gfi1) orchestrates the fidelity of the DP gene expression program and developmental maturation into SP cells. Loss of Gfi1 resulted in premature induction of effector genes and the transcription factors forkhead box protein O1 (Foxo1) and Klf2 in DP thymocytes and the accumulation of postselection intermediate populations and accelerated transition into SP cells. Strikingly, partial loss of Foxo1 function, but not restored survival fitness, rectified the dysregulated gene expression and thymocyte maturation in Gfi1-deficient mice. Our results establish the Gfi1-Foxo1 axis and the transcriptional circuitry that actively maintain DP identity and shape the proper generation of mature T cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein O1/genetics , Gene Expression Regulation/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Transcription Factors/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Thymus Gland/cytology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics
17.
J Neurosci ; 38(10): 2615-2630, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29437892

ABSTRACT

Members of the SCY1-like (SCYL) family of protein kinases are evolutionarily conserved and ubiquitously expressed proteins characterized by an N-terminal pseudokinase domain, centrally located Huntingtin, elongation factor 3, protein phosphatase 2A, yeast kinase TOR1 repeats, and an overall disorganized C-terminal segment. In mammals, three family members encoded by genes Scyl1, Scyl2, and Scyl3 have been described. Studies have pointed to a role for SCYL1 and SCYL2 in regulating neuronal function and viability in mice and humans, but little is known about the biological function of SCYL3. Here, we show that the biochemical and cell biological properties of SCYL3 are similar to those of SCYL1 and both proteins work in conjunction to maintain motor neuron viability. Specifically, although lack of Scyl3 in mice has no apparent effect on embryogenesis and postnatal life, it accelerates the onset of the motor neuron disorder caused by Scyl1 deficiency. Growth abnormalities, motor dysfunction, hindlimb paralysis, muscle wasting, neurogenic atrophy, motor neuron degeneration, and loss of large-caliber axons in peripheral nerves occurred at an earlier age in Scyl1/Scyl3 double-deficient mice than in Scyl1-deficient mice. Disease onset also correlated with the mislocalization of TDP-43 in spinal motor neurons, suggesting that SCYL1 and SCYL3 regulate TDP-43 proteostasis. Together, our results demonstrate an overlapping role for SCYL1 and SCYL3 in vivo and highlight the importance the SCYL family of proteins in regulating neuronal function and survival. Only male mice were used in this study.SIGNIFICANCE STATEMENT SCYL1 and SCYL2, members of the SCY1-like family of pseudokinases, have well established roles in neuronal function. Herein, we uncover the role of SCYL3 in maintaining motor neuron viability. Although targeted disruption of Scyl3 in mice had little or no effect on embryonic development and postnatal life, it accelerated disease onset associated with the loss of Scyl1, a novel motor neuron disease gene in humans. Scyl1 and Scyl3 double-deficient mice had neuronal defects characteristic of amyotrophic lateral sclerosis, including TDP-43 pathology, at an earlier age than did Scyl1-deficient mice. Thus, we show that SCYL1 and SCYL3 play overlapping roles in maintaining motor neuronal viability in vivo and confirm that SCYL family members are critical regulators of neuronal function and survival.


Subject(s)
Cell Survival/genetics , Cell Survival/physiology , Membrane Proteins/physiology , Motor Neurons/physiology , Nerve Tissue Proteins/physiology , Protein Kinases/genetics , Adaptor Proteins, Vesicular Transport , Animals , Atrophy , Axons/pathology , Caspases/metabolism , DNA-Binding Proteins/genetics , Fibroblasts/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Movement Disorders/genetics , Movement Disorders/pathology , Muscle, Skeletal/pathology , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Paralysis/genetics , Paralysis/pathology
18.
Gastroenterology ; 154(4): 948-964.e8, 2018 03.
Article in English | MEDLINE | ID: mdl-29203393

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel diseases (IBD) increase risk for colorectal cancer. Mutations in the Mediterranean fever gene (MEFV or pyrin) are associated with hereditary autoinflammatory disease and severe IBD. Expression of MEFV, a sensor protein that the initiates assembly of the inflammasome complex, is increased in colon biopsies from patients with IBD. We investigated the role of pyrin in intestinal homeostasis in mice. METHODS: Mefv-/- mice and C57/BL6 mice (controls) were given azoxymethane followed by multiple rounds of dextran sodium sulfate (DSS) to induce colitis and tumorigenesis. In some experiments, Mefv-/- mice were given injections of recombinant interleukin 18 (rIL18) or saline (control) during DSS administration. Colon tissues were collected at different time points during colitis development and analyzed by histology, immunohistochemistry, immunoblots, or ELISAs (to measure cytokines). Spleen and mesenteric lymph node were collected, processed, and analyzed by flow cytometry. Colon epithelial permeability was measured in mice with colitis by gavage of fluorescent dextran and quantification of serum levels. RESULTS: MEFV was expressed in colons of control mice and expression increased during chronic and acute inflammation; high levels were detected in colon tumor and adjacent non-tumor tissues. Mefv-/- mice developed more severe colitis than control mice, with a greater extent of epithelial hyperplasia and a larger tumor burden. Levels of inflammatory cytokines (IL6) and chemokines were significantly higher in colons of Mefv-/- mice than control mice following colitis induction, whereas the level IL18, which depends on the inflammasome for maturation and release, was significantly lower in colons of Mefv-/- mice. Mefv-/- mice had increased epithelial permeability following administration of DSS than control mice, and loss of the tight junction proteins occludin and claudin-2 from intercellular junctions. STAT3 was activated (phosphorylated) in inflamed colon tissues from Mefv-/-, which also had increased expression of stem cell markers (OLFM4, BMI1, and MSI1) compared with colons from control mice. Administration of rIL18 to Mefv-/- mice reduced epithelial permeability, intestinal inflammation, the severity of colitis, and colon tumorigenesis. CONCLUSIONS: In studies with DSS-induced colitis, we found that pyrin (MEFV) is required for inflammasome activation and IL18 maturation, which promote intestinal barrier integrity and prevent colon inflammation and tumorigenesis. Strategies to increase activity of MEFV or IL18 might be developed for the treatment of IBD and prevention of colitis-associated tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Inflammasomes/metabolism , Pyrin/metabolism , Tight Junctions/metabolism , Animals , Azoxymethane , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Colon/immunology , Colon/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Dextran Sulfate , Disease Models, Animal , Genetic Predisposition to Disease , Inflammasomes/immunology , Interleukin-18/metabolism , Mice, Inbred C57BL , Mice, Knockout , Permeability , Phenotype , Phosphorylation , Pyrin/deficiency , Pyrin/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Tight Junctions/immunology , Tight Junctions/pathology , Time Factors , Tumor Burden , Tumor Microenvironment
19.
J Immunol ; 199(5): 1555-1560, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733484

ABSTRACT

For the αß or γδTCR chains to integrate extracellular stimuli into the appropriate intracellular cellular response, they must use the 10 ITAMs found within the CD3 subunits (CD3γε, CD3δε, and ζζ) of the TCR signaling complex. However, it remains unclear whether each specific ITAM sequence of the individual subunit (γεδζ) is required for thymocyte development or whether any particular CD3 ITAM motif is sufficient. In this article, we show that mice utilizing a single ITAM sequence (γ, ε, δ, ζa, ζb, or ζc) at each of the 10 ITAM locations exhibit a substantial reduction in thymic cellularity and limited CD4-CD8- (double-negative) to CD4+CD8+ (double-positive) maturation because of low TCR expression and signaling. Together, the data suggest that ITAM sequence diversity is required for optimal TCR signal transduction and subsequent T cell maturation.


Subject(s)
CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Thymus Gland/immunology , Amino Acid Motifs/genetics , Animals , CD3 Complex/genetics , Cell Differentiation , Cells, Cultured , Hematopoiesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction
20.
J Immunol ; 193(1): 258-67, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24899501

ABSTRACT

The TCR:CD3 complex transduces signals that are critical for optimal T cell development and adaptive immunity. In resting T cells, the CD3ε cytoplasmic tail associates with the plasma membrane via a proximal basic-rich stretch (BRS). In this study, we show that mice lacking a functional CD3ε-BRS exhibited substantial reductions in thymic cellularity and limited CD4- CD8- double-negative (DN) 3 to DN4 thymocyte transition, because of enhanced DN4 TCR signaling resulting in increased cell death and TCR downregulation in all subsequent populations. Furthermore, positive, but not negative, T cell selection was affected in mice lacking a functional CD3ε-BRS, which led to limited peripheral T cell function and substantially reduced responsiveness to influenza infection. Collectively, these results indicate that membrane association of the CD3ε signaling domain is required for optimal thymocyte development and peripheral T cell function.


Subject(s)
CD3 Complex/immunology , Cell Membrane/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Thymocytes/immunology , Animals , CD3 Complex/genetics , Cell Membrane/genetics , Mice , Mice, Knockout , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Thymocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL