Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Exp Med ; 221(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38935023

ABSTRACT

Nuclear factor Foxp3 determines regulatory T (Treg) cell fate and function via mechanisms that remain unclear. Here, we investigate the nature of Foxp3-mediated gene regulation in suppressing autoimmunity and antitumor immune response. Contrasting with previous models, we find that Foxp3-chromatin binding is regulated by Treg activation states, tumor microenvironment, and antigen and cytokine stimulations. Proteomics studies uncover dynamic proteins within Foxp3 proximity upon TCR or IL-2 receptor signaling in vitro, reflecting intricate interactions among Foxp3, signal transducers, and chromatin. Pharmacological inhibition and genetic knockdown experiments indicate that NFAT and AP-1 protein Batf are required for enhanced Foxp3-chromatin binding in activated Treg cells and tumor-infiltrating Treg cells to modulate target gene expression. Furthermore, mutations at the Foxp3 DNA-binding domain destabilize the Foxp3-chromatin association. These representative settings delineate context-dependent Foxp3-chromatin interaction, suggesting that Foxp3 associates with chromatin by hijacking DNA-binding proteins resulting from Treg activation or differentiation, which is stabilized by direct Foxp3-DNA binding, to dynamically regulate Treg cell function according to immunological contexts.


Subject(s)
Chromatin , Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Chromatin/metabolism , Animals , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Signal Transduction , Protein Binding , Humans , Gene Expression Regulation , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-2/metabolism , Receptors, Interleukin-2/genetics , Cell Differentiation
2.
Cancer Res Commun ; 3(12): 2430-2446, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37971169

ABSTRACT

Understanding the intricate dynamics between adoptively transferred immune cells and the brain tumor immune microenvironment (TIME) is crucial for the development of effective T cell-based immunotherapies. In this study, we investigated the influence of the TIME and chimeric antigen receptor (CAR) design on the anti-glioma activity of B7-H3-specific CAR T-cells. Using an immunocompetent glioma model, we evaluated a panel of seven fully murine B7-H3 CARs with variations in transmembrane, costimulatory, and activation domains. We then investigated changes in the TIME following CAR T-cell therapy using high-dimensional flow cytometry and single-cell RNA sequencing. Our results show that five out of six B7-H3 CARs with single costimulatory domains demonstrated robust functionality in vitro. However, these CARs had significantly varied levels of antitumor activity in vivo. To enhance therapeutic effectiveness and persistence, we incorporated 41BB and CD28 costimulation through transgenic expression of 41BBL on CD28-based CAR T-cells. This CAR design was associated with significantly improved anti-glioma efficacy in vitro but did not result in similar improvements in vivo. Analysis of the TIME revealed that CAR T-cell therapy influenced the composition of the TIME, with the recruitment and activation of distinct macrophage and endogenous T-cell subsets crucial for successful antitumor responses. Indeed, complete brain macrophage depletion using a CSF1R inhibitor abrogated CAR T-cell antitumor activity. In sum, our study highlights the critical role of CAR design and its modulation of the TIME in mediating the efficacy of adoptive immunotherapy for high-grade glioma. SIGNIFICANCE: CAR T-cell immunotherapies hold great potential for treating brain cancers; however, they are hindered by a challenging immune environment that dampens their effectiveness. In this study, we show that the CAR design influences the makeup of the immune environment in brain tumors, underscoring the need to target specific immune components to improve CAR T-cell performance, and highlighting the significance of using models with functional immune systems to optimize this therapy.


Subject(s)
Brain Neoplasms , Glioma , Receptors, Chimeric Antigen , Mice , Animals , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Tumor-Associated Macrophages/metabolism , CD28 Antigens/genetics , Glioma/therapy , Brain Neoplasms/therapy , Brain/metabolism , Tumor Microenvironment
3.
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
4.
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
5.
Stat Methods Med Res ; 31(8): 1484-1499, 2022 08.
Article in English | MEDLINE | ID: mdl-35450486

ABSTRACT

Spatial data from high-resolution images abound in many scientific disciplines. For example, single-molecule localization microscopy, such as stochastic optical reconstruction microscopy, provides super-resolution images to help scientists investigate co-localization of proteins and hence their interactions inside cells, which are key events in living cells. However, there are few accurate methods for analyzing co-localization in super-resolution images. The current methods and software are prone to produce false-positive errors and are restricted to only 2-dimensional images. In this paper, we develop a novel statistical method to effectively address the problems of unbiased and robust quantification and comparison of protein co-localization for multiple 2- and 3-dimensional image datasets. This method significantly improves the analysis of protein co-localization using super-resolution image data, as shown by its excellent performance in simulation studies and an analysis of co-localization of protein light chain 3 and lysosomal-associated membrane protein 1 in cell autophagy. Moreover, this method is directly applicable to co-localization analyses in other disciplines, such as diagnostic imaging, epidemiology, environmental science, and ecology.


Subject(s)
Software , Computer Simulation
6.
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
7.
Cell Rep ; 38(10): 110482, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263574

ABSTRACT

Infection and vaccination repeatedly expose individuals to antigens that are conserved between influenza virus subtypes. Nevertheless, antibodies recognizing variable influenza epitopes greatly outnumber antibodies reactive against conserved epitopes. Elucidating factors contributing to the paucity of broadly reactive influenza antibodies remains a major obstacle for developing a universal influenza vaccine. Here, we report that inducing broadly reactive influenza antibodies increases autoreactive antibodies in humans and mice and exacerbates disease in four distinct models of autoimmune disease. Importantly, transferring broadly reactive influenza antibodies augments disease in the presence of inflammation or autoimmune susceptibility. Further, broadly reactive influenza antibodies spontaneously arise in mice with defects in B cell tolerance. Together, these data suggest that self-tolerance mechanisms limit the prevalence of broadly reactive influenza antibodies, which can exacerbate disease in the context of additional risk factors.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Antibodies, Neutralizing , Antibodies, Viral , Autoimmunity , Epitopes , Hemagglutinin Glycoproteins, Influenza Virus , Humans , Mice
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
9.
Cancer Res ; 81(19): 5047-5059, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34301764

ABSTRACT

Immune cells regulate tumor growth by mirroring their function as tissue repair organizers in normal tissues. To understand the different facets of immune-tumor collaboration through genetics, spatial transcriptomics, and immunologic manipulation with noninvasive, longitudinal imaging, we generated a penetrant double oncogene-driven autochthonous model of neuroblastoma. Spatial transcriptomic analysis showed that CD4+ and myeloid populations colocalized within the tumor parenchyma, while CD8+ T cells and B cells were peripherally dispersed. Depletion of CD4+ T cells or CCR2+ macrophages, but not B cells, CD8+ T cells, or natural killer (NK) cells, prevented tumor formation. Tumor CD4+ T cells displayed unconventional phenotypes and were clonotypically diverse and antigen independent. Within the myeloid fraction, tumor growth required myeloid cells expressing arginase-1. Overall, these results demonstrate how arginine-metabolizing myeloid cells conspire with pathogenic CD4+ T cells to create permissive conditions for tumor formation, suggesting that these protumorigenic pathways could be disabled by targeting myeloid arginine metabolism. SIGNIFICANCE: A new model of human neuroblastoma provides ways to track tumor formation and expansion in living animals, allowing identification of CD4+ T-cell and macrophage functions required for oncogenesis.


Subject(s)
Arginase/genetics , CD4-Positive T-Lymphocytes/metabolism , Disease Susceptibility , Myeloid Cells/metabolism , Neuroblastoma/etiology , Neuroblastoma/metabolism , Animals , Arginase/metabolism , Biomarkers , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Neuroblastoma/pathology , Oncogenes , Single-Cell Analysis , Transcriptome
12.
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
13.
Sci Adv ; 6(33): eabb9036, 2020 08.
Article in English | MEDLINE | ID: mdl-32851186

ABSTRACT

Noncanonical functions of autophagy proteins have been implicated in neurodegenerative conditions, including Alzheimer's disease (AD). The WD domain of the autophagy protein Atg16L is dispensable for canonical autophagy but required for its noncanonical functions. Two-year-old mice lacking this domain presented with robust ß-amyloid (Aß) pathology, tau hyperphosphorylation, reactive microgliosis, pervasive neurodegeneration, and severe behavioral and memory deficiencies, consistent with human disease. Mechanistically, we found this WD domain was required for the recycling of Aß receptors in primary microglia. Pharmacologic suppression of neuroinflammation reversed established memory impairment and markers of disease pathology in this novel AD model. Therefore, loss of the Atg16L WD domain drives spontaneous AD in mice, and inhibition of neuroinflammation is a potential therapeutic approach for treating neurodegeneration and memory loss. A decline in expression of ATG16L in the brains of human patients with AD suggests the possibility that a similar mechanism may contribute in human disease.


Subject(s)
Alzheimer Disease , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Autophagy , Disease Models, Animal , Humans , Mice , Microglia/metabolism , tau Proteins/genetics , tau Proteins/metabolism
14.
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
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.
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
17.
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
18.
J Clin Invest ; 129(1): 150-162, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30457980

ABSTRACT

Pyrin is an inflammasome sensor that promotes caspase-1-mediated pyroptotic cell death and maturation of proinflammatory cytokines IL-1ß and IL-18. Familial Mediterranean fever (FMF), an autoinflammatory disorder, is associated with mutations in the gene encoding pyrin (MEFV). FMF-knockin (FMF-KI) mice that express chimeric pyrin protein with FMF mutation (MefvV726A/V726A) exhibit an autoinflammatory disorder mediated by autoactivation of the pyrin inflammasome. Increase in the levels of TNF are observed in FMF-KI mice, and many features of FMF overlap with the autoinflammatory disorder associated with TNF receptor signaling. In this study, we assessed the contribution of TNF signaling to pyrin inflammasome activation and its consequent role in distinct FMF pathologies. TNF signaling promoted the expression of pyrin in response to multiple stimuli and was required for inflammasome activation in response to canonical pyrin stimuli and in myeloid cells from FMF-KI mice. TNF signaling promoted systemic wasting, anemia, and neutrophilia in the FMF-KI mice. Further, TNF-induced pathology was induced specifically through the TNFR1 receptor, while TNFR2-mediated signaling was distinctly protective in colitis and ankle joint inflammation. Overall, our data show that TNF is a critical modulator of pyrin expression, inflammasome activation, and pyrin-inflammasomopathy. Further, specific blockade of TNFR1 or activation of TNFR2 could provide substantial protection against FMF pathologies.


Subject(s)
Inflammasomes/immunology , Pyrin/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Gene Knock-In Techniques , Inflammasomes/genetics , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology , Pyrin/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...