Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50.076
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 455-488, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360546

ABSTRACT

Ten-eleven translocation (TET) proteins are iron-dependent and α-ketoglutarate-dependent dioxygenases that sequentially oxidize the methyl group of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). All three epigenetic modifications are intermediates in DNA demethylation. TET proteins are recruited by transcription factors and by RNA polymerase II to modify 5mC at enhancers and gene bodies, thereby regulating gene expression during development, cell lineage specification, and cell activation. It is not yet clear, however, how the established biochemical activities of TET enzymes in oxidizing 5mC and mediating DNA demethylation relate to the known association of TET deficiency with inflammation, clonal hematopoiesis, and cancer. There are hints that the ability of TET deficiency to promote cell proliferation in a signal-dependent manner may be harnessed for cancer immunotherapy. In this review, we draw upon recent findings in cells of the immune system to illustrate established as well as emerging ideas of how TET proteins influence cellular function.


Subject(s)
DNA Demethylation , Dioxygenases , Immunotherapy , Inflammation , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/etiology , Neoplasms/metabolism , Animals , Inflammation/metabolism , Inflammation/immunology , Immunotherapy/methods , Dioxygenases/metabolism , Immune System/metabolism , Immune System/immunology , Epigenesis, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/genetics
2.
Annu Rev Immunol ; 38: 49-77, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340580

ABSTRACT

Mast cells have existed long before the development of adaptive immunity, although they have been given different names. Thus, in the marine urochordate Styela plicata, they have been designated as test cells. However, based on their morphological characteristics (including prominent cytoplasmic granules) and mediator content (including heparin, histamine, and neutral proteases), test cells are thought to represent members of the lineage known in vertebrates as mast cells. So this lineage presumably had important functions that preceded the development of antibodies, including IgE. Yet mast cells are best known, in humans, as key sources of mediators responsible for acute allergic reactions, notably including anaphylaxis, a severe and potentially fatal IgE-dependent immediate hypersensitivity reaction to apparently harmless antigens, including many found in foods and medicines. In this review, we briefly describe the origins of tissue mast cells and outline evidence that these cells can have beneficial as well as detrimental functions, both innately and as participants in adaptive immune responses. We also discuss aspects of mast cell heterogeneity and comment on how the plasticity of this lineage may provide insight into its roles in health and disease. Finally, we consider some currently open questions that are yet unresolved.


Subject(s)
Disease Susceptibility , Inflammation/etiology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Adaptive Immunity , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Humans , Immunity, Innate , Inflammation/diagnosis , Inflammation Mediators/metabolism , Signal Transduction
3.
Annu Rev Immunol ; 38: 567-595, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32017655

ABSTRACT

Caspases are a family of conserved cysteine proteases that play key roles in programmed cell death and inflammation. In multicellular organisms, caspases are activated via macromolecular signaling complexes that bring inactive procaspases together and promote their proximity-induced autoactivation and proteolytic processing. Activation of caspases ultimately results in programmed execution of cell death, and the nature of this cell death is determined by the specific caspases involved. Pioneering new research has unraveled distinct roles and cross talk of caspases in the regulation of programmed cell death, inflammation, and innate immune responses. In-depth understanding of these mechanisms is essential to foster the development of precise therapeutic targets to treat autoinflammatory disorders, infectious diseases, and cancer. This review focuses on mechanisms governing caspase activation and programmed cell death with special emphasis on the recent progress in caspase cross talk and caspase-driven gasdermin D-induced pyroptosis.


Subject(s)
Caspases/metabolism , Cell Death , Inflammation/etiology , Inflammation/metabolism , Neoplasm Proteins/genetics , Pyroptosis/genetics , Animals , Apoptosis , Biomarkers , Caspases/genetics , Cell Death/genetics , Disease Susceptibility , Enzyme Activation , Humans , Inflammation/pathology , Neoplasm Proteins/metabolism , Signal Transduction
4.
Annu Rev Immunol ; 37: 325-347, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30676821

ABSTRACT

ATP, NAD+, and nucleic acids are abundant purines that, in addition to having critical intracellular functions, have evolved extracellular roles as danger signals released in response to cell lysis, apoptosis, degranulation, or membrane pore formation. In general ATP and NAD+ have excitatory and adenosine has anti-inflammatory effects on immune cells. This review focuses on recent advances in our understanding of purine release mechanisms, ectoenzymes that metabolize purines (CD38, CD39, CD73, ENPP1, and ENPP2/autotaxin), and signaling by key P2 purinergic receptors (P2X7, P2Y2, and P2Y12). In addition to metabolizing ATP or NAD+, some purinergic ectoenzymes metabolize other inflammatory modulators, notably lysophosphatidic acid and cyclic GMP-AMP (cGAMP). Also discussed are extracellular signaling effects of NAD+ mediated by ADP-ribosylation, and epigenetic effects of intracellular adenosine mediated by modification of S-adenosylmethionine-dependent DNA methylation.


Subject(s)
Inflammation/immunology , Purines/metabolism , Receptors, Purinergic/metabolism , ADP-Ribosylation , Adenosine Triphosphate/metabolism , Animals , DNA Methylation , Humans , Inflammation/genetics , Inflammation/metabolism , Lysophospholipids/metabolism , Signal Transduction
5.
Annu Rev Immunol ; 36: 73-101, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29144836

ABSTRACT

The cellular degradative pathway of autophagy has a fundamental role in immunity. Here, we review the function of autophagy and autophagy proteins in inflammation. We discuss how the autophagy machinery controls the burden of infectious agents while simultaneously limiting inflammatory pathologies, which often involves processes that are distinct from conventional autophagy. Among the newly emerging processes we describe are LC3-associated phagocytosis and targeting by autophagy proteins, both of which require many of the same proteins that mediate conventional autophagy. We also discuss how autophagy contributes to differentiation of myeloid and lymphoid cell types, coordinates multicellular immunity, and facilitates memory responses. Together, these functions establish an intimate link between autophagy, mucosal immunity, and chronic inflammatory diseases. Finally, we offer our perspective on current challenges and barriers to translation.


Subject(s)
Autophagy , Disease Susceptibility , Inflammation/etiology , Animals , Biomarkers , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunomodulation , Inflammation/diagnosis , Inflammation/metabolism , Signal Transduction
6.
Cell ; 187(4): 981-998.e25, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38325365

ABSTRACT

The female reproductive tract (FRT) undergoes extensive remodeling during reproductive cycling. This recurrent remodeling and how it shapes organ-specific aging remains poorly explored. Using single-cell and spatial transcriptomics, we systematically characterized morphological and gene expression changes occurring in ovary, oviduct, uterus, cervix, and vagina at each phase of the mouse estrous cycle, during decidualization, and into aging. These analyses reveal that fibroblasts play central-and highly organ-specific-roles in FRT remodeling by orchestrating extracellular matrix (ECM) reorganization and inflammation. Our results suggest a model wherein recurrent FRT remodeling over reproductive lifespan drives the gradual, age-related development of fibrosis and chronic inflammation. This hypothesis was directly tested using chemical ablation of cycling, which reduced fibrotic accumulation during aging. Our atlas provides extensive detail into how estrus, pregnancy, and aging shape the organs of the female reproductive tract and reveals the unexpected cost of the recurrent remodeling required for reproduction.


Subject(s)
Aging , Genitalia, Female , Animals , Female , Mice , Pregnancy , Genitalia, Female/cytology , Genitalia, Female/metabolism , Inflammation/metabolism , Uterus/cytology , Vagina/cytology , Single-Cell Analysis
7.
Cell ; 187(8): 2010-2028.e30, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38569542

ABSTRACT

Gut inflammation involves contributions from immune and non-immune cells, whose interactions are shaped by the spatial organization of the healthy gut and its remodeling during inflammation. The crosstalk between fibroblasts and immune cells is an important axis in this process, but our understanding has been challenged by incomplete cell-type definition and biogeography. To address this challenge, we used multiplexed error-robust fluorescence in situ hybridization (MERFISH) to profile the expression of 940 genes in 1.35 million cells imaged across the onset and recovery from a mouse colitis model. We identified diverse cell populations, charted their spatial organization, and revealed their polarization or recruitment in inflammation. We found a staged progression of inflammation-associated tissue neighborhoods defined, in part, by multiple inflammation-associated fibroblasts, with unique expression profiles, spatial localization, cell-cell interactions, and healthy fibroblast origins. Similar signatures in ulcerative colitis suggest conserved human processes. Broadly, we provide a framework for understanding inflammation-induced remodeling in the gut and other tissues.


Subject(s)
Colitis, Ulcerative , Colitis , Animals , Humans , Mice , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , In Situ Hybridization, Fluorescence/methods , Inflammation/metabolism , Inflammation/pathology , Cell Communication , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology
8.
Cell ; 187(15): 4043-4060.e30, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38878778

ABSTRACT

Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.


Subject(s)
Inflammation , Membrane Proteins , Multiple Sclerosis , Neurons , Animals , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice , Humans , Inflammation/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Signal Transduction , Autophagy , Mice, Inbred C57BL , Glutamic Acid/metabolism , Ferroptosis , Disease Models, Animal , Female , Male
9.
Cell ; 187(15): 4061-4077.e17, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38878777

ABSTRACT

NLRs constitute a large, highly conserved family of cytosolic pattern recognition receptors that are central to health and disease, making them key therapeutic targets. NLRC5 is an enigmatic NLR with mutations associated with inflammatory and infectious diseases, but little is known about its function as an innate immune sensor and cell death regulator. Therefore, we screened for NLRC5's role in response to infections, PAMPs, DAMPs, and cytokines. We identified that NLRC5 acts as an innate immune sensor to drive inflammatory cell death, PANoptosis, in response to specific ligands, including PAMP/heme and heme/cytokine combinations. NLRC5 interacted with NLRP12 and PANoptosome components to form a cell death complex, suggesting an NLR network forms similar to those in plants. Mechanistically, TLR signaling and NAD+ levels regulated NLRC5 expression and ROS production to control cell death. Furthermore, NLRC5-deficient mice were protected in hemolytic and inflammatory models, suggesting that NLRC5 could be a potential therapeutic target.


Subject(s)
Inflammation , Intracellular Signaling Peptides and Proteins , NAD , Animals , Mice , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , NAD/metabolism , Humans , Immunity, Innate , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Mice, Knockout , Signal Transduction , HEK293 Cells , Inflammasomes/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Toll-Like Receptors/metabolism , Male , Cytokines/metabolism , Calcium-Binding Proteins
10.
Cell ; 187(9): 2209-2223.e16, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670073

ABSTRACT

Nuclear factor κB (NF-κB) plays roles in various diseases. Many inflammatory signals, such as circulating lipopolysaccharides (LPSs), activate NF-κB via specific receptors. Using whole-genome CRISPR-Cas9 screens of LPS-treated cells that express an NF-κB-driven suicide gene, we discovered that the LPS receptor Toll-like receptor 4 (TLR4) is specifically dependent on the oligosaccharyltransferase complex OST-A for N-glycosylation and cell-surface localization. The tool compound NGI-1 inhibits OST complexes in vivo, but the underlying molecular mechanism remained unknown. We did a CRISPR base-editor screen for NGI-1-resistant variants of STT3A, the catalytic subunit of OST-A. These variants, in conjunction with cryoelectron microscopy studies, revealed that NGI-1 binds the catalytic site of STT3A, where it traps a molecule of the donor substrate dolichyl-PP-GlcNAc2-Man9-Glc3, suggesting an uncompetitive inhibition mechanism. Our results provide a rationale for and an initial step toward the development of STT3A-specific inhibitors and illustrate the power of contemporaneous base-editor and structural studies to define drug mechanism of action.


Subject(s)
CRISPR-Cas Systems , Hexosyltransferases , Lipopolysaccharides , Membrane Proteins , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Hexosyltransferases/metabolism , Hexosyltransferases/genetics , NF-kappa B/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Toll-Like Receptor 4/metabolism , Animals , CRISPR-Cas Systems/genetics , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Mice , HEK293 Cells , Inflammation/metabolism , Inflammation/genetics , Glycosylation , Cryoelectron Microscopy , Catalytic Domain , Clustered Regularly Interspaced Short Palindromic Repeats/genetics
11.
Cell ; 187(8): 1874-1888.e14, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38518773

ABSTRACT

Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.


Subject(s)
Escherichia coli Infections , Escherichia coli , Lung , Polysaccharides, Bacterial , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Female , Male , Mice , Biofilms , Escherichia coli/physiology , Hypothermia/metabolism , Hypothermia/pathology , Inflammation/metabolism , Inflammation/pathology , Lung/microbiology , Lung/pathology , Pneumonia/microbiology , Pneumonia/pathology , Pseudomonas aeruginosa/physiology , Sensory Receptor Cells , Polysaccharides, Bacterial/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Nociceptors/metabolism
12.
Annu Rev Immunol ; 34: 173-202, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26772211

ABSTRACT

The formation and accumulation of crystalline material in tissues is a hallmark of many metabolic and inflammatory conditions. The discovery that the phase transition of physiologically soluble substances to their crystalline forms can be detected by the immune system and activate innate immune pathways has revolutionized our understanding of how crystals cause inflammation. It is now appreciated that crystals are part of the pathogenesis of numerous diseases, including gout, silicosis, asbestosis, and atherosclerosis. In this review we discuss current knowledge of the complex mechanisms of crystal formation in diseased tissues and their interplay with the nutrients, metabolites, and immune cells that account for crystal-induced inflammation.


Subject(s)
Asbestosis/immunology , Atherosclerosis/immunology , Crystallization , Gout/immunology , Immunity, Innate , Inflammation/metabolism , Silicosis/immunology , Animals , Humans , Interleukin-1/metabolism , Nanotechnology , Phase Transition
13.
Annu Rev Immunol ; 33: 49-77, 2015.
Article in English | MEDLINE | ID: mdl-25493334

ABSTRACT

Induction, production, and release of proinflammatory cytokines are essential steps to establish an effective host defense. Cytokines of the interleukin-1 (IL-1) family induce inflammation and regulate T lymphocyte responses while also displaying homeostatic and metabolic activities. With the exception of the IL-1 receptor antagonist, all IL-1 family cytokines lack a signal peptide and require proteolytic processing into an active molecule. One such unique protease is caspase-1, which is activated by protein platforms called the inflammasomes. However, increasing evidence suggests that inflammasomes and caspase-1 are not the only mechanism for processing IL-1 cytokines. IL-1 cytokines are often released as precursors and require extracellular processing for activity. Here we review the inflammasome-independent enzymatic processes that are able to activate IL-1 cytokines, paying special attention to neutrophil-derived serine proteases, which subsequently induce inflammation and modulate host defense. The inflammasome-independent processing of IL-1 cytokines has important consequences for understanding inflammatory diseases, and it impacts the design of IL-1-based modulatory therapies.


Subject(s)
Cytokines/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , Animals , Disease Susceptibility , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism
14.
Annu Rev Immunol ; 33: 417-43, 2015.
Article in English | MEDLINE | ID: mdl-25861977

ABSTRACT

Interleukin-27 (IL-27) is a cytokine with strikingly diverse influences on the immune response. Although it was initially linked with the development of Th1 responses, it is now recognized as a potent antagonist of different classes of inflammation through its ability to directly modify CD4(+) and CD8(+) T cell effector functions, to induce IL-10, and to promote specialized T regulatory cell responses. Although this aspect of IL-27 biology has provided insights into how the immune system prevents hyperactivity in the setting of infectious and autoimmune inflammation, in vaccination and cancer models the stimulatory effects of IL-27 on CD8(+) T cell function appear prominent. Additionally, associations between IL-27 and antibody-mediated disease have led to an interest in defining the impact of IL-27 on innate immunity and humoral responses in different disease states. The maturation of this literature has been accompanied by attempts to translate these findings from experimental models into human diseases and by efforts to define where IL-27 might represent a viable therapeutic target.


Subject(s)
Immunity , Interleukin-27/physiology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Inflammation/etiology , Inflammation/metabolism , Interleukin-27/chemistry , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Translational Research, Biomedical
15.
Annu Rev Immunol ; 33: 715-45, 2015.
Article in English | MEDLINE | ID: mdl-25861980

ABSTRACT

Inflammation is an unstable state. It either resolves or persists. Why inflammation persists and the factors that define tissue tropism remain obscure. Increasing evidence suggests that tissue-resident stromal cells not only provide positional memory but also actively regulate the differential accumulation of inflammatory cells within inflamed tissues. Furthermore, at many sites of chronic inflammation, structures that mimic secondary lymphoid tissues are observed, suggesting that chronic inflammation and lymphoid tissue formation share common activation programs. Similarly, blood and lymphatic endothelial cells contribute to tissue homeostasis and disease persistence in chronic inflammation. This review highlights our increasing understanding of the role of stromal cells in inflammation and summarizes the novel immunological role that stromal cells exert in the persistence of inflammatory diseases.


Subject(s)
Inflammation/immunology , Inflammation/metabolism , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , Animals , Cell Communication , Chronic Disease , Humans , Inflammation/pathology , Organogenesis/immunology , Phenotype
16.
Annu Rev Immunol ; 33: 107-38, 2015.
Article in English | MEDLINE | ID: mdl-25493331

ABSTRACT

Immune responses occur in the midst of a variety of cellular stresses that can severely perturb endoplasmic reticulum (ER) function. The unfolded protein response is a three-pronged signaling axis dedicated to preserving ER homeostasis. In this review, we highlight many important and emerging functional roles for ER stress in immunity, focusing on how the bidirectional cross talk between immunological processes and basic cell biology leads to pleiotropic signaling outcomes and enhanced sensitivity to inflammatory stimuli. We also discuss how dysregulated ER stress responses can provoke many diseases, including autoimmunity, firmly positioning the unfolded protein response as a major therapeutic target in human disease.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Immunity , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmunity , Cell Differentiation/immunology , Endoplasmic Reticulum/metabolism , Humans , Immune System Phenomena , Infections/etiology , Infections/metabolism , Inflammation/immunology , Inflammation/metabolism , Protein Binding , Signal Transduction , Transcription Factors/metabolism , Unfolded Protein Response
17.
Annu Rev Immunol ; 33: 823-74, 2015.
Article in English | MEDLINE | ID: mdl-25706096

ABSTRACT

Patients with autoinflammatory diseases present with noninfectious fever flares and systemic and/or disease-specific organ inflammation. Their excessive proinflammatory cytokine and chemokine responses can be life threatening and lead to organ damage over time. Studying such patients has revealed genetic defects that have helped unravel key innate immune pathways, including excessive IL-1 signaling, constitutive NF-κB activation, and, more recently, chronic type I IFN signaling. Discoveries of monogenic defects that lead to activation of proinflammatory cytokines have inspired the use of anticytokine-directed treatment approaches that have been life changing for many patients and have led to the approval of IL-1-blocking agents for a number of autoinflammatory conditions. In this review, we describe the genetically characterized autoinflammatory diseases, we summarize our understanding of the molecular pathways that drive clinical phenotypes and that continue to inspire the search for novel treatment targets, and we provide a conceptual framework for classification.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immunity, Innate , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferons/metabolism , Interleukin-1/metabolism , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , NF-kappa B/metabolism , Signal Transduction
18.
Annu Rev Immunol ; 33: 79-106, 2015.
Article in English | MEDLINE | ID: mdl-25493335

ABSTRACT

Cell proliferation and cell death are integral elements in maintaining homeostatic balance in metazoans. Disease pathologies ensue when these processes are disturbed. A plethora of evidence indicates that malfunction of cell death can lead to inflammation, autoimmunity, or immunodeficiency. Programmed necrosis or necroptosis is a form of nonapoptotic cell death driven by the receptor interacting protein kinase 3 (RIPK3) and its substrate, mixed lineage kinase domain-like (MLKL). RIPK3 partners with its upstream adaptors RIPK1, TRIF, or DAI to signal for necroptosis in response to death receptor or Toll-like receptor stimulation, pathogen infection, or sterile cell injury. Necroptosis promotes inflammation through leakage of cellular contents from damaged plasma membranes. Intriguingly, many of the signal adaptors of necroptosis have dual functions in innate immune signaling. This unique signature illustrates the cooperative nature of necroptosis and innate inflammatory signaling pathways in managing cell and organismal stresses from pathogen infection and sterile tissue injury.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Necrosis/metabolism , Signal Transduction , Animals , Bacterial Infections/genetics , Bacterial Infections/metabolism , Bacterial Infections/pathology , Biological Evolution , Cell Death , Humans , Inflammasomes/metabolism , Inflammation/genetics , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Parasitic Diseases/genetics , Parasitic Diseases/metabolism , Parasitic Diseases/pathology , Phosphorylation , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitination , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/pathology
19.
Cell ; 185(5): 759-761, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35245478

ABSTRACT

Neutrophil recruitment from blood into tissues is a hallmark of inflammation and anti-microbial host defense. In this issue, De Giovanni et al. describe an unanticipated role for a serotonin metabolite, 5-HIAA, which is produced by activated platelets and mast cells and engages the orphan receptor, GPR35, to recruit neutrophils to inflamed tissues.


Subject(s)
Blood Platelets , Neutrophils , Blood Platelets/metabolism , Humans , Inflammation/metabolism , Mast Cells/metabolism , Neutrophil Infiltration , Neutrophils/metabolism
20.
Cell ; 185(26): 4887-4903.e17, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563662

ABSTRACT

Our bodies turn over billions of cells daily via apoptosis and are in turn cleared by phagocytes via the process of "efferocytosis." Defects in efferocytosis are now linked to various inflammatory diseases. Here, we designed a strategy to boost efferocytosis, denoted "chimeric receptor for efferocytosis" (CHEF). We fused a specific signaling domain within the cytoplasmic adapter protein ELMO1 to the extracellular phosphatidylserine recognition domains of the efferocytic receptors BAI1 or TIM4, generating BELMO and TELMO, respectively. CHEF-expressing phagocytes display a striking increase in efferocytosis. In mouse models of inflammation, BELMO expression attenuates colitis, hepatotoxicity, and nephrotoxicity. In mechanistic studies, BELMO increases ER-resident enzymes and chaperones to overcome protein-folding-associated toxicity, which was further validated in a model of ER-stress-induced renal ischemia-reperfusion injury. Finally, TELMO introduction after onset of kidney injury significantly reduced fibrosis. Collectively, these data advance a concept of chimeric efferocytic receptors to boost efferocytosis and dampen inflammation.


Subject(s)
Macrophages , Phagocytosis , Animals , Mice , Macrophages/metabolism , Inflammation/metabolism , Phagocytes/metabolism , Carrier Proteins/metabolism , Apoptosis , Adaptor Proteins, Signal Transducing/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL