Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 901
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 25(2): 256-267, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38172258

ABSTRACT

The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Animals , Humans , Mice , Alarmins , Antiviral Agents , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , T-Lymphocyte Subsets/metabolism
2.
Cell ; 183(3): 636-649.e18, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33031745

ABSTRACT

Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Nucleotidyltransferases/metabolism , Alarmins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cytoplasm/metabolism , Disease Models, Animal , Disease Progression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NF-kappa B/metabolism , Nerve Degeneration/pathology , Phosphotransferases (Alcohol Group Acceptor) , Protein Subunits/metabolism , Signal Transduction
3.
Cell ; 177(3): 541-555.e17, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955887

ABSTRACT

Neutrophils are attracted to and generate dense swarms at sites of cell damage in diverse tissues, often extending the local disruption of organ architecture produced by the initial insult. Whether the inflammatory damage resulting from such neutrophil accumulation is an inescapable consequence of parenchymal cell death has not been explored. Using a combination of dynamic intravital imaging and confocal multiplex microscopy, we report here that tissue-resident macrophages rapidly sense the death of individual cells and extend membrane processes that sequester the damage, a process that prevents initiation of the feedforward chemoattractant signaling cascade that results in neutrophil swarms. Through this "cloaking" mechanism, the resident macrophages prevent neutrophil-mediated inflammatory damage, maintaining tissue homeostasis in the face of local cell injury that occurs on a regular basis in many organs because of mechanical and other stresses. VIDEO ABSTRACT.


Subject(s)
Macrophages/immunology , Neutrophils/immunology , Alarmins/metabolism , Animals , Endocytosis , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Muscle Fibers, Skeletal/pathology , Neutrophil Activation , Neutrophils/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism
4.
Nat Immunol ; 22(8): 947-957, 2021 08.
Article in English | MEDLINE | ID: mdl-34239121

ABSTRACT

One of most challenging issues in tumor immunology is a better understanding of the dynamics in the accumulation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TIME), as this would lead to the development of new cancer therapeutics. Here, we show that translationally controlled tumor protein (TCTP) released by dying tumor cells is an immunomodulator crucial to full-blown MDSC accumulation in the TIME. We provide evidence that extracellular TCTP mediates recruitment of the polymorphonuclear MDSC (PMN-MDSC) population in the TIME via activation of Toll-like receptor-2. As further proof of principle, we show that inhibition of TCTP suppresses PMN-MDSC accumulation and tumor growth. In human cancers, we find an elevation of TCTP and an inverse correlation of TCTP gene dosage with antitumor immune signatures and clinical prognosis. This study reveals the hitherto poorly understood mechanism of the MDSC dynamics in the TIME, offering a new rationale for cancer immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Chemokine CXCL1/metabolism , Colorectal Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Toll-Like Receptor 2/immunology , Tumor Microenvironment/immunology , Alarmins/genetics , Alarmins/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Tumor Protein, Translationally-Controlled 1
5.
Nat Immunol ; 22(2): 154-165, 2021 02.
Article in English | MEDLINE | ID: mdl-33398185

ABSTRACT

Inflammatory caspase sensing of cytosolic lipopolysaccharide (LPS) triggers pyroptosis and the concurrent release of damage-associated molecular patterns (DAMPs). Collectively, DAMPs are key determinants that shape the aftermath of inflammatory cell death. However, the identity and function of the individual DAMPs released are poorly defined. Our proteomics study revealed that cytosolic LPS sensing triggered the release of galectin-1, a ß-galactoside-binding lectin. Galectin-1 release is a common feature of inflammatory cell death, including necroptosis. In vivo studies using galectin-1-deficient mice, recombinant galectin-1 and galectin-1-neutralizing antibody showed that galectin-1 promotes inflammation and plays a detrimental role in LPS-induced lethality. Mechanistically, galectin-1 inhibition of CD45 (Ptprc) underlies its unfavorable role in endotoxin shock. Finally, we found increased galectin-1 in sera from human patients with sepsis. Overall, we uncovered galectin-1 as a bona fide DAMP released as a consequence of cytosolic LPS sensing, identifying a new outcome of inflammatory cell death.


Subject(s)
Alarmins/metabolism , Endotoxemia/immunology , Galectin 1/metabolism , Inflammation Mediators/metabolism , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Phosphate-Binding Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alarmins/deficiency , Alarmins/genetics , Animals , Case-Control Studies , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/metabolism , Endotoxemia/pathology , Female , Galectin 1/blood , Galectin 1/deficiency , Galectin 1/genetics , HeLa Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Leukocyte Common Antigens/metabolism , Lipopolysaccharides , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Necroptosis , Phosphate-Binding Proteins/deficiency , Phosphate-Binding Proteins/genetics , RAW 264.7 Cells , Sepsis/blood , Sepsis/diagnosis , Signal Transduction , Up-Regulation
6.
Immunity ; 57(4): 752-771, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38599169

ABSTRACT

Damage-associated molecular patterns (DAMPs) are endogenous danger molecules produced in cellular damage or stress, and they can activate the innate immune system. DAMPs contain multiple types of molecules, including nucleic acids, proteins, ions, glycans, and metabolites. Although these endogenous molecules do not trigger immune response under steady-state condition, they may undergo changes in distribution, physical or chemical property, or concentration upon cellular damage or stress, and then they become DAMPs that can be sensed by innate immune receptors to induce inflammatory response. Thus, DAMPs play an important role in inflammation and inflammatory diseases. In this review, we summarize the conversion of homeostatic molecules into DAMPs; the diverse nature and classification, cellular origin, and sensing of DAMPs; and their role in inflammation and related diseases. Furthermore, we discuss the clinical strategies to treat DAMP-associated diseases via targeting DAMP-sensing receptors.


Subject(s)
Inflammation , Nucleic Acids , Humans , Immunity, Innate , Receptors, Immunologic , Alarmins
7.
Nat Immunol ; 21(11): 1319-1326, 2020 11.
Article in English | MEDLINE | ID: mdl-33077953

ABSTRACT

Injury is a key driver of inflammation, a critical yet necessary response involving several mediators that is aimed at restoring tissue homeostasis. Inflammation in the central nervous system can be triggered by a variety of stimuli, some intrinsic to the brain and others arising from peripheral signals. Fine-tuned regulation of this response is crucial in a system that is vulnerable due to, for example, aging and ongoing neurodegeneration. In this context, seemingly harmless interventions like a common surgery to repair a broken limb can overwhelm the immune system and become the driver of further complications such as delirium and other perioperative neurocognitive disorders. Here, we discuss potential mechanisms by which the immune system affects the central nervous system after surgical trauma. Together, these neuroimmune interactions are becoming hallmarks of and potential therapeutic targets for multiple neurologic conditions, including those affecting the perioperative space.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Postoperative Complications , Alarmins/genetics , Alarmins/metabolism , Animals , Blood Coagulation , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Blood-Brain Barrier/metabolism , Combined Modality Therapy , Complement Activation/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Management , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Inflammation/diagnosis , Inflammation/therapy , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy , Neuroglia/immunology , Neuroglia/metabolism , Neuroimmunomodulation , Treatment Outcome
8.
Nat Immunol ; 21(1): 42-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31768073

ABSTRACT

Pathogen-associated molecular patterns (PAMPs) have the capacity to couple inflammatory gene expression to changes in macrophage metabolism, both of which influence subsequent inflammatory activities. Similar to their microbial counterparts, several self-encoded damage-associated molecular patterns (DAMPs) induce inflammatory gene expression. However, whether this symmetry in host responses between PAMPs and DAMPs extends to metabolic shifts is unclear. Here, we report that the self-encoded oxidized phospholipid oxPAPC alters the metabolism of macrophages exposed to lipopolysaccharide. While cells activated by lipopolysaccharide rely exclusively on glycolysis, macrophages exposed to oxPAPC also use mitochondrial respiration, feed the Krebs cycle with glutamine, and favor the accumulation of oxaloacetate in the cytoplasm. This metabolite potentiates interleukin-1ß production, resulting in hyperinflammation. Similar metabolic adaptions occur in vivo in hypercholesterolemic mice and human subjects. Drugs that interfere with oxPAPC-driven metabolic changes reduce atherosclerotic plaque formation in mice, thereby underscoring the importance of DAMP-mediated activities in pathophysiological conditions.


Subject(s)
Alarmins/immunology , Lipopolysaccharides/immunology , Macrophages/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Phosphatidylcholines/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Glycolysis/physiology , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Inflammation/prevention & control , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Oxidative Phosphorylation , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control
9.
Immunity ; 56(3): 467-469, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36921571

ABSTRACT

Timely labor is critical for both infant and maternal health, yet the mechanisms underlying the initiation of childbirth remain unclear. In this issue of Immunity, Siewiera et al. demonstrate a vital role for innate type 2 immune responses in controlling uterus-intrinsic onset of labor in mice.1.


Subject(s)
Alarmins , Interleukin-33 , Female , Mice , Animals , Uterus , Immunity, Innate
10.
Immunity ; 56(11): 2542-2554.e7, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37714152

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are crucial in promoting type 2 inflammation that contributes to both anti-parasite immunity and allergic diseases. However, the molecular checkpoints in ILC2s that determine whether to immediately launch a proinflammatory response are unknown. Here, we found that retinoid X receptor gamma (Rxrg) was highly expressed in small intestinal ILC2s and rapidly suppressed by alarmin cytokines. Genetic deletion of Rxrg did not impact ILC2 development but facilitated ILC2 responses and the tissue inflammation induced by alarmins. Mechanistically, RXRγ maintained the expression of its target genes that support intracellular cholesterol efflux, which in turn reduce ILC2 proliferation. Furthermore, RXRγ expression prevented ILC2 response to mild stimulations, including low doses of alarmin cytokine and mechanical skin injury. Together, we propose that RXRγ expression and its mediated lipid metabolic states function as a cell-intrinsic checkpoint that confers the threshold of ILC2 activation in the small intestine.


Subject(s)
Immunity, Innate , Retinoid X Receptor gamma , Humans , Alarmins , Lymphocytes , Inflammation , Cytokines/metabolism , Intestine, Small/metabolism
11.
Immunity ; 56(4): 813-828.e10, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36809763

ABSTRACT

T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-33 , Lymphocytic Choriomeningitis , Animals , Mice , Alarmins/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Persistent Infection , T Cell Transcription Factor 1/metabolism
12.
Immunity ; 55(4): 575-577, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417668

ABSTRACT

Epithelial cells (tuft and goblet cells) interact with immune cells on the "inside" while secreting effector molecules into the topological "outside." In this issue of Immunity, Zhao et al. investigate an interleukin-33 (IL-33) secretion mechanism in goblet cells dependent on O-GlcNAcylation and gasdermin pores facilitating worm expulsion.


Subject(s)
Alarmins , Nippostrongylus , Animals , Epithelial Cells , Goblet Cells , Interleukin-13
13.
Immunity ; 55(4): 623-638.e5, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385697

ABSTRACT

The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.


Subject(s)
Alarmins , Intestinal Mucosa , Acylation , Alarmins/immunology , Anthelmintics/immunology , Biomarkers, Tumor , Cytokines , DNA-Binding Proteins , Helminthiasis/immunology , Humans , Hyperplasia , Inflammation , Interleukin-33 , Intestinal Mucosa/immunology , Mebendazole , N-Acetylglucosaminyltransferases/immunology , Pore Forming Cytotoxic Proteins , STAT6 Transcription Factor/immunology
14.
Immunity ; 54(5): 1083-1095.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891889

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV-2 infection. We profiled MIS-C, adult COVID-19, and healthy pediatric and adult individuals using single-cell RNA sequencing, flow cytometry, antigen receptor repertoire analysis, and unbiased serum proteomics, which collectively identified a signature in MIS-C patients that correlated with disease severity. Despite having no evidence of active infection, MIS-C patients had elevated S100A-family alarmins and decreased antigen presentation signatures, indicative of myeloid dysfunction. MIS-C patients showed elevated expression of cytotoxicity genes in NK and CD8+ T cells and expansion of specific IgG-expressing plasmablasts. Clinically severe MIS-C patients displayed skewed memory T cell TCR repertoires and autoimmunity characterized by endothelium-reactive IgG. The alarmin, cytotoxicity, TCR repertoire, and plasmablast signatures we defined have potential for application in the clinic to better diagnose and potentially predict disease severity early in the course of MIS-C.


Subject(s)
COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Adolescent , Alarmins/immunology , Autoantibodies/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cytotoxicity, Immunologic/genetics , Endothelium/immunology , Endothelium/pathology , Humans , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Plasma Cells/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Severity of Illness Index
15.
Nature ; 616(7956): 348-356, 2023 04.
Article in English | MEDLINE | ID: mdl-37020026

ABSTRACT

Natural killer (NK) cell kill infected, transformed and stressed cells when an activating NK cell receptor is triggered1. Most NK cells and some innate lymphoid cells express the activating receptor NKp46, encoded by NCR1, the most evolutionarily ancient NK cell receptor2,3. Blockage of NKp46 inhibits NK killing of many cancer targets4. Although a few infectious NKp46 ligands have been identified, the endogenous NKp46 cell surface ligand is unknown. Here we show that NKp46 recognizes externalized calreticulin (ecto-CRT), which translocates from the endoplasmic reticulum (ER) to the cell membrane during ER stress. ER stress and ecto-CRT are hallmarks of chemotherapy-induced immunogenic cell death5,6, flavivirus infection and senescence. NKp46 recognition of the P domain of ecto-CRT triggers NK cell signalling and NKp46 caps with ecto-CRT in NK immune synapses. NKp46-mediated killing is inhibited by knockout or knockdown of CALR, the gene encoding CRT, or CRT antibodies, and is enhanced by ectopic expression of glycosylphosphatidylinositol-anchored CRT. NCR1)-deficient human (and Nrc1-deficient mouse) NK cells are impaired in the killing of ZIKV-infected, ER-stressed and senescent cells and ecto-CRT-expressing cancer cells. Importantly, NKp46 recognition of ecto-CRT controls mouse B16 melanoma and RAS-driven lung cancers and enhances tumour-infiltrating NK cell degranulation and cytokine secretion. Thus, NKp46 recognition of ecto-CRT as a danger-associated molecular pattern eliminates ER-stressed cells.


Subject(s)
Calreticulin , Endoplasmic Reticulum Stress , Killer Cells, Natural , Natural Cytotoxicity Triggering Receptor 1 , Animals , Humans , Mice , Alarmins/metabolism , Calreticulin/immunology , Calreticulin/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Immunity, Innate , Immunological Synapses , Killer Cells, Natural/metabolism , Lung Neoplasms/metabolism , Melanoma, Experimental/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Zika Virus/physiology
16.
Nature ; 618(7967): 1072-1077, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37196676

ABSTRACT

Plasma membrane rupture (PMR) in dying cells undergoing pyroptosis or apoptosis requires the cell-surface protein NINJ11. PMR releases pro-inflammatory cytoplasmic molecules, collectively called damage-associated molecular patterns (DAMPs), that activate immune cells. Therefore, inhibiting NINJ1 and PMR may limit the inflammation that is associated with excessive cell death. Here we describe an anti-NINJ1 monoclonal antibody that specifically targets mouse NINJ1 and blocks oligomerization of NINJ1, preventing PMR. Electron microscopy studies showed that this antibody prevents NINJ1 from forming oligomeric filaments. In mice, inhibition of NINJ1 or Ninj1 deficiency ameliorated hepatocellular PMR induced with TNF plus D-galactosamine, concanavalin A, Jo2 anti-Fas agonist antibody or ischaemia-reperfusion injury. Accordingly, serum levels of lactate dehydrogenase, the liver enzymes alanine aminotransaminase and aspartate aminotransferase, and the DAMPs interleukin 18 and HMGB1 were reduced. Moreover, in the liver ischaemia-reperfusion injury model, there was an attendant reduction in neutrophil infiltration. These data indicate that NINJ1 mediates PMR and inflammation in diseases driven by aberrant hepatocellular death.


Subject(s)
Antibodies, Monoclonal , Cell Membrane , Inflammation , Liver , Nerve Growth Factors , Reperfusion Injury , Animals , Mice , Alanine Transaminase , Alarmins , Antibodies, Monoclonal/immunology , Aspartate Aminotransferases , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/immunology , Cell Adhesion Molecules, Neuronal/ultrastructure , Cell Death , Cell Membrane/pathology , Cell Membrane/ultrastructure , Concanavalin A , Galactosamine , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inflammation/pathology , Lactate Dehydrogenases , Liver/pathology , Microscopy, Electron , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/deficiency , Nerve Growth Factors/immunology , Nerve Growth Factors/ultrastructure , Neutrophil Infiltration , Reperfusion Injury/pathology
17.
EMBO J ; 43(7): 1164-1186, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38396301

ABSTRACT

Ferroptosis is a regulated form of necrotic cell death caused by iron-dependent accumulation of oxidized phospholipids in cellular membranes, culminating in plasma membrane rupture (PMR) and cell lysis. PMR is also a hallmark of other types of programmed necrosis, such as pyroptosis and necroptosis, where it is initiated by dedicated pore-forming cell death-executing factors. However, whether ferroptosis-associated PMR is also actively executed by proteins or driven by osmotic pressure remains unknown. Here, we investigate a potential ferroptosis role of ninjurin-1 (NINJ1), a recently identified executor of pyroptosis-associated PMR. We report that NINJ1 oligomerizes during ferroptosis, and that Ninj1-deficiency protects macrophages and fibroblasts from ferroptosis-associated PMR. Mechanistically, we find that NINJ1 is dispensable for the initial steps of ferroptosis, such as lipid peroxidation, channel-mediated calcium influx, and cell swelling. In contrast, NINJ1 is required for early loss of plasma membrane integrity, which precedes complete PMR. Furthermore, NINJ1 mediates the release of cytosolic proteins and danger-associated molecular pattern (DAMP) molecules from ferroptotic cells, suggesting that targeting NINJ1 could be a therapeutic option to reduce ferroptosis-associated inflammation.


Subject(s)
Alarmins , Ferroptosis , Humans , Necrosis/metabolism , Cell Death , Cell Membrane/metabolism , Nerve Growth Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism
18.
Nat Immunol ; 17(2): 122-31, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26784265

ABSTRACT

Interleukin 33 (IL-33) is a member of the IL-1 family of cytokines with a growing number of target cells and a plethora of biological functions. Although it has commonalities with other IL-1 cytokines, IL-33 exhibits some unique features. Here we review the biology of IL-33 and its receptor and develop a working model that describes two 'lives' for IL-33-one intracellular and one extracellular. Under healthy conditions, constitutively produced, intracellular IL-33 participates in maintaining barrier function by regulating gene expression as a nuclear protein. In parallel, nuclear IL-33 functions as a stored alarmin that is released when barriers are breached. Extracellular IL-33 coordinates immune defense and repair mechanisms while also initiating differentiation of helper T cells as the adaptive immune response is triggered.


Subject(s)
Alarmins/metabolism , Interleukin-33/metabolism , Animals , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-33/chemistry , Interleukin-33/genetics , Protein Binding , Protein Transport , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Signal Transduction
19.
Nat Immunol ; 17(8): 906-13, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27434011

ABSTRACT

Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1ß are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1ß, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.


Subject(s)
Inflammation/physiopathology , Interleukin-1alpha/physiology , Alarmins/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation , Granuloma/etiology , Granuloma/metabolism , Humans , Inflammation/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/genetics , Macrophages/physiology , Mice , Mice, Inbred BALB C , Models, Biological , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/physiopathology , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational , Receptors, Interleukin-1/physiology , Signal Transduction
20.
Immunity ; 51(4): 709-723.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31604686

ABSTRACT

Neuroimmune interactions have emerged as critical modulators of allergic inflammation, and type 2 innate lymphoid cells (ILC2s) are an important cell type for mediating these interactions. Here, we show that ILC2s expressed both the neuropeptide calcitonin gene-related peptide (CGRP) and its receptor. CGRP potently inhibited alarmin-driven type 2 cytokine production and proliferation by lung ILC2s both in vitro and in vivo. CGRP induced marked changes in ILC2 expression programs in vivo and in vitro, attenuating alarmin-driven proliferative and effector responses. A distinct subset of ILCs scored highly for a CGRP-specific gene signature after in vivo alarmin stimulation, suggesting CGRP regulated this response. Finally, we observed increased ILC2 proliferation and type 2 cytokine production as well as exaggerated responses to alarmins in mice lacking the CGRP receptor. Together, these data indicate that endogenous CGRP is a critical negative regulator of ILC2 responses in vivo.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Lymphocytes/physiology , Neuropeptides/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Alarmins/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Cell Proliferation , Cells, Cultured , Feedback, Physiological , Immunity, Innate , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Neuropeptides/genetics , Receptors, Calcitonin Gene-Related Peptide/genetics , Signal Transduction , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL