Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61.321
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 455-485, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32004099

ABSTRACT

Immune cells use a variety of membrane-disrupting proteins [complement, perforin, perforin-2, granulysin, gasdermins, mixed lineage kinase domain-like pseudokinase (MLKL)] to induce different kinds of death of microbes and host cells, some of which cause inflammation. After activation by proteolytic cleavage or phosphorylation, these proteins oligomerize, bind to membrane lipids, and disrupt membrane integrity. These membrane disruptors play a critical role in both innate and adaptive immunity. Here we review our current knowledge of the functions, specificity, activation, and regulation of membrane-disrupting immune proteins and what is known about the mechanisms behind membrane damage, the structure of the pores they form, how the cells expressing these lethal proteins are protected, and how cells targeted for destruction can sometimes escape death by repairing membrane damage.


Subject(s)
Cytotoxicity, Immunologic , Host-Pathogen Interactions/immunology , Immunity , Pore Forming Cytotoxic Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Cell Membrane/immunology , Cell Membrane/metabolism , Complement Membrane Attack Complex , Complement System Proteins/immunology , Complement System Proteins/metabolism , Gene Expression Regulation , Humans , Immune System/immunology , Immune System/metabolism , Lipid Metabolism , Necroptosis/genetics , Necroptosis/immunology , Necrosis/genetics , Necrosis/immunology , Necrosis/metabolism , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Structure-Activity Relationship
2.
Annu Rev Immunol ; 36: 639-665, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29400999

ABSTRACT

Granulomas are organized aggregates of macrophages, often with characteristic morphological changes, and other immune cells. These evolutionarily ancient structures form in response to persistent particulate stimuli-infectious or noninfectious-that individual macrophages cannot eradicate. Granulomas evolved as protective responses to destroy or sequester particles but are frequently pathological in the context of foreign bodies, infections, and inflammatory diseases. We summarize recent findings that suggest that the granulomatous response unfolds in a stepwise program characterized by a series of macrophage activations and transformations that in turn recruit additional cells and produce structural changes. We explore why different granulomas vary and the reasons that granulomas are protective and pathogenic. Understanding the mechanisms and role of granuloma formation may uncover new therapies for the multitude of granulomatous diseases that constitute serious medical problems while enhancing the protective function of granulomas in infections.


Subject(s)
Granuloma/diagnosis , Granuloma/etiology , Animals , Diagnosis, Differential , Fibrosis , Host-Pathogen Interactions/immunology , Humans , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Necrosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
3.
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
4.
Cell ; 184(7): 1757-1774.e14, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33761328

ABSTRACT

The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.


Subject(s)
Granuloma/pathology , Immunity/physiology , Mycobacterium Infections, Nontuberculous/pathology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation , Disease Models, Animal , Epithelioid Cells/cytology , Epithelioid Cells/immunology , Epithelioid Cells/metabolism , Granuloma/immunology , Granuloma/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/isolation & purification , Mycobacterium marinum/physiology , Necrosis , RNA, Guide, Kinetoplastida/metabolism , Receptors, Interleukin-4/antagonists & inhibitors , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction , Zebrafish/growth & development , Zebrafish/metabolism
5.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125891

ABSTRACT

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Immunity , Mycobacterium tuberculosis/physiology , Myelopoiesis , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Susceptibility , Homeostasis , Interferon Type I/metabolism , Iron/metabolism , Kinetics , Lung/microbiology , Lung/pathology , Macrophages/immunology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Necrosis , Signal Transduction , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
6.
Cell ; 180(6): 1115-1129.e13, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200799

ABSTRACT

Influenza A virus (IAV) is a lytic RNA virus that triggers receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated pathways of apoptosis and mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptosis in infected cells. ZBP1 initiates RIPK3-driven cell death by sensing IAV RNA and activating RIPK3. Here, we show that replicating IAV generates Z-RNAs, which activate ZBP1 in the nucleus of infected cells. ZBP1 then initiates RIPK3-mediated MLKL activation in the nucleus, resulting in nuclear envelope disruption, leakage of DNA into the cytosol, and eventual necroptosis. Cell death induced by nuclear MLKL was a potent activator of neutrophils, a cell type known to drive inflammatory pathology in virulent IAV disease. Consequently, MLKL-deficient mice manifest reduced nuclear disruption of lung epithelia, decreased neutrophil recruitment into infected lungs, and increased survival following a lethal dose of IAV. These results implicate Z-RNA as a new pathogen-associated molecular pattern and describe a ZBP1-initiated nucleus-to-plasma membrane "inside-out" death pathway with potentially pathogenic consequences in severe cases of influenza.


Subject(s)
Influenza A virus/genetics , Necroptosis/genetics , RNA-Binding Proteins/metabolism , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Line, Tumor , Female , Influenza A virus/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/metabolism , Phosphorylation , Protein Kinases/metabolism , RNA/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
7.
Cell ; 178(6): 1344-1361.e11, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474371

ABSTRACT

Necrosis of infected macrophages constitutes a critical pathogenetic event in tuberculosis by releasing mycobacteria into the growth-permissive extracellular environment. In zebrafish infected with Mycobacterium marinum or Mycobacterium tuberculosis, excess tumor necrosis factor triggers programmed necrosis of infected macrophages through the production of mitochondrial reactive oxygen species (ROS) and the participation of cyclophilin D, a component of the mitochondrial permeability transition pore. Here, we show that this necrosis pathway is not mitochondrion-intrinsic but results from an inter-organellar circuit initiating and culminating in the mitochondrion. Mitochondrial ROS induce production of lysosomal ceramide that ultimately activates the cytosolic protein BAX. BAX promotes calcium flow from the endoplasmic reticulum into the mitochondrion through ryanodine receptors, and the resultant mitochondrial calcium overload triggers cyclophilin-D-mediated necrosis. We identify ryanodine receptors and plasma membrane L-type calcium channels as druggable targets to intercept mitochondrial calcium overload and necrosis of mycobacterium-infected zebrafish and human macrophages.


Subject(s)
Macrophages/microbiology , Macrophages/pathology , Mitochondria/metabolism , Mycobacterium Infections, Nontuberculous/metabolism , Tuberculosis/immunology , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Calcium/metabolism , Endoplasmic Reticulum/microbiology , Humans , Lysosomes/microbiology , Membrane Potential, Mitochondrial , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium marinum , Mycobacterium tuberculosis , Necrosis , Reactive Oxygen Species/metabolism , THP-1 Cells , Zebrafish
8.
Immunity ; 57(3): 429-445, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38479360

ABSTRACT

Diverse inflammatory conditions, from infections to autoimmune disease, are often associated with cellular damage and death. Apoptotic cell death has evolved to minimize its inflammatory potential. By contrast, necrotic cell death via necroptosis and pyroptosis-driven by membrane-damaging MLKL and gasdermins, respectively-can both initiate and propagate inflammatory responses. In this review, we provide insights into the function and regulation of MLKL and gasdermin necrotic effector proteins and drivers of plasma membrane rupture. We evaluate genetic evidence that MLKL- and gasdermin-driven necrosis may either provide protection against, or contribute to, disease states in a context-dependent manner. These cumulative insights using gene-targeted mice underscore the necessity for future research examining pyroptotic and necroptotic cell death in human tissue, as a basis for developing specific necrotic inhibitors with the potential to benefit a spectrum of pathological conditions.


Subject(s)
Apoptosis , Gasdermins , Humans , Animals , Mice , Necrosis/metabolism , Apoptosis/physiology , Pyroptosis/physiology , Cell Death , Inflammasomes/metabolism , Protein Kinases/metabolism
9.
Cell ; 173(5): 1068-1070, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775589

ABSTRACT

A new solid-state NMR study determines the high-resolution hetero-amyloid structure of the RIPK1-RIPK3 signaling complex that is involved in mediating necroptosis. The structure demonstrates specific formation of hetero-amyloids over homo-amyloids and the structural basis for a functional amyloid to act as a platform to recruit and activate downstream partners in intracellular signaling.


Subject(s)
Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases , Amyloidogenic Proteins , Apoptosis , Humans , Signal Transduction , Tumor Necrosis Factor-alpha
10.
Cell ; 173(5): 1111-1122.e10, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29606355

ABSTRACT

The development of interventions to prevent congenital Zika syndrome (CZS) has been limited by the lack of an established nonhuman primate model. Here we show that infection of female rhesus monkeys early in pregnancy with Zika virus (ZIKV) recapitulates many features of CZS in humans. We infected 9 pregnant monkeys with ZIKV, 6 early in pregnancy (weeks 6-7 of gestation) and 3 later in pregnancy (weeks 12-14 of gestation), and compared findings with uninfected controls. 100% (6 of 6) of monkeys infected early in pregnancy exhibited prolonged maternal viremia and fetal neuropathology, including fetal loss, smaller brain size, and histopathologic brain lesions, including microcalcifications, hemorrhage, necrosis, vasculitis, gliosis, and apoptosis of neuroprogenitor cells. High-resolution MRI demonstrated concordant lesions indicative of deep gray matter injury. We also observed spinal, ocular, and neuromuscular pathology. Our data show that vascular compromise and neuroprogenitor cell dysfunction are hallmarks of CZS pathogenesis, suggesting novel strategies to prevent and to treat this disease.


Subject(s)
Fetus/virology , Neurons/pathology , Zika Virus Infection/pathology , Zika Virus/pathogenicity , Animals , Animals, Newborn , Apoptosis , Brain/diagnostic imaging , Brain/pathology , Calcinosis/pathology , Calcinosis/veterinary , Female , Gestational Age , Macaca mulatta , Magnetic Resonance Imaging , Necrosis , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Neurons/virology , Pregnancy , Severity of Illness Index , Vasculitis/pathology , Vasculitis/veterinary , Zika Virus Infection/veterinary , Zika Virus Infection/virology
11.
Immunity ; 56(10): 2325-2341.e15, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37652021

ABSTRACT

Maladaptive, non-resolving inflammation contributes to chronic inflammatory diseases such as atherosclerosis. Because macrophages remove necrotic cells, defective macrophage programs can promote chronic inflammation with persistent tissue injury. Here, we investigated the mechanisms sustaining vascular macrophages. Intravital imaging revealed a spatiotemporal macrophage niche across vascular beds alongside mural cells (MCs)-pericytes and smooth muscle cells. Single-cell transcriptomics, co-culture, and genetic deletion experiments revealed MC-derived expression of the chemokines CCL2 and MIF, which actively preserved macrophage survival and their homeostatic functions. In atherosclerosis, this positioned macrophages in viable plaque areas, away from the necrotic core, and maintained a homeostatic macrophage phenotype. Disruption of this MC-macrophage unit via MC-specific deletion of these chemokines triggered detrimental macrophage relocalizing, exacerbated plaque necrosis, inflammation, and atheroprogression. In line, CCL2 inhibition at advanced stages of atherosclerosis showed detrimental effects. This work presents a MC-driven safeguard toward maintaining the homeostatic vascular macrophage niche.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Macrophages/metabolism , Atherosclerosis/metabolism , Plaque, Atherosclerotic/metabolism , Chemokines/metabolism , Inflammation/metabolism , Necrosis/metabolism
12.
Cell ; 169(2): 186-187, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388403

ABSTRACT

Necroptosis is a highly inflammatory form of programmed cell death that results from MLKL-mediated disruption of the cell membrane. In this issue of Cell, Gong et al. challenge the notion that MLKL activation is a point of no return by identifying mechanisms to counterbalance necroptosis, sustain plasma membrane integrity, and prolong cell viability.


Subject(s)
Protein Kinases , Receptor-Interacting Protein Serine-Threonine Kinases , Apoptosis , Necrosis , Phosphorylation
13.
Cell ; 169(2): 301-313.e11, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28366204

ABSTRACT

Receptor-interacting protein kinase-3 (RIPK3) is an activator of necroptotic cell death, but recent work has implicated additional roles for RIPK3 in inflammatory signaling independent of cell death. However, while necroptosis has been shown to contribute to antiviral immunity, death-independent roles for RIPK3 in host defense have not been demonstrated. Using a mouse model of West Nile virus (WNV) encephalitis, we show that RIPK3 restricts WNV pathogenesis independently of cell death. Ripk3-/- mice exhibited enhanced mortality compared to wild-type (WT) controls, while mice lacking the necroptotic effector MLKL, or both MLKL and caspase-8, were unaffected. The enhanced susceptibility of Ripk3-/- mice arose from suppressed neuronal chemokine expression and decreased central nervous system (CNS) recruitment of T lymphocytes and inflammatory myeloid cells, while peripheral immunity remained intact. These data identify pleiotropic functions for RIPK3 in the restriction of viral pathogenesis and implicate RIPK3 as a key coordinator of immune responses within the CNS.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , West Nile Fever/immunology , West Nile virus/physiology , Animals , Central Nervous System/metabolism , Chemokines/immunology , Leukocytes/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Necrosis , Neurons/metabolism
14.
Cell ; 169(2): 286-300.e16, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388412

ABSTRACT

The activation of mixed lineage kinase-like (MLKL) by receptor-interacting protein kinase-3 (RIPK3) results in plasma membrane (PM) disruption and a form of regulated necrosis, called necroptosis. Here, we show that, during necroptosis, MLKL-dependent calcium (Ca2+) influx and phosphatidylserine (PS) exposure on the outer leaflet of the plasma membrane preceded loss of PM integrity. Activation of MLKL results in the generation of broken, PM "bubbles" with exposed PS that are released from the surface of the otherwise intact cell. The ESCRT-III machinery is required for formation of these bubbles and acts to sustain survival of the cell when MLKL activation is limited or reversed. Under conditions of necroptotic cell death, ESCRT-III controls the duration of plasma membrane integrity. As a consequence of the action of ESCRT-III, cells undergoing necroptosis can express chemokines and other regulatory molecules and promote antigenic cross-priming of CD8+ T cells.


Subject(s)
Cell Membrane/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Necrosis/metabolism , Animals , Calcium/metabolism , Cell Survival , HT29 Cells , Humans , Jurkat Cells , Mice , NIH 3T3 Cells , Phosphatidylserines , Protein Kinases/metabolism , Signal Transduction
15.
Annu Rev Cell Dev Biol ; 34: 311-332, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30089222

ABSTRACT

Balancing cell death and survival is essential for normal development and homeostasis and for preventing diseases, especially cancer. Conventional cell death pathways include apoptosis, a form of programmed cell death controlled by a well-defined biochemical pathway, and necrosis, the lysis of acutely injured cells. New types of regulated cell death include necroptosis, pyroptosis, ferroptosis, phagoptosis, and entosis. Autophagy can promote survival or can cause death. Newly described processes of anastasis and resuscitation show that, remarkably, cells can recover from the brink of apoptosis or necroptosis. Important new work shows that epithelia achieve homeostasis by extruding excess cells, which then die by anoikis due to loss of survival signals. This mechanically regulated process both maintains barrier function as cells die and matches rates of proliferation and death. In this review, we describe these unconventional ways in which cells have evolved to die or survive, as well as the contributions that these processes make to homeostasis and cancer.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Necrosis/genetics , Neoplasms/genetics , Anoikis/genetics , Cell Proliferation/genetics , Entosis/genetics , Homeostasis/genetics , Humans , Pyroptosis/genetics , Signal Transduction/genetics
16.
Annu Rev Biochem ; 85: 743-63, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-26865533

ABSTRACT

Necroptosis is a regulated form of necrosis, with the dying cell rupturing and releasing intracellular components that can trigger an innate immune response. Toll-like receptor 3 and 4 agonists, tumor necrosis factor, certain viral infections, or the T cell receptor can trigger necroptosis if the activity of the protease caspase-8 is compromised. Necroptosis signaling is modulated by the kinase RIPK1 and requires the kinase RIPK3 and the pseudokinase MLKL. Either RIPK3 deficiency or RIPK1 inhibition confers resistance in various animal disease models, suggesting that inflammation caused by necroptosis contributes to tissue damage and that inhibitors of these kinases could have therapeutic potential. Recent studies have revealed unexpected complexity in the regulation of cell death programs by RIPK1 and RIPK3 with the possibility that necroptosis is but one mechanism by which these kinases promote inflammation.


Subject(s)
Gene Expression Regulation , Necrosis/genetics , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis , Caspase 8/genetics , Caspase 8/immunology , Gene Expression Profiling , Humans , Immunity, Innate , Inflammation , Necrosis/immunology , Necrosis/pathology , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
17.
Cell ; 167(7): 1693-1704, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984721

ABSTRACT

Caspases were originally identified as important mediators of inflammatory response and apoptosis. Recent discoveries, however, have unveiled their roles in mediating and suppressing two regulated forms of necrotic cell death, termed pyroptosis and necroptosis, respectively. These recent advances have significantly expanded our understanding of the roles of caspases in regulating development, adult homeostasis, and host defense response.


Subject(s)
Caspases/metabolism , Necrosis/metabolism , Animals , Apoptosis , Humans , Infections/enzymology , Infections/metabolism , Infections/pathology , Inflammation/enzymology , Inflammation/metabolism , Inflammation/pathology , Necrosis/enzymology , Pyroptosis
18.
Mol Cell ; 83(15): 2621-2623, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541218

ABSTRACT

Red blood cell rupture (hemolysis) activates innate immunity and inflammation by releasing heme. Sundaram et al.1 implicate the immune sensor NLRP12 in hemolytic disease, showing that it controls necrotic cell death induction in response to heme combined with pathogen-associated molecules.


Subject(s)
Heme , Pathogen-Associated Molecular Pattern Molecules , Humans , Heme/metabolism , Inflammation/metabolism , Immunity, Innate , Hemolysis , Necrosis , Cell Death , Intracellular Signaling Peptides and Proteins
19.
Cell ; 160(1-2): 48-61, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594174

ABSTRACT

How the genomic landscape of a tumor shapes and is shaped by anti-tumor immunity has not been systematically explored. Using large-scale genomic data sets of solid tissue tumor biopsies, we quantified the cytolytic activity of the local immune infiltrate and identified associated properties across 18 tumor types. The number of predicted MHC Class I-associated neoantigens was correlated with cytolytic activity and was lower than expected in colorectal and other tumors, suggesting immune-mediated elimination. We identified recurrently mutated genes that showed positive association with cytolytic activity, including beta-2-microglobulin (B2M), HLA-A, -B and -C and Caspase 8 (CASP8), highlighting loss of antigen presentation and blockade of extrinsic apoptosis as key strategies of resistance to cytolytic activity. Genetic amplifications were also associated with high cytolytic activity, including immunosuppressive factors such as PDL1/2 and ALOX12B/15B. Our genetic findings thus provide evidence for immunoediting in tumors and uncover mechanisms of tumor-intrinsic resistance to cytolytic activity.


Subject(s)
Neoplasms/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , Apoptosis , DNA Copy Number Variations , Databases, Genetic , Endogenous Retroviruses , Genome, Human , Humans , Mutation , Necrosis , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology
20.
Nature ; 630(8015): 158-165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38693268

ABSTRACT

The liver has a unique ability to regenerate1,2; however, in the setting of acute liver failure (ALF), this regenerative capacity is often overwhelmed, leaving emergency liver transplantation as the only curative option3-5. Here, to advance understanding of human liver regeneration, we use paired single-nucleus RNA sequencing combined with spatial profiling of healthy and ALF explant human livers to generate a single-cell, pan-lineage atlas of human liver regeneration. We uncover a novel ANXA2+ migratory hepatocyte subpopulation, which emerges during human liver regeneration, and a corollary subpopulation in a mouse model of acetaminophen (APAP)-induced liver regeneration. Interrogation of necrotic wound closure and hepatocyte proliferation across multiple timepoints following APAP-induced liver injury in mice demonstrates that wound closure precedes hepatocyte proliferation. Four-dimensional intravital imaging of APAP-induced mouse liver injury identifies motile hepatocytes at the edge of the necrotic area, enabling collective migration of the hepatocyte sheet to effect wound closure. Depletion of hepatocyte ANXA2 reduces hepatocyte growth factor-induced human and mouse hepatocyte migration in vitro, and abrogates necrotic wound closure following APAP-induced mouse liver injury. Together, our work dissects unanticipated aspects of liver regeneration, demonstrating an uncoupling of wound closure and hepatocyte proliferation and uncovering a novel migratory hepatocyte subpopulation that mediates wound closure following liver injury. Therapies designed to promote rapid reconstitution of normal hepatic microarchitecture and reparation of the gut-liver barrier may advance new areas of therapeutic discovery in regenerative medicine.


Subject(s)
Liver Failure, Acute , Liver Regeneration , Animals , Female , Humans , Male , Mice , Acetaminophen/pharmacology , Cell Lineage , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/cytology , Liver/drug effects , Liver/pathology , Liver Failure, Acute/pathology , Liver Failure, Acute/chemically induced , Liver Regeneration/drug effects , Mice, Inbred C57BL , Necrosis/chemically induced , Regenerative Medicine , Single-Cell Gene Expression Analysis , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL