Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
Immunity ; 49(1): 42-55.e6, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021146

ABSTRACT

The execution of shock following high dose E. coli lipopolysaccharide (LPS) or bacterial sepsis in mice required pro-apoptotic caspase-8 in addition to pro-pyroptotic caspase-11 and gasdermin D. Hematopoietic cells produced MyD88- and TRIF-dependent inflammatory cytokines sufficient to initiate shock without any contribution from caspase-8 or caspase-11. Both proteases had to be present to support tumor necrosis factor- and interferon-ß-dependent tissue injury first observed in the small intestine and later in spleen and thymus. Caspase-11 enhanced the activation of caspase-8 and extrinsic cell death machinery within the lower small intestine. Neither caspase-8 nor caspase-11 was individually sufficient for shock. Both caspases collaborated to amplify inflammatory signals associated with tissue damage. Therefore, combined pyroptotic and apoptotic signaling mediated endotoxemia independently of RIPK1 kinase activity and RIPK3 function. These observations bring to light the relevance of tissue compartmentalization to disease processes in vivo where cytokines act in parallel to execute diverse cell death pathways.


Subject(s)
Caspase 8/metabolism , Caspases/metabolism , Escherichia coli Infections/enzymology , Escherichia coli Infections/physiopathology , Shock, Septic/enzymology , Shock, Septic/physiopathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Caspase 8/genetics , Caspases/genetics , Caspases, Initiator , Cells, Cultured , Female , Inflammation/metabolism , Inflammation/pathology , Interferon Regulatory Factor-3/genetics , Interferon-beta/blood , Interferon-beta/metabolism , Intestine, Small/pathology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spleen/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
2.
Mol Cell ; 73(3): 413-428.e7, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30598363

ABSTRACT

Receptor-interacting protein kinase (RIPK) 1 functions as a key mediator of tissue homeostasis via formation of Caspase-8 activating ripoptosome complexes, positively and negatively regulating apoptosis, necroptosis, and inflammation. Here, we report an unanticipated cell-death- and inflammation-independent function of RIPK1 and Caspase-8, promoting faithful chromosome alignment in mitosis and thereby ensuring genome stability. We find that ripoptosome complexes progressively form as cells enter mitosis, peaking at metaphase and disassembling as cells exit mitosis. Genetic deletion and mitosis-specific inhibition of Ripk1 or Caspase-8 results in chromosome alignment defects independently of MLKL. We found that Polo-like kinase 1 (PLK1) is recruited into mitotic ripoptosomes, where PLK1's activity is controlled via RIPK1-dependent recruitment and Caspase-8-mediated cleavage. A fine balance of ripoptosome assembly is required as deregulated ripoptosome activity modulates PLK1-dependent phosphorylation of downstream effectors, such as BUBR1. Our data suggest that ripoptosome-mediated regulation of PLK1 contributes to faithful chromosome segregation during mitosis.


Subject(s)
Caspase 8/metabolism , Chromosomal Instability , Colonic Neoplasms/enzymology , Fibroblasts/enzymology , Mitosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Aneuploidy , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/pathology , HT29 Cells , Humans , Inflammation/enzymology , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Polo-Like Kinase 1
3.
Mol Cell ; 70(5): 936-948.e7, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29883610

ABSTRACT

Necroptosis is an important form of lytic cell death triggered by injury and infection, but whether mixed lineage kinase domain-like (MLKL) is sufficient to execute this pathway is unknown. In a genetic selection for human cell mutants defective for MLKL-dependent necroptosis, we identified mutations in IPMK and ITPK1, which encode inositol phosphate (IP) kinases that regulate the IP code of soluble molecules. We show that IP kinases are essential for necroptosis triggered by death receptor activation, herpesvirus infection, or a pro-necrotic MLKL mutant. In IP kinase mutant cells, MLKL failed to oligomerize and localize to membranes despite proper receptor-interacting protein kinase-3 (RIPK3)-dependent phosphorylation. We demonstrate that necroptosis requires IP-specific kinase activity and that a highly phosphorylated product, but not a lowly phosphorylated precursor, potently displaces the MLKL auto-inhibitory brace region. These observations reveal control of MLKL-mediated necroptosis by a metabolite and identify a key molecular mechanism underlying regulated cell death.


Subject(s)
Colonic Neoplasms/enzymology , Inositol Phosphates/metabolism , Protein Kinases/metabolism , Binding Sites , Cell Death/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/virology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HT29 Cells , Herpesvirus 1, Human/pathogenicity , Humans , Jurkat Cells , Mutation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
4.
Immunity ; 45(1): 46-59, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27396959

ABSTRACT

Macrophages are a crucial component of the innate immune system in sensing pathogens and promoting local and systemic inflammation. RIPK1 and RIPK3 are homologous kinases, previously linked to activation of necroptotic death. In this study, we have described roles for these kinases as master regulators of pro-inflammatory gene expression induced by lipopolysaccharide, independent of their well-documented cell death functions. In primary macrophages, this regulation was elicited in the absence of caspase-8 activity, required the adaptor molecule TRIF, and proceeded in a cell autonomous manner. RIPK1 and RIPK3 kinases promoted sustained activation of Erk, cFos, and NF-κB, which were required for inflammatory changes. Utilizing genetic and pharmacologic tools, we showed that RIPK1 and RIPK3 account for acute inflammatory responses induced by lipopolysaccharide in vivo; notably, this regulation did not require exogenous manipulation of caspases. These findings identified a new pharmacologically accessible pathway that may be relevant to inflammatory pathologies.


Subject(s)
Immunity, Innate , Inflammation/immunology , Macrophages/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cells, Cultured , Female , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcriptome
6.
Nature ; 564(7736): 439-443, 2018 12.
Article in English | MEDLINE | ID: mdl-30405246

ABSTRACT

Stimulator of interferon genes (STING) is a receptor in the endoplasmic reticulum that propagates innate immune sensing of cytosolic pathogen-derived and self DNA1. The development of compounds that modulate STING has recently been the focus of intense research for the treatment of cancer and infectious diseases and as vaccine adjuvants2. To our knowledge, current efforts are focused on the development of modified cyclic dinucleotides that mimic the endogenous STING ligand cGAMP; these have progressed into clinical trials in patients with solid accessible tumours amenable to intratumoral delivery3. Here we report the discovery of a small molecule STING agonist that is not a cyclic dinucleotide and is systemically efficacious for treating tumours in mice. We developed a linking strategy to synergize the effect of two symmetry-related amidobenzimidazole (ABZI)-based compounds to create linked ABZIs (diABZIs) with enhanced binding to STING and cellular function. Intravenous administration of a diABZI STING agonist to immunocompetent mice with established syngeneic colon tumours elicited strong anti-tumour activity, with complete and lasting regression of tumours. Our findings represent a milestone in the rapidly growing field of immune-modifying cancer therapies.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Drug Design , Membrane Proteins/agonists , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Humans , Ligands , Membrane Proteins/immunology , Mice , Models, Molecular , Nucleotides, Cyclic/metabolism
7.
Mol Cell ; 60(1): 63-76, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26344099

ABSTRACT

TNF is a master pro-inflammatory cytokine. Activation of TNFR1 by TNF can result in both RIPK1-independent apoptosis and RIPK1 kinase-dependent apoptosis or necroptosis. These cell death outcomes are regulated by two distinct checkpoints during TNFR1 signaling. TNF-mediated NF-κB-dependent induction of pro-survival or anti-apoptotic molecules is a well-known late checkpoint in the pathway, protecting cells from RIPK1-independent death. On the other hand, the molecular mechanism regulating the contribution of RIPK1 to cell death is far less understood. We demonstrate here that the IKK complex phosphorylates RIPK1 at TNFR1 complex I and protects cells from RIPK1 kinase-dependent death, independent of its function in NF-κB activation. We provide in vitro and in vivo evidence that inhibition of IKKα/IKKß or its upstream activators sensitizes cells to death by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We therefore report on an unexpected, NF-κB-independent role for the IKK complex in protecting cells from RIPK1-dependent death downstream of TNFR1.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Caspase 8/metabolism , Cell Death , Cell Line , Embryo, Mammalian/cytology , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Phosphorylation , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
8.
J Immunol ; 204(9): 2337-2348, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213560

ABSTRACT

The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB-dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow-derived mast cells from MALT1 knockout (MALT1-/-) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1-/- and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell-derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.


Subject(s)
Endothelial Cells/metabolism , Hypersensitivity/metabolism , Mast Cells/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line , Cytokines/immunology , Cytokines/metabolism , Endothelial Cells/immunology , Female , Histamine/immunology , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/immunology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism
9.
Mol Cell ; 56(4): 481-95, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25459880

ABSTRACT

Receptor-interacting protein kinase 3 (RIP3 or RIPK3) has emerged as a central player in necroptosis and a potential target to control inflammatory disease. Here, three selective small-molecule compounds are shown to inhibit RIP3 kinase-dependent necroptosis, although their therapeutic value is undermined by a surprising, concentration-dependent induction of apoptosis. These compounds interact with RIP3 to activate caspase 8 (Casp8) via RHIM-driven recruitment of RIP1 (RIPK1) to assemble a Casp8-FADD-cFLIP complex completely independent of pronecrotic kinase activities and MLKL. RIP3 kinase-dead D161N mutant induces spontaneous apoptosis independent of compound, whereas D161G, D143N, and K51A mutants, like wild-type, only trigger apoptosis when compound is present. Accordingly, RIP3-K51A mutant mice (Rip3(K51A/K51A)) are viable and fertile, in stark contrast to the perinatal lethality of Rip3(D161N/D161N) mice. RIP3 therefore holds both necroptosis and apoptosis in balance through a Ripoptosome-like platform. This work highlights a common mechanism unveiling RHIM-driven apoptosis by therapeutic or genetic perturbation of RIP3.


Subject(s)
Apoptosis , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Knock-In Techniques , HT29 Cells , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Necrosis/enzymology , Nuclear Pore Complex Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
10.
Am J Respir Crit Care Med ; 201(11): 1358-1371, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32105156

ABSTRACT

Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown.Objectives: To determine whether necroptosis contributes to RSV bronchiolitis pathogenesis via HMGB1 (high mobility group box 1) release.Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice.Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGB1 translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life.Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma.


Subject(s)
Bronchiolitis/virology , Epithelial Cells/metabolism , Epithelial Cells/pathology , HMGB1 Protein/metabolism , Necroptosis , Respiratory Mucosa/cytology , Respiratory Syncytial Virus Infections/metabolism , Animals , Child, Preschool , Humans , Infant , Mice , Prospective Studies
11.
Nature ; 513(7516): 95-9, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25186904

ABSTRACT

Receptor interacting protein kinase 1 (RIPK1) has an essential role in the signalling triggered by death receptors and pattern recognition receptors. RIPK1 is believed to function as a node driving NF-κB-mediated cell survival and inflammation as well as caspase-8 (CASP8)-dependent apoptotic or RIPK3/MLKL-dependent necroptotic cell death. The physiological relevance of this dual function has remained elusive because of the perinatal death of RIPK1 full knockout mice. To circumvent this problem, we generated RIPK1 conditional knockout mice, and show that mice lacking RIPK1 in intestinal epithelial cells (IECs) spontaneously develop severe intestinal inflammation associated with IEC apoptosis leading to early death. This early lethality was rescued by antibiotic treatment, MYD88 deficiency or tumour-necrosis factor (TNF) receptor 1 deficiency, demonstrating the importance of commensal bacteria and TNF in the IEC Ripk1 knockout phenotype. CASP8 deficiency, but not RIPK3 deficiency, rescued the inflammatory phenotype completely, indicating the indispensable role of RIPK1 in suppressing CASP8-dependent apoptosis but not RIPK3-dependent necroptosis in the intestine. RIPK1 kinase-dead knock-in mice did not exhibit any sign of inflammation, suggesting that RIPK1-mediated protection resides in its kinase-independent platform function. Depletion of RIPK1 in intestinal organoid cultures sensitized them to TNF-induced apoptosis, confirming the in vivo observations. Unexpectedly, TNF-mediated NF-κB activation remained intact in these organoids. Our results demonstrate that RIPK1 is essential for survival of IECs, ensuring epithelial homeostasis by protecting the epithelium from CASP8-mediated IEC apoptosis independently of its kinase activity and NF-κB activation.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestines/cytology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Caspase 8/genetics , Caspase 8/metabolism , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/pathology , Female , Gene Deletion , Homeostasis/drug effects , Inflammation/metabolism , Inflammation/pathology , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , NF-kappa B/metabolism , Necrosis , Organoids/cytology , Organoids/drug effects , Organoids/enzymology , Organoids/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Survival Analysis , Tumor Necrosis Factors/pharmacology
12.
J Immunol ; 198(11): 4435-4447, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28461567

ABSTRACT

The innate immune response is a central element of the initial defense against bacterial and viral pathogens. Macrophages are key innate immune cells that upon encountering pathogen-associated molecular patterns respond by producing cytokines, including IFN-ß. In this study, we identify a novel role for RIPK1 and RIPK3, a pair of homologous serine/threonine kinases previously implicated in the regulation of necroptosis and pathologic tissue injury, in directing IFN-ß production in macrophages. Using genetic and pharmacologic tools, we show that catalytic activity of RIPK1 directs IFN-ß synthesis induced by LPS in mice. Additionally, we report that RIPK1 kinase-dependent IFN-ß production may be elicited in an analogous fashion using LPS in bone marrow-derived macrophages upon inhibition of caspases. Notably, this regulation requires kinase activities of both RIPK1 and RIPK3, but not the necroptosis effector protein, MLKL. Mechanistically, we provide evidence that necrosome-like RIPK1 and RIPK3 aggregates facilitate canonical TRIF-dependent IFN-ß production downstream of the LPS receptor TLR4. Intriguingly, we also show that RIPK1 and RIPK3 kinase-dependent synthesis of IFN-ß is markedly induced by avirulent strains of Gram-negative bacteria, Yersinia and Klebsiella, and less so by their wild-type counterparts. Overall, these observations identify unexpected roles for RIPK1 and RIPK3 kinases in the production of IFN-ß during the host inflammatory responses to bacterial infection and suggest that the axis in which these kinases operate may represent a target for bacterial virulence factors.


Subject(s)
Interferon-beta/biosynthesis , Lipopolysaccharides/immunology , Macrophages/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/immunology , Gram-Negative Bacteria/immunology , Interferon-beta/immunology , Klebsiella/immunology , Macrophages/microbiology , Mice , Necrosis/immunology , Phosphorylation , Toll-Like Receptor 4/immunology , Yersinia/immunology
13.
J Immunol ; 197(10): 4110-4117, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27733552

ABSTRACT

Innate immune responses that are crucial for control of infection are often targeted by microbial pathogens. Blockade of NF-κB and MAPK signaling by the Yersinia virulence factor YopJ inhibits cytokine production by innate immune cells but also triggers cell death. This cell death requires RIPK1 kinase activity and caspase-8, which are engaged by TLR4 and the adaptor protein TRIF. Nevertheless, TLR4- and TRIF-deficient cells undergo significant apoptosis, implicating TLR4/TRIF-independent pathways in the death of Yersinia-infected cells. In this article, we report a key role for TNF/TNFR1 in Yersinia-induced cell death of murine macrophages, which occurs despite the blockade of NF-κB and MAPK signaling imposed by Yersinia on infected cells. Intriguingly, direct analysis of YopJ injection revealed a heterogeneous population of injection-high and injection-low cells, and demonstrated that TNF expression came from the injection-low population. Moreover, TNF production by this subpopulation was necessary for maximal apoptosis in the population of highly injected cells, and TNFR-deficient mice displayed enhanced susceptibility to Yersinia infection. These data demonstrate an important role for collaboration between TNF and pattern recognition receptor signals in promoting maximal apoptosis during bacterial infection, and demonstrate that heterogeneity in virulence factor injection and cellular responses play an important role in promoting anti-Yersinia immune defense.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Apoptosis , Macrophages/microbiology , Macrophages/physiology , Tumor Necrosis Factors/metabolism , Yersinia Infections/immunology , Yersinia pseudotuberculosis/pathogenicity , Animals , Bacterial Proteins/genetics , Caspase 1/metabolism , Cell Death , Immunity, Innate , L-Lactate Dehydrogenase/metabolism , Mice , Plasmids/genetics , Signal Transduction , Toll-Like Receptor 4/immunology , Tumor Necrosis Factors/deficiency , Tumor Necrosis Factors/immunology , Yersinia pseudotuberculosis/immunology
14.
J Immunol ; 196(1): 407-15, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26582950

ABSTRACT

The cytokine IL-1ß is intimately linked to many pathological inflammatory conditions. Mature IL-1ß secretion requires cleavage by the inflammasome. Recent evidence indicates that many cell death signal adaptors have regulatory roles in inflammasome activity. These include the apoptosis inducers FADD and caspase 8, and the necroptosis kinases receptor interacting protein kinase 1 (RIPK1) and RIPK3. PGAM5 is a mitochondrial phosphatase that has been reported to function downstream of RIPK3 to promote necroptosis and IL-1ß secretion. To interrogate the biological function of PGAM5, we generated Pgam5(-/-) mice. We found that Pgam5(-/-) mice were smaller compared with wild type littermates, and male Pgam5(-/-) mice were born at sub-Mendelian ratio. Despite these growth and survival defects, Pgam5(-/-) cells responded normally to multiple inducers of apoptosis and necroptosis. Rather, we found that PGAM5 is critical for IL-1ß secretion in response to NLRP3 and AIM2 inflammasome agonists. Moreover, vesicular stomatosis virus-induced IL-1ß secretion was impaired in Pgam5(-/-) bone marrow-derived macrophages, but not in Ripk3(-/-) bone marrow-derived dendritic cells, indicating that PGAM5 functions independent of RIPK3 to promote inflammasome activation. Mechanistically, PGAM5 promotes ASC polymerization, maintenance of mitochondrial integrity, and optimal reactive oxygen species production in response to inflammasome signals. Hence PGAM5 is a novel regulator of inflammasome and caspase 1 activity that functions independently of RIPK3.


Subject(s)
Apoptosis/immunology , Inflammasomes/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Phosphoric Monoester Hydrolases/genetics , Animals , Carrier Proteins/immunology , Caspase 1/immunology , Caspase 8/immunology , Cells, Cultured , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Fas-Associated Death Domain Protein/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphoprotein Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , Vesicular stomatitis Indiana virus/immunology
15.
J Immunol ; 196(12): 5056-63, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183605

ABSTRACT

Leishmaniasis is an important parasitic disease found in the tropics and subtropics. Cutaneous and visceral leishmaniasis affect an estimated 1.5 million people worldwide. Despite its human health relevance, relatively little is known about the cell death pathways that control Leishmania replication in the host. Necroptosis is a recently identified form of cell death with potent antiviral effects. Receptor interacting protein kinase 1 (RIPK1) is a critical kinase that mediates necroptosis downstream of death receptors and TLRs. Heme, a product of hemoglobin catabolism during certain intracellular pathogen infections, is also a potent inducer of macrophage necroptosis. We found that human visceral leishmaniasis patients exhibit elevated serum levels of heme. Therefore, we examined the impact of heme and necroptosis on Leishmania replication. Indeed, heme potently inhibited Leishmania replication in bone marrow-derived macrophages. Moreover, we found that inhibition of RIPK1 kinase activity also enhanced parasite replication in the absence of heme. We further found that the mitochondrial phosphatase phosphoglycerate mutase family member 5 (PGAM5), a putative downstream effector of RIPK1, was also required for inhibition of Leishmania replication. In mouse infection, both PGAM5 and RIPK1 kinase activity are required for IL-1ß expression in response to Leishmania However, PGAM5, but not RIPK1 kinase activity, was directly responsible for Leishmania-induced IL-1ß secretion and NO production in bone marrow-derived macrophages. Collectively, these results revealed that RIPK1 and PGAM5 function independently to exert optimal control of Leishmania replication in the host.


Subject(s)
Host-Parasite Interactions , Leishmania/growth & development , Leishmania/immunology , Leishmaniasis/parasitology , Phosphoprotein Phosphatases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death , Heme/analysis , Heme/pharmacology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Leishmania/drug effects , Leishmaniasis/blood , Leishmaniasis/immunology , Leishmaniasis/microbiology , Leishmaniasis, Visceral/blood , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Macrophages/physiology , Mice , Nitric Oxide/biosynthesis , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
16.
Stroke ; 48(4): 1033-1043, 2017 04.
Article in English | MEDLINE | ID: mdl-28250197

ABSTRACT

BACKGROUND AND PURPOSE: Intracerebral hemorrhage leads to disability or death with few established treatments. Adverse outcomes after intracerebral hemorrhage result from irreversible damage to neurons resulting from primary and secondary injury. Secondary injury has been attributed to hemoglobin and its oxidized product hemin from lysed red blood cells. The aim of this study was to identify the underlying cell death mechanisms attributable to secondary injury by hemoglobin and hemin to broaden treatment options. METHODS: We investigated cell death mechanisms in cultured neurons exposed to hemoglobin or hemin. Chemical inhibitors implicated in all known cell death pathways were used. Identified cell death mechanisms were confirmed using molecular markers and electron microscopy. RESULTS: Chemical inhibitors of ferroptosis and necroptosis protected against hemoglobin- and hemin-induced toxicity. By contrast, inhibitors of caspase-dependent apoptosis, protein or mRNA synthesis, autophagy, mitophagy, or parthanatos had no effect. Accordingly, molecular markers of ferroptosis and necroptosis were increased after intracerebral hemorrhage in vitro and in vivo. Electron microscopy showed that hemin induced a necrotic phenotype. Necroptosis and ferroptosis inhibitors each abrogated death by >80% and had similar therapeutic windows in vitro. CONCLUSIONS: Experimental intracerebral hemorrhage shares features of ferroptotic and necroptotic cell death, but not caspase-dependent apoptosis or autophagy. We propose that ferroptosis or necroptotic signaling induced by lysed blood is sufficient to reach a threshold of death that leads to neuronal necrosis and that inhibition of either of these pathways can bring cells below that threshold to survival.


Subject(s)
Apoptosis , Cerebral Hemorrhage/metabolism , Hemin/metabolism , Hemoglobins/metabolism , Necrosis/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
17.
Stroke ; 48(9): 2549-2556, 2017 09.
Article in English | MEDLINE | ID: mdl-28765287

ABSTRACT

BACKGROUND AND PURPOSE: Recent studies using cultured cells and rodent intracerebral hemorrhage (ICH) models have implicated RIPK1 (receptor interacting protein kinase-1) as a driver of programmed necrosis and secondary injury based on use of chemical inhibitors. However, these inhibitors have off-target effects and cannot be used alone to prove a role for RIPK1. The aim of the current study was to examine the effect of genetic inhibition of the kinase domain of RIPK1 in a mouse ICH model. METHODS: We subjected 2 lines of mice with RIPK1 point mutations of the kinase domain (K45A and D138N), rendering them kinase inactive, to autologous blood ICH and measured acute cell death and functional outcome. RESULTS: Compared with wild-type controls, RIPK1K45A/K45A and RIPK1D138N/D138N had significantly less cells with plasmalemma permeability, less acute neuronal cell death, less weight loss and more rapid weight gain to baseline, and improved performance in a Morris water maze paradigm after autologous blood ICH. In addition, mice systemically administered GSK'963, a potent, specific, brain penetrant small molecule RIPK1 inhibitor, had reduced acute neuronal death at 24 hours after ICH. CONCLUSIONS: The data show that the kinase domain of RIPK1 is a disease driver of ICH, mediating both acute cell death and functional outcome, and support development of RIPK1 inhibitors as therapeutic agents for human ICH.


Subject(s)
Apoptosis/genetics , Brain/metabolism , Cerebral Hemorrhage/genetics , Maze Learning/physiology , Necrosis/genetics , Neurons/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/drug effects , Behavior, Animal , Blotting, Western , Brain/pathology , Cell Death/drug effects , Cell Death/genetics , Cerebral Hemorrhage/physiopathology , Immunohistochemistry , Maze Learning/drug effects , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
18.
J Immunol ; 194(4): 1938-44, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25567679

ABSTRACT

Caspase 8, the initiator caspase for death receptor-induced apoptosis, functions as a negative regulator of receptor interacting protein kinase 3 (RIPK3), an essential factor for TNF-, TLR3-, and TLR4-induced necroptosis. In certain situations, caspase 8 can also participate in pro-IL-1ß processing. However, the biochemical complex that mediates caspase 8-mediated processing is not defined. In this study, we show that RIPK3 is crucial for caspase 1- and caspase 8-mediated pro-IL-1ß and pro-IL-18 processing in bone marrow-derived dendritic cells (BMDCs) in response to LPS stimulation. Caspase 8-mediated pro-IL-1ß processing requires intact RIPK1, RIPK3, TRIF, and FADD. In response to LPS, a complex that contains RIPK1, RIPK3, FADD, and caspase 8 is formed. Surprisingly, RIPK3-specific kinase inhibitors strongly enhanced caspase 8 activation and pro-IL-1ß processing in LPS-stimulated BMDCs. However, studies in BMDCs expressing the kinase-inactive RIPK3-K51A mutant or RIPK1-K45A mutant showed that the kinase activity of neither RIPK1 nor RIPK3 is required for LPS-induced caspase 8 activation and IL-1ß secretion. Hence, RIPK3 is an unexpected positive regulator of caspase 8 activity that promotes IL-1ß maturation in BMDCs.


Subject(s)
Caspase 8/immunology , Dendritic Cells/immunology , Interleukin-1beta/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Blotting, Western , Bone Marrow Cells/immunology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Gene Knock-In Techniques , Immunoprecipitation , Inflammation/immunology , Mice , Mice, Knockout , Transfection
19.
Proc Natl Acad Sci U S A ; 111(21): 7753-8, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821786

ABSTRACT

The pronecrotic kinase, receptor interacting protein (RIP1, also called RIPK1) mediates programmed necrosis and, together with its partner, RIP3 (RIPK3), drives midgestational death of caspase 8 (Casp8)-deficient embryos. RIP1 controls a second vital step in mammalian development immediately after birth, the mechanism of which remains unresolved. Rip1(-/-) mice display perinatal lethality, accompanied by gross immune system abnormalities. Here we show that RIP1 K45A (kinase dead) knockin mice develop normally into adulthood, indicating that development does not require RIP1 kinase activity. In the face of complete RIP1 deficiency, cells develop sensitivity to RIP3-mixed lineage kinase domain-like-mediated necroptosis as well as to Casp8-mediated apoptosis activated by diverse innate immune stimuli (e.g., TNF, IFN, double-stranded RNA). When either RIP3 or Casp8 is disrupted in combination with RIP1, the resulting double knockout mice exhibit slightly prolonged survival over RIP1-deficient animals. Surprisingly, triple knockout mice with combined RIP1, RIP3, and Casp8 deficiency develop into viable and fertile adults, with the capacity to produce normal levels of myeloid and lymphoid lineage cells. Despite the combined deficiency, these mice sustain a functional immune system that responds robustly to viral challenge. A single allele of Rip3 is tolerated in Rip1(-/-)Casp8(-/-)Rip3(+/-) mice, contrasting the need to eliminate both alleles of either Rip1 or Rip3 to rescue midgestational death of Casp8-deficient mice. These observations reveal a vital kinase-independent role for RIP1 in preventing pronecrotic as well as proapoptotic signaling events associated with life-threatening innate immune activation at the time of mammalian parturition.


Subject(s)
Apoptosis/immunology , GTPase-Activating Proteins/immunology , Immunity, Innate/immunology , Necrosis/immunology , Parturition/immunology , Signal Transduction/immunology , Animals , Caspase 8/immunology , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Knock-In Techniques , Immunoblotting , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/immunology
20.
Proc Natl Acad Sci U S A ; 111(20): 7391-6, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799678

ABSTRACT

A number of pathogens cause host cell death upon infection, and Yersinia pestis, infamous for its role in large pandemics such as the "Black Death" in medieval Europe, induces considerable cytotoxicity. The rapid killing of macrophages induced by Y. pestis, dependent upon type III secretion system effector Yersinia outer protein J (YopJ), is minimally affected by the absence of caspase-1, caspase-11, Fas ligand, and TNF. Caspase-8 is known to mediate apoptotic death in response to infection with several viruses and to regulate programmed necrosis (necroptosis), but its role in bacterially induced cell death is poorly understood. Here we provide genetic evidence for a receptor-interacting protein (RIP) kinase-caspase-8-dependent macrophage apoptotic death pathway after infection with Y. pestis, influenced by Toll-like receptor 4-TIR-domain-containing adapter-inducing interferon-ß (TLR4-TRIF). Interestingly, macrophages lacking either RIP1, or caspase-8 and RIP3, also had reduced infection-induced production of IL-1ß, IL-18, TNF, and IL-6; impaired activation of the transcription factor NF-κB; and greatly compromised caspase-1 processing. Cleavage of the proform of caspase-1 is associated with triggering inflammasome activity, which leads to the maturation of IL-1ß and IL-18, cytokines important to host responses against Y. pestis and many other infectious agents. Our results identify a RIP1-caspase-8/RIP3-dependent caspase-1 activation pathway after Y. pestis challenge. Mice defective in caspase-8 and RIP3 were also highly susceptible to infection and displayed reduced proinflammatory cytokines and myeloid cell death. We propose that caspase-8 and the RIP kinases are key regulators of macrophage cell death, NF-κB and inflammasome activation, and host resistance after Y. pestis infection.


Subject(s)
Caspase 8/metabolism , Cell Death , Immunity, Innate , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Bacterial Proteins/genetics , Bone Marrow Cells/cytology , Cytokines/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Yersinia Infections/microbiology , Yersinia pestis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL