Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Biol Chem ; 295(19): 6543-6560, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32245890

ABSTRACT

Optic atrophy 1 (OPA1) is a dynamin protein that mediates mitochondrial fusion at the inner membrane. OPA1 is also necessary for maintaining the cristae and thus essential for supporting cellular energetics. OPA1 exists as membrane-anchored long form (L-OPA1) and short form (S-OPA1) that lacks the transmembrane region and is generated by cleavage of L-OPA1. Mitochondrial dysfunction and cellular stresses activate the inner membrane-associated zinc metallopeptidase OMA1 that cleaves L-OPA1, causing S-OPA1 accumulation. The prevailing notion has been that L-OPA1 is the functional form, whereas S-OPA1 is an inactive cleavage product in mammals, and that stress-induced OPA1 cleavage causes mitochondrial fragmentation and sensitizes cells to death. However, S-OPA1 contains all functional domains of dynamin proteins, suggesting that it has a physiological role. Indeed, we recently demonstrated that S-OPA1 can maintain cristae and energetics through its GTPase activity, despite lacking fusion activity. Here, applying oxidant insult that induces OPA1 cleavage, we show that cells unable to generate S-OPA1 are more sensitive to this stress under obligatory respiratory conditions, leading to necrotic death. These findings indicate that L-OPA1 and S-OPA1 differ in maintaining mitochondrial function. Mechanistically, we found that cells that exclusively express L-OPA1 generate more superoxide and are more sensitive to Ca2+-induced mitochondrial permeability transition, suggesting that S-OPA1, and not L-OPA1, protects against cellular stress. Importantly, silencing of OMA1 expression increased oxidant-induced cell death, indicating that stress-induced OPA1 cleavage supports cell survival. Our findings suggest that S-OPA1 generation by OPA1 cleavage is a survival mechanism in stressed cells.


Subject(s)
GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/enzymology , Oxidative Stress , Animals , Calcium/metabolism , Cell Line , Cell Survival , GTP Phosphohydrolases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Permeability , Superoxides/metabolism
2.
J Biol Chem ; 294(32): 11994-12006, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31217278

ABSTRACT

Phosphatidylserine (PS), an anionic phospholipid enriched in the inner leaflet of the plasma membrane, is exposed to the outer leaflet during apoptosis. PS exposure was recently shown to be induced during tumor necrosis factor-induced necroptosis. We herein demonstrated that interferon (IFN)-γ induced necroptosis in Caspase-8-knockout mouse-derived embryonic fibroblasts (C8KO MEFs), as well as in WT MEFs co-treated with the pan-caspase inhibitor, z-VAD-fmk. PS exposure and necroptosis were significant after 6- and 24-h treatments with IFN-γ, respectively. To elucidate the molecular mechanisms underlying IFN-γ-induced PS exposure, we generated C8KO MEF-derived cell lines without the expression of RIPK3 (receptor-interacting protein kinase 3), an essential molecule in tumor necrosis factor-induced necroptosis, and IFN-γ-induced PS exposure and necrotic cell death were shown to be specifically inhibited by the loss of RIPK3 expression. Furthermore, the down-regulated expression of MLKL (mixed lineage kinase domain-like protein), a key molecule for inducing membrane rupture downstream of RIPK3 in necroptosis, abolished IFN-γ-induced PS exposure in C8KO MEFs. In human colorectal adenocarcinoma-derived HT29 cells, PS exposure and necroptosis were similarly induced by treatment with IFN-γ in the presence of Smac mimetics and z-VAD-fmk. The removal of IFN-γ from PS-exposing MEFs after a 6-h treatment completely inhibited necroptotic cell death but not the subsequent increase in the number of PS-exposing cells. Therefore, PS exposure mediated by RIPK3-activated MLKL oligomers was induced by a treatment with IFN-γ for a significant interval of time before the induction of necroptosis by membrane rupture.


Subject(s)
Caspase 8/genetics , Interferon-gamma/pharmacology , Necroptosis/drug effects , Phosphatidylserines/metabolism , Protein Kinases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase 8/metabolism , Cell Line , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Kinases/chemistry , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
J Biol Chem ; 294(2): 502-519, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30420430

ABSTRACT

Formation of membrane pores/channels regulates various cellular processes, such as necroptosis or stem cell niche signaling. However, the roles of membrane lipids in the formation of pores and their biological functions are largely unknown. Here, using the cellular stress model evoked by the sphingolipid analog drug FTY720, we show that formation of ceramide-enriched membrane pores, referred to here as ceramidosomes, is initiated by a receptor-interacting Ser/Thr kinase 1 (RIPK1)-ceramide complex transported to the plasma membrane by nonmuscle myosin IIA-dependent trafficking in human lung cancer cells. Molecular modeling/simulation coupled with site-directed mutagenesis revealed that Asp147 or Asn169 of RIPK1 are key for ceramide binding and that Arg258 or Leu293 residues are involved in the myosin IIA interaction, leading to ceramidosome formation and necroptosis. Moreover, generation of ceramidosomes independently of any external drug/stress stimuli was also detected in the plasma membrane of germ line stem cells in ovaries during the early stages of oogenesis in Drosophila melanogaster Inhibition of ceramidosome formation via myosin IIA silencing limited germ line stem cell signaling and abrogated oogenesis. In conclusion, our findings indicate that the RIPK1-ceramide complex forms large membrane pores we named ceramidosomes. They further suggest that, in addition to their roles in stress-mediated necroptosis, these ceramide-enriched pores also regulate membrane integrity and signaling and might also play a role in D. melanogaster ovary development.


Subject(s)
Cell Membrane/metabolism , Ceramides/metabolism , Lung Neoplasms/metabolism , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Necrosis/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , A549 Cells , Animals , Cell Line , Cell Membrane/pathology , Drosophila melanogaster/growth & development , Female , Humans , Lung Neoplasms/pathology , Molecular Docking Simulation , Necrosis/pathology , Oogenesis , Ovary/growth & development
4.
J Biol Chem ; 293(42): 16348-16363, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30190323

ABSTRACT

Immune therapy of cancer is among the most promising recent advances in medicine. Whether the immune system can keep cancer in check depends on, among other factors, the efficiency of immune cells to recognize and eliminate cancer cells. We describe a time-resolved single-cell assay that reports the quality, quantity, and kinetics of target cell death induced by single primary human natural killer (NK) cells. The assay reveals that single NK cells induce cancer cell death by apoptosis and necrosis but also by mixed forms. Inhibition of either one of the two major cytotoxic pathways, perforin/granzyme release or FasL/FasR interaction, unmasked the parallel activity of the other one. Ca2+ influx through Orai channels is important for tuning killer cell function. We found that the apoptosis/necrosis ratio of cancer cell death by NK cells is controlled by the magnitude of Ca2+ entry and furthermore by the relative concentrations of perforin and granzyme B. The possibility to change the apoptosis/necrosis ratio employed by NK cells offers an intriguing possibility to modulate the immunogenicity of the tumor microenvironment.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms/immunology , Calcium/metabolism , Calcium/pharmacology , Cell Death , Granzymes/analysis , Humans , Neoplasms/pathology , Perforin/analysis , Single-Cell Analysis
5.
J Biol Chem ; 293(21): 8032-8047, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29626097

ABSTRACT

Mitochondrial dysfunction lies at the core of acute pancreatitis (AP). Diverse AP stimuli induce Ca2+-dependent formation of the mitochondrial permeability transition pore (MPTP), a solute channel modulated by cyclophilin D (CypD), the formation of which causes ATP depletion and necrosis. Oxidative stress reportedly triggers MPTP formation and is elevated in clinical AP, but how reactive oxygen species influence cell death is unclear. Here, we assessed potential MPTP involvement in oxidant-induced effects on pancreatic acinar cell bioenergetics and fate. H2O2 application promoted acinar cell apoptosis at low concentrations (1-10 µm), whereas higher levels (0.5-1 mm) elicited rapid necrosis. H2O2 also decreased the mitochondrial NADH/FAD+ redox ratio and ΔΨm in a concentration-dependent manner (10 µm to 1 mm H2O2), with maximal effects at 500 µm H2O2 H2O2 decreased the basal O2 consumption rate of acinar cells, with no alteration of ATP turnover at <50 µm H2O2 However, higher H2O2 levels (≥50 µm) diminished spare respiratory capacity and ATP turnover, and bioenergetic collapse, ATP depletion, and cell death ensued. Menadione exerted detrimental bioenergetic effects similar to those of H2O2, which were inhibited by the antioxidant N-acetylcysteine. Oxidant-induced bioenergetic changes, loss of ΔΨm, and cell death were not ameliorated by genetic deletion of CypD or by its acute inhibition with cyclosporine A. These results indicate that oxidative stress alters mitochondrial bioenergetics and modifies pancreatic acinar cell death. A shift from apoptosis to necrosis appears to be associated with decreased mitochondrial spare respiratory capacity and ATP production, effects that are independent of CypD-sensitive MPTP formation.


Subject(s)
Apoptosis , Cyclophilins/physiology , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/physiology , Necrosis , Oxidative Stress , Pancreas/pathology , Acinar Cells/metabolism , Acinar Cells/pathology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Energy Metabolism , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Permeability Transition Pore , Pancreas/metabolism , Reactive Oxygen Species/metabolism
6.
J Biol Chem ; 293(30): 11913-11927, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29899110

ABSTRACT

Monocytes differentiate into macrophages, which deactivate invading pathogens. Macrophages can be resistant to cell death mechanisms in some situations, and the mechanisms involved are not clear. Here, using mouse immune cells, we investigated whether the differentiation of macrophages affects their susceptibility to cell death by the ripoptosome/necrosome pathways. We show that treatment of macrophages with a mimetic of second mitochondrial activator of caspases (SMAC) resulted in ripoptosome-driven cell death that specifically depended on tumor necrosis factor α (TNFα) expression and the receptor-interacting serine/threonine protein kinase 1 (RipK1)-RipK3-caspase-8 interaction in activated and cycling macrophages. Differentiation of macrophages increased the expression of pro-inflammatory cytokines but reduced RipK1-dependent cell death and the RipK3-caspase-8 interaction. The expression of the anti-apoptotic mediators, X-linked inhibitor of apoptosis protein (XIAP) and caspase-like apoptosis regulatory protein (cFLIPL), also increased in differentiated macrophages, which inhibited caspase activation. The resistance to cell death was abrogated in XIAP-deficient macrophages. However, even in the presence of increased XIAP expression, inhibition of the mitogen-activated protein kinase (MAPK) p38 and MAPK-activated protein kinase 2 (MK2) made differentiated macrophages susceptible to cell death. These results suggest that the p38/MK2 pathway overrides apoptosis inhibition by XIAP and that acquisition of resistance to cell death by increased expression of XIAP and cFLIPL may allow inflammatory macrophages to participate in pathogen control for a longer duration.


Subject(s)
Inflammation/immunology , Inhibitor of Apoptosis Proteins/immunology , Macrophages/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Apoptosis , Cell Differentiation , Cells, Cultured , Macrophages/cytology , Mice, Inbred C57BL
7.
J Biol Chem ; 292(42): 17514-17524, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28878015

ABSTRACT

Necroptosis is an immunogenic cell death program that is associated with a host of human diseases, including inflammation, infections, and cancer. Receptor-interacting protein kinase 3 (RIPK3) and its substrate mixed lineage kinase domain-like protein (MLKL) are required for necroptosis activation. Specifically, RIPK3-dependent MLKL phosphorylation promotes the assembly of disulfide bond-dependent MLKL polymers that drive the execution of necroptosis. However, how MLKL disulfide bond formation is regulated is not clear. In this study we discovered that the MLKL-modifying compound necrosulfonamide cross-links cysteine 86 of human MLKL to cysteine 32 of the thiol oxidoreductase thioredoxin-1 (Trx1). Recombinant Trx1 preferentially binds to monomeric MLKL and blocks MLKL disulfide bond formation and polymerization in vitro Inhibition of MLKL polymer formation requires the reducing activity of Trx1. Importantly, shRNA-mediated knockdown of Trx1 promotes MLKL polymerization and sensitizes cells to necroptosis. Furthermore, pharmacological inhibition of Trx1 with compound PX-12 induces necroptosis in multiple cancer cell lines. Altogether, these findings demonstrate that Trx1 is a critical regulator of necroptosis that suppresses cell death by maintaining MLKL in a reduced inactive state. Our results further suggest new directions for targeted cancer therapy in which thioredoxin inhibitors like PX-12 could potentially be used to specifically target cancers expressing high levels of MLKL or MLKL short isoforms.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/metabolism , Protein Kinases/metabolism , Protein Multimerization , Thioredoxins/metabolism , Cell Death/drug effects , Cell Death/genetics , Disulfides/pharmacology , HeLa Cells , Humans , Imidazoles/pharmacology , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinases/genetics , Thioredoxins/genetics
8.
J Biol Chem ; 291(38): 20247-59, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27502281

ABSTRACT

Recent studies have shown that myocardial ischemia/reperfusion (I/R)-induced necrosis can be controlled by multiple genes. In this study, we observed that both strands (5p and 3p) of miR-223 were remarkably dysregulated in mouse hearts upon I/R. Precursor miR-223 (pre-miR-223) transgenic mouse hearts exhibited better recovery of contractile performance over reperfusion period and lesser degree of myocardial necrosis than wild type hearts upon ex vivo and in vivo myocardial ischemia. Conversely, pre-miR-223 knock-out (KO) mouse hearts displayed opposite effects. Furthermore, we found that the RIP1/RIP3/MLKL necroptotic pathway and inflammatory response were suppressed in transgenic hearts, whereas they were activated in pre-miR-223 KO hearts upon I/R compared with wild type controls. Accordingly, treatment of pre-miR-223 KO mice with necrostatin-1s, a potent necroptosis inhibitor, significantly decreased I/R-triggered cardiac necroptosis, infarction size, and dysfunction. Mechanistically, we identified two critical cell death receptors, TNFR1 and DR6, as direct targets of miR-223-5p, whereas miR-223-3p directly suppressed the expression of NLRP3 and IκB kinase α, two important mediators known to be involved in I/R-induced inflammation and cell necroptosis. Our findings indicate that miR-223-5p/-3p duplex works together and cooperatively inhibits I/R-induced cardiac necroptosis at multiple layers. Thus, pre-miR-223 may constitute a new therapeutic agent for the treatment of ischemic heart disease.


Subject(s)
MicroRNAs/biosynthesis , Myocardial Reperfusion Injury/metabolism , Animals , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Mice, Knockout , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Necrosis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/genetics
9.
J Biol Chem ; 291(39): 20602-16, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27514746

ABSTRACT

Injury to the barrier tissue initiates a rapid distribution of myeloid immune cells from bone marrow, which guide sound wound healing. Bisphosphonates, a widely used anti-bone resorptive drug with minimal systemic side effects, have been linked to an abnormal wound healing in the oral barrier tissue leading to, in some cases, osteonecrosis of the jaw (ONJ). Here we report that the development of ONJ may involve abnormal phenotypic plasticity of Ly6G+/Gr1+ myeloid cells in the oral barrier tissue undergoing tooth extraction wound healing. A bolus intravenous zoledronate (ZOL) injection to female C57Bl/6 mice followed by maxillary first molar extraction resulted in the development of ONJ-like lesion during the second week of wound healing. The multiplex assay of dissociated oral barrier cells exhibited the secretion of cytokines and chemokines, which was significantly modulated in ZOL mice. Tooth extraction-induced distribution of Ly6G+/Gr1+ cells in the oral barrier tissue increased in ZOL mice at week 2. ONJ-like lesion in ZOL mice contained Ly6G+/Gr1+ cells with abnormal size and morphology as well as different flow cytometric staining intensity. When anti-Ly6G (Gr1) antibody was intraperitoneally injected for 5 days during the second week of tooth extraction, CD11b+GR1(hi) cells in bone marrow and Ly6G+ cells in the oral barrier tissue were depleted, and the development of ONJ-like lesion was significantly attenuated. This study suggests that local modulation of myeloid cell plasticity in the oral barrier tissue may provide the basis for pathogenesis and thus therapeutic as well as preventive strategy of ONJ.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw/immunology , Myeloid Cells/immunology , Wound Healing/immunology , Animals , Antigens, Ly/immunology , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Female , Mice , Mouth/pathology , Myeloid Cells/pathology , Tooth Extraction
10.
J Biol Chem ; 291(11): 5948-5959, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26786097

ABSTRACT

Receptor-interacting protein kinase 3 (RIPK3) is a serine/threonine kinase with essential function in necroptosis. The activity of RIPK3 is controlled by phosphorylation. Once activated, RIPK3 phosphorylates and activates the downstream effector mixed lineage kinase domain-like (MLKL) to induce necroptosis. In certain situations, RIPK3 has also been shown to promote apoptosis or cytokine expression in a necroptosis and kinase-independent manner. The ubiquitin-proteasome system is the major pathway for selective degradation of cellular proteins and thus has a critical role in many cellular processes such as cell survival and cell death. Clinically, proteasome inhibition has shown promise as an anti-cancer agent. Here we show that the proteasome inhibitors MG132 and bortezomib activate the RIPK3-MLKL necroptotic pathway in mouse fibroblasts as well as human leukemia cells. Unlike necroptosis induced by classical TNF-like cytokines, necroptosis induced by proteasome inhibitors does not require caspase inhibition. However, an intact RIP homotypic interaction motif (RHIM) is essential. Surprisingly, when recruitment of MLKL to RIPK3 is restricted, proteasome inhibitors induced RIPK3-dependent apoptosis. Proteasome inhibition led to accumulation of K48-linked ubiquitinated RIPK3, which was partially reduced when Lys-264 was mutated. Taken together, these results reveal the ubiquitin-proteasome system as a novel regulatory mechanism for RIPK3-dependent necroptosis.


Subject(s)
Cell Death/drug effects , Leupeptins/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Cell Line , Cell Line, Tumor , Humans , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinases/metabolism , Ubiquitination/drug effects
11.
J Biol Chem ; 291(44): 22936-22948, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27587392

ABSTRACT

We investigated the in vitro and in vivo anticancer effect of combining lysosomal membrane permeabilization (LMP)-inducing agent N-dodecylimidazole (NDI) with glycolytic inhibitor 2-deoxy-d-glucose (2DG). NDI-triggered LMP and 2DG-mediated glycolysis block synergized in inducing rapid ATP depletion, mitochondrial damage, and reactive oxygen species production, eventually leading to necrotic death of U251 glioma cells but not primary astrocytes. NDI/2DG-induced death of glioma cells was partly prevented by lysosomal cathepsin inhibitor E64 and antioxidant α-tocopherol, suggesting the involvement of LMP and oxidative stress in the observed cytotoxicity. LMP-inducing agent chloroquine also displayed a synergistic anticancer effect with 2DG, whereas glucose deprivation or glycolytic inhibitors iodoacetate and sodium fluoride synergistically cooperated with NDI, thus further indicating that the anticancer effect of NDI/2DG combination was indeed due to LMP and glycolysis block. The two agents synergistically induced ATP depletion, mitochondrial depolarization, oxidative stress, and necrotic death also in B16 mouse melanoma cells. Moreover, the combined oral administration of NDI and 2DG reduced in vivo melanoma growth in C57BL/6 mice by inducing necrotic death of tumor cells, without causing liver, spleen, or kidney toxicity. Based on these results, we propose that NDI-triggered LMP causes initial mitochondrial damage that is further increased by 2DG due to the lack of glycolytic ATP required to maintain mitochondrial health. This leads to a positive feedback cycle of mitochondrial dysfunction, ATP loss, and reactive oxygen species production, culminating in necrotic cell death. Therefore, the combination of LMP-inducing agents and glycolysis inhibitors seems worthy of further exploration as an anticancer strategy.


Subject(s)
Deoxyglucose/pharmacology , Glioma/metabolism , Glycolysis/drug effects , Imidazoles/pharmacology , Lysosomes/drug effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Drug Synergism , Glioma/drug therapy , Glioma/physiopathology , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects
12.
J Biol Chem ; 291(26): 13753-61, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129772

ABSTRACT

Necroptosis is a RIP1-dependent programmed cell death (PCD) pathway that is distinct from apoptosis. Downstream effector pathways of necroptosis include formation of advanced glycation end products (AGEs) and reactive oxygen species (ROS), both of which depend on glycolysis. This suggests that increased cellular glucose may prime necroptosis. Here we show that exposure to hyperglycemic levels of glucose enhances necroptosis in primary red blood cells (RBCs), Jurkat T cells, and U937 monocytes. Pharmacologic or siRNA inhibition of RIP1 prevented the enhanced death, confirming it as RIP1-dependent necroptosis. Hyperglycemic enhancement of necroptosis depends upon glycolysis with AGEs and ROS playing a role. Total levels of RIP1, RIP3, and mixed lineage kinase domain-like (MLKL) proteins were increased following treatment with high levels of glucose in Jurkat and U937 cells and was not due to transcriptional regulation. The observed increase in RIP1, RIP3, and MLKL protein levels suggests a potential positive feedback mechanism in nucleated cell types. Enhanced PCD due to hyperglycemia was specific to necroptosis as extrinsic apoptosis was inhibited by exposure to high levels of glucose. Hyperglycemia resulted in increased infarct size in a mouse model of brain hypoxia-ischemia injury. The increased infarct size was prevented by treatment with nec-1s, strongly suggesting that increased necroptosis accounts for exacerbation of this injury in conditions of hyperglycemia. This work reveals that hyperglycemia represents a condition in which cells are extraordinarily susceptible to necroptosis, that local glucose levels alter the balance of PCD pathways, and that clinically relevant outcomes may depend on glucose-mediated effects on PCD.


Subject(s)
Erythrocytes/metabolism , GTPase-Activating Proteins/metabolism , Hyperglycemia/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Death , Disease Models, Animal , Erythrocytes/pathology , GTPase-Activating Proteins/genetics , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/pathology , Jurkat Cells , Mice , Nuclear Pore Complex Proteins/genetics , RNA-Binding Proteins/genetics , U937 Cells
13.
J Biol Chem ; 290(18): 11635-48, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25778401

ABSTRACT

Necroptosis is an alternate programmed cell death pathway that is unleashed by caspase-8 compromise and mediated by receptor-interacting protein kinase 3 (RIP3). Murine cytomegalovirus (CMV) and herpes simplex virus (HSV) encode caspase-8 inhibitors that prevent apoptosis together with competitors of RIP homotypic interaction motif (RHIM)-dependent signal transduction to interrupt the necroptosis. Here, we show that pro-necrotic murine CMV M45 mutant virus drives virus-induced necroptosis during nonproductive infection of RIP3-expressing human fibroblasts, whereas WT virus does not. Thus, M45-encoded RHIM competitor, viral inhibitor of RIP activation, sustains viability of human cells like it is known to function in infected mouse cells. Importantly, human CMV is shown to block necroptosis induced by either TNF or M45 mutant murine CMV in RIP3-expressing human cells. Human CMV blocks TNF-induced necroptosis after RIP3 activation and phosphorylation of the mixed lineage kinase domain-like (MLKL) pseudokinase. An early, IE1-regulated viral gene product acts on a necroptosis step that follows MLKL phosphorylation prior to membrane leakage. This suppression strategy is distinct from RHIM signaling competition by murine CMV or HSV and interrupts an execution process that has not yet been fully elaborated.


Subject(s)
Cytomegalovirus/physiology , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cytomegalovirus/metabolism , Evolution, Molecular , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Immediate-Early Proteins/metabolism , Mice , Muromegalovirus/physiology , Phosphorylation , Protein Transport , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Transduction, Genetic
14.
J Biol Chem ; 290(34): 20841-20855, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26124276

ABSTRACT

Apoptosis is triggered by the activation of caspases and characterized by chromatin condensation and nuclear fragmentation (type II nuclear morphology). Necrosis is depicted by a gain in cell volume (oncosis), swelling of organelles, plasma membrane leakage, and subsequent loss of intracellular contents. Although considered as different cell death entities, there is an overlap between apoptosis and necrosis. In this sense, mounting evidence suggests that both processes can be morphological expressions of a common biochemical network known as "apoptosis-necrosis continuum." To gain insight into the events driving the apoptosis-necrosis continuum, apoptotically proficient cells were screened facing several apoptotic inducers for the absence of type II apoptotic nuclear morphologies. Chelerythrine was selected for further studies based on its cytotoxicity and the lack of apoptotic nuclear alterations. Chelerythrine triggered an early plasma membrane leakage without condensed chromatin aggregates. Ultrastructural analysis revealed that chelerythrine-mediated cytotoxicity was compatible with a necrotic-like type of cell death. Biochemically, chelerythrine induced the activation of caspases. Moreover, the inhibition of caspases prevented chelerythrine-triggered necrotic-like cell death. Compared with staurosporine, chelerythrine induced stronger caspase activation detectable at earlier times. After using a battery of chemicals, we found that high concentrations of thiolic antioxidants fully prevented chelerythrine-driven caspase activation and necrotic-like cell death. Lower amounts of thiolic antioxidants partially prevented chelerythrine-mediated cytotoxicity and allowed cells to display type II apoptotic nuclear morphology correlating with a delay in caspase-3 activation. Altogether, these data support that an early and pronounced activation of caspases can drive cells to undergo a form of necrotic-like regulated cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Caspases/metabolism , Chromatin/drug effects , Enzyme Inhibitors/pharmacology , Necrosis/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Benzophenanthridines/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspases/genetics , Cell Line, Tumor , Chromatin/metabolism , Chromatin/ultrastructure , Colchicine/pharmacology , Enzyme Activation/drug effects , Gene Expression Regulation , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Necrosis/chemically induced , Necrosis/genetics , Neurons , Nocodazole/pharmacology , Peptidomimetics/pharmacology , Quinolines/pharmacology , Rotenone/pharmacology , Signal Transduction , Staurosporine/pharmacology , Thapsigargin/pharmacology
15.
J Biol Chem ; 290(36): 22030-48, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26100636

ABSTRACT

It is widely accepted that overactivation of NMDA receptors, resulting in calcium overload and consequent mitochondrial dysfunction in retinal ganglion neurons, plays a significant role in promoting neurodegenerative disorders such as glaucoma. Calcium has been shown to initiate a transient hyperpolarization of the mitochondrial membrane potential triggering a burst of reactive oxygen species leading to apoptosis. Strategies that enhance cell survival signaling pathways aimed at preventing this adverse hyperpolarization of the mitochondrial membrane potential may provide a novel therapeutic intervention in retinal disease. In the retina, brain-derived neurotrophic factor has been shown to be neuroprotective, and our group previously reported a PSD-95/PDZ-binding cyclic peptide (CN2097) that augments brain-derived neurotrophic factor-induced pro-survival signaling. Here, we examined the neuroprotective properties of CN2097 using an established retinal in vivo NMDA toxicity model. CN2097 completely attenuated NMDA-induced caspase 3-dependent and -independent cell death and PARP-1 activation pathways, blocked necrosis, and fully prevented the loss of long term ganglion cell viability. Although neuroprotection was partially dependent upon CN2097 binding to the PDZ domain of PSD-95, our results show that the polyarginine-rich transport moiety C-R(7), linked to the PDZ-PSD-95-binding cyclic peptide, was sufficient to mediate short and long term protection via a mitochondrial targeting mechanism. C-R(7) localized to mitochondria and was found to reduce mitochondrial respiration, mitochondrial membrane hyperpolarization, and the generation of reactive oxygen species, promoting survival of retinal neurons.


Subject(s)
Membrane Potential, Mitochondrial/drug effects , N-Methylaspartate/pharmacology , Peptides/pharmacology , Retinal Neurons/drug effects , Animals , Blotting, Western , Cell Death/drug effects , Disks Large Homolog 4 Protein , Excitatory Amino Acid Agonists/pharmacology , Guanylate Kinases/metabolism , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Peptides/metabolism , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Protein Binding , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retina/cytology , Retina/drug effects , Retina/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Neurons/metabolism , Stress, Physiological/drug effects , Stress, Physiological/physiology
16.
J Biol Chem ; 290(41): 25188-96, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26324711

ABSTRACT

Inflammation is a key instigator of the immune responses that drive atherosclerosis and allograft rejection. IL-1α, a powerful cytokine that activates both innate and adaptive immunity, induces vessel inflammation after release from necrotic vascular smooth muscle cells (VSMCs). Similarly, IL-1α released from endothelial cells (ECs) damaged during transplant drives allograft rejection. However, IL-1α requires cleavage for full cytokine activity, and what controls cleavage in necrotic ECs is currently unknown. We find that ECs have very low levels of IL-1α activity upon necrosis. However, TNFα or IL-1 induces significant levels of active IL-1α in EC necrotic lysates without alteration in protein levels. Increased activity requires cleavage of IL-1α by calpain to the more active mature form. Immunofluorescence and proximity ligation assays show that IL-1α associates with interleukin-1 receptor-2, and this association is decreased by TNFα or IL-1 and requires caspase activity. Thus, TNFα or IL-1 treatment of ECs leads to caspase proteolytic activity that cleaves interleukin-1 receptor-2, allowing IL-1α dissociation and subsequent processing by calpain. Importantly, ECs could be primed by IL-1α from adjacent damaged VSMCs, and necrotic ECs could activate neighboring normal ECs and VSMCs, causing them to release inflammatory cytokines and up-regulate adhesion molecules, thus amplifying inflammation. These data unravel the molecular mechanisms and interplay between damaged ECs and VSMCs that lead to activation of IL-1α and, thus, initiation of adaptive responses that cause graft rejection.


Subject(s)
Allografts/immunology , Caspase 1/metabolism , Graft Rejection/metabolism , Graft Rejection/pathology , Human Umbilical Vein Endothelial Cells/pathology , Interleukin-1alpha/metabolism , Receptors, Interleukin-1 Type II/metabolism , Calpain/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-1/pharmacology , Necrosis/immunology , Proteolysis , Tumor Necrosis Factor-alpha/pharmacology
17.
J Biol Chem ; 289(12): 8337-52, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24519941

ABSTRACT

Degenerative loss of photoreceptors occurs in inherited and age-related retinal degenerative diseases. A chemical screen facilitates development of new testing routes for neuroprotection and mechanistic investigation. Herein, we conducted a mouse-derived photoreceptor (661W cell)-based high throughput screen of the Food and Drug Administration-approved Prestwick drug library to identify putative cytoprotective compounds against light-induced, synthetic visual chromophore-precipitated cell death. Different classes of hit compounds were identified, some of which target known genes or pathways pathologically associated with retinitis pigmentosa. Sulfaphenazole (SFZ), a selective inhibitor of human cytochrome P450 (CYP) 2C9 isozyme, was identified as a novel and leading cytoprotective compound. Expression of CYP2C proteins was induced by light. Gene-targeted knockdown of CYP2C55, the homologous gene of CYP2C9, demonstrated viability rescue to light-induced cell death, whereas stable expression of functional CYP2C9-GFP fusion protein further exacerbated light-induced cell death. Mechanistically, SFZ inhibited light-induced necrosis and mitochondrial stress-initiated apoptosis. Light elicited calcium influx, which was mitigated by SFZ. Light provoked the release of arachidonic acid from membrane phospholipids and production of non-epoxyeicosatrienoic acid metabolites. Administration of SFZ further stimulated the production of non-epoxyeicosatrienoic acid metabolites, suggesting a metabolic shift of arachidonic acid under inhibition of the CYP2C pathway. Together, our findings indicate that CYP2C genes play a direct causative role in photochemical stress-induced death of photoreceptors and suggest that the CYP monooxygenase system is a risk factor for retinal photodamage, especially in individuals with Stargardt disease and age-related macular degeneration that deposit condensation products of retinoids.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytoprotection/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Sulfaphenazole/pharmacology , Amino Acid Sequence , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/genetics , Cell Death/drug effects , Cell Death/radiation effects , Cell Line , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 2 , Drug Evaluation, Preclinical , Gene Expression , Gene Silencing , Humans , Light , Mice , Molecular Sequence Data , Photoreceptor Cells, Vertebrate/enzymology , Sequence Alignment
18.
J Biol Chem ; 289(37): 25699-710, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25096578

ABSTRACT

Cell death by necrosis is emerging not merely as a passive phenomenon but as a cell-regulated process. Here, by using different necrotic triggers, we prove the existence of two distinct necrotic pathways. The mitochondrial reactive oxygen species generator 2,3-dimethoxy-1,4-naphthoquinone elicits necrosis characterized by the involvement of RIP1 and Drp1. However, G5, a non-selective isopeptidase inhibitor, triggers a distinct necrotic pathway that depends on the protein phosphatase PP2A and the actin cytoskeleton. PP2A catalytic subunit is stabilized by G5 treatment, and its activity is increased. Furthermore, PP2Ac accumulates into the cytoplasm during necrosis similarly to HMGB1. We have also defined in the actin-binding protein cofilin-1 a link between PP2A, actin cytoskeleton, and necrotic death. Cofilin-1-severing/depolymerization activity is negatively regulated by phosphorylation of serine 3. PP2A contributes to the dephosphorylation of serine 3 elicited by G5. Finally, a cofilin mutant that mimics phosphorylated Ser-3 can partially rescue necrosis in response to G5.


Subject(s)
Actin Cytoskeleton/metabolism , Cofilin 1/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Phosphatase 2/metabolism , RNA-Binding Proteins/metabolism , Actin Cytoskeleton/ultrastructure , Actin Depolymerizing Factors/chemistry , Cell Membrane Structures/chemistry , Cell Membrane Structures/drug effects , Cofilin 1/chemistry , HT29 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis/genetics , Necrosis/metabolism , Nuclear Pore Complex Proteins/chemistry , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Proteolysis , Pyrans/pharmacology , RNA-Binding Proteins/chemistry , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/pharmacology
19.
J Biol Chem ; 289(24): 16711-26, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24764300

ABSTRACT

Intracellular peptides are constantly produced by the ubiquitin-proteasome system, and many are probably functional. Here, the peptide WELVVLGKL (pep5) from G1/S-specific cyclin D2 showed a 2-fold increase during the S phase of HeLa cell cycle. pep5 (25-100 µm) induced cell death in several tumor cells only when it was fused to a cell-penetrating peptide (pep5-cpp), suggesting its intracellular function. In vivo, pep5-cpp reduced the volume of the rat C6 glioblastoma by almost 50%. The tryptophan at the N terminus of pep5 is essential for its cell death activity, and N terminus acetylation reduced the potency of pep5-cpp. WELVVL is the minimal active sequence of pep5, whereas Leu-Ala substitutions totally abolished pep5 cell death activity. Findings from the initial characterization of the cell death/signaling mechanism of pep5 include caspase 3/7 and 9 activation, inhibition of Akt2 phosphorylation, activation of p38α and -γ, and inhibition of proteasome activity. Further pharmacological analyses suggest that pep5 can trigger cell death by distinctive pathways, which can be blocked by IM-54 or a combination of necrostatin-1 and q-VD-OPh. These data further support the biological and pharmacological potential of intracellular peptides.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclin D2/pharmacology , Oligopeptides/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Motifs , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle , Cyclin D2/chemistry , Glioblastoma/drug therapy , HeLa Cells , Humans , Imidazoles/pharmacology , Indoles/pharmacology , MCF-7 Cells , Male , Maleimides/pharmacology , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Quinolines/pharmacology , Rats , Rats, Wistar
20.
J Biol Chem ; 289(23): 15942-50, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24790078

ABSTRACT

The cytokine interleukin-1 (IL-1) has two main pro-inflammatory forms, IL-1α and IL-1ß, which are central to host responses to infection and to damaging sterile inflammation. Processing of IL-1 precursor proteins to active cytokines commonly occurs through activation of proteases, notably caspases and calpains. These proteases are instrumental in cell death, and inflammation and cell death are closely associated, hence we sought to determine the impact of cell death pathways on IL-1 processing and release. We discovered that apoptotic regulation of caspase-8 specifically induced the processing and release of IL-1ß. Conversely, necroptosis caused the processing and release of IL-1α, and this was independent of IL-1ß processing and release. These data suggest that the mechanism through which an IL-1-expressing cell dies dictates the nature of the inflammatory mechanism that follows. These insights may allow modification of inflammation through the selective targeting of cell death mechanisms during disease.


Subject(s)
Apoptosis , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Animals , Calcium/metabolism , Calpain/metabolism , Caspase 8/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Necrosis
SELECTION OF CITATIONS
SEARCH DETAIL