Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Cell Dev Biol ; 30: 337-56, 2014.
Article in English | MEDLINE | ID: mdl-25150011

ABSTRACT

Cell turnover is a fundamental feature in metazoans. Cells can die passively, as a consequence of severe damage to their structural integrity, or actively, owing to a more confined biological disruption such as DNA damage. Passive cell death is uncontrolled and often harmful to the organism. In contrast, active cell death is tightly regulated and serves to support the organism's life. Apoptosis-the primary form of regulated cell death-is relatively well defined. Necroptosis-an alternative, distinct kind of regulated cell death discovered more recently-is less well understood. Apoptosis and necroptosis can be triggered either from within the cell or by extracellular stimuli. Certain signaling components, including several death ligands and receptors, can regulate both processes. Whereas apoptosis is triggered and executed via intracellular proteases called caspases, necroptosis is suppressed by caspase activity. Here we highlight current understanding of the key signaling mechanisms that control regulated cell death.


Subject(s)
Cell Death/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/physiology , Caspases/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Enzyme Activation , Humans , Models, Biological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factors/physiology
2.
Biochem J ; 479(3): 357-384, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35147165

ABSTRACT

Regulated cell death is a vital and dynamic process in multicellular organisms that maintains tissue homeostasis and eliminates potentially dangerous cells. Apoptosis, one of the better-known forms of regulated cell death, is activated when cell-surface death receptors like Fas are engaged by their ligands (the extrinsic pathway) or when BCL-2-family pro-apoptotic proteins cause the permeabilization of the mitochondrial outer membrane (the intrinsic pathway). Both the intrinsic and extrinsic pathways of apoptosis lead to the activation of a family of proteases, the caspases, which are responsible for the final cell demise in the so-called execution phase of apoptosis. In this review, I will first discuss the most common types of regulated cell death on a morphological basis. I will then consider in detail the molecular pathways of intrinsic and extrinsic apoptosis, discussing how they are activated in response to specific stimuli and are sometimes overlapping. In-depth knowledge of the cellular mechanisms of apoptosis is becoming more and more important not only in the field of cellular and molecular biology but also for its translational potential in several pathologies, including neurodegeneration and cancer.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Animals , Apoptosomes/physiology , Apoptosomes/ultrastructure , Autophagy , Caspases/physiology , Humans , Invertebrates/cytology , Ligands , Lysosomes/physiology , Macrophages/physiology , Mitochondrial Membranes/physiology , Necrosis , Neoplasm Proteins/physiology , Permeability , Phagocytosis , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Death Domain/physiology
3.
Nat Rev Mol Cell Biol ; 11(10): 700-14, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20823910

ABSTRACT

For a long time, apoptosis was considered the sole form of programmed cell death during development, homeostasis and disease, whereas necrosis was regarded as an unregulated and uncontrollable process. Evidence now reveals that necrosis can also occur in a regulated manner. The initiation of programmed necrosis, 'necroptosis', by death receptors (such as tumour necrosis factor receptor 1) requires the kinase activity of receptor-interacting protein 1 (RIP1; also known as RIPK1) and RIP3 (also known as RIPK3), and its execution involves the active disintegration of mitochondrial, lysosomal and plasma membranes. Necroptosis participates in the pathogenesis of diseases, including ischaemic injury, neurodegeneration and viral infection, thereby representing an attractive target for the avoidance of unwarranted cell death.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Receptors, Death Domain/physiology , Animals , Cell Membrane/pathology , Cell Membrane/physiology , Humans , Lysosomes/pathology , Lysosomes/physiology , Macrophages/microbiology , Macrophages/pathology , Mitochondria/pathology , Mitochondria/physiology , Morphogenesis/physiology , Necrosis , Protein Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/physiology , Shigella flexneri/pathogenicity
4.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295631

ABSTRACT

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Fas-Associated Death Domain Protein/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF Receptor-Associated Death Domain Protein/physiology , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/physiology , Fas-Associated Death Domain Protein/immunology , Humans , Immunity, Active , Immunity, Innate , Receptors, Death Domain/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Death Domain Protein/immunology
5.
Med Sci Monit ; 24: 8722-8733, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30504760

ABSTRACT

BACKGROUND Hypoxia induces cell apoptosis in the uterosacral ligaments of patients with pelvic organ prolapse by upregulation of hypoxia-inducible factor-1α (HIF-1α). This study aimed to investigate the effects of HIF-1α on human uterosacral ligament fibroblasts (hUSLFs) following treatment with the chemical inducer of hypoxia, cobalt chloride (CoCl2), and to explore the underlying mechanisms. MATERIAL AND METHODS Ten women who underwent hysterectomy for benign disease provided uterosacral ligament tissue for cell extraction. Following CoCl2 treatment, cell viability of isolated and cultured hUSLFs was evaluated by the MTT assay. JC-1 fluorescence mitochondrial imaging was used to study the change in mitochondrial membrane potential. Cell apoptosis and expression of apoptosis-associated proteins and collagen type I alpha 1 (COL1A1) were measured by flow cytometry, TUNEL and Western blot, respectively. RESULTS Hypoxia increased the expression of HIF-1a and increased cell apoptosis, decreased cell viability and expression levels of COL1A1. The JC-1 assay showed that the mitochondrial membrane potential was reduced and caspase-8, and -9 inhibitors partly reduced hUSLF apoptosis. HIF-1α treatment downregulated the expression of cellular FLICE inhibitory protein (c-FLIP), decoy receptor 2 (DcR2), and the ratio of Bcl-2 to Bax, and upregulated the expression tumor necrosis factor related apoptosis-inducing ligand (TRAIL), death receptor 5 (DR5) or TRAIL-R2, Fas, Bcl-2 interacting protein 3 (BNIP3), and cytochrome C, and increased the activation of caspase-3, caspase-8, and caspase-9, all of which were reversed by knockdown of HIF-1α. CONCLUSIONS HIF-1α significantly induced apoptosis of hUSLFs through both the cell death receptor and the mitochondrial-associated apoptosis pathways.


Subject(s)
Fibroblasts/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Receptors, Death Domain/physiology , Adult , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cell Hypoxia/physiology , Cell Survival , China , Cobalt/pharmacology , Collagen Type I/genetics , Collagen Type I/physiology , Collagen Type I, alpha 1 Chain , Female , Fibroblasts/metabolism , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ligaments , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Pelvic Organ Prolapse/complications , Primary Cell Culture , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Uterus
6.
Proc Natl Acad Sci U S A ; 112(34): 10732-7, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261303

ABSTRACT

The diphthamide on human eukaryotic translation elongation factor 2 (eEF2) is the target of ADP ribosylating diphtheria toxin (DT) and Pseudomonas exotoxin A (PE). This modification is synthesized by seven dipthamide biosynthesis proteins (DPH1-DPH7) and is conserved among eukaryotes and archaea. We generated MCF7 breast cancer cell line-derived DPH gene knockout (ko) cells to assess the impact of complete or partial inactivation on diphthamide synthesis and toxin sensitivity, and to address the biological consequence of diphthamide deficiency. Cells with heterozygous gene inactivation still contained predominantly diphthamide-modified eEF2 and were as sensitive to PE and DT as parent cells. Thus, DPH gene copy number reduction does not affect overall diphthamide synthesis and toxin sensitivity. Complete inactivation of DPH1, DPH2, DPH4, and DPH5 generated viable cells without diphthamide. DPH1ko, DPH2ko, and DPH4ko harbored unmodified eEF2 and DPH5ko ACP- (diphthine-precursor) modified eEF2. Loss of diphthamide prevented ADP ribosylation of eEF2, rendered cells resistant to PE and DT, but does not affect sensitivity toward other protein synthesis inhibitors, such as saporin or cycloheximide. Surprisingly, cells without diphthamide (independent of which the DPH gene compromised) were presensitized toward nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) and death-receptor pathways without crossing lethal thresholds. In consequence, loss of diphthamide rendered cells hypersensitive toward TNF-mediated apoptosis. This finding suggests a role of diphthamide in modulating NF-κB, death receptor, or apoptosis pathways.


Subject(s)
Apoptosis/physiology , Histidine/analogs & derivatives , NF-kappa B/physiology , Peptide Elongation Factor 2/chemistry , Receptors, Death Domain/physiology , Apoptosis/drug effects , Apoptosis/genetics , Bacterial Proteins/pharmacology , Breast Neoplasms/pathology , Carbon-Nitrogen Ligases/deficiency , Carbon-Nitrogen Ligases/physiology , Cell Line, Tumor , Diphtheria Toxin/pharmacology , Female , Gene Dosage , Gene Knockout Techniques , Histidine/biosynthesis , Histidine/deficiency , Humans , Neoplasm Proteins/physiology , Protein Processing, Post-Translational
7.
Proc Natl Acad Sci U S A ; 111(46): 16520-5, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368155

ABSTRACT

Colorectal tumorigenesis is driven by genetic alterations in the adenomatous polyposis coli (APC) tumor suppressor pathway and effectively inhibited by nonsteroidal antiinflammatory drugs (NSAIDs). However, how NSAIDs prevent colorectal tumorigenesis has remained obscure. We found that the extrinsic apoptotic pathway and the BH3 interacting-domain death agonist (BID) are activated in adenomas from NSAID-treated patients. Loss of BID abolishes NSAID-mediated tumor suppression, survival benefit, and apoptosis in tumor-initiating stem cells in APC(Min/+) mice. BID-mediated cross-talk between the extrinsic and intrinsic apoptotic pathways is responsible for selective killing of neoplastic cells by NSAIDs. We further demonstrate that NSAIDs induce death receptor signaling in both cancer and normal cells, but only activate BID in cells with APC deficiency and ensuing c-Myc activation. Our results suggest that NSAIDs suppress intestinal tumorigenesis through BID-mediated synthetic lethality triggered by death receptor signaling and gatekeeper mutations, and provide a rationale for developing more effective cancer prevention strategies and agents.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis , BH3 Interacting Domain Death Agonist Protein/physiology , Genes, APC , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis Regulatory Proteins/physiology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , Caspases/physiology , Cell Line, Tumor , Colon/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Indomethacin/pharmacology , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Organ Specificity , Pyrazoles/pharmacology , RNA, Small Interfering/pharmacology , Receptors, Death Domain/physiology , Stem Cells/metabolism , Stem Cells/pathology , Sulfonamides/pharmacology , Sulindac/pharmacology
8.
Adv Exp Med Biol ; 930: 89-112, 2016.
Article in English | MEDLINE | ID: mdl-27558818

ABSTRACT

Apoptosis, a major form of programmed cell death, is an important mechanism to remove extra or unwanted cells during development. In tissue homeostasis apoptosis also acts as a monitoring machinery to eliminate damaged cells in response to environmental stresses. During these processes, caspases, a group of proteases, have been well defined as key drivers of cell death. However, a wealth of evidence is emerging which supports the existence of many other non-apoptotic functions of these caspases, which are essential not only in proper organism development but also in tissue homeostasis and post-injury recovery. In particular, apoptotic caspases in stress-induced dying cells can activate mitogenic signals leading to proliferation of neighbouring cells, a phenomenon termed apoptosis-induced proliferation. Apparently, such non-apoptotic functions of caspases need to be controlled and restrained in a context-dependent manner during development to prevent their detrimental effects. Intriguingly, accumulating studies suggest that cancer cells are able to utilise these functions of caspases to their advantage to enable their survival, proliferation and metastasis in order to grow and progress. This book chapter will review non-apoptotic functions of the caspases in development and tissue homeostasis with focus on how these cellular processes can be hijacked by cancer cells and contribute to tumourigenesis.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Neoplasm Proteins/physiology , Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/physiology , Carcinogenesis , Cell Division , DNA Damage , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Homeostasis , Humans , Mammals/physiology , Mitosis , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/physiopathology , Receptors, Death Domain/physiology , Signal Transduction/physiology
9.
Adv Exp Med Biol ; 930: 1-23, 2016.
Article in English | MEDLINE | ID: mdl-27558815

ABSTRACT

Cell death is a major mechanism to eliminate cells in which DNA is damaged, organelles are stressed, or oncogenes are overexpressed, all events that would otherwise predispose cells to oncogenic transformation. The pathways that initiate and execute cell death are complex, genetically encoded, and subject to significant regulation. Consequently, while these pathways are often mutated in malignancy, there is considerable interest in inducing cell death in tumor cells as therapy. This chapter addresses our current understanding of molecular mechanisms contributing to two cell death pathways, apoptotic cell death and necroptosis, a regulated form of necrotic cell death. Apoptosis can be induced by a wide variety of signals, leading to protease activation that dismantles the cell. We discuss the physiological importance of each apoptosis pathway and summarize their known roles in cancer suppression and the current efforts at targeting each pathway therapeutically. The intricate mechanistic link between death receptor-mediated apoptosis and necroptosis is described, as well as the potential opportunities for utilizing necroptosis in the treatment of malignancy.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Necrosis , Neoplasm Proteins/physiology , Neoplasms/pathology , Animals , Apoptosis Regulatory Proteins/physiology , Caspases/physiology , Cytokines/physiology , Humans , Inflammasomes/physiology , Mice , Mice, Knockout , Mitochondria/physiology , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/physiology , Signal Transduction
10.
Diabetologia ; 58(1): 140-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25301392

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes results from T cell-mediated destruction of pancreatic beta cells. The mechanisms of beta cell destruction in vivo, however, remain unclear. We aimed to test the relative roles of the main cell death pathways: apoptosis, necrosis and necroptosis, in beta cell death in the development of CD4(+) T cell-mediated autoimmune diabetes. METHODS: We altered expression levels of critical cell death proteins in mouse islets and tested their ability to survive CD4(+) T cell-mediated attack using an in vivo graft model. RESULTS: Loss of the B cell leukaemia/lymphoma 2 (BCL-2) homology domain 3-only proteins BIM, PUMA or BID did not protect beta cells from this death. Overexpression of the anti-apoptotic protein BCL-2 or combined deficiency of the pro-apoptotic multi-BCL2 homology domain proteins BAX and BAK also failed to prevent beta cell destruction. Furthermore, loss of function of the death receptor Fas or its essential downstream signalling molecule Fas-associated death domain (FADD) in islets was also not protective. Using electron microscopy we observed that dying beta cells showed features of necrosis. However, islets deficient in receptor-interacting serine/threonine protein kinase 3 (RIPK3), a critical initiator of necroptosis, were still normally susceptible to CD4(+) T cell-mediated destruction. Remarkably, simultaneous inhibition of apoptosis and necroptosis by combining loss of RIPK3 and overexpression of BCL-2 in islets did not protect them against immune attack either. CONCLUSIONS/INTERPRETATION: Collectively, our data indicate that beta cells die by necrosis in autoimmune diabetes and that the programmed cell death pathways apoptosis and necroptosis are both dispensable for this process.


Subject(s)
Autoimmunity/physiology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/pathology , T-Lymphocytes/immunology , Animals , Apoptosis/genetics , Apoptosis/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Necrosis/genetics , Necrosis/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/genetics , Receptors, Death Domain/physiology
11.
Apoptosis ; 20(2): 216-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25528554

ABSTRACT

Death receptors such as Tumor necrosis factor receptor 1, FAS and TNF-associated apoptosis-inducing ligand-R1/2 play a major role in counteracting with bacterial pathogen infection through regulation of inflammation and programmed cell death. The highly regulated death receptor signaling is frequently targeted by gram-negative bacterial pathogens such as Salmonella, Shigella, enteropathogenic Escherichia coli and enterohamorrhagic Escherichia coli, which harbor a conserved type III secretion system that delivers a repertoire of effector proteins to manipulate host signal transductions for their own benefit. This review focuses on how bacterial gut pathogens hijack death receptor signaling to inhibit host NF-κB and programmed cell death pathways.


Subject(s)
Apoptosis , Bacterial Proteins/physiology , Receptors, Death Domain/physiology , Animals , Bacterial Secretion Systems , Enterobacteriaceae/physiology , Host-Pathogen Interactions , Humans , NF-kappa B/physiology , Signal Transduction
12.
Apoptosis ; 20(2): 174-95, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25420757

ABSTRACT

CARD subfamily is the second largest subfamily in the DD superfamily that plays important roles in regulating various signaling pathways, including but not limited to NF-kB activation signaling, apoptosis signaling and inflammatory signaling. The CARD subfamily contains 33 human CARD-containing proteins, regulating the assembly of many signaling complexes, including apoptosome, inflammsome, nodosome, the CBM complex, PIDDosome, the TRAF2 complex, and the MAVS signalosome, by homotypic CARD-CARD interactions. The mechanism of how CARDs find the right binding partner to form a specific complex remains unclear. This review uses different classification schemes to update the classification of CARD-containing proteins. Combining the classification based on domain structures, functions, associated signaling complexes, and roles would help better understand the structural and function diversity of CARD-containing proteins. This review also summarizes recent structural studies on CARDs. Especially, the CARD-containing complexes can be divided into the homodimeric, heterodimeric, oligomeric, filamentous CARD complexes and the CARD-ubiquitin complex. This review will give an overview of the versatile roles of CARDs in regulating signaling transduction, as well as the therapeutic drugs targeting CARD-containing proteins.


Subject(s)
Apoptosis , CARD Signaling Adaptor Proteins/physiology , NF-kappa B/metabolism , Humans , Inflammation/metabolism , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Receptors, Death Domain/physiology , Signal Transduction
13.
Nat Med ; 13(9): 1070-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17767167

ABSTRACT

Apo2L/TRAIL stimulates cancer cell death through the proapoptotic receptors DR4 and DR5, but the determinants of tumor susceptibility to this ligand are not fully defined. mRNA expression of the peptidyl O-glycosyltransferase GALNT14 correlated with Apo2L/TRAIL sensitivity in pancreatic carcinoma, non-small-cell lung carcinoma and melanoma cell lines, and up to 30% of samples from various human malignancies showed GALNT14 overexpression. RNA interference of GALNT14 reduced cellular Apo2L/TRAIL sensitivity, whereas overexpression increased responsiveness. Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine-galactose-sialic acid) structures. Sequence comparison predicted conserved extracellular DR4 and DR5 O-glycosylation sites; progressive mutation of the DR5 sites attenuated apoptotic signaling. O-glycosylation promoted ligand-stimulated clustering of DR4 and DR5, which mediated recruitment and activation of the apoptosis-initiating protease caspase-8. These results uncover a new link between death-receptor O-glycosylation and apoptotic signaling, providing potential predictive biomarkers for Apo2L/TRAIL-based cancer therapy.


Subject(s)
Receptors, Death Domain/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Amino Acid Sequence , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Cell Survival , Genetic Predisposition to Disease , Glycosylation , Humans , Lung Neoplasms , Melanoma , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms , RNA, Messenger/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transplantation, Heterologous
14.
IUBMB Life ; 65(8): 651-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824949

ABSTRACT

Loss of cardiomyocytes plays a critical role in the pathogenesis of heart failure. With fewer myocytes, the heart is unable to sustain efficient contraction. Much attention has been focused on understanding mechanisms of cell death in myocytes with the ultimate goal being to reduce the extent of injury and improve function in the failing myocardium. Both necrosis and apoptosis contribute to loss of myocytes, and this loss of cells is a hallmark of cardiac pathologies, including ischemia/reperfusion, myocardial infarction, and heart failure. Apoptosis is a highly regulated process that is activated via death receptors in the plasma membrane or via permeabilization of the mitochondria. Necrosis is generally viewed as an uncontrolled process that leads to mitochondrial swelling, cell rupture, and subsequent inflammation. However, recent studies have uncovered a signaling pathway that mediates regulated necrosis or necroptosis. Mitochondria play an important role in both apoptosis and necrosis, and changes in their morphology can affect the cells' susceptibility to stress. This review focuses on the various modes of cell death in the myocardium and highlights how they contribute to loss of myocytes in response to stress.


Subject(s)
Cell Death/physiology , Myocardium/pathology , Myocytes, Cardiac/pathology , Animals , Apoptosis/physiology , Heart Failure/pathology , Humans , MicroRNAs/physiology , Mitochondria, Heart/pathology , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore , Necrosis/pathology , Receptors, Death Domain/physiology , Signal Transduction/physiology
15.
Exp Cell Res ; 318(11): 1269-77, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22542855

ABSTRACT

Cell death is critical to the normal functioning of multi-cellular organisms, playing a central role in development, immunity, inflammation, and cancer progression. Two cell death mechanisms, apoptosis and necroptosis, are dependent on the formation of distinct multi-protein complexes including the DISC, Apoptosome, Piddosome and Necrosome following the induction of cell death by specific stimuli. The role of several of these key multi-protein signalling platforms, namely the DISC, TNFR1 complex I/II, the Necrosome and Ripoptosome, in mediating these pathways will be discussed, as well as the open questions and potential therapeutic benefits of understanding their underlying mechanisms.


Subject(s)
Cell Death , Receptors, Death Domain/physiology , Signal Transduction , Animals , Humans , Multiprotein Complexes , Receptors, Death Domain/metabolism
16.
Mol Pharmacol ; 81(5): 701-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22328720

ABSTRACT

In cervical cancer, the p53 and retinoblastoma (pRb) tumor suppressor pathways are disrupted by the human papilloma virus (HPV) E6 and E7 oncoproteins, because E6 targets p53 and E7 targets pRb for rapid proteasome-mediated degradation. We have investigated whether E6 suppression with small interfering RNA (siRNA) restores p53 functionality and sensitizes the HPV16-positive cervical cancer cell line SiHa to apoptosis by cisplatin, irradiation, recombinant human tumor necrosis factor-related apoptosis-inducing ligand (rhTRAIL), or agonistic anti-Fas antibody. E6 siRNA resulted in decreased E6 mRNA levels and enhanced p53 and p21 expression, demonstrating the restoration of p53 functionality in SiHa cells, without inducing high levels of apoptosis (<10%). Cell surface expression of the proapoptotic death receptors (DRs) DR4, DR5, and Fas was not affected by E6 suppression. E6 suppression conferred susceptibility to cisplatin-induced apoptosis but not to irradiation-, rhTRAIL-, or anti-Fas antibody-induced apoptosis. Combining cisplatin with rhTRAIL or anti-Fas antibody induced even higher apoptosis levels in E6-suppressed cells. At the molecular level, cisplatin treatment resulted in elevated p53 levels, enhanced caspase-3 activation, and reduced p21 levels in E6-suppressed cells. Cisplatin in combination with death receptor ligands enhanced caspase-8 and caspase-3 activation and reduced X-linked inhibitor-of-apoptosis protein (XIAP) levels in these cells. We showed using siRNA that the enhanced apoptosis in E6-supressed cells was related to reduced XIAP levels and not due to reduced p21 levels. In conclusion, targeting E6 or XIAP in combination with cisplatin can efficiently potentiate rhTRAIL-induced apoptosis in HPV-positive cervical cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Oncogene Proteins, Viral/physiology , Receptors, Death Domain/physiology , Repressor Proteins/physiology , Uterine Cervical Neoplasms/drug therapy , Caspase 3/metabolism , Caspase 8/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , HeLa Cells , Humans , Oncogene Proteins, Viral/genetics , RNA Interference , Repressor Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Suppressor Protein p53/analysis , Uterine Cervical Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/physiology
17.
J Neuroinflammation ; 9: 82, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22540228

ABSTRACT

BACKGROUND: The mechanisms of progressive dopaminergic neuronal loss in Parkinson's disease (PD) remain poorly understood, largely due to the complex etiology and multifactorial nature of disease pathogenesis. Several lines of evidence from human studies and experimental models over the last decade have identified neuroinflammation as a potential pathophysiological mechanism contributing to disease progression. Tumor necrosis factor α (TNF) has recently emerged as the primary neuroinflammatory mediator that can elicit dopaminergic cell death in PD. However, the signaling pathways by which TNF mediates dopaminergic cell death have not been completely elucidated. METHODS: In this study we used a dopaminergic neuronal cell model and recombinant TNF to characterize intracellular signaling pathways activated during TNF-induced dopaminergic neurotoxicity. Etanercept and neutralizing antibodies to tumor necrosis factor receptor 1 (TNFR1) were used to block TNF signaling. We confirmed the results from our mechanistic studies in primary embryonic mesencephalic cultures and in vivo using the stereotaxic lipopolysaccharide (LPS) model of nigral dopaminergic degeneration. RESULTS: TNF signaling in dopaminergic neuronal cells triggered the activation of protein kinase Cδ (PKCδ), an isoform of the novel PKC family, by caspase-3 and caspase-8 dependent proteolytic cleavage. Both TNFR1 neutralizing antibodies and the soluble TNF receptor Etanercept blocked TNF-induced PKCδ proteolytic activation. Proteolytic activation of PKCδ was accompanied by translocation of the kinase to the nucleus. Notably, inhibition of PKCδ signaling by small interfering (si)RNA or overexpression of a PKCδ cleavage-resistant mutant protected against TNF-induced dopaminergic neuronal cell death. Further, primary dopaminergic neurons obtained from PKCδ knockout (-/-) mice were resistant to TNF toxicity. The proteolytic activation of PKCδ in the mouse substantia nigra in the neuroinflammatory LPS model was also observed. CONCLUSIONS: Collectively, these results identify proteolytic activation of PKCδ proapoptotic signaling as a key downstream effector of dopaminergic cell death induced by TNF. These findings also provide a rationale for therapeutically targeting PKCδ to mitigate progressive dopaminergic degeneration resulting from chronic neuroinflammatory processes.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Dopaminergic Neurons/enzymology , Encephalitis/enzymology , Encephalitis/pathology , Protein Kinase C-delta/metabolism , Receptors, Death Domain/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/toxicity , Cell Death/physiology , Cells, Cultured , Dopaminergic Neurons/pathology , Encephalitis/etiology , Enzyme Activation/physiology , Mice , Protein Kinase C-delta/physiology , Proteolysis , Rats , Tumor Necrosis Factor-alpha/toxicity
18.
Toxicol Appl Pharmacol ; 258(1): 72-81, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22027265

ABSTRACT

We investigated whether bee venom and melittin, a major component of bee venom, inhibit cell growth through enhancement of death receptor expressions in the human ovarian cancer cells, SKOV3 and PA-1. Bee venom (1-5 µg/ml) and melittin (0.5-2 µg/ml) inhibited the growth of SKOV3 and PA-1 ovarian cancer cells by the induction of apoptotic cell death in a dose dependent manner. Consistent with apoptotic cell death, expression of death receptor (DR) 3 and DR6 was increased in both cancer cells, but expression of DR4 was increased only in PA-1 cells. Expression of DR downstream pro-apoptotic proteins including caspase-3, 8, and Bax was concomitantly increased, but the phosphorylation of JAK2 and STAT3 and the expression of Bcl-2 were inhibited by treatment with bee venom and melittin in SKOV3 and PA-1 cells. Expression of cleaved caspase-3 was increased in SKOV3, but cleaved caspase-8 was increased in PA-1 cells. Moreover, deletion of DR3, DR4, and DR6 by small interfering RNA significantly reversed bee venom and melittin-induced cell growth inhibitory effect as well as down regulation of STAT3 by bee venom and melittin in SKOV3 and PA-1 ovarian cancer cell. These results suggest that bee venom and melittin induce apoptotic cell death in ovarian cancer cells through enhancement of DR3, DR4, and DR6 expression and inhibition of STAT3 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Bee Venoms/pharmacology , Janus Kinase 2/antagonists & inhibitors , Melitten/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Death Domain/physiology , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Apoptosis/drug effects , Female , Humans , Janus Kinase 2/physiology , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/physiology
19.
J Immunol ; 184(8): 4447-54, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20220092

ABSTRACT

L-selectin is an adhesion molecule expressed by neutrophils that broadly directs their infiltration in to sites of inflammation. It is also present at relatively high levels in the serum of normal individuals. It is well established that L-selectin is efficiently shed from the surface of neutrophils upon their activation, a process that regulates its density and binding activity. Neutrophil programmed cell death is critical for the resolution of inflammation, and L-selectin downregulation is induced during this process as well. The mechanisms underpinning this latter process are much less understood, and were investigated in this study. Using a disintegrin and metalloprotease (ADAM)-17 radiation chimeric mice, we demonstrate for the first time that during early events of death receptor-mediated neutrophil apoptosis, L-selectin downregulation occurs primarily by ADAM17-mediated shedding. This was observed as well upon using shRNA to knock down ADAM17 expression in Jurkat cells, a well-studied cell line in terms of the molecular processes involved in the induction of apoptosis. These findings directly reveal that ADAM17 activity occurs during programmed cell death. Hence, the cleavage of particular ADAM17 substrates may be an additional component of the anti-inflammatory program initiated by apoptotic neutrophils. Of interest was that during later stages of induced leukocyte apoptosis, soluble L-selectin production occurred independent of ADAM17, as well as membrane events, such as blebbing and microparticle production. This process may provide an explanation for the lack of diminished serum L-selectin levels in ADAM17-null mice, and suggests a mechanism for the homeostatic maintenance of soluble L-selectin levels in the blood of healthy individuals.


Subject(s)
ADAM Proteins/physiology , Apoptosis/immunology , L-Selectin/biosynthesis , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Death Domain/physiology , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAM17 Protein , Animals , Apoptosis/genetics , Cells, Cultured , Humans , Inflammation Mediators/blood , Inflammation Mediators/physiology , Jurkat Cells , L-Selectin/blood , L-Selectin/metabolism , Mice , Mice, Knockout , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/cytology , Radiation Chimera/genetics , Radiation Chimera/immunology , Receptors, Death Domain/blood , Receptors, Death Domain/genetics , Solubility , Time Factors
20.
Gen Comp Endocrinol ; 176(3): 461-4, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22245291

ABSTRACT

The tumor necrosis factor (TNF) superfamily includes death receptor (DR) ligands, such as TNF-α, FasL, and TRAIL. Death receptors (DRs) induce intracellular signaling upon engagement of their cognate DR ligands, either leading to apoptosis, survival, or proinflammatory responses. The DR signaling is mediated by the recruitment of several death domain (DD)-containing molecules such as Fas-associated death domain (FADD) and receptor-interacting protein (RIP) 1. In this review, we describe DR signaling in mammals, and describe recent findings of DR signaling during metamorphosis in the African clawed frog Xenopus laevis. Specifically, we focus on the cell fate (apoptosis or survival) mediated through a DR ligand, TNF-α or TRAIL in endothelial cells or red blood cells (RBCs). In addition, we discuss relationships between thyroid hormone-induced metamorphosis and DR signaling.


Subject(s)
Apoptosis/physiology , Receptors, Death Domain/physiology , Xenopus laevis/physiology , Animals , Metamorphosis, Biological/physiology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/physiology , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL