51
|
Rodriguez DA, Weinlich R, Brown S, Guy C, Fitzgerald P, Dillon CP, Oberst A, Quarato G, Low J, Cripps JG, Chen T, Green DR. Characterization of RIPK3-mediated phosphorylation of the activation loop of MLKL during necroptosis. Cell Death Differ 2015; 23:76-88. [PMID: 26024392 DOI: 10.1038/cdd.2015.70] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/28/2015] [Accepted: 04/28/2015] [Indexed: 12/14/2022] Open
Abstract
Mixed lineage kinase domain-like pseudokinase (MLKL) mediates necroptosis by translocating to the plasma membrane and inducing its rupture. The activation of MLKL occurs in a multimolecular complex (the 'necrosome'), which is comprised of MLKL, receptor-interacting serine/threonine kinase (RIPK)-3 (RIPK3) and, in some cases, RIPK1. Within this complex, RIPK3 phosphorylates the activation loop of MLKL, promoting conformational changes and allowing the formation of MLKL oligomers, which migrate to the plasma membrane. Previous studies suggested that RIPK3 could phosphorylate the murine MLKL activation loop at Ser345, Ser347 and Thr349. Moreover, substitution of the Ser345 for an aspartic acid creates a constitutively active MLKL, independent of RIPK3 function. Here we examine the role of each of these residues and found that the phosphorylation of Ser345 is critical for RIPK3-mediated necroptosis, Ser347 has a minor accessory role and Thr349 seems to be irrelevant. We generated a specific monoclonal antibody to detect phospho-Ser345 in murine cells. Using this antibody, a series of MLKL mutants and a novel RIPK3 inhibitor, we demonstrate that the phosphorylation of Ser345 is not required for the interaction between RIPK3 and MLKL in the necrosome, but is essential for MLKL translocation, accumulation in the plasma membrane, and consequent necroptosis.
Collapse
Affiliation(s)
- D A Rodriguez
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - R Weinlich
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - S Brown
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - C Guy
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - P Fitzgerald
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - C P Dillon
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - A Oberst
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - G Quarato
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J Low
- Department Chemical Biology & Therapeutics, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J G Cripps
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202, USA
| | - T Chen
- Department Chemical Biology & Therapeutics, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - D R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
52
|
Zhang M, Harashima N, Moritani T, Huang W, Harada M. The Roles of ROS and Caspases in TRAIL-Induced Apoptosis and Necroptosis in Human Pancreatic Cancer Cells. PLoS One 2015; 10:e0127386. [PMID: 26000607 PMCID: PMC4441514 DOI: 10.1371/journal.pone.0127386] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/15/2015] [Indexed: 01/22/2023] Open
Abstract
Death signaling provided by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) can induce death in cancer cells with little cytotoxicity to normal cells; this cell death has been thought to involve caspase-dependent apoptosis. Reactive oxygen species (ROS) are also mediators that induce cell death, but their roles in TRAIL-induced apoptosis have not been elucidated fully. In the current study, we investigated ROS and caspases in human pancreatic cancer cells undergoing two different types of TRAIL-induced cell death, apoptosis and necroptosis. TRAIL treatment increased ROS in two TRAIL-sensitive pancreatic cancer cell lines, MiaPaCa-2 and BxPC-3, but ROS were involved in TRAIL-induced apoptosis only in MiaPaCa-2 cells. Unexpectedly, inhibition of ROS by either N-acetyl-L-cysteine (NAC), a peroxide inhibitor, or Tempol, a superoxide inhibitor, increased the annexin V-/propidium iodide (PI)+ early necrotic population in TRAIL-treated cells. Additionally, both necrostatin-1, an inhibitor of receptor-interacting protein kinase 1 (RIP1), and siRNA-mediated knockdown of RIP3 decreased the annexin V-/PI+ early necrotic population after TRAIL treatment. Furthermore, an increase in early apoptosis was induced in TRAIL-treated cancer cells under inhibition of either caspase-2 or -9. Caspase-2 worked upstream of caspase-9, and no crosstalk was observed between ROS and caspase-2/-9 in TRAIL-treated cells. Together, these results indicate that ROS contribute to TRAIL-induced apoptosis in MiaPaCa-2 cells, and that ROS play an inhibitory role in TRAIL-induced necroptosis of MiaPaCa-2 and BxPC-3 cells, with caspase-2 and -9 playing regulatory roles in this process.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Shengli Street, Yinchuan, China
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Nanae Harashima
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Tamami Moritani
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
| | - Weidong Huang
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Shengli Street, Yinchuan, China
| | - Mamoru Harada
- Department of Immunology, Shimane University Faculty of Medicine, Izumi, Shimane, Japan
- * E-mail:
| |
Collapse
|
53
|
Abstract
Receptor‐interacting protein kinase‐1 (RIPK1) sits at a signaling node controlling a number of functional pathways. These include both positive and negative control of apoptosis and necroptosis (a form of regulated necrosis). In this issue of EMBO Reports, Yonekawa and colleagues describe another function for RIPK1, the inhibition of autophagy via ERK‐mediated phosphorylation of the transcription factor, TFEB [1]. Their findings are considered in the context of RIPK1 signaling, and how it is engaged.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
54
|
Brown MF, Leibowitz BJ, Chen D, He K, Zou F, Sobol RW, Beer-Stolz D, Zhang L, Yu J. Loss of caspase-3 sensitizes colon cancer cells to genotoxic stress via RIP1-dependent necrosis. Cell Death Dis 2015; 6:e1729. [PMID: 25906152 PMCID: PMC4650537 DOI: 10.1038/cddis.2015.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 01/08/2023]
Abstract
Caspase-3 is the best known executioner caspase in apoptosis. We generated caspase-3 knockout (C3KO) and knockdown human colorectal cancer cells, and found that they are unexpectedly sensitized to DNA-damaging agents including 5-fluorouracil (5-FU), etoposide, and camptothecin. C3KO xenograft tumors also displayed enhanced therapeutic response and cell death to 5-FU. C3KO cells showed intact apoptosis and activation of caspase-7 and -9, impaired processing of caspase-8, and induction of necrosis in response to DNA-damaging agents. This form of necrosis is associated with HMGB1 release and ROS production, and suppressed by genetic or pharmacological inhibition of RIP1, MLKL1, or caspase-8, but not inhibitors of pan-caspases or RIP3. 5-FU treatment led to the formation of a z-VAD-resistant pro-caspase-8/RIP1/FADD complex, which was strongly stabilized by caspase-3 KO. These data demonstrate a key role of caspase-3 in caspase-8 processing and suppression of DNA damage-induced necrosis, and provide a potentially novel way to chemosensitize cancer cells.
Collapse
Affiliation(s)
- M F Brown
- Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
| | - B J Leibowitz
- Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
| | - D Chen
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
| | - K He
- Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
| | - F Zou
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
| | - R W Sobol
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
| | - D Beer-Stolz
- Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
| | - L Zhang
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
| | - J Yu
- Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center Research Pavilion, 5117 Centre Ave., Pittsburgh, PA, USA
| |
Collapse
|
55
|
Andrukov BG, Somova LM, Timchenko NF. STRATEGY OF PROGRAMMED CELL DEATH IN PROKARYOTES. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2015. [DOI: 10.15789/2220-7619-2015-1-15-26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Programmed cell death (PCD) was first studied in eukaryotic organisms. This system also operates in the development life cycle of prokaryotes. The system PCD in microorganisms is activated a wide range of signals in response to the stresses associated with adverse environmental conditions or exposure to antibacterial agents. The results of numerous studies in the past decade allow considering the system PCD in prokaryotes as an evolutionary conservation of the species. These results significantly expanded understanding of the role of PCD in microorganisms and opened a number of important areas of research of the morphological and molecular genetic approaches to the study of death strategies for the survival in bacterial populations. The purpose of the review is to summarize the morphological and molecular genetic characteristics of PCD in prokaryotes which are real manifestations of the mechanisms of this phenomenon.
Collapse
|
56
|
Tait SWG, Ichim G, Green DR. Die another way--non-apoptotic mechanisms of cell death. J Cell Sci 2015; 127:2135-44. [PMID: 24833670 DOI: 10.1242/jcs.093575] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Regulated, programmed cell death is crucial for all multicellular organisms. Cell death is essential in many processes, including tissue sculpting during embryogenesis, development of the immune system and destruction of damaged cells. The best-studied form of programmed cell death is apoptosis, a process that requires activation of caspase proteases. Recently it has been appreciated that various non-apoptotic forms of cell death also exist, such as necroptosis and pyroptosis. These non-apoptotic cell death modalities can be either triggered independently of apoptosis or are engaged should apoptosis fail to execute. In this Commentary, we discuss several regulated non-apoptotic forms of cell death including necroptosis, autophagic cell death, pyroptosis and caspase-independent cell death. We outline what we know about their mechanism, potential roles in vivo and define outstanding questions. Finally, we review data arguing that the means by which a cell dies actually matters, focusing our discussion on inflammatory aspects of cell death.
Collapse
Affiliation(s)
- Stephen W G Tait
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Gabriel Ichim
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
57
|
Wang HY, Zhang B. Cobalt Chloride Induces Necroptosis in Human Colon Cancer HT-29 Cells. Asian Pac J Cancer Prev 2015; 16:2569-74. [DOI: 10.7314/apjcp.2015.16.6.2569] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
58
|
Kang TB, Yang SH, Toth B, Kovalenko A, Wallach D. Activation of the NLRP3 inflammasome by proteins that signal for necroptosis. Methods Enzymol 2015; 545:67-81. [PMID: 25065886 DOI: 10.1016/b978-0-12-801430-1.00003-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Necroptosis-a form of programmed necrotic cell death-and its resulting release of damage-associated molecular patterns (DAMPs) are believed to participate in the triggering of inflammatory processes. To assess the relative contribution of this cell death mode to inflammation, we need to know what other cellular effects can be exerted by molecules shown to trigger necrotic death, and the extent to which those effects might themselves contribute to inflammation. Here, we describe the technical approaches that have been applied to assess the impact of the main signaling molecules known to mediate activation of necroptosis upon generation of inflammatory cytokines in LPS-treated mouse bone marrow-derived dendritic cells. The findings obtained by this assessment indicated that signaling molecules known to initiate necroptosis can also initiate activation of the NLRP3 inflammasome, thereby inducing inflammation independently of cell death by triggering the generation of proinflammatory cytokines such as IL-1β.
Collapse
Affiliation(s)
- Tae-Bong Kang
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel; Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chung-Ju, Republic of Korea
| | - Seung-Hoon Yang
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - Beata Toth
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - Andrew Kovalenko
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - David Wallach
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
59
|
Abstract
Mucosal healing has been a central issue in inflammatory bowel disease (IBD) for the last years, and has been proposed to be included as the new treatment goal in IBD. The molecular understanding of both the disruption and the healing of the intestinal epithelial cell lining and the mucosal barrier in IBD is complex and only partly understood. There is no general agreement on how to define healed mucosa, but there is a general acceptance that clinicians should use endoscopy and imaging technique in their assessments. Mucosal healing is an old concept that has been actualized in the present era of the highly effective biological agents. Randomized clinical studies with mucosal healing as end-point parameters have been reported, and early mucosal healing has been associated with low complication rates. We are waiting for documentation of whether treatment to healed mucosa can change the natural course of IBD. The concept of immunological remission has recently been introduced and can be the new treatment goal and one of several criteria for discontinuation of biological treatment in IBD. In conclusion, mucosal healing is a fairly novel concept and goal for biological treatment of IBD. There is a need for a standardization of its assessment and validation of the prognostic value.
Collapse
Affiliation(s)
- Jon Florholmen
- Research group of Gastroenterology and Nutrition, Institute of Clinicel Medicine, Arctic University of Norway , Tromsø , Norway
| |
Collapse
|
60
|
Myeloid-derived suppressor activity is mediated by monocytic lineages maintained by continuous inhibition of extrinsic and intrinsic death pathways. Immunity 2014; 41:947-59. [PMID: 25500368 DOI: 10.1016/j.immuni.2014.10.020] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/31/2014] [Indexed: 12/31/2022]
Abstract
Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.
Collapse
|
61
|
Creagh EM. Caspase crosstalk: integration of apoptotic and innate immune signalling pathways. Trends Immunol 2014; 35:631-640. [PMID: 25457353 DOI: 10.1016/j.it.2014.10.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 01/30/2023]
Abstract
The caspase family of cysteine proteases has been functionally divided into two groups: those involved in apoptosis and those involved in innate immune signalling. Recent findings have identified 'apoptotic' caspases within inflammasome complexes and revealed that 'inflammatory' caspases are capable of inducing cell death, suggesting that the earlier view of caspase function may have been overly simplistic. Here, I review evidence attributing nonclassical functions to many caspases and propose that caspases serve as critical mediators in the integration of apoptotic and inflammatory pathways, thereby forming an integrated signalling system that regulates cell death and innate immune responses during development, infection, and homeostasis.
Collapse
Affiliation(s)
- Emma M Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.
| |
Collapse
|
62
|
Peltzer N, Rieser E, Taraborrelli L, Draber P, Darding M, Pernaute B, Shimizu Y, Sarr A, Draberova H, Montinaro A, Martinez-Barbera JP, Silke J, Rodriguez TA, Walczak H. HOIP deficiency causes embryonic lethality by aberrant TNFR1-mediated endothelial cell death. Cell Rep 2014; 9:153-165. [PMID: 25284787 DOI: 10.1016/j.celrep.2014.08.066] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/31/2014] [Accepted: 08/26/2014] [Indexed: 10/25/2022] Open
Abstract
Linear ubiquitination is crucial for innate and adaptive immunity. The linear ubiquitin chain assembly complex (LUBAC), consisting of HOIL-1, HOIP, and SHARPIN, is the only known ubiquitin ligase that generates linear ubiquitin linkages. HOIP is the catalytically active LUBAC component. Here, we show that both constitutive and Tie2-Cre-driven HOIP deletion lead to aberrant endothelial cell death, resulting in defective vascularization and embryonic lethality at midgestation. Ablation of tumor necrosis factor receptor 1 (TNFR1) prevents cell death, vascularization defects, and death at midgestation. HOIP-deficient cells are more sensitive to death induction by both tumor necrosis factor (TNF) and lymphotoxin-α (LT-α), and aberrant complex-II formation is responsible for sensitization to TNFR1-mediated cell death in the absence of HOIP. Finally, we show that HOIP's catalytic activity is necessary for preventing TNF-induced cell death. Hence, LUBAC and its linear-ubiquitin-forming activity are required for maintaining vascular integrity during embryogenesis by preventing TNFR1-mediated endothelial cell death.
Collapse
Affiliation(s)
- Nieves Peltzer
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Eva Rieser
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Peter Draber
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Maurice Darding
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Barbara Pernaute
- British Heart Foundation Centre for Research Excellence, National Heart and Lung Institute (NHLI), Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Yutaka Shimizu
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Aida Sarr
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Helena Draberova
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Juan Pedro Martinez-Barbera
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Tristan A Rodriguez
- British Heart Foundation Centre for Research Excellence, National Heart and Lung Institute (NHLI), Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
63
|
Abstract
Beyond their contribution to basic metabolism, the major cellular organelles, in particular mitochondria, can determine whether cells respond to stress in an adaptive or suicidal manner. Thus, mitochondria can continuously adapt their shape to changing bioenergetic demands as they are subjected to quality control by autophagy, or they can undergo a lethal permeabilization process that initiates apoptosis. Along similar lines, multiple proteins involved in metabolic circuitries, including oxidative phosphorylation and transport of metabolites across membranes, may participate in the regulated or catastrophic dismantling of organelles. Many factors that were initially characterized as cell death regulators are now known to physically or functionally interact with metabolic enzymes. Thus, several metabolic cues regulate the propensity of cells to activate self-destructive programs, in part by acting on nutrient sensors. This suggests the existence of "metabolic checkpoints" that dictate cell fate in response to metabolic fluctuations. Here, we discuss recent insights into the intersection between metabolism and cell death regulation that have major implications for the comprehension and manipulation of unwarranted cell loss.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, F-75006 Paris, France. Université Paris Descartes/Paris V; Sorbonne Paris Cité; F-75005 Paris, France. INSERM, U1138, F-94805 Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, F-75006 Paris, France. Université Paris Descartes/Paris V; Sorbonne Paris Cité; F-75005 Paris, France. INSERM, U1138, F-94805 Villejuif, France. Metabolomics and Cell Biology Platforms, Gustave Roussy, F-94805 Villejuif, France. Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, F-75015 Paris, France.
| |
Collapse
|
64
|
Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, Baehrecke EH, Bazan NG, Bertrand MJ, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Campanella M, Candi E, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR, Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ, Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ, Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Muñoz-Pinedo C, Nuñez G, Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y, Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden Berghe T, Vandenabeele P, Villunger A, Wagner EF, et alGalluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, Baehrecke EH, Bazan NG, Bertrand MJ, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Campanella M, Candi E, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR, Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ, Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ, Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Muñoz-Pinedo C, Nuñez G, Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y, Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden Berghe T, Vandenabeele P, Villunger A, Wagner EF, Walczak H, White E, Wood WG, Yuan J, Zakeri Z, Zhivotovsky B, Melino G, Kroemer G. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ 2014; 22:58-73. [PMID: 25236395 PMCID: PMC4262782 DOI: 10.1038/cdd.2014.137] [Show More Authors] [Citation(s) in RCA: 718] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as ‘accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. ‘Regulated cell death' (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death.
Collapse
Affiliation(s)
- L Galluzzi
- 1] Gustave Roussy Cancer Center, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - J M Bravo-San Pedro
- 1] Gustave Roussy Cancer Center, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] INSERM, U1138, Gustave Roussy, Paris, France
| | - I Vitale
- Regina Elena National Cancer Institute, Rome, Italy
| | - S A Aaronson
- Department of Oncological Sciences, The Tisch Cancer Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - J M Abrams
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - D Adam
- Institute of Immunology, Christian-Albrechts University, Kiel, Germany
| | - E S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - L Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Napoli, Italy
| | - D Andrews
- Department of Biochemistry and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - M Annicchiarico-Petruzzelli
- Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata - Istituto Ricovero Cura Carattere Scientifico (IDI-IRCCS), Rome, Italy
| | - E H Baehrecke
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - N G Bazan
- Neuroscience Center of Excellence, School of Medicine, New Orleans, LA, USA
| | - M J Bertrand
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - K Bianchi
- 1] Barts Cancer Institute, Cancer Research UK Centre of Excellence, London, UK [2] Queen Mary University of London, John Vane Science Centre, London, UK
| | - M V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K Blomgren
- Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - C Borner
- Institute of Molecular Medicine and Spemann Graduate School of Biology and Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - D E Bredesen
- 1] Buck Institute for Research on Aging, Novato, CA, USA [2] Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - C Brenner
- 1] INSERM, UMRS769, Châtenay Malabry, France [2] LabEx LERMIT, Châtenay Malabry, France [3] Université Paris Sud/Paris XI, Orsay, France
| | - M Campanella
- Department of Comparative Biomedical Sciences and Consortium for Mitochondrial Research, University College London (UCL), London, UK
| | - E Candi
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - F Cecconi
- 1] Laboratory of Molecular Neuroembryology, IRCCS Fondazione Santa Lucia, Rome, Italy [2] Department of Biology, University of Rome Tor Vergata; Rome, Italy [3] Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - F K Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - N S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - E H Cheng
- Human Oncology and Pathogenesis Program and Department of Pathology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - J E Chipuk
- Department of Oncological Sciences, The Tisch Cancer Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - J A Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), National Institute of Health (NIH), North Carolina, NC, USA
| | - A Ciechanover
- Tumor and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel
| | - T M Dawson
- 1] Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (ICE), Departments of Neurology, Pharmacology and Molecular Sciences, Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - V L Dawson
- 1] Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (ICE), Departments of Neurology, Pharmacology and Molecular Sciences, Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - V De Laurenzi
- Department of Experimental and Clinical Sciences, Gabriele d'Annunzio University, Chieti, Italy
| | - R De Maria
- Regina Elena National Cancer Institute, Rome, Italy
| | - K-M Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - N Di Daniele
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - V M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA, USA
| | - B D Dynlacht
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY, USA
| | - W S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | - G M Fimia
- 1] Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy [2] Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases Lazzaro Spallanzani, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Rome, Italy
| | - R A Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University, Frankfurt, Germany
| | - C Garrido
- 1] INSERM, U866, Dijon, France [2] Faculty of Medicine, University of Burgundy, Dijon, France
| | - M-L Gougeon
- Antiviral Immunity, Biotherapy and Vaccine Unit, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - D R Green
- Department of Immunology, St Jude's Children's Research Hospital, Memphis, TN, USA
| | - H Gronemeyer
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | - G Hajnoczky
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J M Hardwick
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - M O Hengartner
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - H Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - B Joseph
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institute, Stockholm, Sweden
| | - P J Jost
- Medical Department for Hematology, Technical University of Munich, Munich, Germany
| | - T Kaufmann
- Institute of Pharmacology, Medical Faculty, University of Bern, Bern, Switzerland
| | - O Kepp
- 1] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [2] INSERM, U1138, Gustave Roussy, Paris, France [3] Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - D J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - R A Knight
- 1] Medical Molecular Biology Unit, Institute of Child Health, University College London (UCL), London, UK [2] Medical Research Council Toxicology Unit, Leicester, UK
| | - S Kumar
- 1] Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia [2] School of Medicine and School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - J J Lemasters
- Departments of Drug Discovery and Biomedical Sciences and Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - B Levine
- 1] Center for Autophagy Research, University of Texas, Southwestern Medical Center, Dallas, TX, USA [2] Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA
| | - A Linkermann
- Division of Nephrology and Hypertension, Christian-Albrechts University, Kiel, Germany
| | - S A Lipton
- 1] The Scripps Research Institute, La Jolla, CA, USA [2] Sanford-Burnham Center for Neuroscience, Aging, and Stem Cell Research, La Jolla, CA, USA [3] Salk Institute for Biological Studies, La Jolla, CA, USA [4] University of California, San Diego (UCSD), San Diego, CA, USA
| | - R A Lockshin
- Department of Biological Sciences, St. John's University, Queens, NY, USA
| | - C López-Otín
- Department of Biochemistry and Molecular Biology, Faculty of Medecine, Instituto Universitario de Oncología (IUOPA), University of Oviedo, Oviedo, Spain
| | - E Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - F Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - W Malorni
- 1] Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanita (ISS), Roma, Italy [2] San Raffaele Institute, Sulmona, Italy
| | - J-C Marine
- 1] Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, Leuven, Belgium [2] Laboratory for Molecular Cancer Biology, Center of Human Genetics, Leuven, Belgium
| | - S J Martin
- Department of Genetics, The Smurfit Institute, Trinity College, Dublin, Ireland
| | - J-C Martinou
- Department of Cell Biology, University of Geneva, Geneva, Switzerland
| | - J P Medema
- Laboratory for Experiments Oncology and Radiobiology (LEXOR), Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Meier
- Institute of Cancer Research, The Breakthrough Toby Robins Breast Cancer Research Centre, London, UK
| | - S Melino
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - N Mizushima
- Graduate School and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - U Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - C Muñoz-Pinedo
- Cell Death Regulation Group, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - G Nuñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - A Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - T Panaretakis
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institute, Stockholm, Sweden
| | - J M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - M E Peter
- Department of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M Piacentini
- 1] Department of Biology, University of Rome Tor Vergata; Rome, Italy [2] Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases Lazzaro Spallanzani, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Rome, Italy
| | - P Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - J H Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons, Dublin, Ireland
| | - H Puthalakath
- Department of Biochemistry, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Australia
| | - G A Rabinovich
- Laboratory of Immunopathology, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - K S Ravichandran
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - R Rizzuto
- Department Biomedical Sciences, University of Padova, Padova, Italy
| | - C M Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - D C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - T Rudel
- Department of Microbiology, University of Würzburg; Würzburg, Germany
| | - Y Shi
- Soochow Institute for Translational Medicine, Soochow University, Suzhou, China
| | - H-U Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - B R Stockwell
- 1] Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA [2] Departments of Biological Sciences and Chemistry, Columbia University, New York, NY, USA
| | - G Szabadkai
- 1] Department Biomedical Sciences, University of Padova, Padova, Italy [2] Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London (UCL), London, UK
| | - S W Tait
- 1] Cancer Research UK Beatson Institute, Glasgow, UK [2] Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - H L Tang
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - N Tavernarakis
- 1] Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece [2] Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Y Tsujimoto
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - T Vanden Berghe
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - P Vandenabeele
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium [3] Methusalem Program, Ghent University, Ghent, Belgium
| | - A Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - E F Wagner
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London (UCL), London, UK
| | - E White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - W G Wood
- 1] Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, MN, USA [2] Geriatric Research, Education and Clinical Center, VA Medical Center, Minneapolis, MN, USA
| | - J Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Z Zakeri
- 1] Department of Biology, Queens College, Queens, NY, USA [2] Graduate Center, City University of New York (CUNY), Queens, NY, USA
| | - B Zhivotovsky
- 1] Division of Toxicology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden [2] Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - G Melino
- 1] Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy [2] Medical Research Council Toxicology Unit, Leicester, UK
| | - G Kroemer
- 1] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [2] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France [3] INSERM, U1138, Gustave Roussy, Paris, France [4] Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France [5] Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
65
|
Patel S, Murphy D, Haralambieva E, Abdulla ZA, Wong KK, Chen H, Gould E, Roncador G, Hatton C, Anderson AP, Banham AH, Pulford K. Increased Expression of Phosphorylated FADD in Anaplastic Large Cell and Other T-Cell Lymphomas. Biomark Insights 2014; 9:77-84. [PMID: 25232277 PMCID: PMC4159367 DOI: 10.4137/bmi.s16553] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/03/2014] [Indexed: 01/02/2023] Open
Abstract
FAS-associated protein with death domain (FADD) is a major adaptor protein involved in extrinsic apoptosis, embryogenesis, and lymphocyte homeostasis. Although abnormalities of the FADD/death receptor apoptotic pathways have been established in tumorigenesis, fewer studies have analyzed the expression and role of phosphorylated FADD (pFADD). Our identification of FADD as a lymphoma-associated autoantigen in T-cell lymphoma patients raises the possibility that pFADD, with its correlation with cell cycle, may possess role(s) in human T-cell lymphoma development. This immunohistochemical study investigated pFADD protein expression in a range of normal tissues and lymphomas, particularly T-cell lymphomas that require improved therapies. Whereas pFADD was expressed only in scattered normal T cells, it was detected at high levels in T-cell lymphomas (eg, 84% anaplastic large cell lymphoma and 65% peripheral T cell lymphomas, not otherwise specified). The increased expression of pFADD supports further study of its clinical relevance and role in lymphomagenesis, highlighting phosphorylation of FADD as a potential therapeutic target.
Collapse
Affiliation(s)
- Suketu Patel
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Derek Murphy
- Center for Human Proteomics, Royal College of Surgeons in Ireland, Dublin, Ireland. ; School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland. ; Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Hong Chen
- Center for Human Proteomics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Edith Gould
- Center for Human Proteomics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Giovanna Roncador
- Monoclonal Antibodies Unit, Biotechnology Programme, Spanish National Cancer Research Center, Madrid, Spain
| | - Chris Hatton
- Department of Hematology, John Radcliffe Hospital, Oxford, UK
| | - Amanda P Anderson
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Alison H Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Karen Pulford
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| |
Collapse
|
66
|
Abstract
Apoptosis is a fundamental process contributing to tissue homeostasis, immune response, and development. CD95, also called Fas, is a member of the tumor necrosis factor receptor (TNF-R) superfamily. Its ligand, CD95L, was initially detected at the plasma membrane of activated T lymphocytes and natural killer (NK) cells where it contributes to the elimination of transformed and infected cells. Given its implication in immune homeostasis and immune surveillance combined with the fact that various lineages of malignant cells exhibit loss-of-function mutations, CD95 was initially classified as a tumor suppressor gene. Nonetheless, in different pathophysiological contexts, this receptor is able to transmit non-apoptotic signals and promote inflammation and carcinogenesis. Although the different non-apoptotic signaling pathways (NF-κB, MAPK, and PI3K) triggered by CD95 are known, the initial molecular events leading to these signals, the mechanisms by which the receptor switches from an apoptotic function to an inflammatory role, and, more importantly, the biological functions of these signals remain elusive.
Collapse
Affiliation(s)
- Nima Rezaei
- Children's Medical Center Hospital, Tehran University of Medical Sciences Research Center for Immunodeficiencies, Tehran, Iran
| |
Collapse
|
67
|
Häcker G. ER-stress and apoptosis: molecular mechanisms and potential relevance in infection. Microbes Infect 2014; 16:805-10. [PMID: 25172397 DOI: 10.1016/j.micinf.2014.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 02/07/2023]
Abstract
During ER-stress, one of the responses a cell can choose is apoptosis. Apoptosis generally is a cell's preferred response when other control mechanisms are overwhelmed. We now have a reasonably clear molecular picture what is happening once the apoptotic apparatus has been started. Unclear however are the majority of the upstream pathways that connect other signalling to apoptosis. During ER-stress, confirmed apoptosis-regulating targets are pro- and anti-apoptotic proteins of the Bcl-2-family, whose concerted action induces apoptosis. I will here discuss how mitochondrial apoptosis is triggered, how this is linked to the ER-stress response and in what way this may be relevant during microbial infections.
Collapse
Affiliation(s)
- Georg Häcker
- Institute for Medical Microbiology and Hygiene, University Medical Centre Freiburg, Hermann Herder-Str. 11, D-79104 Freiburg, Germany.
| |
Collapse
|
68
|
TAK1 control of cell death. Cell Death Differ 2014; 21:1667-76. [PMID: 25146924 DOI: 10.1038/cdd.2014.123] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 07/09/2014] [Accepted: 07/13/2014] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death, a physiologic process for removing cells, is critically important in normal development and for elimination of damaged cells. Conversely, unattended cell death contributes to a variety of human disease pathogenesis. Thus, precise understanding of molecular mechanisms underlying control of cell death is important and relevant to public health. Recent studies emphasize that transforming growth factor-β-activated kinase 1 (TAK1) is a central regulator of cell death and is activated through a diverse set of intra- and extracellular stimuli. The physiologic importance of TAK1 and TAK1-binding proteins in cell survival and death has been demonstrated using a number of genetically engineered mice. These studies uncover an indispensable role of TAK1 and its binding proteins for maintenance of cell viability and tissue homeostasis in a variety of organs. TAK1 is known to control cell viability and inflammation through activating downstream effectors such as NF-κB and mitogen-activated protein kinases (MAPKs). It is also emerging that TAK1 regulates cell survival not solely through NF-κB but also through NF-κB-independent pathways such as oxidative stress and receptor-interacting protein kinase 1 (RIPK1) kinase activity-dependent pathway. Moreover, recent studies have identified TAK1's seemingly paradoxical role to induce programmed necrosis, also referred to as necroptosis. This review summarizes the consequences of TAK1 deficiency in different cell and tissue types from the perspective of cell death and also focuses on the mechanism by which TAK1 complex inhibits or promotes programmed cell death. This review serves to synthesize our current understanding of TAK1 in cell survival and death to identify promising directions for future research and TAK1's potential relevance to human disease pathogenesis.
Collapse
|
69
|
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.
Collapse
Affiliation(s)
- Avi Ashkenazi
- Genentech Inc., South San Francisco, California 94080;
| | | |
Collapse
|
70
|
Fouqué A, Debure L, Legembre P. The CD95/CD95L signaling pathway: a role in carcinogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1846:130-141. [PMID: 24780723 DOI: 10.1016/j.bbcan.2014.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/17/2014] [Accepted: 04/19/2014] [Indexed: 11/18/2022]
Abstract
Apoptosis is a fundamental process that contributes to tissue homeostasis, immune responses, and development. The receptor CD95, also called Fas, is a member of the tumor necrosis factor receptor (TNF-R) superfamily. Its cognate ligand, CD95L, is implicated in immune homeostasis and immune surveillance, and various lineages of malignant cells exhibit loss-of-function mutations in this pathway; therefore, CD95 was initially classified as a tumor suppressor gene. However, more recent data indicate that in different pathophysiological contexts, this receptor can transmit non-apoptotic signals, promote inflammation, and contribute to carcinogenesis. A comparison with the initial molecular events of the TNF-R signaling pathway leading to non-apoptotic, apoptotic, and necrotic pathways reveals that CD95 is probably using different molecular mechanisms to transmit its non-apoptotic signals (NF-κB, MAPK, and PI3K). As discussed in this review, the molecular process by which the receptor switches from an apoptotic function to an inflammatory role is unknown. More importantly, the biological functions of these signals remain elusive.
Collapse
Affiliation(s)
- Amélie Fouqué
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France
| | - Laure Debure
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France
| | - Patrick Legembre
- Université Rennes-1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; INSERM U1085, IRSET, 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Equipe Labellisée Ligue Contre Le Cancer "Death Receptors and Tumor Escape", 2 Avenue du Professeur Léon Bernard, 35043 Rennes, France; Centre Eugène Marquis, rue bataille Flandres Dunkerque, Rennes, France.
| |
Collapse
|
71
|
|
72
|
Rodrigue-Gervais IG, Labbé K, Dagenais M, Dupaul-Chicoine J, Champagne C, Morizot A, Skeldon A, Brincks EL, Vidal SM, Griffith TS, Saleh M. Cellular inhibitor of apoptosis protein cIAP2 protects against pulmonary tissue necrosis during influenza virus infection to promote host survival. Cell Host Microbe 2014; 15:23-35. [PMID: 24439895 DOI: 10.1016/j.chom.2013.12.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/10/2013] [Accepted: 12/10/2013] [Indexed: 12/22/2022]
Abstract
Cellular inhibitors of apoptosis proteins (cIAPs) are essential regulators of cell death and immunity. The corresponding contributions of IAPs to infectious disease outcomes are relatively unexplored. We find that mice deficient in cIAP2 exhibit increased susceptibility and mortality to influenza A virus infection. The lethality was not due to impaired antiviral immune functions, but rather because of death-receptor-induced programmed necrosis of airway epithelial cells that led to severe bronchiole epithelial degeneration, despite control of viral replication. Pharmacological inhibition of RIPK1 or genetic deletion of Ripk3, both kinases involved in programmed necrosis, rescued cIAP2-deficient mice from influenza-induced lethality. Genetic deletion of the death receptor agonists Fas ligand or TRAIL from the hematopoietic compartment also reversed the susceptibility of cIAP2-deficient mice. Thus, cIAP2-dependent antagonism of RIPK3-mediated programmed necrosis critically protects the host from influenza infection through maintenance of pulmonary tissue homeostasis rather than through pathogen control by the immune system.
Collapse
Affiliation(s)
| | - Katherine Labbé
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 1B1, Canada
| | - Maryse Dagenais
- Department of Biochemistry, McGill University, Montréal, QC H3A 1B1, Canada
| | | | - Claudia Champagne
- Department of Medicine, McGill University, Montréal, QC H3A 1B1, Canada
| | - Alexandre Morizot
- Department of Medicine, McGill University, Montréal, QC H3A 1B1, Canada
| | - Alexander Skeldon
- Department of Biochemistry, McGill University, Montréal, QC H3A 1B1, Canada
| | - Erik L Brincks
- Department of Urology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Silvia M Vidal
- Department of Human Genetics, McGill University, Montréal, QC H3A 1B1, Canada
| | - Thomas S Griffith
- Department of Urology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Maya Saleh
- Department of Medicine, McGill University, Montréal, QC H3A 1B1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 1B1, Canada; Department of Biochemistry, McGill University, Montréal, QC H3A 1B1, Canada.
| |
Collapse
|
73
|
Morotomi-Yano K, Akiyama H, Yano KI. Different involvement of extracellular calcium in two modes of cell death induced by nanosecond pulsed electric fields. Arch Biochem Biophys 2014; 555-556:47-54. [PMID: 24893145 DOI: 10.1016/j.abb.2014.05.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/25/2014] [Accepted: 05/22/2014] [Indexed: 11/15/2022]
Abstract
Exposure of cultured cells to nanosecond pulsed electric fields (nsPEFs) induces various cellular responses, including the influx of extracellular Ca2+ and cell death. Recently, nsPEFs have been regarded as a novel means of cancer therapy, but their molecular mechanism of action remains to be fully elucidated. Here, we demonstrate the involvement of extracellular Ca2+ in nsPEF-induced cell death. Extracellular Ca2+ was essential for necrosis and consequent poly(ADP-ribose) (PAR) formation in HeLa S3 cells. Treatment with a Ca2+ ionophore enhanced necrosis as well as PAR formation in nsPEF-exposed HeLa S3 cells. In the absence of extracellular Ca2+, HeLa S3 cells were less susceptible to nsPEFs and exhibited apoptotic proteolysis of caspase 3 and PARP-1. HeLa S3 cells retained the ability to undergo apoptosis even after nsPEF exposure but instead underwent necrosis, suggesting that necrosis is the preferential mode of cell death. In K562 and HEK293 cells, exposure to nsPEFs resulted in the formation of necrosis-associated PAR, whereas Jurkat cells exclusively underwent apoptosis independently of extracellular Ca2+. These observations demonstrate that the mode of cell death induced by nsPEFs is cell-type dependent and that extracellular Ca2+ is a critical factor for nsPEF-induced necrosis.
Collapse
Affiliation(s)
- Keiko Morotomi-Yano
- Department of Bioelectrics, Institute of Pulsed Power Science, Kumamoto University, Japan
| | - Hidenori Akiyama
- Department of Pulsed Power Infrastructure, Institute of Pulsed Power Science, Kumamoto University, Japan
| | - Ken-ichi Yano
- Department of Bioelectrics, Institute of Pulsed Power Science, Kumamoto University, Japan.
| |
Collapse
|
74
|
Salvesen GS, Walsh CM. Functions of caspase 8: the identified and the mysterious. Semin Immunol 2014; 26:246-52. [PMID: 24856110 DOI: 10.1016/j.smim.2014.03.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/27/2014] [Indexed: 02/07/2023]
Abstract
Initially discovered as an initiator protease in apoptosis mediated by death receptors, caspase-8 is now known to have an apparently confounding opposing effect in securing cell survival. It is required to allow mouse embryo survival, and the survival of hematopoietic cells during their development and activation. Classic models in which caspase-8 is depleted or inhibited frequently result in inhibition of apoptosis, and conversion to death through a necrotic pathway. This bewildering switch is now known to be driven by activation of a pathway dependent on protein kinases of the RIP family, which engage a pathway known as necroptosis. If caspase-8 does not control this pathway, necrotic death results. The pro-apoptotic and pro-survival functions of caspase-8 are regulated by a specific interaction with the pseudo-caspase cFLIP, and it is thought that the heterocomplex between these two partners alters the substrate specificity of caspase-8 in favor of inactivating components of the RIP kinase pathway. The description of how caspase-8 and cFLIP coordinate the switch between apoptosis and survival is just beginning. The mechanism is not known, the differential targets are not known, and the reason of why an apoptotic initiator has been co-opted as a critical survival factor is only guessed at. Elucidating these unknowns will be important in understanding mechanisms and possible therapeutic targets in autoimmune, inflammatory, and metastatic diseases.
Collapse
Affiliation(s)
- Guy S Salvesen
- Program in Cell Death and Survival Networks, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
75
|
RIP1 suppresses innate immune necrotic as well as apoptotic cell death during mammalian parturition. Proc Natl Acad Sci U S A 2014; 111:7753-8. [PMID: 24821786 DOI: 10.1073/pnas.1401857111] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
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.
Collapse
|
76
|
Leppkes M, Roulis M, Neurath MF, Kollias G, Becker C. Pleiotropic functions of TNF-α in the regulation of the intestinal epithelial response to inflammation. Int Immunol 2014; 26:509-15. [PMID: 24821262 DOI: 10.1093/intimm/dxu051] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
An important function of intestinal epithelial cells (IECs) is to maintain the integrity of the mucosal barrier. Inflammation challenges the integrity of the mucosal barrier and the intestinal epithelium needs to adapt to a multitude of signals in order to perform the complex process of maintenance and restitution of its barrier function. Dysfunctions in epithelial barrier integrity and restoration contribute to the pathogenesis of inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis. Mucosal healing has developed to a significant treatment goal in IBD. In this review, we would like to highlight physiologic and pathologic adaptations of the intestinal epithelium to inflammation, exemplified by its responses to TNF-α. A large body of literature exists that highlights the diverse effects of this cytokine on IECs. TNF-α modulates intestinal mucus secretion and constitution. TNF-α stimulation modulates paracellular flow via tight junctional control. TNF-α induces intracellular signaling cascades that determine significant cell fate decisions such as survival, cell death or proliferation. TNF-α impacts epithelial wound healing in ErbB- and Wnt-dependent pathways while also importantly guiding immune cell attraction and function. We selected important studies from recent years with a focus on functional in vivo data providing crucial insights into the complex process of intestinal homeostasis.
Collapse
Affiliation(s)
- Moritz Leppkes
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen 91052, Germany
| | - Manolis Roulis
- Institute of Immunology, Biomedical Sciences Research Centre "Alexander Fleming," 34 Fleming Street, 16672 Vari, Greece
| | - Markus F Neurath
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen 91052, Germany
| | - George Kollias
- Institute of Immunology, Biomedical Sciences Research Centre "Alexander Fleming," 34 Fleming Street, 16672 Vari, Greece
| | - Christoph Becker
- Medical Clinic 1, University Clinic, Friedrich Alexander University, Erlangen 91052, Germany
| |
Collapse
|
77
|
RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell 2014; 157:1189-202. [PMID: 24813850 DOI: 10.1016/j.cell.2014.04.018] [Citation(s) in RCA: 543] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 04/07/2014] [Accepted: 04/14/2014] [Indexed: 11/22/2022]
Abstract
Receptor-interacting protein kinase (RIPK)-1 is involved in RIPK3-dependent and -independent signaling pathways leading to cell death and/or inflammation. Genetic ablation of ripk1 causes postnatal lethality, which was not prevented by deletion of ripk3, caspase-8, or fadd. However, animals that lack RIPK1, RIPK3, and either caspase-8 or FADD survived weaning and matured normally. RIPK1 functions in vitro to limit caspase-8-dependent, TNFR-induced apoptosis, and animals lacking RIPK1, RIPK3, and TNFR1 survive to adulthood. The role of RIPK3 in promoting lethality in ripk1(-/-) mice suggests that RIPK3 activation is inhibited by RIPK1 postbirth. Whereas TNFR-induced RIPK3-dependent necroptosis requires RIPK1, cells lacking RIPK1 were sensitized to necroptosis triggered by poly I:C or interferons. Disruption of TLR (TRIF) or type I interferon (IFNAR) signaling delayed lethality in ripk1(-/-)tnfr1(-/-) mice. These results clarify the complex roles for RIPK1 in postnatal life and provide insights into the regulation of FADD-caspase-8 and RIPK3-MLKL signaling by RIPK1.
Collapse
|
78
|
Caspase-8 mediates caspase-1 processing and innate immune defense in response to bacterial blockade of NF-κB and MAPK signaling. Proc Natl Acad Sci U S A 2014; 111:7385-90. [PMID: 24799700 DOI: 10.1073/pnas.1403252111] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Toll-like receptor signaling and subsequent activation of NF-κB- and MAPK-dependent genes during infection play an important role in antimicrobial host defense. The YopJ protein of pathogenic Yersinia species inhibits NF-κB and MAPK signaling, resulting in blockade of NF-κB-dependent cytokine production and target cell death. Nevertheless, Yersinia infection induces inflammatory responses in vivo. Moreover, increasing the extent of YopJ-dependent cytotoxicity induced by Yersinia pestis and Yersinia pseudotuberculosis paradoxically leads to decreased virulence in vivo, suggesting that cell death promotes anti-Yersinia host defense. However, the specific pathways responsible for YopJ-induced cell death and how this cell death mediates immune defense against Yersinia remain poorly defined. YopJ activity induces processing of multiple caspases, including caspase-1, independently of inflammasome components or the adaptor protein ASC. Unexpectedly, caspase-1 activation in response to the activity of YopJ required caspase-8, receptor-interacting serine/threonine kinase 1 (RIPK1), and Fas-associated death domain (FADD), but not RIPK3. Furthermore, whereas RIPK3 deficiency did not affect YopJ-induced cell death or caspase-1 activation, deficiency of both RIPK3 and caspase-8 or FADD completely abrogated Yersinia-induced cell death and caspase-1 activation. Mice lacking RIPK3 and caspase-8 in their hematopoietic compartment showed extreme susceptibility to Yersinia and were deficient in monocyte and neutrophil-derived production of proinflammatory cytokines. Our data demonstrate for the first time to our knowledge that RIPK1, FADD, and caspase-8 are required for YopJ-induced cell death and caspase-1 activation and suggest that caspase-8-mediated cell death overrides blockade of immune signaling by YopJ to promote anti-Yersinia immune defense.
Collapse
|
79
|
Mocarski ES, Kaiser WJ, Livingston-Rosanoff D, Upton JW, Daley-Bauer LP. True grit: programmed necrosis in antiviral host defense, inflammation, and immunogenicity. THE JOURNAL OF IMMUNOLOGY 2014; 192:2019-26. [PMID: 24563506 DOI: 10.4049/jimmunol.1302426] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Programmed necrosis mediated by receptor interacting protein kinase (RIP)3 (also called RIPK3) has emerged as an alternate death pathway triggered by TNF family death receptors, pathogen sensors, IFNRs, Ag-specific TCR activation, and genotoxic stress. Necrosis leads to cell leakage and acts as a "trap door," eliminating cells that cannot die by apoptosis because of the elaboration of pathogen-encoded caspase inhibitors. Necrotic signaling requires RIP3 binding to one of three partners-RIP1, DAI, or TRIF-via a common RIP homotypic interaction motif. Once activated, RIP3 kinase targets the pseudokinase mixed lineage kinase domain-like to drive cell lysis. Although necrotic and apoptotic death can enhance T cell cross-priming during infection, mice that lack these extrinsic programmed cell death pathways are able to produce Ag-specific T cells and control viral infection. The entwined relationship of apoptosis and necrosis evolved in response to pathogen-encoded suppressors to support host defense and contribute to inflammation.
Collapse
Affiliation(s)
- Edward S Mocarski
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | | | | |
Collapse
|
80
|
Morioka S, Broglie P, Omori E, Ikeda Y, Takaesu G, Matsumoto K, Ninomiya-Tsuji J. TAK1 kinase switches cell fate from apoptosis to necrosis following TNF stimulation. ACTA ACUST UNITED AC 2014; 204:607-23. [PMID: 24535827 PMCID: PMC3926964 DOI: 10.1083/jcb.201305070] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activation of the TAK1 kinase drives RIPK3-dependent necrosis and inhibits apoptosis downstream of TNF-α stimulation. TNF activates three distinct intracellular signaling cascades leading to cell survival, caspase-8–mediated apoptosis, or receptor interacting protein kinase 3 (RIPK3)–dependent necrosis, also called necroptosis. Depending on the cellular context, one of these pathways is activated upon TNF challenge. When caspase-8 is activated, it drives the apoptosis cascade and blocks RIPK3-dependent necrosis. Here we report the biological event switching to activate necrosis over apoptosis. TAK1 kinase is normally transiently activated upon TNF stimulation. We found that prolonged and hyperactivation of TAK1 induced phosphorylation and activation of RIPK3, leading to necrosis without caspase activation. In addition, we also demonstrated that activation of RIPK1 and RIPK3 promoted TAK1 activation, suggesting a positive feedforward loop of RIPK1, RIPK3, and TAK1. Conversely, ablation of TAK1 caused caspase-dependent apoptosis, in which Ripk3 deletion did not block cell death either in vivo or in vitro. Our results reveal that TAK1 activation drives RIPK3-dependent necrosis and inhibits apoptosis. TAK1 acts as a switch between apoptosis and necrosis.
Collapse
Affiliation(s)
- Sho Morioka
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695
| | | | | | | | | | | | | |
Collapse
|
81
|
Wallach D, Kang TB, Yang SH, Kovalenko A. The in vivo significance of necroptosis: Lessons from exploration of caspase-8 function. Cytokine Growth Factor Rev 2014; 25:157-65. [DOI: 10.1016/j.cytogfr.2013.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/15/2013] [Indexed: 01/28/2023]
|
82
|
Bao C, Shao Y, Li X. Hepatocyte Necroptosis Induced by Ischemic Acute Kidney Injury in Rats. Ultrastruct Pathol 2014; 38:217-23. [DOI: 10.3109/01913123.2014.895788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
83
|
Brentnall M, Weir DB, Rongvaux A, Marcus AI, Boise LH. Procaspase-3 regulates fibronectin secretion and influences adhesion, migration and survival independently of catalytic function. J Cell Sci 2014; 127:2217-26. [PMID: 24610949 DOI: 10.1242/jcs.135137] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Caspase-3 is an effector caspase that is activated downstream of mitochondrial outer-membrane permeabilization (MOMP) during apoptosis. However, previous work has demonstrated that caspase-3-deficient mouse embryonic fibroblasts (MEFs) are resistant to mitochondrially mediated cell death and display a delay in the mitochondrial events of apoptosis, including Bax activation, MOMP and release of cytochrome c. Here, we show that caspase-3 regulates fibronectin secretion and impacts on cell morphology, adhesion and migration. Surprisingly, the catalytic activity of caspase-3 is not required for these non-apoptotic functions. Moreover, we found that caspase-3-deficient MEFs are not resistant to death by anoikis and that exogenous fibronectin protects wild-type MEFs from cell death induced by serum withdrawal. Taken together, our data indicate that procaspase-3 has a non-apoptotic function; it regulates the secretion of fibronectin and influences morphology, adhesion and migration. Furthermore, this novel procaspase-3 function might alter the apoptotic threshold of the cell.
Collapse
Affiliation(s)
- Matthew Brentnall
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David B Weir
- Cancer Biology Graduate Program, Emory University, Atlanta, GA 30322, USA
| | - Anthony Rongvaux
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Adam I Marcus
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Lawrence H Boise
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
84
|
IL-15 maintains T-cell survival via S-nitrosylation-mediated inhibition of caspase-3. Cell Death Differ 2014; 21:904-14. [PMID: 24510126 DOI: 10.1038/cdd.2014.10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/27/2013] [Accepted: 01/08/2014] [Indexed: 12/18/2022] Open
Abstract
Caspase activity is critical for both T-cell survival and death. However, little is known regarding what determines caspase activity in cycling T cells. Interleukin (IL)-2 and IL-15 confer very different susceptibilities to T-cell death. We therefore considered that IL-2 and IL-15 differentially regulate caspase activity to influence T-cell survival. We observed that IL-2-cultured primary murine effector T cells manifested elevated levels of caspase-3 activity compared with IL-15-cultured T cells. T cell receptor (TCR) restimulation further increased caspase activity and induced considerable cell death in IL-2-cultured T cells, but provoked only a minimal increase of caspase activity and cell death in IL-15-cultured T cells. IL-2 sensitization to cell death was caspase-3 mediated. Interestingly, increased active caspase-3 levels with IL-2 were independent of active initiator caspase-8 and caspase-9 that were similar with IL-2 and IL-15. Rather, caspase-3 activity was inhibited by posttranslational S-nitrosylation in IL-15-cultured T cells, but not in the presence of IL-2. This paralleled increased reactive nitrogen and oxygen species with IL-15 and reduced glycolysis. Taken together, these data suggest that the metabolic state conferred by IL-15 inhibits T-cell apoptosis in part by maintaining low levels of active caspase-3 via S-nitrosylation.
Collapse
|
85
|
Graf RP, Keller N, Barbero S, Stupack D. Caspase-8 as a regulator of tumor cell motility. Curr Mol Med 2014; 14:246-54. [PMID: 24467204 PMCID: PMC4106798 DOI: 10.2174/1566524014666140128111951] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/05/2013] [Accepted: 12/02/2013] [Indexed: 01/31/2023]
Abstract
The caspases are a family of ubiquitously expressed cysteine proteases best known for their roles in programmed cell death. However, caspases play a number of other roles in vertebrates. In the case of caspase-8, loss of expression is an embryonic lethal phenotype, and caspase-8 plays roles in suppressing cellular necrosis, promoting differentiation and immune signaling, regulating autophagy, and promoting cellular migration. Apoptosis and migration require localization of caspase-8 in the periphery of the cells, where caspase-8 acts as part of distinct biosensory complexes that either promote migration in appropriate cellular microenvironments, or cell death in inappropriate settings. In the cellular periphery, caspase-8 interacts with components of the focal adhesion complex in a tyrosine-kinase dependent manner, promoting both cell migration in vitro and metastasis in vivo. Mechanistically, caspase-8 interacts with components of both focal adhesions and early endosomes, enhancing focal adhesion turnover and promoting rapid integrin recycling to the cell surface. Clinically, this suggests that the expression of caspase-8 may not always be a positive prognostic sign, and that the role of caspase-8 in cancer progression is likely context-dependent.
Collapse
Affiliation(s)
| | | | | | - D Stupack
- University of California San Diego, Moores Cancer Center, Department of Reproductive Medicine, 0803, 3855 Health Sciences Dr., La Jolla, CA 92093, USA.
| |
Collapse
|
86
|
Gurung P, Anand PK, Malireddi RKS, Vande Walle L, Van Opdenbosch N, Dillon CP, Weinlich R, Green DR, Lamkanfi M, Kanneganti TD. FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. THE JOURNAL OF IMMUNOLOGY 2014; 192:1835-46. [PMID: 24453255 DOI: 10.4049/jimmunol.1302839] [Citation(s) in RCA: 540] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Nlrp3 inflammasome is critical for host immunity, but the mechanisms controlling its activation are enigmatic. In this study, we show that loss of FADD or caspase-8 in a RIP3-deficient background, but not RIP3 deficiency alone, hampered transcriptional priming and posttranslational activation of the canonical and noncanonical Nlrp3 inflammasome. Deletion of caspase-8 in the presence or absence of RIP3 inhibited caspase-1 and caspase-11 activation by Nlrp3 stimuli but not the Nlrc4 inflammasome. In addition, FADD deletion prevented caspase-8 maturation, positioning FADD upstream of caspase-8. Consequently, FADD- and caspase-8-deficient mice had impaired IL-1β production when challenged with LPS or infected with the enteropathogen Citrobacter rodentium. Thus, our results reveal FADD and caspase-8 as apical mediators of canonical and noncanonical Nlrp3 inflammasome priming and activation.
Collapse
Affiliation(s)
- Prajwal Gurung
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Matsuda I, Matsuo K, Matsushita Y, Haruna Y, Niwa M, Kataoka T. The C-terminal domain of the long form of cellular FLICE-inhibitory protein (c-FLIPL) inhibits the interaction of the caspase 8 prodomain with the receptor-interacting protein 1 (RIP1) death domain and regulates caspase 8-dependent nuclear factor κB (NF-κB) activation. J Biol Chem 2014; 289:3876-87. [PMID: 24398693 DOI: 10.1074/jbc.m113.506485] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caspase 8 plays an essential role in the regulation of apoptotic and non-apoptotic signaling pathways. The long form of cellular FLICE-inhibitory protein (c-FLIPL) has been shown previously to regulate caspase 8-dependent nuclear factor κB (NF-κB) activation by receptor-interacting protein 1 (RIP1) and TNF receptor-associated factor 2 (TRAF2). In this study, the molecular mechanism by which c-FLIPL regulates caspase 8-dependent NF-κB activation was further explored in the human embryonic kidney cell line HEK 293 and variant cells barely expressing caspase 8. The caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone greatly diminished caspase 8-dependent NF-κB activation induced by Fas ligand (FasL) when c-FLIPL, but not its N-terminal fragment c-FLIP(p43), was expressed. The prodomain of caspase 8 was found to interact with the RIP1 death domain and to be sufficient to mediate NF-κB activation induced by FasL or c-FLIP(p43). The interaction of the RIP1 death domain with caspase 8 was inhibited by c-FLIPL but not c-FLIP(p43). Thus, these results reveal that the C-terminal domain of c-FLIPL specifically inhibits the interaction of the caspase 8 prodomain with the RIP1 death domain and, thereby, regulates caspase 8-dependent NF-κB activation.
Collapse
Affiliation(s)
- Iyo Matsuda
- From the Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan and
| | | | | | | | | | | |
Collapse
|
88
|
Nomura M, Ueno A, Saga K, Fukuzawa M, Kaneda Y. Accumulation of cytosolic calcium induces necroptotic cell death in human neuroblastoma. Cancer Res 2013; 74:1056-66. [PMID: 24371227 DOI: 10.1158/0008-5472.can-13-1283] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Necrosis has been studied extensively since the early days of medicine, with some patterns of necrosis found to be programmed like apoptotic cell death. However, mechanisms of programmed necrosis (necroptosis) are yet to be fully elucidated. In this study, we investigated how the hemagglutinating virus of Japan-envelope (HVJ-E) induces necrosis in mouse xenografts of human neuroblastoma cells. HVJ-E-induced necrosis in this system was found to depend on phosphorylation of the death receptor kinase receptor interacting protein kinase 1 (RIP1) and on the production of reactive oxygen species. This process was interpreted as necroptosis, based on its suppression by the small molecule necrostatin-1, and it did not involve the TNF-α receptor pathway. We also demonstrated that increased concentrations of cytoplasmic calcium triggered necroptosis by activating calcium-calmodulin kinase (CaMK) II. Finally, we determined that RIP1 phosphorylation was mediated by CaMK II activation. Together, our results define an upstream pathway for the activation of necroptosis in neuroblastoma cells, with potential therapeutic implications.
Collapse
Affiliation(s)
- Motonari Nomura
- Authors' Affiliations: Division of Gene Therapy Science and Department of Pediatric Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | |
Collapse
|
89
|
Cabon L, Martinez-Torres AC, Susin SA. [Programmed cell death comes in many flavors]. Med Sci (Paris) 2013; 29:1117-24. [PMID: 24356142 DOI: 10.1051/medsci/20132912015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Apoptosis is nowadays what comes first to your scientist mind when someone mentions cellular suicide. However this is not the sole form of programmed cell death and many other alternative or atypical pathways have now been described. These pathways are indeed rather preferred to apoptosis in some instances based on tissue origin, cell type or development stage of the target cell. In this review, we describe many different programmed cell death subtypes according to their characteristics. Discrete, brutal, final or singular cell death pathways all participate in the elimination of unwanted, damaged or dangerous cells in organisms hence contributing to our knowledge of this particular feature of living beings: dying! Through description of anoikis, necroptosis, entosis, netosis, pyroptosis or ferroptosis, we have no choice but to realize that programmed cell death comes in many flavors.
Collapse
Affiliation(s)
- Lauriane Cabon
- Inserm U872, équipe 19 « mort cellulaire programmée et physiopathologie des cellules tumorales », Centre de recherche des Cordeliers, Université Pierre et Marie Curie, Université Paris-Descartes, Sorbonne-Universités, UMRS 872, 15, rue de l'École de Médecine, 75006 Paris, France
| | - Ana-Carolina Martinez-Torres
- Inserm U872, équipe 19 « mort cellulaire programmée et physiopathologie des cellules tumorales », Centre de recherche des Cordeliers, Université Pierre et Marie Curie, Université Paris-Descartes, Sorbonne-Universités, UMRS 872, 15, rue de l'École de Médecine, 75006 Paris, France
| | - Santos A Susin
- Inserm U872, équipe 19 « mort cellulaire programmée et physiopathologie des cellules tumorales », Centre de recherche des Cordeliers, Université Pierre et Marie Curie, Université Paris-Descartes, Sorbonne-Universités, UMRS 872, 15, rue de l'École de Médecine, 75006 Paris, France
| |
Collapse
|
90
|
Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol 2013; 16:55-65. [PMID: 24316671 DOI: 10.1038/ncb2883] [Citation(s) in RCA: 1130] [Impact Index Per Article: 94.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 10/28/2013] [Indexed: 11/08/2022]
Abstract
The mixed lineage kinase domain-like protein (MLKL) has recently been identified as a key RIP3 (receptor interacting protein 3) downstream component of tumour necrosis factor (TNF)-induced necroptosis. MLKL is phosphorylated by RIP3 and is recruited to the necrosome through its interaction with RIP3. However, it is still unknown how MLKL mediates TNF-induced necroptosis. Here, we report that MLKL forms a homotrimer through its amino-terminal coiled-coil domain and locates to the cell plasma membrane during TNF-induced necroptosis. By generating different MLKL mutants, we demonstrated that the plasma membrane localization of trimerized MLKL is critical for mediating necroptosis. Importantly, we found that the membrane localization of MLKL is essential for Ca(2+) influx, which is an early event of TNF-induced necroptosis. Furthermore, we identified that TRPM7 (transient receptor potential melastatin related 7) is a MLKL downstream target for the mediation of Ca(2+) influx and TNF-induced necroptosis. Hence, our study reveals a crucial mechanism of MLKL-mediated TNF-induced necroptosis.
Collapse
|
91
|
Choi EJ, Kim SM, Shin JH, Kim S, Song KJ, Kee SH. Involvement of caspase-2 activation in aurora kinase inhibitor-induced cell death in axin-expressing L929 cells. Apoptosis 2013; 19:657-67. [DOI: 10.1007/s10495-013-0951-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
92
|
Tait SWG, Oberst A, Quarato G, Milasta S, Haller M, Wang R, Karvela M, Ichim G, Yatim N, Albert ML, Kidd G, Wakefield R, Frase S, Krautwald S, Linkermann A, Green DR. Widespread mitochondrial depletion via mitophagy does not compromise necroptosis. Cell Rep 2013; 5:878-85. [PMID: 24268776 PMCID: PMC4005921 DOI: 10.1016/j.celrep.2013.10.034] [Citation(s) in RCA: 340] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/02/2013] [Accepted: 10/21/2013] [Indexed: 10/26/2022] Open
Abstract
Programmed necrosis (or necroptosis) is a form of cell death triggered by the activation of receptor interacting protein kinase-3 (RIPK3). Several reports have implicated mitochondria and mitochondrial reactive oxygen species (ROS) generation as effectors of RIPK3-dependent cell death. Here, we directly test this idea by employing a method for the specific removal of mitochondria via mitophagy. Mitochondria-deficient cells were resistant to the mitochondrial pathway of apoptosis, but efficiently died via tumor necrosis factor (TNF)-induced, RIPK3-dependent programmed necrosis or as a result of direct oligomerization of RIPK3. Although the ROS scavenger butylated hydroxyanisole (BHA) delayed TNF-induced necroptosis, it had no effect on necroptosis induced by RIPK3 oligomerization. Furthermore, although TNF-induced ROS production was dependent on mitochondria, the inhibition of TNF-induced necroptosis by BHA was observed in mitochondria-depleted cells. Our data indicate that mitochondrial ROS production accompanies, but does not cause, RIPK3-dependent necroptotic cell death.
Collapse
Affiliation(s)
- Stephen W G Tait
- CR-UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK.
| | - Andrew Oberst
- Department of Immunology, University of Washington, 750 Republican Street, Seattle, WA 98109, USA
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Sandra Milasta
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Martina Haller
- CR-UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University School of Medicine, 700 Children's Drive, Columbus, OH 43205, USA
| | - Maria Karvela
- CR-UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Gabriel Ichim
- CR-UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Nader Yatim
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; Unité d'Immunobiologie des Cellules Dendritiques, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France; INSERM U818, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| | - Matthew L Albert
- Unité d'Immunobiologie des Cellules Dendritiques, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France; INSERM U818, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| | - Grahame Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Randall Wakefield
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Sharon Frase
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stefan Krautwald
- Division of Nephrology and Hypertension, Christian Albrechts University, Kiel 24105, Germany
| | - Andreas Linkermann
- Division of Nephrology and Hypertension, Christian Albrechts University, Kiel 24105, Germany
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
93
|
Abstract
Necroptosis is mediated by engagement of RIP kinases and a downstream pseudokinase, MLKL. In this issue of Immunity, Murphy et al. (2013) show that it operates at or close to the final execution mechanism of the death process.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38139, USA.
| |
Collapse
|
94
|
Lau A, Wang S, Jiang J, Haig A, Pavlosky A, Linkermann A, Zhang ZX, Jevnikar AM. RIPK3-mediated necroptosis promotes donor kidney inflammatory injury and reduces allograft survival. Am J Transplant 2013; 13:2805-2818. [PMID: 24103001 DOI: 10.1111/ajt.12447] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/12/2013] [Accepted: 07/02/2013] [Indexed: 01/25/2023]
Abstract
Kidney transplant injury occurs with ischemia and alloimmunity. Members of the receptor interacting protein kinase family (RIPK1,3) are key regulators of "necroptosis," a newly recognized, regulated form of necrosis. Necroptosis and apoptosis death appear to be counterbalanced as caspase-8 inhibition can divert death from apoptosis to necrosis. Inhibition of necroptosis in donor organs to limit injury has not been studied in transplant models. In this study, necroptosis was triggered in caspase inhibited tubular epithelial cells (TEC) exposed to tumor necrosis factor alpha in vitro, while RIPK1 inhibition with necrostatin-1 or use of RIPK3(-/-) TEC, prevented necroptosis. In vivo, short hairpin RNA silencing of caspase-8 in donor B6 mouse kidneys increased necroptosis, enhanced high-mobility group box 1 release, reduced renal function and accelerated rejection when transplanted into BALB/c recipients. Using ethidium homodimer perfusion to assess necrosis in vivo, necrosis was abrogated in RIPK3(-/-) kidneys postischemia. Following transplantation, recipients receiving RIPK3(-/-) kidneys had longer survival (p = 0.002) and improved renal function (p = 0.03) when compared to controls. In summary, we show for the first time that RIPK3-mediated necroptosis in donor kidneys can promote inflammatory injury, and has a major impact on renal ischemia-reperfusion injury and transplant survival. We suggest inhibition of necroptosis in donor organs may similarly provide a major clinical benefit.
Collapse
Affiliation(s)
- A Lau
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada; Department of Pathology, Western University, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Moquin DM, McQuade T, Chan FKM. CYLD deubiquitinates RIP1 in the TNFα-induced necrosome to facilitate kinase activation and programmed necrosis. PLoS One 2013; 8:e76841. [PMID: 24098568 PMCID: PMC3788787 DOI: 10.1371/journal.pone.0076841] [Citation(s) in RCA: 253] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 09/04/2013] [Indexed: 12/21/2022] Open
Abstract
Background Necroptosis/programmed necrosis is initiated by a macro-molecular protein complex termed the necrosome. Receptor interacting protein kinase 1 (RIPK1/RIP1) and RIP3 are key components of the necrosome. TNFα is a prototypic inducer of necrosome activation, and it is widely believed that deubiquitination of RIP1 at the TNFR-1 signaling complex precedes transition of RIP1 into the cytosol where it forms the RIP1-RIP3 necrosome. Cylindromatosis (CYLD) is believed to promote programmed necrosis by facilitating RIP1 deubiquitination at this membrane receptor complex. Methodology/Principal Findings We demonstrate that RIP1 is indeed the primary target of CYLD in TNFα-induced programmed necrosis. We observed that CYLD does not regulate RIP1 ubiquitination at the TNF receptor. TNF and zVAD-induced programmed necrosis was highly attenuated in CYLD-/- cells. However, in the presence of cycloheximide or SMAC mimetics, programmed necrosis was only moderately reduced in CYLD-/- cells. Under the latter conditions, RIP1-RIP3 necrosome formation is only delayed, but not abolished in CYLD-/- cells. We further demonstrate that RIP1 within the NP-40 insoluble necrosome is ubiquitinated and that CYLD regulates RIP1 ubiquitination in this compartment. Hence, RIP1 ubiquitination in this late-forming complex is greatly increased in CYLD-/- cells. Increased RIP1 ubiquitination impairs RIP1 and RIP3 phosphorylation, a signature of kinase activation. Conclusions/Significance Our results show that CYLD regulates RIP1 ubiquitination in the TNFα-induced necrosome, but not in the TNFR-1 signaling complex. In cells sensitized to programmed necrosis with SMAC mimetics, CYLD is not essential for necrosome assembly. Since SMAC mimetics induces the loss of the E3 ligases cIAP1 and cIAP2, reduced RIP1 ubiquitination could lead to reduced requirement for CYLD to remove ubiquitin chains from RIP1 in the TNFR-1 complex. As increased RIP1 ubiquitination in the necrosome correlates with impaired RIP1 and RIP3 phosphorylation and function, these results suggest that CYLD controls RIP1 kinase activity during necrosome assembly.
Collapse
Affiliation(s)
- David M. Moquin
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Thomas McQuade
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Francis Ka-Ming Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
96
|
Fan Q, Huang ZM, Boucher M, Shang X, Zuo L, Brinks H, Lau WB, Zhang J, Chuprun JK, Gao E. Inhibition of Fas-associated death domain-containing protein (FADD) protects against myocardial ischemia/reperfusion injury in a heart failure mouse model. PLoS One 2013; 8:e73537. [PMID: 24058479 PMCID: PMC3772851 DOI: 10.1371/journal.pone.0073537] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 07/30/2013] [Indexed: 11/29/2022] Open
Abstract
Aim As technological interventions treating acute myocardial infarction (MI) improve, post-ischemic heart failure increasingly threatens patient health. The aim of the current study was to test whether FADD could be a potential target of gene therapy in the treatment of heart failure. Methods Cardiomyocyte-specific FADD knockout mice along with non-transgenic littermates (NLC) were subjected to 30 minutes myocardial ischemia followed by 7 days of reperfusion or 6 weeks of permanent myocardial ischemia via the ligation of left main descending coronary artery. Cardiac function were evaluated by echocardiography and left ventricular (LV) catheterization and cardiomyocyte death was measured by Evans blue-TTC staining, TUNEL staining, and caspase-3, -8, and -9 activities. In vitro, H9C2 cells transfected with ether scramble siRNA or FADD siRNA were stressed with chelerythrin for 30 min and cleaved caspase-3 was assessed. Results FADD expression was significantly decreased in FADD knockout mice compared to NLC. Ischemia/reperfusion (I/R) upregulated FADD expression in NLC mice, but not in FADD knockout mice at the early time. FADD deletion significantly attenuated I/R-induced cardiac dysfunction, decreased myocardial necrosis, and inhibited cardiomyocyte apoptosis. Furthermore, in 6 weeks long term permanent ischemia model, FADD deletion significantly reduced the infarct size (from 41.20±3.90% in NLC to 26.83±4.17% in FADD deletion), attenuated myocardial remodeling, improved cardiac function and improved survival. In vitro, FADD knockdown significantly reduced chelerythrin-induced the level of cleaved caspase-3. Conclusion Taken together, our results suggest FADD plays a critical role in post-ischemic heart failure. Inhibition of FADD retards heart failure progression. Our data supports the further investigation of FADD as a potential target for genetic manipulation in the treatment of heart failure.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Benzophenanthridines/pharmacology
- Caspase 3/genetics
- Caspase 3/metabolism
- Caspase 8/genetics
- Caspase 8/metabolism
- Caspase 9/genetics
- Caspase 9/metabolism
- Cells, Cultured
- Coronary Vessels/surgery
- Disease Models, Animal
- Fas-Associated Death Domain Protein/antagonists & inhibitors
- Fas-Associated Death Domain Protein/deficiency
- Fas-Associated Death Domain Protein/genetics
- Gene Expression Regulation
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Male
- Mice
- Mice, Knockout
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proteolysis
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Severity of Illness Index
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
Collapse
Affiliation(s)
- Qian Fan
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Gerontology, Beijing Chaoyang Hospital-Affiliate of Beijing Capital Medical University, Beijing, China
| | - Zheng M. Huang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Matthieu Boucher
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xiying Shang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Lin Zuo
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Henriette Brinks
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, United States of America
| | - Jianke Zhang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - J. Kurt Chuprun
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
97
|
Murakami Y, Notomi S, Hisatomi T, Nakazawa T, Ishibashi T, Miller JW, Vavvas DG. Photoreceptor cell death and rescue in retinal detachment and degenerations. Prog Retin Eye Res 2013; 37:114-40. [PMID: 23994436 DOI: 10.1016/j.preteyeres.2013.08.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/08/2013] [Accepted: 08/10/2013] [Indexed: 02/08/2023]
Abstract
Photoreceptor cell death is the ultimate cause of vision loss in various retinal disorders, including retinal detachment (RD). Photoreceptor cell death has been thought to occur mainly through apoptosis, which is the most characterized form of programmed cell death. The caspase family of cysteine proteases plays a central role for inducing apoptosis, and in experimental models of RD, dying photoreceptor cells exhibit caspase activation; however, there is a paradox that caspase inhibition alone does not provide a sufficient protection against photoreceptor cell loss, suggesting that other mechanisms of cell death are involved. Recent accumulating evidence demonstrates that non-apoptotic forms of cell death, such as autophagy and necrosis, are also regulated by specific molecular machinery, such as those mediated by autophagy-related proteins and receptor-interacting protein kinases, respectively. Here we summarize the current knowledge of cell death signaling and its roles in photoreceptor cell death after RD and other retinal degenerative diseases. A body of studies indicate that not only apoptotic but also autophagic and necrotic signaling are involved in photoreceptor cell death, and that combined targeting of these pathways may be an effective neuroprotective strategy for retinal diseases associated with photoreceptor cell loss.
Collapse
Affiliation(s)
- Yusuke Murakami
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
98
|
Coates CJ, Whalley T, Wyman M, Nairn J. A putative link between phagocytosis-induced apoptosis and hemocyanin-derived phenoloxidase activation. Apoptosis 2013; 18:1319-1331. [DOI: 10.1007/s10495-013-0891-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
99
|
The transcription factor NFAT1 induces apoptosis through cooperation with Ras/Raf/MEK/ERK pathway and upregulation of TNF-α expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2016-28. [DOI: 10.1016/j.bbamcr.2013.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/20/2013] [Accepted: 04/02/2013] [Indexed: 12/26/2022]
|
100
|
Interferon-induced RIP1/RIP3-mediated necrosis requires PKR and is licensed by FADD and caspases. Proc Natl Acad Sci U S A 2013; 110:E3109-18. [PMID: 23898178 DOI: 10.1073/pnas.1301218110] [Citation(s) in RCA: 378] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interferons (IFNs) are cytokines with powerful immunomodulatory and antiviral properties, but less is known about how they induce cell death. Here, we show that both type I (α/β) and type II (γ) IFNs induce precipitous receptor-interacting protein (RIP)1/RIP3 kinase-mediated necrosis when the adaptor protein Fas-associated death domain (FADD) is lost or disabled by phosphorylation, or when caspases (e.g., caspase 8) are inactivated. IFN-induced necrosis proceeds via progressive assembly of a RIP1-RIP3 "necrosome" complex that requires Jak1/STAT1-dependent transcription, but does not need the kinase activity of RIP1. Instead, IFNs transcriptionally activate the RNA-responsive protein kinase PKR, which then interacts with RIP1 to initiate necrosome formation and trigger necrosis. Although IFNs are powerful activators of necrosis when FADD is absent, these cytokines are likely not the dominant inducers of RIP kinase-driven embryonic lethality in FADD-deficient mice. We also identify phosphorylation on serine 191 as a mechanism that disables FADD and collaborates with caspase inactivation to allow IFN-activated necrosis. Collectively, these findings outline a mechanism of IFN-induced RIP kinase-dependent necrotic cell death and identify FADD and caspases as negative regulators of this process.
Collapse
|