451
|
Stieg DC, Willis SD, Ganesan V, Ong KL, Scuorzo J, Song M, Grose J, Strich R, Cooper KF. A complex molecular switch directs stress-induced cyclin C nuclear release through SCF Grr1-mediated degradation of Med13. Mol Biol Cell 2017; 29:363-375. [PMID: 29212878 PMCID: PMC5996960 DOI: 10.1091/mbc.e17-08-0493] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/22/2017] [Accepted: 12/01/2017] [Indexed: 02/03/2023] Open
Abstract
In response to oxidative stress, cells must choose either to live or to die. Here we show that the E3 ligase SCFGrr1 mediates the destruction of Med13, which releases cyclin C into the cytoplasm and results in cell death. The Med13 SCF degron is most likely primed by the Cdk8 kinase and marked for destruction by the MAPK Slt2. In response to oxidative stress, cells decide whether to mount a survival or cell death response. The conserved cyclin C and its kinase partner Cdk8 play a key role in this decision. Both are members of the Cdk8 kinase module, which, with Med12 and Med13, associate with the core mediator complex of RNA polymerase II. In Saccharomyces cerevisiae, oxidative stress triggers Med13 destruction, which thereafter releases cyclin C into the cytoplasm. Cytoplasmic cyclin C associates with mitochondria, where it induces hyperfragmentation and regulated cell death. In this report, we show that residues 742–844 of Med13’s 600–amino acid intrinsic disordered region (IDR) both directs cyclin C-Cdk8 association and serves as the degron that mediates ubiquitin ligase SCFGrr1-dependent destruction of Med13 following oxidative stress. Here, cyclin C-Cdk8 phosphorylation of Med13 most likely primes the phosphodegron for destruction. Next, pro-oxidant stimulation of the cell wall integrity pathway MAP kinase Slt2 initially phosphorylates cyclin C to trigger its release from Med13. Thereafter, Med13 itself is modified by Slt2 to stimulate SCFGrr1-mediated destruction. Taken together, these results support a model in which this IDR of Med13 plays a key role in controlling a molecular switch that dictates cell fate following exposure to adverse environments.
Collapse
Affiliation(s)
- David C Stieg
- Department of Molecular Biology, Graduate School of Biological Sciences, Rowan University, Stratford, NJ 08084
| | - Stephen D Willis
- Department of Molecular Biology, Graduate School of Biological Sciences, Rowan University, Stratford, NJ 08084
| | - Vidyaramanan Ganesan
- Department of Molecular Biology, Graduate School of Biological Sciences, Rowan University, Stratford, NJ 08084
| | - Kai Li Ong
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602
| | - Joseph Scuorzo
- School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Mia Song
- School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Julianne Grose
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602
| | - Randy Strich
- Department of Molecular Biology, Graduate School of Biological Sciences, Rowan University, Stratford, NJ 08084
| | - Katrina F Cooper
- Department of Molecular Biology, Graduate School of Biological Sciences, Rowan University, Stratford, NJ 08084
| |
Collapse
|
452
|
Benfica PL, de Ávila RI, Rodrigues BDS, Cortez AP, Batista AC, Gaeti MPN, Lima EM, Rezende KR, Valadares MC. 4-Nerolidylcatechol: apoptosis by mitochondrial mechanisms with reduction in cyclin D1 at G0/G1 stage of the chronic myelogenous K562 cell line. PHARMACEUTICAL BIOLOGY 2017; 55:1899-1908. [PMID: 28644062 PMCID: PMC6130688 DOI: 10.1080/13880209.2017.1311351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 11/01/2016] [Accepted: 03/22/2017] [Indexed: 05/07/2023]
Abstract
CONTEXT 4-Nerolidylcatechol (4-NRC) has showed antitumor potential through apoptosis. However, its apoptotic mechanisms are still unclear, especially in leukemic cells. OBJECTIVES To evaluate the cytotoxic potential of 4-NRC and its cell death pathways in p53-null K562 leukemic cells. MATERIALS AND METHODS Cytotoxicity of 4-NRC (4.17-534.5 μM) over 24 h of exposure was evaluated by MTT assay. 4-NRC-induced apoptosis in K562 cells was investigated by phosphatidylserine (PS) externalization, cell cycle, sub-G1, mitochondrial evaluation, cytochrome c, cyclin D1 and intracellular reactive oxygen species (ROS) levels, and caspase activity analysis. RESULTS IC50 values obtained were 11.40, 27.31, 15.93 and 15.70 μM for lymphocytes, K562, HL-60 and Jurkat cells, respectively. In K562 cells, 4-NRC (27 μM) promoted apoptosis as verified by cellular morphological changes, a significant increase in PS externalization and sub-G1 cells. Moreover, it significantly arrested the cells at the G0/G1 phase due to a reduction in cyclin D1 expression. These effects of 4-NRC also significantly promoted a reduction in mitochondrial activity and membrane depolarization, accumulation of cytosolic cytochrome c and ROS overproduction. Additionally, it triggered an increase in caspases -3/7, -8 and -9 activities. When the cells were pretreated with N-acetyl-l-cysteine ROS scavenger, 4-NRC-induced apoptosis was partially blocked, which suggests that it exerts cytotoxicity though not exclusively through ROS-mediated mechanisms. DISCUSSION AND CONCLUSION 4-NRC has antileukemic properties, inducing apoptosis mediated by mitochondrial-dependent mechanisms with cyclin D1 inhibition. Given that emerging treatment concepts include novel combinations of well-known agents, 4-NRC could offer a promising alternative for chemotherapeutic combinations to maximize tumour suppression.
Collapse
Affiliation(s)
- Polyana Lopes Benfica
- Laboratório de Farmacologia e Toxicologia Celular-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Renato Ivan de Ávila
- Laboratório de Farmacologia e Toxicologia Celular-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Bruna dos Santos Rodrigues
- Laboratório de Farmacologia e Toxicologia Celular-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Alane Pereira Cortez
- Laboratório de Farmacologia e Toxicologia Celular-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Aline Carvalho Batista
- Laboratório de Patologia Bucal, Faculdade de Odontologia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Eliana Martins Lima
- Laboratório de Tecnologia Farmacêutica-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Kênnia Rocha Rezende
- Laboratório de Biofarmácia e Farmacocinética de Substâncias Bioativas, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marize Campos Valadares
- Laboratório de Farmacologia e Toxicologia Celular-FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| |
Collapse
|
453
|
Chatziandreou N, Farsakoglu Y, Palomino-Segura M, D'Antuono R, Pizzagalli DU, Sallusto F, Lukacs-Kornek V, Uguccioni M, Corti D, Turley SJ, Lanzavecchia A, Carroll MC, Gonzalez SF. Macrophage Death following Influenza Vaccination Initiates the Inflammatory Response that Promotes Dendritic Cell Function in the Draining Lymph Node. Cell Rep 2017; 18:2427-2440. [PMID: 28273457 DOI: 10.1016/j.celrep.2017.02.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/14/2016] [Accepted: 02/07/2017] [Indexed: 10/20/2022] Open
Abstract
The mechanism by which inflammation influences the adaptive response to vaccines is not fully understood. Here, we examine the role of lymph node macrophages (LNMs) in the induction of the cytokine storm triggered by inactivated influenza virus vaccine. Following vaccination, LNMs undergo inflammasome-independent necrosis-like death that is reliant on MyD88 and Toll-like receptor 7 (TLR7) expression and releases pre-stored interleukin-1α (IL-1α). Furthermore, activated medullary macrophages produce interferon-β (IFN-β) that induces the autocrine secretion of IL-1α. We also found that macrophage depletion promotes lymph node-resident dendritic cell (LNDC) relocation and affects the capacity of CD11b+ LNDCs to capture virus and express co-stimulatory molecules. Inhibition of the IL-1α-induced inflammatory cascade reduced B cell responses, while co-administration of recombinant IL-1α increased the humoral response. Stimulation of the IL-1α inflammatory pathway might therefore represent a strategy to enhance antigen presentation by LNDCs and improve the humoral response against influenza vaccines.
Collapse
Affiliation(s)
- Nikolaos Chatziandreou
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Yagmur Farsakoglu
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Rocco D'Antuono
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland; Institute of Computational Science, Università della Svizzera Italiana, via G. Buffi 13, 6900 Lugano, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland; Institute for Microbiology, ETH Zurich, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Veronika Lukacs-Kornek
- Department of Internal Medicine II, Saarland University Medical Centre, 66424 Homburg, Germany
| | - Mariagrazia Uguccioni
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland; Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, 20089 Rozzano-Milan, Italy
| | | | - Shannon J Turley
- Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland; Institute for Microbiology, ETH Zurich, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Michael C Carroll
- Department of Pediatrics, Harvard Medical School and PCMM, Boston Childrens Hospital, Boston, MA 02115, USA
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, via Vincenzo Vela 6, 6500 Bellinzona, Switzerland.
| |
Collapse
|
454
|
O'Toole JF, Schilling W, Kunze D, Madhavan SM, Konieczkowski M, Gu Y, Luo L, Wu Z, Bruggeman LA, Sedor JR. ApoL1 Overexpression Drives Variant-Independent Cytotoxicity. J Am Soc Nephrol 2017; 29:869-879. [PMID: 29180397 DOI: 10.1681/asn.2016121322] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
Coding variants in the APOL1 gene are associated with kidney diseases in African ancestral populations; yet, the underlying biologic mechanisms remain uncertain. Variant-dependent autophagic and cytotoxic cell death have been proposed as pathogenic pathways mediating kidney injury. To examine this possibility, we conditionally expressed APOL1-G0 (reference), -G1, and -G2 (variants) using a tetracycline-regulated system in HEK293 cells. Autophagy was monitored biochemically and cell death was measured using multiple assays. We measured intracellular Na+ and K+ content with atomic absorption spectroscopy and APOL1-dependent currents with whole-cell patch clamping. Neither reference nor variant APOL1s induced autophagy. At high expression levels, APOL1-G0, -G1, and -G2 inserted into the plasma membrane and formed pH-sensitive cation channels, causing collapse of cellular Na+ and K+ gradients, phosphorylation of p38 mitogen-activated protein kinase, and cell death, without variant-dependent differences. APOL1-G0 and -G2 exhibited similar channel properties in whole-cell patch clamp experiments. At low expression levels, neither reference nor variant APOL1s localized on the plasma membrane, Na+ and K+ gradients were maintained, and cells remained viable. Our results indicate that APOL1-mediated pore formation is critical for the trypanolytic activity of APOL1 and drives APOL1-mediated cytotoxicity in overexpression systems. The absence of cytotoxicity at physiologic expression levels suggests variant-dependent intracellular K+ loss and cytotoxicity does not drive kidney disease progression.
Collapse
Affiliation(s)
- John F O'Toole
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and
| | - William Schilling
- Rammelkamp Center, MetroHealth System.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | | | | | - Yaping Gu
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Liping Luo
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Zhenzhen Wu
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Leslie A Bruggeman
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and
| | - John R Sedor
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
455
|
Hatem E, El Banna N, Huang ME. Multifaceted Roles of Glutathione and Glutathione-Based Systems in Carcinogenesis and Anticancer Drug Resistance. Antioxid Redox Signal 2017; 27:1217-1234. [PMID: 28537430 DOI: 10.1089/ars.2017.7134] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE Glutathione is the most abundant antioxidant molecule in living organisms and has multiple functions. Intracellular glutathione homeostasis, through its synthesis, consumption, and degradation, is an intricately balanced process. Glutathione levels are often high in tumor cells before treatment, and there is a strong correlation between elevated levels of intracellular glutathione/sustained glutathione-mediated redox activity and resistance to pro-oxidant anticancer therapy. Recent Advances: Ample evidence demonstrates that glutathione and glutathione-based systems are particularly relevant in cancer initiation, progression, and the development of anticancer drug resistance. CRITICAL ISSUES This review highlights the multifaceted roles of glutathione and glutathione-based systems in carcinogenesis, anticancer drug resistance, and clinical applications. FUTURE DIRECTIONS The evidence summarized here underscores the important role played by glutathione and the glutathione-based systems in carcinogenesis and anticancer drug resistance. Future studies should address mechanistic questions regarding the distinct roles of glutathione in different stages of cancer development and cancer cell death. It will be important to study how metabolic alterations in cancer cells can influence glutathione homeostasis. Sensitive approaches to monitor glutathione dynamics in subcellular compartments will be an indispensible step. Therapeutic perspectives should focus on mechanism-based rational drug combinations that are directed against multiple redox targets using effective, specific, and clinically safe inhibitors. This new strategy is expected to produce a synergistic effect, prevent drug resistance, and diminish doses of single drugs. Antioxid. Redox Signal. 27, 1217-1234.
Collapse
Affiliation(s)
- Elie Hatem
- 1 CNRS UMR3348, Institut Curie, PSL Research University , Orsay, France .,2 CNRS UMR3348, Université Paris Sud, Université Paris-Saclay , Orsay, France
| | - Nadine El Banna
- 1 CNRS UMR3348, Institut Curie, PSL Research University , Orsay, France .,2 CNRS UMR3348, Université Paris Sud, Université Paris-Saclay , Orsay, France
| | - Meng-Er Huang
- 1 CNRS UMR3348, Institut Curie, PSL Research University , Orsay, France .,2 CNRS UMR3348, Université Paris Sud, Université Paris-Saclay , Orsay, France
| |
Collapse
|
456
|
Isopropyl quinoxaline-7-carboxylate 1,4-di-N-oxide derivatives induce regulated necrosis-like cell death on Leishmania (Leishmania) mexicana. Parasitol Res 2017; 117:45-58. [PMID: 29159705 DOI: 10.1007/s00436-017-5635-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/25/2017] [Indexed: 02/03/2023]
Abstract
Leishmaniasis is a neglected tropical disease caused by the parasite of the genus Leishmania. About 13 million people are infected worldwide, and it is estimated that 350 million are at risk of infection. Clinical manifestations depend on the parasite species and factors related to the host such as the immune system, nutrition, housing, and financial resources. Available treatments have severe side effects; therefore, research currently focuses on finding more active and less toxic compounds. Quinoxalines have been described as promising alternatives. In this context, 17 isopropyl quinoxaline-7-carboxylate 1,4-di-N-oxide derivatives were evaluated as potential leishmanicidal agents. Their effect on the cell metabolism of Leishmania mexicana promastigotes and their cytotoxic effects on the J774.A1 cell line and on erythrocytes were evaluated, and their selectivity index was calculated. Compounds T-069 (IC50 = 1.49 μg/mL), T-070 (IC50 = 1.71 μg/mL), T-072 (IC50 = 6.62 μg/mL), T-073 (IC50 = 1.25 μg/mL), T-085 (IC50 = 0.74 μg/mL), and T-116 (IC50 = 0.88 μg/mL) were the most active against L. mexicana promastigotes and their mechanism of action was characterized by flow cytometry and microscopy. Compound T-073, the most selective quinoxaline derivative, induced cell membrane damage, phosphatidylserine exposition, reactive oxygen species production, disruption of the mitochondrion membrane potential, and DNA fragmentation, all in a dose-dependent manner, indicating the induction of regulated necrosis. Light and transmission electron microscopy showed the drastic morphological changes induced and the mitochondrion as the most sensitive organelle in response to T-073. This study describes the mechanism by which active isopropyl quinoxaline-7-carboxylate 1,4-di-N-oxide quinoxalines affect the parasite.
Collapse
|
457
|
Masalova OV, Lesnova EI, Solyev PN, Zakirova NF, Prassolov VS, Kochetkov SN, Ivanov AV, Kushch AA. Modulation of Cell Death Pathways by Hepatitis C Virus Proteins in Huh7.5 Hepatoma Cells. Int J Mol Sci 2017; 18:2346. [PMID: 29113144 PMCID: PMC5713315 DOI: 10.3390/ijms18112346] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/27/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022] Open
Abstract
The hepatitis C virus (HCV) causes chronic liver disease leading to fibrosis, cirrhosis, and hepatocellular carcinoma. HCV infection triggers various types of cell death which contribute to hepatitis C pathogenesis. However, much is still unknown about the impact of viral proteins on them. Here we present the results of simultaneous immunocytochemical analysis of markers of apoptosis, autophagy, and necrosis in Huh7.5 cells expressing individual HCV proteins or their combinations, or harboring the virus replicon. Stable replication of the full-length HCV genome or transient expression of its core, Е1/Е2, NS3 and NS5B led to the death of 20-47% cells, 72 h posttransfection, whereas the expression of the NS4A/B, NS5A or NS3-NS5B polyprotein did not affect cell viability. HCV proteins caused different impacts on the activation of caspases-3, -8 and -9 and on DNA fragmentation. The structural core and E1/E2 proteins promoted apoptosis, whereas non-structural NS4A/B, NS5A, NS5B suppressed apoptosis by blocking various members of the caspase cascade. The majority of HCV proteins also enhanced autophagy, while NS5A also induced necrosis. As a result, the death of Huh7.5 cells expressing the HCV core was induced via apoptosis, the cells expressing NS3 and NS5B via autophagy-associated death, and the cells expressing E1/E2 glycoproteins or harboring HCV the replicon via both apoptosis and autophagy.
Collapse
Affiliation(s)
- Olga V Masalova
- Ivanovsky Institute of Virology, Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia.
| | - Ekaterina I Lesnova
- Ivanovsky Institute of Virology, Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia.
| | - Pavel N Solyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| | - Natalia F Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| | - Vladimir S Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| | - Alla A Kushch
- Ivanovsky Institute of Virology, Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow 123098, Russia.
| |
Collapse
|
458
|
Sato A. [Novel Anticancer Strategy Targeting Switch Mechanisms in Two Types of Cell Death: Necrosis and Apoptosis]. YAKUGAKU ZASSHI 2017; 137:1315-1321. [PMID: 29093367 DOI: 10.1248/yakushi.17-00131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two types of cell death, necrosis and apoptosis, are defined in terms of cell death morphological features. We have been studying the mechanisms by which cell death processes are switched during the treatment of mouse tumor FM3A with anticancer, 5-fluoro-2'-deoxyuridine (FUdR): it induces original clone F28-7 to necrosis, but its sub-clone F28-7-A to apoptosis. We identified several such switch regulators of cell death: heat shock protein 90 (HSP90), lamin-B1, cytokeratin-19, and activating transcription factor 3 (ATF3), by using transcriptomic, proteomic analyses and siRNA screening. For example, the inhibition of HSP90 by its inhibitor geldanamycin in F28-7 caused a shift from necrosis to apoptosis. We also observed that the knockdown of lamin-B1, cytokeratin-19, or ATF3 expression in F28-7 resulted in a shift from necrosis to apoptosis. Recently, we used microRNA (miRNA, miR) microarray analyses to investigate the miRNA expression profiles in these sister cells. The miR-351 and miR-743a were expressed at higher levels in F28-7-A than in F28-7. Higher expression of miR-351 or miR-743a in F28-7, induced by transfecting the miR mimics, resulted in a switch of cell death mode: necrosis to apoptosis. Furthermore, transfection of an miR-351 inhibitor into F28-7-A resulted in morphological changes, and mode of cell death from apoptosis to necrosis. These findings suggest that the identified cell death regulators may have key roles in switching cell death mode. Possible mechanisms involving cell death regulators in the switch of necrosis or apoptosis are discussed. We propose a novel anticancer strategy targeting the switch regulators of necrosis or apoptosis.
Collapse
Affiliation(s)
- Akira Sato
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
459
|
Fuchslocher Chico J, Saggau C, Adam D. Proteolytic control of regulated necrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2147-2161. [DOI: 10.1016/j.bbamcr.2017.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 12/20/2022]
|
460
|
Kaźmierczak A, Doniak M, Kunikowska A. Proteolytic activities in cortex of apical parts of Vicia faba ssp. minor seedling roots during kinetin-induced programmed cell death. PROTOPLASMA 2017; 254:2273-2285. [PMID: 28501974 DOI: 10.1007/s00709-017-1119-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/02/2017] [Indexed: 06/07/2023]
Abstract
Programmed cell death (PCD) is a crucial process in plant development. In this paper, proteolytically related aspects of kinetin-induced PCD in cortex cells of Vicia faba ssp. minor seedlings were examined using morphological, fluorometric, spectrophotometric, and fluorescence microscopic analyses. Cell viability estimation after 46 μM kinetin treatment of seedling roots showed that the number of dying cortex cells increased with treatment duration, reaching maximum after 72 h. Weight of the apical root segments increased with time and was about 2.5-fold greater after 96 h, while the protein content remained unchanged, compared to the control. The total and cysteine-dependent proteolytic activities fluctuated during 1-96-h treatment, which was not accompanied by the changes in the protein amount, indicating that the absolute protein amounts decreased during kinetin-induced PCD. N-ethylmaleimide (NEM), phenylmethylsulfonyl fluoride (PMSF), and Z-Leu-Leu-Nva-H (MG115), the respective cysteine, serine, and proteasome inhibitors, suppressed kinetin-induced PCD. PMSF significantly decreased serine-dependent proteolytic activities without changing the amount of proteins, unlike NEM and MG115. More pronounced effect of PMSF over NEM indicated that in the root apical segments, the most important proteolytic activity during kinetin-induced PCD was that of serine proteases, while that of cysteine proteases may be important for protein degradation in the last phase of the process. Both NEM and PMSF inhibited apoptotic-like structure formation during kinetin-induced PCD. The level of caspase-3-like activity of β1 proteasome subunit increased after kinetin treatment. Addition of proteasome inhibitor MG-115 reduced the number of dying cells, suggesting that proteasomes might play an important role during kinetin-induced PCD.
Collapse
Affiliation(s)
- Andrzej Kaźmierczak
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, The University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland.
| | - Magdalena Doniak
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, The University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| | - Anita Kunikowska
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, The University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| |
Collapse
|
461
|
Lou JS, Bi WC, Chan GKL, Jin Y, Wong CW, Zhou ZY, Wang HY, Yao P, Dong TTX, Tsim KWK. Ginkgetin induces autophagic cell death through p62/SQSTM1-mediated autolysosome formation and redox setting in non-small cell lung cancer. Oncotarget 2017; 8:93131-93148. [PMID: 29190983 PMCID: PMC5696249 DOI: 10.18632/oncotarget.21862] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/27/2017] [Indexed: 12/19/2022] Open
Abstract
Promoting cell death by autophagy could be a novel treatment for cancer. The major player in autophagy, p62, serves as a good therapeutic target. Ginkgetin, a biflavonoid from Ginkgo biloba leaves, exhibited promising anticancer activity in non-small cell lung cancer cell lines, with an IC50 lower than that of cisplatin. This anticancer effect of ginkgetin was illustrated in a xenograft nude mouse model. Ginkgetin induced autophagic cell death in A549 cells, and this effect was markedly reversed by chemical and genetic approaches. Ginkgetin showed potential binding affinity to p62. Upregulation of p62 through chemical and genetic means decreased cell death, lysosome acidification, and autophagosome formation, which consequently disrupted autolysosome formation. In addition, the decreased autophagy induced by p62 overexpression increased Nrf2/ARE activity and the oxygen consumption rate and decreased on formation of reactive oxygen species. These phenomena were exhibited in a reciprocal manner when p62 was knocked down. Thus, p62 may be a potential target in ginkgetin-induced autophagic cell death, and ginkgetin could be developed as a novel anticancer drug.
Collapse
Affiliation(s)
- Jian-Shu Lou
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Wen-Chuan Bi
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Gallant K L Chan
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yan Jin
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Chau-Wing Wong
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zhong-Yu Zhou
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Huai-You Wang
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ping Yao
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Tina T X Dong
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Karl W K Tsim
- Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| |
Collapse
|
462
|
How do we fit ferroptosis in the family of regulated cell death? Cell Death Differ 2017; 24:1991-1998. [PMID: 28984871 DOI: 10.1038/cdd.2017.149] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/18/2017] [Accepted: 08/06/2017] [Indexed: 02/07/2023] Open
Abstract
In the last few years many new cell death modalities have been described. To classify different types of cell death, the term 'regulated cell death' was introduced to discriminate it from 'accidental cell death'. Regulated cell death involves the activation of genetically encoded molecular machinery that couples the presence of some signal to cell death. These forms of cell death, like apoptosis, necroptosis and pyroptosis have important physiological roles in development, tissue repair, and immunity. Accidental cell death occurs in response to physical or chemical insults and occurs independently of molecular signalling pathways. Ferroptosis, an emerging and recently (re)discovered type of regulated cell death occurs through Fe(II)-dependent lipid peroxidation when the reduction capacity of a cell is insufficient. Ferroptosis is coined after the requirement for free ferrous iron. Here, we will consider the extent to which ferroptosis is similar to other regulated cell deaths and explore emerging ideas about the physiological role of ferroptosis.
Collapse
|
463
|
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, Noel K, Jiang X, Linkermann A, Murphy ME, Overholtzer M, Oyagi A, Pagnussat GC, Park J, Ran Q, Rosenfeld CS, Salnikow K, Tang D, Torti FM, Torti SV, Toyokuni S, Woerpel KA, Zhang DD. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017; 171:273-285. [PMID: 28985560 PMCID: PMC5685180 DOI: 10.1016/j.cell.2017.09.021] [Citation(s) in RCA: 4818] [Impact Index Per Article: 602.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a form of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides to lethal levels. Emerging evidence suggests that ferroptosis represents an ancient vulnerability caused by the incorporation of polyunsaturated fatty acids into cellular membranes, and cells have developed complex systems that exploit and defend against this vulnerability in different contexts. The sensitivity to ferroptosis is tightly linked to numerous biological processes, including amino acid, iron, and polyunsaturated fatty acid metabolism, and the biosynthesis of glutathione, phospholipids, NADPH, and coenzyme Q10. Ferroptosis has been implicated in the pathological cell death associated with degenerative diseases (i.e., Alzheimer's, Huntington's, and Parkinson's diseases), carcinogenesis, stroke, intracerebral hemorrhage, traumatic brain injury, ischemia-reperfusion injury, and kidney degeneration in mammals and is also implicated in heat stress in plants. Ferroptosis may also have a tumor-suppressor function that could be harnessed for cancer therapy. This Primer reviews the mechanisms underlying ferroptosis, highlights connections to other areas of biology and medicine, and recommends tools and guidelines for studying this emerging form of regulated cell death.
Collapse
Affiliation(s)
- Brent R Stockwell
- Department of Biological Sciences, Columbia University, 550 West 120(th) Street, MC 4846, New York, NY 10027, USA; Department of Chemistry, Columbia University, 550 West 120(th) Street, MC 4846, New York, NY 10027, USA.
| | - José Pedro Friedmann Angeli
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), München, Germany
| | - Hülya Bayir
- Department of Critical Care Medicine, Safar Center for Resuscitation Research and Center for Free Radical and Antioxidant Health, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), München, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, German Cancer Consortium (DKTK), partner site Frankfurt, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sergio Gascón
- Ludwig-Maximilians University of Munich, Physiological Genomics, Biomedical Center (BMC), Planegg-Martinsried, Germany; Institute for Stem Cell Research, Helmholtz Center Munich at the Biomedical Center (BMC), Grosshaderner Strasse 9, 82152 Planegg-Martinsried, Germany
| | - Stavroula K Hatzios
- Department of Molecular, Cellular and Developmental Biology and Department of Chemistry, Yale University, New Haven, CT 06511, USA; Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kay Noel
- Collaborative Medicinal Development, LLC, Sausalito, CA, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andreas Linkermann
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Gabriela C Pagnussat
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| | | | - Qitao Ran
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Konstantin Salnikow
- Division of Cancer Biology, National Cancer Institute, NIH, Rockville, MD 20850, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Protein Modification and Degradation Laboratory, Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K A Woerpel
- Department of Chemistry, New York University, New York, NY, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
464
|
Bai T, Wang S, Zhao Y, Zhu R, Wang W, Sun Y. Haloperidol, a sigma receptor 1 antagonist, promotes ferroptosis in hepatocellular carcinoma cells. Biochem Biophys Res Commun 2017; 491:919-925. [PMID: 28756230 DOI: 10.1016/j.bbrc.2017.07.136] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023]
Abstract
Ferroptosis is a novel form of cell death, which is characterized by accumulation of reactive oxygen species (ROS). Sigma 1 receptor (S1R) has been suggested to function in oxidative stress metabolism. Both erastin and sorafenib significantly induced S1R protein expression. Haloperidol strongly promoted erastin- and sorafenib-induced cell death, which was blocked by ferrostatin-1 but not ZVAD-FMK or necrosulfonamide. During ferroptosis, haloperidol substantially increased the cellular levels of Fe2+, GSH and lipid peroxidation. Furthermore, several ferroptosis-related protein targets were up-regulated in the absence of haloperidol. Thus, Our study identified an association between haloperidol and ferroptosis for the first time. Our analyses of a combination of drugs may provide a novel strategy of hepatocellular carcinoma (HCC) therapy.
Collapse
Affiliation(s)
- Tao Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China
| | - Shuai Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China
| | - Yipu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China
| | - Rongtao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China
| | - Weijie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China
| | - Yuling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, School of Medicine, Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, PR China.
| |
Collapse
|
465
|
Lee JS. Immunologic Mechanism of Ischemia Reperfusion Injury in Transplantation. KOREAN JOURNAL OF TRANSPLANTATION 2017. [DOI: 10.4285/jkstn.2017.31.3.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Jong Soo Lee
- Division of Nephrology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
- Biomedical Research Center, Ulsan, Korea
| |
Collapse
|
466
|
Saeed WK, Jun DW, Jang K, Chae YJ, Lee JS, Kang HT. Does necroptosis have a crucial role in hepatic ischemia-reperfusion injury? PLoS One 2017; 12:e0184752. [PMID: 28957350 PMCID: PMC5619711 DOI: 10.1371/journal.pone.0184752] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/30/2017] [Indexed: 02/01/2023] Open
Abstract
Background Previous studies have demonstrated protective effects of anti-receptor interacting protein kinase 1 (RIP1), a key necroptosis molecule. However, it is uncertain whether necroptosis has a crucial role in hepatic IR injury. Therefore, we evaluated the role of necroptosis in hepatic IR injury. Method The IR mice underwent 70% segmental IR injury induced by the clamping of the hepatic artery and portal vein for 1 hr followed by reperfusion for 4 hr. The key necroptosis molecules (RIP1, RIP3, and MLKL) and other key molecules of regulated necrosis (PGAM5 and caspase-1) were evaluated in the warm IR injury model. A RIP1 inhibitor (necrostain-1s) and/or an anti-mitochondrial permeability transition (MPT)-mediated necrosis mediator (cyclosporine A, CyA) were administered before clamping. Necrotic injury was quantified using Suzuki’s scoring system. qRT-PCR and western blot were performed to evaluate RIP1, RIP3, MLKL and PGAM5 expressions. Results RIP1, RIP3, MLKL and PGAM5 expression did not change in the hepatic IR injury model. Moreover, Nec1s pretreatment did not improve histology or biochemical markers. The overall Suzuki score (cytoplasmic vacuolization, sinusoidal congestion and hepatocytes necrosis) was increased in the RIP3(-/-) mice compared to the IR group (3.5 vs. 5, p = 0.026). CyA pretreatment and/or RIP3(-/-) mice decreased Bax/Bcl2 expression; however, it did lead to an overall change in the levels of AST, ALT and LDH or necrotic injury. The Bax/Bcl2 ratio and the expression of caspase-1 and caspase-3 did not increase in our hepatic IR injury model. Conclusion Key necroptosis molecules did not increase in the necrosis-dominant hepatic IR injury model. Anti-necroptosis and/or cyclosporine-A treatment did not have an overall protective effect on necrosis-dominant hepatic IR injury.
Collapse
Affiliation(s)
- Waqar K. Saeed
- Department of Internal Medicine, Hanyang University School of Medicine, Seoul, South Korea
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University School of Medicine, Seoul, South Korea
- Department of Translational Medicine, Hanyang University Graduate school of Biomedical Science and Engineering, Seoul, South Korea
- * E-mail:
| | - Kiseok Jang
- Department of Pathology, Hanyang University School of Medicine, Seoul, South Korea
| | - Yeon Ji Chae
- Department of Translational Medicine, Hanyang University Graduate school of Biomedical Science and Engineering, Seoul, South Korea
| | - Jai Sun Lee
- Department of Translational Medicine, Hanyang University Graduate school of Biomedical Science and Engineering, Seoul, South Korea
| | - Hyeon Tae Kang
- Department of Translational Medicine, Hanyang University Graduate school of Biomedical Science and Engineering, Seoul, South Korea
| |
Collapse
|
467
|
Bustos PL, Perrone AE, Milduberger NA, Bua J. Mitochondrial permeability transition in protozoan parasites: what we learned from Trypanosoma cruzi. Cell Death Dis 2017; 8:e3057. [PMID: 28933785 PMCID: PMC5636976 DOI: 10.1038/cddis.2017.431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- P L Bustos
- Instituto Nacional de Parasitología ‘‘Dr. Mario Fatala Chabén’’- A.N.L.I.S. Malbrán, 568 Paseo Colon Avenue, C1063AC S, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - A E Perrone
- Instituto Nacional de Parasitología ‘‘Dr. Mario Fatala Chabén’’- A.N.L.I.S. Malbrán, 568 Paseo Colon Avenue, C1063AC S, Buenos Aires, Argentina
| | - N A Milduberger
- Instituto Nacional de Parasitología ‘‘Dr. Mario Fatala Chabén’’- A.N.L.I.S. Malbrán, 568 Paseo Colon Avenue, C1063AC S, Buenos Aires, Argentina
- CAECIHS, Universidad Abierta Interamericana, Av. Montes de Oca 745, 2º piso, Buenos Aires C1270AAH, Argentina
| | - J Bua
- Instituto Nacional de Parasitología ‘‘Dr. Mario Fatala Chabén’’- A.N.L.I.S. Malbrán, 568 Paseo Colon Avenue, C1063AC S, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- CAECIHS, Universidad Abierta Interamericana, Av. Montes de Oca 745, 2º piso, Buenos Aires C1270AAH, Argentina
| |
Collapse
|
468
|
Gonçalves AP, Heller J, Daskalov A, Videira A, Glass NL. Regulated Forms of Cell Death in Fungi. Front Microbiol 2017; 8:1837. [PMID: 28983298 PMCID: PMC5613156 DOI: 10.3389/fmicb.2017.01837] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 09/07/2017] [Indexed: 12/15/2022] Open
Abstract
Cell death occurs in all domains of life. While some cells die in an uncontrolled way due to exposure to external cues, other cells die in a regulated manner as part of a genetically encoded developmental program. Like other eukaryotic species, fungi undergo programmed cell death (PCD) in response to various triggers. For example, exposure to external stress conditions can activate PCD pathways in fungi. Calcium redistribution between the extracellular space, the cytoplasm and intracellular storage organelles appears to be pivotal for this kind of cell death. PCD is also part of the fungal life cycle, in which it occurs during sexual and asexual reproduction, aging, and as part of development associated with infection in phytopathogenic fungi. Additionally, a fungal non-self-recognition mechanism termed heterokaryon incompatibility (HI) also involves PCD. Some of the molecular players mediating PCD during HI show remarkable similarities to major constituents involved in innate immunity in metazoans and plants. In this review we discuss recent research on fungal PCD mechanisms in comparison to more characterized mechanisms in metazoans. We highlight the role of PCD in fungi in response to exogenic compounds, fungal development and non-self-recognition processes and discuss identified intracellular signaling pathways and molecules that regulate fungal PCD.
Collapse
Affiliation(s)
- A Pedro Gonçalves
- Plant and Microbial Biology Department, University of California, BerkeleyBerkeley, CA, United States
| | - Jens Heller
- Plant and Microbial Biology Department, University of California, BerkeleyBerkeley, CA, United States
| | - Asen Daskalov
- Plant and Microbial Biology Department, University of California, BerkeleyBerkeley, CA, United States
| | - Arnaldo Videira
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do PortoPorto, Portugal.,I3S - Instituto de Investigação e Inovação em SaúdePorto, Portugal
| | - N Louise Glass
- Plant and Microbial Biology Department, University of California, BerkeleyBerkeley, CA, United States
| |
Collapse
|
469
|
Lei-Leston AC, Murphy AG, Maloy KJ. Epithelial Cell Inflammasomes in Intestinal Immunity and Inflammation. Front Immunol 2017; 8:1168. [PMID: 28979266 PMCID: PMC5611393 DOI: 10.3389/fimmu.2017.01168] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022] Open
Abstract
Pattern recognition receptors (PRR), such as NOD-like receptors (NLRs), sense conserved microbial signatures, and host danger signals leading to the coordination of appropriate immune responses. Upon activation, a subset of NLR initiate the assembly of a multimeric protein complex known as the inflammasome, which processes pro-inflammatory cytokines and mediates a specialized form of cell death known as pyroptosis. The identification of inflammasome-associated genes as inflammatory bowel disease susceptibility genes implicates a role for the inflammasome in intestinal inflammation. Despite the fact that the functional importance of inflammasomes within immune cells has been well established, the contribution of inflammasome expression in non-hematopoietic cells remains comparatively understudied. Given that intestinal epithelial cells (IEC) act as a barrier between the host and the intestinal microbiota, inflammasome expression by these cells is likely important for intestinal immune homeostasis. Accumulating evidence suggests that the inflammasome plays a key role in shaping epithelial responses at the host-lumen interface with many inflammasome components highly expressed by IEC. Recent studies have exposed functional roles of IEC inflammasomes in mucosal immune defense, inflammation, and tumorigenesis. In this review, we present the main features of the predominant inflammasomes and their effector mechanisms contributing to intestinal homeostasis and inflammation. We also discuss existing controversies in the field and open questions related to their implications in disease. A comprehensive understanding of the molecular basis of intestinal inflammasome signaling could hold therapeutic potential for clinical translation.
Collapse
Affiliation(s)
- Andrea C Lei-Leston
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alison G Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
470
|
Taxane-mediated radiosensitization derives from chromosomal missegregation on tripolar mitotic spindles orchestrated by AURKA and TPX2. Oncogene 2017; 37:52-62. [DOI: 10.1038/onc.2017.304] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 06/29/2017] [Accepted: 07/20/2017] [Indexed: 12/13/2022]
|
471
|
Probst L, Dächert J, Schenk B, Fulda S. Lipoxygenase inhibitors protect acute lymphoblastic leukemia cells from ferroptotic cell death. Biochem Pharmacol 2017; 140:41-52. [DOI: 10.1016/j.bcp.2017.06.112] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/02/2017] [Indexed: 12/11/2022]
|
472
|
Lee SH, Kwon JY, Kim SY, Jung K, Cho ML. Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci Rep 2017; 7:10133. [PMID: 28860618 PMCID: PMC5579272 DOI: 10.1038/s41598-017-09767-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023] Open
Abstract
Interferon γ (IFN-γ) induces an inflammatory response and apoptotic cell death. Rheumatoid arthritis (RA) is a systemic inflammatory disease associated with increased levels of inflammatory mediators, including tumour necrosis factor α (TNF-α) and T helper (Th) 17 cells, and downregulation of apoptosis of inflammatory cells. We hypothesized that IFN-γ would reduce inflammatory cell death in vitro and that loss of IFN-γ would aggravate inflammation in vivo. IFN-γ downregulated necroptosis and the expression of cellular FLICE-like inhibitory protein (cFLIPL) and mixed lineage kinase domain-like (MLKL). However, loss of IFN-γ promoted the production of cFLIPL and MLKL, and necroptosis. IFN-γ deficiency increased Th17 cell number and upregulated the expression of IL-17 and TNF-α. Expression of MLKL, receptor interacting protein kinase (RIPK)1, and RIPK3 was increased in the joints of mice with collagen-induced arthritis (CIA). Compared with wild-type mice with CIA, IFN-γ−/− CIA mice showed exacerbation of cartilage damage and joint inflammation, and acceleration of MLKL, RIPK1, and RIPK3 production in the joints. IFN-γ deficiency induced the activation of signal transducer and activator of transcription 3. These results suggest that IFN-γ regulates inflammatory cell death and may have potential for use in the treatment of RA.
Collapse
Affiliation(s)
- Seung Hoon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Ye Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,Laboratory of Immune Network, Conversant Research Consortium in Immunologic disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
473
|
Thornton C, Leaw B, Mallard C, Nair S, Jinnai M, Hagberg H. Cell Death in the Developing Brain after Hypoxia-Ischemia. Front Cell Neurosci 2017; 11:248. [PMID: 28878624 PMCID: PMC5572386 DOI: 10.3389/fncel.2017.00248] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023] Open
Abstract
Perinatal insults such as hypoxia–ischemia induces secondary brain injury. In order to develop the next generation of neuroprotective therapies, we urgently need to understand the underlying molecular mechanisms leading to cell death. The cell death mechanisms have been shown to be quite different in the developing brain compared to that in the adult. The aim of this review is update on what cell death mechanisms that are operating particularly in the setting of the developing CNS. In response to mild stress stimuli a number of compensatory mechanisms will be activated, most often leading to cell survival. Moderate-to-severe insults trigger regulated cell death. Depending on several factors such as the metabolic situation, cell type, nature of the stress stimulus, and which intracellular organelle(s) are affected, the cell undergoes apoptosis (caspase activation) triggered by BAX dependent mitochondrial permeabilzation, necroptosis (mixed lineage kinase domain-like activation), necrosis (via opening of the mitochondrial permeability transition pore), autophagic cell death (autophagy/Na+, K+-ATPase), or parthanatos (poly(ADP-ribose) polymerase 1, apoptosis-inducing factor). Severe insults cause accidental cell death that cannot be modulated genetically or by pharmacologic means. However, accidental cell death leads to the release of factors (damage-associated molecular patterns) that initiate systemic effects, as well as inflammation and (regulated) secondary brain injury in neighboring tissue. Furthermore, if one mode of cell death is inhibited, another route may step in at least in a scenario when upstream damaging factors predominate over protective responses. The provision of alternative routes through which the cell undergoes death has to be taken into account in the hunt for novel brain protective strategies.
Collapse
Affiliation(s)
- Claire Thornton
- Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom
| | - Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Carina Mallard
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Syam Nair
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Masako Jinnai
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Henrik Hagberg
- Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom.,Department of Clinical Sciences and Physiology and Neuroscience, Perinatal Center, Sahlgrenska Academy, Gothenburg UniversityGothenburg, Sweden
| |
Collapse
|
474
|
Apoptotic resistance of human skin mast cells is mediated by Mcl-1. Cell Death Discov 2017; 3:17048. [PMID: 28845295 PMCID: PMC5563844 DOI: 10.1038/cddiscovery.2017.48] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/03/2017] [Indexed: 12/16/2022] Open
Abstract
Mast cells (MCs) are major effector cells of allergic reactions and contribute to multiple other pathophysiological processes. MCs are long-lived in the tissue microenvironment, in which they matured, but it remains ill-defined how longevity is established by the natural habitat, as research on human MCs chiefly employs cells generated and expanded in culture. In this study, we report that naturally differentiated skin MCs exhibit substantial resilience to cell death with considerable portions surviving up to 3 days in the complete absence of growth factors (GF). This was evidenced by kinetic resolution of membrane alterations (Annexin-V, YoPro), DNA degradation (propidium iodide), mitochondrial membrane disruption (Depsipher), and Caspase-3 activity. Because of the high basal survival, further protection by SCF was modest. Conversely, survival was severely compromised by staurosporine, implying functional caspase machinery. Contrary to the resistance of freshly purified MCs, their culture-expanded counterpart readily underwent cell death upon GF deprivation. Searching for the molecular underpinnings explaining the difference, we identified Mcl-1 as a critical protector. In fact, silencing Mcl-1 by RNAi led to impaired survival in skin MCs ex vivo, but not their cultured equivalent. Therefore, MCs matured in the skin have not only higher expression of Mcl-1 than proliferating MCs, but also greater reliance on Mcl-1 for their survival. Collectively, we report that human skin MCs display low susceptibility to cell death through vast expression of Mcl-1, which protects from mortality and may contribute to MC longevity in the tissue.
Collapse
|
475
|
Pires WC, Lima BAV, de Castro Pereira F, Lima AP, Mello-Andrade F, Silva HD, da Silva MM, Colina-Vegas L, Ellena J, Batista AA, de Paul Silveira-Lacerda E. Ru(II)/diphenylphosphine/pyridine-6-thiolate complexes induce S-180 cell apoptosis through intrinsic mitochondrial pathway involving inhibition of Bcl-2 and p53/Bax activation. Mol Cell Biochem 2017; 438:199-217. [PMID: 28795366 DOI: 10.1007/s11010-017-3129-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/26/2017] [Indexed: 01/06/2023]
Abstract
The aim of this work was the synthesis, characterization, and cytotoxicity evaluation of three new Ru(II) complexes with a general formula [Ru(Spy)(bipy)(P-P)]PF6 [Spy = pyridine-6-thiolate; bipy = 2,2'-bipyridine; P-P = 1,2-bis(diphenylphosphine)ethane (1); 1,3-bis(diphenylphosphine) propane (2); and 1,1'-bis(diphenylphosphino)ferrocene] (4). Complex (3) with the 1,4-bis(diphenylphosphine)butane ligand, already known from the literature, was also synthesized, to be better studied here. The cytotoxicities of the complexes toward two kinds of cancerous cells (K562 and S-180 cells) were evaluated and compared to normal cells (L-929 and PBMC) by MTT assay. The complex [Ru(Spy)(bipy)(dppb)]PF6 (3) was selected to study both the cellular and molecular mechanisms underlying its promising anticancer action in S-180 cells. The results obtained from this study indicated that complex (3) induces cell cycle arrest in the G0/G1 phase in S-180 cells associated with a decrease in the number of cells in S phase. After 24 and 48 h of exposure to complex (3), the cell viability decreased when compared to the negative control. Complex (3) does not appear to be involved in the DNA damage, but induced changes in the mitochondrial membrane potential in S-180 cells. Furthermore, there was also an increase in the gene expression of Bax, Caspase 9, and Tp53. According to our results, complex (3) induces cell apoptosis through p53/Bax-dependent intrinsic pathway and suppresses the expression of active antiapoptotic Bcl-2 protein.
Collapse
Affiliation(s)
- Wanessa Carvalho Pires
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | | | - Flávia de Castro Pereira
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - Aliny Pereira Lima
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - Francyelli Mello-Andrade
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - Hugo Delleon Silva
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil
| | - Monize Martins da Silva
- Departamento de Química, Universidade Federal de São Carlos, P.O. Box 676, São Carlos, 13565-905, Brazil
| | - Legna Colina-Vegas
- Departamento de Química, Universidade Federal de São Carlos, P.O. Box 676, São Carlos, 13565-905, Brazil
| | - Javier Ellena
- Instituto de Física de São Carlos, Universidade de São Paulo, P.O. Box 369, São Carlos, SP, 13560-970, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, P.O. Box 676, São Carlos, 13565-905, Brazil.
| | - Elisângela de Paul Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil.
| |
Collapse
|
476
|
Conte A, Paladino S, Bianco G, Fasano D, Gerlini R, Tornincasa M, Renna M, Fusco A, Tramontano D, Pierantoni GM. High mobility group A1 protein modulates autophagy in cancer cells. Cell Death Differ 2017; 24:1948-1962. [PMID: 28777374 PMCID: PMC5635219 DOI: 10.1038/cdd.2017.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 06/01/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
High Mobility Group A1 (HMGA1) is an architectural chromatin protein whose overexpression is a feature of malignant neoplasias with a causal role in cancer initiation and progression. HMGA1 promotes tumor growth by several mechanisms, including increase of cell proliferation and survival, impairment of DNA repair and induction of chromosome instability. Autophagy is a self-degradative process that, by providing energy sources and removing damaged organelles and misfolded proteins, allows cell survival under stress conditions. On the other hand, hyper-activated autophagy can lead to non-apoptotic programmed cell death. Autophagy deregulation is a common feature of cancer cells in which has a complex role, showing either an oncogenic or tumor suppressor activity, depending on cellular context and tumor stage. Here, we report that depletion of HMGA1 perturbs autophagy by different mechanisms. HMGA1-knockdown increases autophagosome formation by constraining the activity of the mTOR pathway, a major regulator of autophagy, and transcriptionally upregulating the autophagy-initiating kinase Unc-51-like kinase 1 (ULK1). Consistently, functional experiments demonstrate that HMGA1 binds ULK1 promoter region and negatively regulates its transcription. On the other hand, the increase in autophagosomes is not associated to a proportionate increase in their maturation. Overall, the effects of HMGA1 depletion on autophagy are associated to a decrease in cell proliferation and ultimately impact on cancer cells viability. Importantly, silencing of ULK1 prevents the effects of HMGA1-knockdown on cellular proliferation, viability and autophagic activity, highlighting how these effects are, at least in part, mediated by ULK1. Interestingly, this phenomenon is not restricted to skin cancer cells, as similar results have been observed also in HeLa cells silenced for HMGA1. Taken together, these results clearly indicate HMGA1 as a key regulator of the autophagic pathway in cancer cells, thus suggesting a novel mechanism through which HMGA1 can contribute to cancer progression.
Collapse
Affiliation(s)
- Andrea Conte
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Gaia Bianco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Dominga Fasano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Raffaele Gerlini
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Mara Tornincasa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Maurizio Renna
- Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome Trust, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Alfredo Fusco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Donatella Tramontano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II' and Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS) of CNR, Naples, Italy
| |
Collapse
|
477
|
Filippova SN, Vinogradova KA. Programmed cell death as one of the stages of streptomycete differentiation. Microbiology (Reading) 2017. [DOI: 10.1134/s0026261717040075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
478
|
Fond AM, Ravichandran KS. Clearance of Dying Cells by Phagocytes: Mechanisms and Implications for Disease Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 930:25-49. [PMID: 27558816 PMCID: PMC6721615 DOI: 10.1007/978-3-319-39406-0_2] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The efficient clearance of apoptotic cells is an evolutionarily conserved process crucial for homeostasis in multicellular organisms. The clearance involves a series of steps that ultimately facilitates the recognition of the apoptotic cell by the phagocytes and the subsequent uptake and processing of the corpse. These steps include the phagocyte sensing of "find-me" signals released by the apoptotic cell, recognizing "eat-me" signals displayed on the apoptotic cell surface, and then intracellular signaling within the phagocyte to mediate phagocytic cup formation around the corpse and corpse internalization, and the processing of the ingested contents. The engulfment of apoptotic cells by phagocytes not only eliminates debris from tissues but also produces an anti-inflammatory response that suppresses local tissue inflammation. Conversely, impaired corpse clearance can result in loss of immune tolerance and the development of various inflammation-associated disorders such as autoimmunity, atherosclerosis, and airway inflammation but can also affect cancer progression. Recent studies suggest that the clearance process can also influence antitumor immune responses. In this review, we will discuss how apoptotic cells interact with their engulfing phagocytes to generate important immune responses, and how modulation of such responses can influence pathology.
Collapse
Affiliation(s)
- Aaron M Fond
- Center for Cell Clearance, and the Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, and the Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
479
|
Magrath JW, Kim Y. Salinomycin's potential to eliminate glioblastoma stem cells and treat glioblastoma multiforme (Review). Int J Oncol 2017; 51:753-759. [PMID: 28766685 DOI: 10.3892/ijo.2017.4082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/12/2017] [Indexed: 12/09/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and deadliest form of primary brain tumor. Despite treatment with surgery, radiotherapy, and chemotherapy with the drug temozolomide, the expected survival after diagnosis remains low. The median survival is only 14.6 months and the two-year survival is a mere 30%. One reason for this is the heterogeneity of GBM including the presence of glioblastoma cancer stem cells (GSCs). GSCs are a subset of cells with the unique ability to proliferate, differentiate, and create tumors. GSCs are resistant to chemotherapy and radiation and thought to play an important role in recurrence. In order to effectively treat GBM, a drug must be identified that can kill GSCs. The ionophore salinomycin has been shown to kill cancer stem cells and is therefore a promising future treatment for GBM. This study focuses on salinomycin's potential to treat GBM including its ability to reduce the CSC population, its toxicity to normal brain cells, its mechanism of action, and its potential for combination treatment.
Collapse
Affiliation(s)
- Justin W Magrath
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| |
Collapse
|
480
|
Basso P, Wallet P, Elsen S, Soleilhac E, Henry T, Faudry E, Attrée I. Multiple Pseudomonas species secrete exolysin-like toxins and provoke Caspase-1-dependent macrophage death. Environ Microbiol 2017; 19:4045-4064. [PMID: 28654176 DOI: 10.1111/1462-2920.13841] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/02/2017] [Accepted: 06/19/2017] [Indexed: 12/19/2022]
Abstract
Pathogenic bacteria secrete protein toxins that provoke apoptosis or necrosis of eukaryotic cells. Here, we developed a live-imaging method, based on incorporation of a DNA-intercalating dye into membrane-damaged host cells, to study the kinetics of primary bone marrow-derived macrophages (BMDMs) mortality induced by opportunistic pathogen Pseudomonas aeruginosa expressing either Type III Secretion System (T3SS) toxins or the pore-forming toxin, Exolysin (ExlA). We found that ExlA promotes the activation of Caspase-1 and maturation of interleukin-1β. BMDMs deficient for Caspase-1 and Caspase-11 were resistant to ExlA-induced death. Furthermore, by using KO BMDMs, we determined that the upstream NLRP3/ASC complex leads to the Caspase-1 activation. We also demonstrated that Pseudomonas putida and Pseudomonas protegens and the Drosophila pathogen Pseudomonas entomophila, which naturally express ExlA-like toxins, are cytotoxic toward macrophages and provoke the same type of pro-inflammatory death as does ExlA+ P. aeruginosa. These results demonstrate that ExlA-like toxins of two-partner secretion systems from diverse Pseudomonas species activate the NLRP3 inflammasome and provoke inflammatory pyroptotic death of macrophages.
Collapse
Affiliation(s)
- Pauline Basso
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Pierre Wallet
- CIRI, Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, F-69007, France
| | - Sylvie Elsen
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Emmanuelle Soleilhac
- CMBA Platform, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, CEA, INSERM; Genetics & Chemogenomics, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, F-69007, France
| | - Eric Faudry
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Ina Attrée
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| |
Collapse
|
481
|
Mikhail M, Vachon PH, D'Orléans-Juste P, Jacques D, Bkaily G. Role of endothelin-1 and its receptors, ET A and ET B, in the survival of human vascular endothelial cells. Can J Physiol Pharmacol 2017; 95:1298-1305. [PMID: 28732172 DOI: 10.1139/cjpp-2017-0412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our previous work showed the presence of endothelin-1 (ET-1) receptors, ETA and ETB, in human vascular endothelial cells (hVECs). In this study, we wanted to verify whether ET-1 plays a role in the survival of hVECs via the activation of its receptors ETA and (or) ETB (ETAR and ETBR, respectively). Our results showed that treatment of hVECs with ET-1 prevented apoptosis induced by genistein, an effect that was mimicked by treatment with ETBR-specific agonist IRL1620. Furthermore, blockade of ETBR with the selective ETBR antagonist A-192621 prevented the anti-apoptotic effect of ET-1 in hVECs. However, activation of ETA receptor alone did not seem to contribute to the anti-apoptotic effect of ET-1. In addition, the anti-apoptotic effect of ETBR was found to be associated with caspase 3 inhibition and does not depend on the density of this type of receptor. In conclusion, our results showed that ET-1 possesses an anti-apoptotic effect in hVECs and that this effect is mediated, to a great extent, via the activation of ETBR. This study revealed a new role for ETBR in the survival of hVECs.
Collapse
Affiliation(s)
- Marianne Mikhail
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pierre H Vachon
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pedro D'Orléans-Juste
- b Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Danielle Jacques
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Ghassan Bkaily
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
482
|
Lei Y, Liu K, Hou L, Ding L, Li Y, Liu L. Small chaperons and autophagy protected neurons from necrotic cell death. Sci Rep 2017; 7:5650. [PMID: 28720827 PMCID: PMC5515951 DOI: 10.1038/s41598-017-05995-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/07/2017] [Indexed: 01/22/2023] Open
Abstract
Neuronal necrosis occurs during early phase of ischemic insult. However, our knowledge of neuronal necrosis is still inadequate. To study the mechanism of neuronal necrosis, we previously established a Drosophila genetic model of neuronal necrosis by calcium overloading through expression of a constitutively opened cation channel mutant. Here, we performed further genetic screens and identified a suppressor of neuronal necrosis, CG17259, which encodes a seryl-tRNA synthetase. We found that loss-of-function (LOF) CG17259 activated eIF2α phosphorylation and subsequent up-regulation of chaperons (Hsp26 and Hsp27) and autophagy. Genetically, down-regulation of eIF2α phosphorylation, Hsp26/Hsp27 or autophagy reduced the protective effect of LOF CG17259, indicating they function downstream of CG17259. The protective effect of these protein degradation pathways indicated activation of a toxic protein during neuronal necrosis. Our data indicated that p53 was likely one such protein, because p53 was accumulated in the necrotic neurons and down-regulation of p53 rescued necrosis. In the SH-SY5Y human cells, tunicamycin (TM), a PERK activator, promoted transcription of hsp27; and necrosis induced by glutamate could be rescued by TM, associated with reduced p53 accumulation. In an ischemic stroke model in rats, p53 protein was also increased, and TM treatment could reduce the p53 accumulation and brain damage.
Collapse
Affiliation(s)
- Ye Lei
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China.,Aging and Disease lab of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Kai Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lin Hou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China.,Aging and Disease lab of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Lianggong Ding
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China.,Aging and Disease lab of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Yuhong Li
- Aging and Disease lab of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Lei Liu
- Aging and Disease lab of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing, 100069, China.
| |
Collapse
|
483
|
Dludla PV, Joubert E, Muller CJF, Louw J, Johnson R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2- O-β-D-glucoside. Nutr Metab (Lond) 2017; 14:45. [PMID: 28702068 PMCID: PMC5504778 DOI: 10.1186/s12986-017-0200-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022] Open
Abstract
Diabetic patients are at an increased risk of developing heart failure when compared to their non-diabetic counter parts. Accumulative evidence suggests chronic hyperglycemia to be central in the development of myocardial infarction in these patients. At present, there are limited therapies aimed at specifically protecting the diabetic heart at risk from hyperglycemia-induced injury. Oxidative stress, through over production of free radical species, has been hypothesized to alter mitochondrial function and abnormally augment the activity of the NADPH oxidase enzyme system resulting in accelerated myocardial injury within a diabetic state. This has led to a dramatic increase in the exploration of plant-derived materials known to possess antioxidative properties. Several edible plants contain various natural constituents, including polyphenols that may counteract oxidative-induced tissue damage through their modulatory effects of intracellular signaling pathways. Rooibos, an indigenous South African plant, well-known for its use as herbal tea, is increasingly studied for its metabolic benefits. Prospective studies linking diet rich in polyphenols from rooibos to reduced diabetes associated cardiovascular complications have not been extensively assessed. Aspalathin, a flavonoid, and phenylpyruvic acid-2-O-β-D-glucoside, a phenolic precursor, are some of the major compounds found in rooibos that can ameliorate hyperglycemia-induced cardiomyocyte damage in vitro. While the latter has demonstrated potential to protect against cell apoptosis, the proposed mechanism of action of aspalathin is linked to its capacity to enhance the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression, an intracellular antioxidant response element. Thus, here we review literature on the potential cardioprotective properties of flavonoids and a phenylpropenoic acid found in rooibos against diabetes-induced oxidative injury.
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Elizabeth Joubert
- Plant Bioactives Group, Post-Harvest and Wine Technology Division, Agricultural Research Council (ARC) Infruitec- Nietvoorbij, Stellenbosch, South Africa.,Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
484
|
Peña-Oyarzun D, Troncoso R, Kretschmar C, Hernando C, Budini M, Morselli E, Lavandero S, Criollo A. Hyperosmotic stress stimulates autophagy via polycystin-2. Oncotarget 2017; 8:55984-55997. [PMID: 28915568 PMCID: PMC5593539 DOI: 10.18632/oncotarget.18995] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/21/2017] [Indexed: 12/13/2022] Open
Abstract
Various intracellular mechanisms are activated in response to stress, leading to adaptation or death. Autophagy, an intracellular process that promotes lysosomal degradation of proteins, is an adaptive response to several types of stress. Osmotic stress occurs under both physiological and pathological conditions, provoking mechanical stress and activating various osmoadaptive mechanisms. Polycystin-2 (PC2), a membrane protein of the polycystin family, is a mechanical sensor capable of activating the cell signaling pathways required for cell adaptation and survival. Here we show that hyperosmotic stress provoked by treatment with hyperosmolar concentrations of sorbitol or mannitol induces autophagy in HeLa and HCT116 cell lines. In addition, we show that mTOR and AMPK, two stress sensor proteins involved modulating autophagy, are downregulated and upregulated, respectively, when cells are subjected to hyperosmotic stress. Finally, our findings show that PC2 is required to promote hyperosmotic stress-induced autophagy. Downregulation of PC2 prevents inhibition of hyperosmotic stress-induced mTOR pathway activation. In conclusion, our data provide new insight into the role of PC2 as a mechanosensor that modulates autophagy under hyperosmotic stress conditions.
Collapse
Affiliation(s)
- Daniel Peña-Oyarzun
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Catalina Kretschmar
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Cecilia Hernando
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Mauricio Budini
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Eugenia Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| |
Collapse
|
485
|
Moreno-Gonzalez G, Vandenabeele P, Krysko DV. Necroptosis: A Novel Cell Death Modality and Its Potential Relevance for Critical Care Medicine. Am J Respir Crit Care Med 2017; 194:415-28. [PMID: 27285640 DOI: 10.1164/rccm.201510-2106ci] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cell death is intertwined with life in development, homeostasis, pathology, and aging. Until recently, apoptosis was the best known form of programmed cell death, whereas necrosis was for a long time considered accidental owing to physicochemical injury. However, identification of crucial signaling and execution molecules, which are highly regulated, revealed that necrosis encompasses several cell death modalities that can be therapeutically targeted. The best understood form of regulated necrosis is necroptosis, which is transduced by the kinase activities of receptor interacting protein kinase-1 and receptor interacting protein kinase-3, eventually leading to the activation of mixed lineage kinase domain-like and plasma membrane permeabilization. We are only beginning to appreciate the role of necroptosis in different pathological conditions, including critical illnesses. In this review, we discuss the molecular mechanisms of necroptosis and analyze the effect of inhibiting necroptosis in experimental models of critical illnesses. In view of the identification of an increasing number of cell death modalities, we also briefly discuss the simultaneous targeting of multiple cell death modalities because, depending on the cell type and cellular conditions, various types of cell death may contribute to the pathology.
Collapse
Affiliation(s)
- Gabriel Moreno-Gonzalez
- 1 Molecular Signaling and Cell Death Unit, VIB Inflammation Research Center, Ghent, Belgium.,2 Department of Biomedical Molecular Biology, and.,3 Intensive Care Unit, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Peter Vandenabeele
- 1 Molecular Signaling and Cell Death Unit, VIB Inflammation Research Center, Ghent, Belgium.,2 Department of Biomedical Molecular Biology, and.,4 Methusalem Program, Ghent University, Ghent, Belgium; and
| | - Dmitri V Krysko
- 1 Molecular Signaling and Cell Death Unit, VIB Inflammation Research Center, Ghent, Belgium.,2 Department of Biomedical Molecular Biology, and
| |
Collapse
|
486
|
Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, Cuervo AM, Debnath J, Deretic V, Dikic I, Eskelinen EL, Fimia GM, Fulda S, Gewirtz DA, Green DR, Hansen M, Harper JW, Jäättelä M, Johansen T, Juhasz G, Kimmelman AC, Kraft C, Ktistakis NT, Kumar S, Levine B, Lopez-Otin C, Madeo F, Martens S, Martinez J, Melendez A, Mizushima N, Münz C, Murphy LO, Penninger JM, Piacentini M, Reggiori F, Rubinsztein DC, Ryan KM, Santambrogio L, Scorrano L, Simon AK, Simon HU, Simonsen A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Kroemer G. Molecular definitions of autophagy and related processes. EMBO J 2017; 36:1811-1836. [PMID: 28596378 PMCID: PMC5494474 DOI: 10.15252/embj.201796697] [Citation(s) in RCA: 1222] [Impact Index Per Article: 152.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/15/2022] Open
Abstract
Over the past two decades, the molecular machinery that underlies autophagic responses has been characterized with ever increasing precision in multiple model organisms. Moreover, it has become clear that autophagy and autophagy-related processes have profound implications for human pathophysiology. However, considerable confusion persists about the use of appropriate terms to indicate specific types of autophagy and some components of the autophagy machinery, which may have detrimental effects on the expansion of the field. Driven by the overt recognition of such a potential obstacle, a panel of leading experts in the field attempts here to define several autophagy-related terms based on specific biochemical features. The ultimate objective of this collaborative exchange is to formulate recommendations that facilitate the dissemination of knowledge within and outside the field of autophagy research.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Université Paris Descartes/Paris V, Paris, France
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - José Manuel Bravo-San Pedro
- Université Paris Descartes/Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Francesco Cecconi
- Department of Biology, University of Tor Vergata, Rome, Italy
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Augustine M Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Charleen T Chu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrice Codogno
- Université Paris Descartes/Paris V, Paris, France
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM, U1151, Paris, France
- CNRS, UMR8253, Paris, France
| | - Maria Isabel Colombo
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología (IHEM)-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jayanta Debnath
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ivan Dikic
- Institute of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt Main, Germany
- Department of Immunology and Medical Genetics, University of Split School of Medicine, Split, Croatia
| | | | - Gian Maria Fimia
- National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David A Gewirtz
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Gabor Juhasz
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
- Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Alec C Kimmelman
- Department of Radiation Oncology, Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, USA
| | - Claudine Kraft
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
| | | | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute (HHMI), Dallas, TX, USA
| | - Carlos Lopez-Otin
- Department de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación en Red de Cáncer, Oviedo, Spain
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sascha Martens
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Alicia Melendez
- Department of Biology, Queens College, Queens, NY, USA
- Graduate Center, City University of New York, New York, NY, USA
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Leon O Murphy
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Campus Vienna BioCentre, Vienna, Austria
| | - Mauro Piacentini
- Department of Biology, University of Tor Vergata, Rome, Italy
- National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Fulvio Reggiori
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| |
Collapse
|
487
|
Ciavatta ML, Lefranc F, Carbone M, Mollo E, Gavagnin M, Betancourt T, Dasari R, Kornienko A, Kiss R. Marine Mollusk-Derived Agents with Antiproliferative Activity as Promising Anticancer Agents to Overcome Chemotherapy Resistance. Med Res Rev 2017; 37:702-801. [PMID: 27925266 PMCID: PMC5484305 DOI: 10.1002/med.21423] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022]
Abstract
The chemical investigation of marine mollusks has led to the isolation of a wide variety of bioactive metabolites, which evolved in marine organisms as favorable adaptations to survive in different environments. Most of them are derived from food sources, but they can be also biosynthesized de novo by the mollusks themselves, or produced by symbionts. Consequently, the isolated compounds cannot be strictly considered as "chemotaxonomic markers" for the different molluscan species. However, the chemical investigation of this phylum has provided many compounds of interest as potential anticancer drugs that assume particular importance in the light of the growing literature on cancer biology and chemotherapy. The current review highlights the diversity of chemical structures, mechanisms of action, and, most importantly, the potential of mollusk-derived metabolites as anticancer agents, including those biosynthesized by mollusks and those of dietary origin. After the discussion of dolastatins and kahalalides, compounds previously studied in clinical trials, the review covers potentially promising anticancer agents, which are grouped based on their structural type and include terpenes, steroids, peptides, polyketides and nitrogen-containing compounds. The "promise" of a mollusk-derived natural product as an anticancer agent is evaluated on the basis of its ability to target biological characteristics of cancer cells responsible for poor treatment outcomes. These characteristics include high antiproliferative potency against cancer cells in vitro, preferential inhibition of the proliferation of cancer cells over normal ones, mechanism of action via nonapoptotic signaling pathways, circumvention of multidrug resistance phenotype, and high activity in vivo, among others. The review also includes sections on the targeted delivery of mollusk-derived anticancer agents and solutions to their procurement in quantity.
Collapse
Affiliation(s)
- Maria Letizia Ciavatta
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital ErasmeUniversité Libre de Bruxelles (ULB)1070BrusselsBelgium
| | - Marianna Carbone
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Ernesto Mollo
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Margherita Gavagnin
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Tania Betancourt
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Ramesh Dasari
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Alexander Kornienko
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie ExpérimentaleFaculté de Pharmacie, Université Libre de Bruxelles (ULB)1050BrusselsBelgium
| |
Collapse
|
488
|
Grootjans S, Vanden Berghe T, Vandenabeele P. Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ 2017; 24:1184-1195. [PMID: 28498367 PMCID: PMC5520172 DOI: 10.1038/cdd.2017.65] [Citation(s) in RCA: 422] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/23/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022] Open
Abstract
Necroptosis is a form of regulated cell death, which is induced by ligand binding to TNF family death domain receptors, pattern recognizing receptors and virus sensors. The common feature of these receptor systems is the implication of proteins, which contain a receptor interaction protein kinase (RIPK) homology interaction motif (RHIM) mediating recruitment and activation of receptor-interacting protein kinase 3 (RIPK3), which ultimately activates the necroptosis executioner mixed lineage kinase domain-like (MLKL). In case of the TNF family members, the initiator is the survival- and cell death-regulating RIPK1 kinase, in the case of Toll-like receptor 3/4 (TLR3/4), a RHIM-containing adaptor, called TRIF, while in the case of Z-DNA-binding protein ZBP1/DAI, the cytosolic viral sensor itself contains a RHIM domain. In this review, we discuss the different protein complexes that serve as nucleation platforms for necroptosis and the mechanism of execution of necroptosis. Transgenic models (knockout, kinase-dead knock-in) and pharmacologic inhibition indicate that RIPK1, RIPK3 or MLKL are implicated in many inflammatory, degenerative and infectious diseases. However, the conclusion of necroptosis being solely involved in the etiology of diseases is blurred by the pleiotropic roles of RIPK1 and RIPK3 in other cellular processes such as apoptosis and inflammasome activation.
Collapse
Affiliation(s)
- Sasker Grootjans
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Tom Vanden Berghe
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
489
|
Kim JS, Ha JY, Yang SJ, Son JH. A Novel Non-Apoptotic Role of Procaspase-3 in the Regulation of Mitochondrial Biogenesis Activators. J Cell Biochem 2017; 119:347-357. [PMID: 28585712 DOI: 10.1002/jcb.26186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/05/2017] [Indexed: 12/28/2022]
Abstract
The executioner caspase-3 has been proposed as a pharmacological intervention target to preserve degenerating dopaminergic (DA) neurons because apoptotic mechanisms involving caspase-3 contribute, at least in part, to the loss of DA neurons in patients and experimental models of Parkinson's disease (PD). Here, we determined that genetic intervention of caspase-3 was sufficient to prevent cell death against oxidative stress (OS), accompanied by unexpected severe mitochondrial dysfunction. Specifically, as we expected, caspase-3-deficient DA neuronal cells were very significantly resistant to OS-induced cell death, while the activation of the initiator caspase-9 by OS was preserved. Moreover, detailed phenotypic characterization of caspase-3-deficient DA cells revealed severe mitochondrial dysfunction, including an accumulation of damaged mitochondria with a characteristic swollen structure and broken cristae, reduced membrane potential, increased levels of reactive oxygen species (ROS), and deficits in mitochondrial oxidative phosphorylation (OXPHOS) enzymes. Of great interest, we found that mitochondrial biogenesis was dramatically decreased in caspase-3-deficient DA cells, whereas their capability of mitophagy was normal. In accordance with this observation, caspase-3 gene knock down (KD) resulted in dramatically decreased expression of the key transcriptional activators of mitochondrial biogenesis, such as Tfam and Nrf-1, implicating a non-apoptotic role of procaspase-3 in mitochondrial biogenesis. Therefore, a prolonged anti-apoptotic intervention targeting caspase-3 should be considered with caution due to the potential adverse effects in mitochondria dynamics resulting from a novel potential functional role of procaspase-3 in mitochondrial biogenesis via regulating the expression of mitochondrial biogenesis activators. J. Cell. Biochem. 119: 347-357, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ji-Soo Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha W. University, Seoul, 120-750, Republic of Korea
| | - Ji-Young Ha
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha W. University, Seoul, 120-750, Republic of Korea
| | - Sol-Ji Yang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha W. University, Seoul, 120-750, Republic of Korea
| | - Jin H Son
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha W. University, Seoul, 120-750, Republic of Korea
| |
Collapse
|
490
|
Cippà PE, Fehr T. Pharmacological modulation of cell death in organ transplantation. Transpl Int 2017; 30:851-859. [PMID: 28480540 DOI: 10.1111/tri.12977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/20/2017] [Accepted: 04/29/2017] [Indexed: 12/22/2022]
Abstract
New options to pharmacologically modulate fundamental mechanisms of regulated cell death are rapidly evolving and found first clinical applications in cancer therapy. Here, we present an overview on how the recent advances in the understanding of the biology and pharmacology of cell death might influence research and clinical practice in solid organ transplantation. Of particular interest are the novel opportunities related to organ preservation and immunomodulation, which might contribute to promote organ repair and to develop more selective ways to modulate allogeneic immune responses to prevent rejection and induce immunological tolerance.
Collapse
Affiliation(s)
- Pietro E Cippà
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Fehr
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
| |
Collapse
|
491
|
Zargarian S, Shlomovitz I, Erlich Z, Hourizadeh A, Ofir-Birin Y, Croker BA, Regev-Rudzki N, Edry-Botzer L, Gerlic M. Phosphatidylserine externalization, "necroptotic bodies" release, and phagocytosis during necroptosis. PLoS Biol 2017. [PMID: 28650960 PMCID: PMC5501695 DOI: 10.1371/journal.pbio.2002711] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Necroptosis is a regulated, nonapoptotic form of cell death initiated by receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL) proteins. It is considered to be a form of regulated necrosis, and, by lacking the “find me” and “eat me” signals that are a feature of apoptosis, necroptosis is considered to be inflammatory. One such “eat me” signal observed during apoptosis is the exposure of phosphatidylserine (PS) on the outer plasma membrane. Here, we demonstrate that necroptotic cells also expose PS after phosphorylated mixed lineage kinase-like (pMLKL) translocation to the membrane. Necroptotic cells that expose PS release extracellular vesicles containing proteins and pMLKL to their surroundings. Furthermore, inhibition of pMLKL after PS exposure can reverse the process of necroptosis and restore cell viability. Finally, externalization of PS by necroptotic cells drives recognition and phagocytosis, and this may limit the inflammatory response to this nonapoptotic form of cell death. The exposure of PS to the outer membrane and to extracellular vesicles is therefore a feature of necroptotic cell death and may serve to provide an immunologically-silent window by generating specific “find me” and “eat me” signals. Necroptosis, a recently discovered regulated form of cell death, is widely considered to be inflammatory due to the absence of specific “find me” and “eat me” signals prior to lytic death. Here, we demonstrate that necroptotic cells generate “find me” and “eat me” signals by exposure of phosphatidylserine on their outer plasma membrane. This was further associated with the release of extracellular vesicles (“necroptotic bodies”) that contain phosphatidylserine, pMLKL (a key necroptotic marker), as well as other proteins. These signals drive recognition and phagocytosis of necroptotic cells to modulate the immune response. The exposure of phosphatidylserine and release of “necroptotic bodies” indicate that apoptosis and necroptosis share some common biochemical and cellular features and highlight the need for new biomarkers to distinguish apoptotic and necroptotic cell death.
Collapse
Affiliation(s)
- Sefi Zargarian
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Shlomovitz
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ziv Erlich
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aria Hourizadeh
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yifat Ofir-Birin
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ben A. Croker
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Liat Edry-Botzer
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
492
|
Abstract
Autophagy represents a catabolic program involved in the degradation of cellular components via lysosomes. It serves to mitigate cellular stress and to provide metabolic precursors especially upon starvation. Thereby, autophagy can support the survival of cancer cells. In addition, there is now convincing evidence showing that under certain conditions autophagy can also foster cell death. This dual function of autophagy is also relevant upon anticancer treatment, as many chemotherapeutic agents engage autophagy. A better understanding of the molecular mechanisms that are critical for mediating autophagic cell death in cancer cells will be instrumental to selectively interfere with this cellular program in order to increase the cancer cell’s response to cytotoxic drugs. This review illustrates how anticancer drug-induced autophagy is involved in mediating cell death.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
493
|
Goodall ML, Fitzwalter BE, Zahedi S, Wu M, Rodriguez D, Mulcahy-Levy JM, Green DR, Morgan M, Cramer SD, Thorburn A. The Autophagy Machinery Controls Cell Death Switching between Apoptosis and Necroptosis. Dev Cell 2017; 37:337-349. [PMID: 27219062 DOI: 10.1016/j.devcel.2016.04.018] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 03/29/2016] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Although autophagy controls cell death and survival, underlying mechanisms are poorly understood, and it is unknown whether autophagy affects only whether or not cells die or also controls other aspects of programmed cell death. MAP3K7 is a tumor suppressor gene associated with poor disease-free survival in prostate cancer. Here, we report that Map3k7 deletion in mouse prostate cells sensitizes to cell death by TRAIL (TNF-related apoptosis-inducing ligand). Surprisingly, this death occurs primarily through necroptosis, not apoptosis, due to assembly of the necrosome in association with the autophagy machinery, mediated by p62/SQSTM1 recruitment of RIPK1. The mechanism of cell death switches to apoptosis if p62-dependent recruitment of the necrosome to the autophagy machinery is blocked. These data show that the autophagy machinery can control the mechanism of programmed cell death by serving as a scaffold rather than by degrading cargo.
Collapse
Affiliation(s)
- Megan L Goodall
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Brent E Fitzwalter
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Shadi Zahedi
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Min Wu
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA; Department of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Diego Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jean M Mulcahy-Levy
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael Morgan
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Scott D Cramer
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
494
|
Yang Y, Yu Y, Wang J, Li Y, Li Y, Wei J, Zheng T, Jin M, Sun Z. Silica nanoparticles induced intrinsic apoptosis in neuroblastoma SH-SY5Y cells via CytC/Apaf-1 pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 52:161-169. [PMID: 28426994 DOI: 10.1016/j.etap.2017.01.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/11/2016] [Accepted: 01/18/2017] [Indexed: 06/07/2023]
Abstract
The present study was to investigate effects of Silica nanoparticles (SiNPs) on nervous system and explore potential mechanisms in human neuroblastoma cells (SH-SY5Y). Cytotoxicity was detected by cell viability and Lactate dehydrogenase (LDH) release. Flow cytometry analysis was applied to assess mitochondrial membrane potential (MMP) loss, intracellular Ca2+ and apoptosis. To clarify the mechanism of SiNPs-induced apoptosis, intrinsic apoptosis-related proteins were detected. Our results showed that SiNPs caused cytotoxicity, cell membrane damage and Ca2+ increase in a dose-dependent manner in SH-SY5Y cells. Both the mitochondrial membrane potential (MMP) loss and potential mitochondria damage resulted in Cyt C release to the cytoplasm. The elevated Cyt C and Apaf1 further triggered intrinsic apoptosis via executive molecular caspase-9 and caspase-3. The present study confirmed that SiNPs induced intrinsic apoptosis in neuroblastoma SH-SY5Y cells via CytC/Apaf-1 pathway and provided a better understanding of the potential toxicity induced by SiNPs on human neurocyte.
Collapse
Affiliation(s)
- Yanyan Yang
- School of Public Health, Jilin University, Changchun, PR China
| | - Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, PR China; School of Public Health, Capital Medical University, Beijing, PR China
| | - Jiahui Wang
- School of Public Health, Jilin University, Changchun, PR China
| | - Yanbo Li
- School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Yang Li
- School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Jia Wei
- College of Clinical Medicine, Norman Bethune Health Science Center, Jilin University, Changchun, PR China
| | - Tong Zheng
- School of Public Health, Jilin University, Changchun, PR China
| | - Minghua Jin
- School of Public Health, Jilin University, Changchun, PR China.
| | - Zhiwei Sun
- School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China.
| |
Collapse
|
495
|
Abstract
PURPOSE OF REVIEW Autophagy is a conserved intracellular degradation system and plays a dual role in cell death, depending on context and phase. Ferroptosis is a new form of regulated cell death that mainly depends on iron accumulation and lipid peroxidation. In this review, we summarize the processes of autophagy and ferroptosis and discuss their crosstalk mechanisms at the molecular level. RECENT FINDINGS The original study shows that ferroptosis is morphologically, biochemically, and genetically distinct from autophagy and other types of cell death. However, recent studies demonstrate that activation of ferroptosis is indeed dependent on the induction of autophagy. Additionally, many ferroptosis regulators such as SLC7A11, GPX4, NRF2, p53, HSPB1, CISD1, FANCD2, and ACSL4 have been identified as potential regulators of autophagy. SUMMARY This review not only highlights the importance of autophagy as an emerging mechanism of ferroptosis, but also raises new insights regarding regulated cell death.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| |
Collapse
|
496
|
Bonora M, Morganti C, Morciano G, Pedriali G, Lebiedzinska-Arciszewska M, Aquila G, Giorgi C, Rizzo P, Campo G, Ferrari R, Kroemer G, Wieckowski MR, Galluzzi L, Pinton P. Mitochondrial permeability transition involves dissociation of F 1F O ATP synthase dimers and C-ring conformation. EMBO Rep 2017; 18:1077-1089. [PMID: 28566520 DOI: 10.15252/embr.201643602] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/22/2017] [Accepted: 04/25/2017] [Indexed: 11/09/2022] Open
Abstract
The impact of the mitochondrial permeability transition (MPT) on cellular physiology is well characterized. In contrast, the composition and mode of action of the permeability transition pore complex (PTPC), the supramolecular entity that initiates MPT, remain to be elucidated. Specifically, the precise contribution of the mitochondrial F1FO ATP synthase (or subunits thereof) to MPT is a matter of debate. We demonstrate that F1FO ATP synthase dimers dissociate as the PTPC opens upon MPT induction. Stabilizing F1FO ATP synthase dimers by genetic approaches inhibits PTPC opening and MPT Specific mutations in the F1FO ATP synthase c subunit that alter C-ring conformation sensitize cells to MPT induction, which can be reverted by stabilizing F1FO ATP synthase dimers. Destabilizing F1FO ATP synthase dimers fails to trigger PTPC opening in the presence of mutants of the c subunit that inhibit MPT The current study does not provide direct evidence that the C-ring is the long-sought pore-forming subunit of the PTPC, but reveals that PTPC opening requires the dissociation of F1FO ATP synthase dimers and involves the C-ring.
Collapse
Affiliation(s)
- Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Claudia Morganti
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Giampaolo Morciano
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Gaia Pedriali
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | - Giorgio Aquila
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Cardiovascular Institute, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Cardiovascular Institute, University of Ferrara, Ferrara, Italy
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,INSERM, U1138, Paris, France.,Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France .,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy .,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
497
|
Cimini E, Viola D, Cabeza-Cabrerizo M, Romanelli A, Tumino N, Sacchi A, Bordoni V, Casetti R, Turchi F, Martini F, Bore JA, Koundouno FR, Duraffour S, Michel J, Holm T, Zekeng EG, Cowley L, Garcia Dorival I, Doerrbecker J, Hetzelt N, Baum JHJ, Portmann J, Wölfel R, Gabriel M, Miranda O, Díaz G, Díaz JE, Fleites YA, Piñeiro CA, Castro CM, Koivogui L, Magassouba N, Diallo B, Ruibal P, Oestereich L, Wozniak DM, Lüdtke A, Becker-Ziaja B, Capobianchi MR, Ippolito G, Carroll MW, Günther S, Di Caro A, Muñoz-Fontela C, Agrati C. Different features of Vδ2 T and NK cells in fatal and non-fatal human Ebola infections. PLoS Negl Trop Dis 2017; 11:e0005645. [PMID: 28558022 PMCID: PMC5472323 DOI: 10.1371/journal.pntd.0005645] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/15/2017] [Accepted: 05/17/2017] [Indexed: 01/08/2023] Open
Abstract
Background Human Ebola infection is characterized by a paralysis of the immune system. A signature of αβ T cells in fatal Ebola infection has been recently proposed, while the involvement of innate immune cells in the protection/pathogenesis of Ebola infection is unknown. Aim of this study was to analyze γδ T and NK cells in patients from the Ebola outbreak of 2014–2015 occurred in West Africa, and to assess their association with the clinical outcome. Methodology/Principal findings Nineteen Ebola-infected patients were enrolled at the time of admission to the Ebola Treatment Centre in Guinea. Patients were divided in two groups on the basis of the clinical outcome. The analysis was performed by using multiparametric flow cytometry established by the European Mobile Laboratory in the field. A low frequency of Vδ2 T-cells was observed during Ebola infection, independently from the clinical outcome. Moreover, Vδ2 T-cells from Ebola patients massively expressed CD95 apoptotic marker, suggesting the involvement of apoptotic mechanisms in Vδ2 T-cell loss. Interestingly, Vδ2 T-cells from survivors expressed an effector phenotype and presented a lower expression of the CTLA-4 exhaustion marker than fatalities, suggesting a role of effector Vδ2 T-cells in the protection. Furthermore, patients with fatal Ebola infection were characterized by a lower NK cell frequency than patients with non fatal infection. In particular, both CD56bright and CD56dim NK frequency were very low both in fatal and non fatal infections, while a higher frequency of CD56neg NK cells was associated to non-fatal infections. Finally, NK activation and expression of NKp46 and CD158a were independent from clinical outcome. Conclusions/Significances Altogether, the data suggest that both effector Vδ2 T-cells and NK cells may play a role in the complex network of protective response to EBOV infection. Further studies are required to characterize the protective effector functions of Vδ2 and NK cells. Human Ebola infection presents a high lethality rate and is characterized by a paralysis of the immune response. The definition of the protective immune profile during Ebola infection represents a main challenge useful in vaccine and therapy design. In particular, the protective/pathogenetic involvement of innate immune cells during Ebola infection in humans remains to be clarified. Nineteen Ebola-infected patients were enrolled at the time of admission to the Ebola Treatment Center in Guinea, and the profiling of innate immunity was correlated with the clinical outcome. Our results show that both effector Vδ2 T-cells and NK cells were associated with survival, suggesting their involvement in the complex network of protective response to EBOV infection.
Collapse
Affiliation(s)
- Eleonora Cimini
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Domenico Viola
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Mar Cabeza-Cabrerizo
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Antonella Romanelli
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Nicola Tumino
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Alessandra Sacchi
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Veronica Bordoni
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Rita Casetti
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Federica Turchi
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Federico Martini
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Joseph A Bore
- European Mobile Laboratory Consortium, Hamburg, Germany
| | | | - Sophie Duraffour
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Janine Michel
- European Mobile Laboratory Consortium, Hamburg, Germany.,Robert Koch Institute, Berlin, Germany
| | - Tobias Holm
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Elsa Gayle Zekeng
- European Mobile Laboratory Consortium, Hamburg, Germany.,Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Lauren Cowley
- European Mobile Laboratory Consortium, Hamburg, Germany.,National Infection Service, Public Health England, Porton Down and Colindale, United Kingdom
| | - Isabel Garcia Dorival
- European Mobile Laboratory Consortium, Hamburg, Germany.,Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Juliane Doerrbecker
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,Centre for Experimental and Clinical Infection Research (TWINCORE), Institute for Experimental Virology, Hannover, Germany
| | - Nicole Hetzelt
- European Mobile Laboratory Consortium, Hamburg, Germany.,Robert Koch Institute, Berlin, Germany
| | - Jonathan H J Baum
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany
| | - Jasmine Portmann
- European Mobile Laboratory Consortium, Hamburg, Germany.,Federal Office for Civil Protection, Spiez Laboratory, Switzerland
| | - Roman Wölfel
- European Mobile Laboratory Consortium, Hamburg, Germany.,Bundeswehr Institute of Microbiology, Munich, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | - Martin Gabriel
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | | | | | - José E Díaz
- Hospital Militar Central Dr. Carlos J. Finlay, Havana, Cuba
| | - Yoel A Fleites
- Hospital Militar Central Dr. Carlos J. Finlay, Havana, Cuba
| | | | | | | | - N'Faly Magassouba
- Laboratoire des Fièvres Hémorragiques en Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Boubacar Diallo
- World Health Organization, Geneva, Switzerland. (Boubacar is separate: World Health Organization, Conakry, Guinea)
| | - Paula Ruibal
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Lisa Oestereich
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | - David M Wozniak
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | - Anja Lüdtke
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Beate Becker-Ziaja
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | - Maria R Capobianchi
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Giuseppe Ippolito
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Miles W Carroll
- European Mobile Laboratory Consortium, Hamburg, Germany.,National Infection Service, Public Health England, Porton Down and Colindale, United Kingdom.,University of Southampton, South General Hospital, Southampton, United Kingdom
| | - Stephan Günther
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany
| | - Antonino Di Caro
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy.,European Mobile Laboratory Consortium, Hamburg, Germany
| | - César Muñoz-Fontela
- European Mobile Laboratory Consortium, Hamburg, Germany.,Department of Virology, Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Chiara Agrati
- Department of Epidemiology and Pre-clinical research, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| |
Collapse
|
498
|
Abstract
Autophagy contributes to the maintenance of intracellular homeostasis in most cells of cardiovascular origin, including cardiomyocytes, endothelial cells, and arterial smooth muscle cells. Mitophagy is an autophagic response that specifically targets damaged, and hence potentially cytotoxic, mitochondria. As these organelles occupy a critical position in the bioenergetics of the cardiovascular system, mitophagy is particularly important for cardiovascular homeostasis in health and disease. Consistent with this notion, genetic defects in autophagy or mitophagy have been shown to exacerbate the propensity of laboratory animals to spontaneously develop cardiodegenerative disorders. Moreover, pharmacological or genetic maneuvers that alter the autophagic or mitophagic flux have been shown to influence disease outcome in rodent models of several cardiovascular conditions, such as myocardial infarction, various types of cardiomyopathy, and atherosclerosis. In this review, we discuss the intimate connection between autophagy, mitophagy, and cardiovascular disorders.
Collapse
|
499
|
Lerner LK, Francisco G, Soltys DT, Rocha CRR, Quinet A, Vessoni AT, Castro LP, David TIP, Bustos SO, Strauss BE, Gottifredi V, Stary A, Sarasin A, Chammas R, Menck CFM. Predominant role of DNA polymerase eta and p53-dependent translesion synthesis in the survival of ultraviolet-irradiated human cells. Nucleic Acids Res 2017; 45:1270-1280. [PMID: 28180309 PMCID: PMC5388406 DOI: 10.1093/nar/gkw1196] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 01/19/2023] Open
Abstract
Genome lesions trigger biological responses that help cells manage damaged DNA, improving cell survival. Pol eta is a translesion synthesis (TLS) polymerase that bypasses lesions that block replicative polymerases, avoiding continued stalling of replication forks, which could lead to cell death. p53 also plays an important role in preventing cell death after ultraviolet (UV) light exposure. Intriguingly, we show that p53 does so by favoring translesion DNA synthesis by pol eta. In fact, the p53-dependent induction of pol eta in normal and DNA repair-deficient XP-C human cells after UV exposure has a protective effect on cell survival after challenging UV exposures, which was absent in p53- and Pol H-silenced cells. Viability increase was associated with improved elongation of nascent DNA, indicating the protective effect was due to more efficient lesion bypass by pol eta. This protection was observed in cells proficient or deficient in nucleotide excision repair, suggesting that, from a cell survival perspective, proper bypass of DNA damage can be as relevant as removal. These results indicate p53 controls the induction of pol eta in DNA damaged human cells, resulting in improved TLS and enhancing cell tolerance to DNA damage, which parallels SOS responses in bacteria.
Collapse
Affiliation(s)
- Leticia K Lerner
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Guilherme Francisco
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of São Paulo-ICESP, São Paulo, Brazil
| | - Daniela T Soltys
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Clarissa R R Rocha
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Annabel Quinet
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexandre T Vessoni
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ligia P Castro
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Taynah I P David
- Viral Vector Laboratory, Heart Institute, University of São Paulo, São Paulo, Brazil
| | - Silvina O Bustos
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of São Paulo-ICESP, São Paulo, Brazil
| | - Bryan E Strauss
- Viral Vector Laboratory, Heart Institute, University of São Paulo, São Paulo, Brazil
| | - Vanesa Gottifredi
- Cell Cycle and Genomic Stability Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Anne Stary
- CNRS-UMR8200, Université Paris-Sud, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Alain Sarasin
- CNRS-UMR8200, Université Paris-Sud, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Roger Chammas
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of São Paulo-ICESP, São Paulo, Brazil
| | - Carlos F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
500
|
Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat Rev Drug Discov 2017; 16:487-511. [PMID: 28529316 DOI: 10.1038/nrd.2017.22] [Citation(s) in RCA: 644] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autophagy is central to the maintenance of organismal homeostasis in both physiological and pathological situations. Accordingly, alterations in autophagy have been linked to clinically relevant conditions as diverse as cancer, neurodegeneration and cardiac disorders. Throughout the past decade, autophagy has attracted considerable attention as a target for the development of novel therapeutics. However, such efforts have not yet generated clinically viable interventions. In this Review, we discuss the therapeutic potential of autophagy modulators, analyse the obstacles that have limited their development and propose strategies that may unlock the full therapeutic potential of autophagy modulation in the clinic.
Collapse
|