201
|
Identification of a Transcription Factor That Regulates Host Cell Exit and Virulence of Mycobacterium tuberculosis. PLoS Pathog 2016; 12:e1005652. [PMID: 27191591 PMCID: PMC4871555 DOI: 10.1371/journal.ppat.1005652] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/01/2016] [Indexed: 02/04/2023] Open
Abstract
The interaction of Mycobacterium tuberculosis (Mtb) with host cell death signaling pathways is characterized by an initial anti-apoptotic phase followed by a pro-necrotic phase to allow for host cell exit of the bacteria. The bacterial modulators regulating necrosis induction are poorly understood. Here we describe the identification of a transcriptional repressor, Rv3167c responsible for regulating the escape of Mtb from the phagosome. Increased cytosolic localization of MtbΔRv3167c was accompanied by elevated levels of mitochondrial reactive oxygen species and reduced activation of the protein kinase Akt, and these events were critical for the induction of host cell necrosis and macroautophagy. The increase in necrosis led to an increase in bacterial virulence as reflected in higher bacterial burden and reduced survival of mice infected with MtbΔRv3167c. The regulon of Rv3167c thus contains the bacterial mediators involved in escape from the phagosome and host cell necrosis induction, both of which are crucial steps in the intracellular lifecycle and virulence of Mtb. Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is a highly successful human pathogen. Following entry into host phagocytic cells, Mtb resides within a modified phagosomal compartment and inhibits apoptotic host cell death. Recent studies have demonstrated that Mtb eventually translocates from the phagosomal compartment to the cytosol. This event is followed by the induction of necrotic host cell death allowing the bacteria to exit the host cell and infect naive cell populations. Our study adds to this relatively unexplored aspect of Mtb pathogenesis by revealing that the transcriptional repressor Rv3167c of Mtb negatively regulates phagosomal escape and host cell necrosis. We furthermore demonstrate that the increased necrosis induction by the Mtb mutant strain deficient in Rv3167c required elevated reactive oxygen species levels within host cell mitochondria and reduced activation of the protein kinase Akt. In addition, the increased virulence of the Mtb mutant strain observed after aerosol infection of mice strengthens the link between the ability of the bacteria to induce host cell necrosis and virulence. The Mtb genes negatively regulated by Rv3167c are thus potential virulence factors that can be targeted for drug and vaccine development.
Collapse
|
202
|
Seldon CS, Colbert LE, Hall WA, Fisher SB, Yu DS, Landry JC. Chromodomain-helicase-DNA binding protein 5, 7 and pronecrotic mixed lineage kinase domain-like protein serve as potential prognostic biomarkers in patients with resected pancreatic adenocarcinomas. World J Gastrointest Oncol 2016; 8:358-365. [PMID: 27096031 PMCID: PMC4824714 DOI: 10.4251/wjgo.v8.i4.358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/05/2015] [Accepted: 01/11/2016] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer is one of the deadliest cancers with a very poor prognosis. Recently, there has been a significant increase in research directed towards identifying potential biomarkers that can be used to diagnose and provide prognostic information for pancreatic cancer. These markers can be used clinically to optimize and personalize therapy for individual patients. In this review, we focused on 3 biomarkers involved in the DNA damage response pathway and the necroptosis pathway: Chromodomain-helicase-DNA binding protein 5, chromodomain-helicase-DNA binding protein 7, and mixed lineage kinase domain-like protein. The aim of this article is to review present literature provided for these biomarkers and current studies in which their effectiveness as prognostic biomarkers are analyzed in order to determine their future use as biomarkers in clinical medicine. Based on the data presented, these biomarkers warrant further investigation, and should be validated in future studies.
Collapse
|
203
|
Wang S, Zhang C, Hu L, Yang C. Necroptosis in acute kidney injury: a shedding light. Cell Death Dis 2016; 7:e2125. [PMID: 26938298 PMCID: PMC4823938 DOI: 10.1038/cddis.2016.37] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/27/2022]
Abstract
Acute kidney injury (AKI) is a common and severe clinical condition with a heavy healthy burden around the world. In spite of supportive therapies, the mortality associated with AKI remains high. Our limited understanding of the complex cell death mechanism in the process of AKI impedes the development of desirable therapeutics. Necroptosis is a recently identified novel form of cell death contributing to numerable diseases and tissue damages. Increasing evidence has suggested that necroptosis has an important role in the pathogenesis of various types of AKI. Therefore, we present here the signaling pathways and main regulators of necroptosis that are potential candidate for therapeutic strategies. Moreover, we emphasize on the potential role and corresponding mechanisms of necroptosis in AKI based on recent advances, and also discuss the possible therapeutic regimens based on manipulating necroptosis. Taken together, the progress in this field sheds new light into the prevention and management of AKI in clinical practice.
Collapse
Affiliation(s)
- S Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - C Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - L Hu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - C Yang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
204
|
Ghanem CI, Pérez MJ, Manautou JE, Mottino AD. Acetaminophen from liver to brain: New insights into drug pharmacological action and toxicity. Pharmacol Res 2016; 109:119-31. [PMID: 26921661 DOI: 10.1016/j.phrs.2016.02.020] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/19/2016] [Accepted: 02/21/2016] [Indexed: 01/13/2023]
Abstract
Acetaminophen (APAP) is a well-known analgesic and antipyretic drug. It is considered to be safe when administered within its therapeutic range, but in cases of acute intoxication, hepatotoxicity can occur. APAP overdose is the leading cause of acute liver failure in the northern hemisphere. Historically, studies on APAP toxicity have been focused on liver, with alterations in brain function attributed to secondary effects of acute liver failure. However, in the last decade the pharmacological mechanism of APAP as a cannabinoid system modulator has been documented and some articles have reported "in situ" toxicity by APAP in brain tissue at high doses. Paradoxically, low doses of APAP have been reported to produce the opposite, neuroprotective effects. In this paper we present a comprehensive, up-to-date overview of hepatic toxicity as well as a thorough review of both toxic and beneficial effects of APAP in brain.
Collapse
Affiliation(s)
- Carolina I Ghanem
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina; Cátedra de Fisiopatología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - María J Pérez
- Cátedra de Química Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), UBA-CONICET, Buenos Aires, Argentina
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Aldo D Mottino
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| |
Collapse
|
205
|
Zhang L, Ding Y, Yuan Z, Liu J, Sun J, Lei F, Wu S, Li S, Zhang D. MicroRNA-500 sustains nuclear factor-κB activation and induces gastric cancer cell proliferation and resistance to apoptosis. Oncotarget 2016; 6:2483-95. [PMID: 25595906 PMCID: PMC4385865 DOI: 10.18632/oncotarget.2800] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 11/20/2015] [Indexed: 11/25/2022] Open
Abstract
Ubiquitin deconjugation of key signalling molecules by deubiquitinases (DUBs) such as cylindromatosis (CYLD), A20, and OTU deubiquitinase 7B (OTUD7B) has emerged as an important regulatory mechanism in the downregulation of NF-κB signalling and homeostasis. However, how these serial negative regulations are simultaneously disrupted to result in constitutive activation of NF-κB signalling in cancers remains puzzling. Here, we report that the miR-500 directly repressed the expression of CYLD, OTUD7B, and the A20 complex component Tax1-binding protein 1 (TAX1BP1), leading to ubiquitin conjugation of receptor-interacting protein 1 (RIP1) and sustained NF-ĸB activation. Furthermore, we found that miR-500 promoted gastric cancer cell proliferation, survival, and tumorigenicity. Importantly, miR-500 was upregulated in gastric cancer and was highly correlated with malignant progression and poor survival. Hence, we report the uncovering of a novel mechanism for constitutive NF-κB activation, indicating the potentially pivotal role of miR-500 in the progression of gastric cancer.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Department of Diagnostic Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ya Ding
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Junling Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jian Sun
- Clinical Trial Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Fangyong Lei
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shu Wu
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Su Li
- Clinical Trial Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Dongsheng Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
206
|
Sun X, Shi X, Lu L, Jiang Y, Liu B. Stimulus-dependent neuronal cell responses in SH-SY5Y neuroblastoma cells. Mol Med Rep 2016; 13:2215-20. [PMID: 26781445 DOI: 10.3892/mmr.2016.4759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 08/25/2015] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to elucidate the intracellular mechanisms that cause neuronal cell death following exposure to excitatory neurotransmitter‑induced neurotoxicity, neurotoxins and oxidative stress. Human SH‑SY5Y neuroblastoma cells were exposed to various stimuli, including glutamate, 6‑hydroxydopamine (6‑OHDA), and glucose oxidase, and cell viability was determined by MTT assay. Early apoptosis and necrosis were examined by Annexin V/propidium iodide double staining and flow cytometric analysis. Intracellular calcium ion concentration and mitochondrial membrane potential were assessed by Fluo‑3a and JC‑1 staining, respectively. In addition, protein expression of receptor‑interacting protein (RIP) kinase 1 and RIP kinase 3 were evaluated by western blotting. Glutamate, 6‑OHDA and glucose oxidase treatment decreased cell viability. Glutamate induced apoptosis and necrosis, whereas, 6‑OHDA induced cell necrosis and glucose oxidase induced apoptosis. Furthermore, glutamate, 6‑OHDA or glucose oxidase treatment significantly increased intracellular calcium concentrations (P<0.05). The effect of glutamate on mitochondrial membrane potential varied with high and low concentrations, whereas 6‑OHDA and glucose oxidase significantly increased the mitochondrial membrane potential in the SH‑SY5Y cells (P<0.05). Glutamate significantly upregulated expression levels of RIP kinase 1 (P<0.05), but not RIP kinase 3. These findings demonstrate that the response of SH‑SY5Y cells varies with the stimuli. Furthermore, RIP kinase 1 may specifically regulate programmed necrosis in glutamate‑mediated excitatory toxicity, but not in cell damage induced by either 6-OHDA or glucose oxidase.
Collapse
Affiliation(s)
- Xiguang Sun
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xu Shi
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Laijing Lu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
207
|
Mathis BJ, Lai Y, Qu C, Janicki JS, Cui T. CYLD-mediated signaling and diseases. Curr Drug Targets 2016; 16:284-94. [PMID: 25342597 DOI: 10.2174/1389450115666141024152421] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/26/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
The conserved cylindromatosis (CYLD) codes for a deubiquitinating enzyme and is a crucial regulator of diverse cellular processes such as immune responses, inflammation, death, and proliferation. It directly regulates multiple key signaling cascades, such as the Nuclear Factor kappa B [NFkB] and the Mitogen-Activated Protein Kinase (MAPK) pathways, by its catalytic activity on polyubiquitinated key intermediates. Several lines of emerging evidence have linked CYLD to the pathogenesis of various maladies, including cancer, poor infection control, lung fibrosis, neural development, and now cardiovascular dysfunction. While CYLD-mediated signaling is cell type and stimuli specific, the activity of CYLD is tightly controlled by phosphorylation and other regulators such as Snail. This review explores a broad selection of current and past literature regarding CYLD's expression, function and regulation with emerging reports on its role in cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | | | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
208
|
|
209
|
Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe 2015; 17:243-51. [PMID: 25674983 DOI: 10.1016/j.chom.2015.01.003] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/08/2014] [Accepted: 01/09/2015] [Indexed: 11/22/2022]
Abstract
Herpes simplex virus (HSV)-1 and HSV-2 are significant human pathogens causing recurrent disease. During infection, HSV modulates cell death pathways using the large subunit (R1) of ribonucleotide reductase (RR) to suppress apoptosis by binding to and blocking caspase-8. Here, we demonstrate that HSV-1 and HSV-2 R1 proteins (ICP6 and ICP10, respectively) also prevent necroptosis in human cells by inhibiting the interaction between receptor-interacting protein kinase 1 (RIP1) and RIP3, a key step in tumor necrosis factor (TNF)-induced necroptosis. We show that suppression of this cell death pathway requires an N-terminal RIP homotypic interaction motif (RHIM) within R1, acting in concert with the caspase-8-binding domain, which unleashes necroptosis independent of RHIM function. Thus, necroptosis is a human host defense pathway against two important viral pathogens that naturally subvert multiple death pathways via a single evolutionarily conserved gene product.
Collapse
|
210
|
Matty MA, Roca FJ, Cronan MR, Tobin DM. Adventures within the speckled band: heterogeneity, angiogenesis, and balanced inflammation in the tuberculous granuloma. Immunol Rev 2015; 264:276-87. [PMID: 25703566 DOI: 10.1111/imr.12273] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent work in a variety of animal models, including mice, zebrafish, and macaques, as well as in humans, has led to a reassessment of several tenets of mycobacterial infection. In this review, we describe new findings about the composition and dynamics of the tuberculous granuloma, the central host structure in mycobacterial infection, as well as inflammatory mediators that drive a successful anti-microbial response on one hand and pathological inflammation on the other. We highlight granuloma heterogeneity that emerges in the context of infection, the functional consequences of angiogenesis in tuberculous granulomas, and data that balanced inflammation in humans, with a central role for tumor necrosis factor, appears to play a key role in optimal defense against mycobacterial infection. These findings have suggested new and specific host-directed therapies that await further clinical exploration.
Collapse
Affiliation(s)
- Molly A Matty
- Department of Molecular Genetics and Microbiology, Center for Host-Microbial Interactions, Duke University Medical Center, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | | | | | | |
Collapse
|
211
|
Delgado ME, Grabinger T, Brunner T. Cell death at the intestinal epithelial front line. FEBS J 2015; 283:2701-19. [PMID: 26499289 DOI: 10.1111/febs.13575] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/23/2015] [Accepted: 10/21/2015] [Indexed: 12/25/2022]
Abstract
The intestinal epithelium represents the largest epithelial surface in our body. This single-cell-layer epithelium mediates important functions in the absorption of nutrients and in the maintenance of barrier function, preventing luminal microorganisms from invading the body. Due to its constant regeneration the intestinal epithelium is a tissue not only with very high proliferation rates but also with very prominent physiological and pathophysiological cell death induction. The normal physiological differentiation and maturation of intestinal epithelial cells leads to their shedding and apoptotic cell death within a few days, without disturbing the epithelial barrier integrity. In contrast excessive intestinal epithelial cell death induced by irradiation, drugs and inflammation severely impairs the vital functions of this tissue. In this review we discuss cell death processes in the intestinal epithelium in health and disease, with special emphasis on cell death triggered by the tumour necrosis factor receptor family.
Collapse
Affiliation(s)
- Maria Eugenia Delgado
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Germany
| | - Thomas Grabinger
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Germany
| | - Thomas Brunner
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Germany
| |
Collapse
|
212
|
Dhingra R, Lin J, Kirshenbaum LA. Disruption of RIP1-FADD Complexes by MicroRNA-103/107 Provokes Necrotic Cardiac Cell Death. Circ Res 2015; 117:314-6. [PMID: 26227876 DOI: 10.1161/circresaha.115.307023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Rimpy Dhingra
- From the Department of Physiology and Pathophysiology, Faculty of Health Sciences, Institute of Cardiovascular Sciences, College of Medicine, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Junjun Lin
- From the Department of Physiology and Pathophysiology, Faculty of Health Sciences, Institute of Cardiovascular Sciences, College of Medicine, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lorrie A Kirshenbaum
- From the Department of Physiology and Pathophysiology, Faculty of Health Sciences, Institute of Cardiovascular Sciences, College of Medicine, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
213
|
Ko AR, Hyun HW, Min SJ, Kim JE, Kang TC. Endothelin-1 induces LIMK2-mediated programmed necrotic neuronal death independent of NOS activity. Mol Brain 2015; 8:58. [PMID: 26438559 PMCID: PMC4595180 DOI: 10.1186/s13041-015-0149-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/18/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Recently, we have reported that LIM kinase 2 (LIMK2) involves programmed necrotic neuronal deaths induced by aberrant cyclin D1 expression following status epilepticus (SE). Up-regulation of LIMK2 expression induces neuronal necrosis by impairment of dynamin-related protein 1 (DRP1)-mediated mitochondrial fission. However, we could not elucidate the upstream effecter for LIMK2-mediated neuronal death. Thus, we investigated the role of endothelin-1 (ET-1) in LIMK2-mediated neuronal necrosis, since ET-1 involves neuronal death via various pathways. RESULTS Following SE, ET-1 concentration and its mRNA were significantly increased in the hippocampus with up-regulation of ETB receptor expression. BQ788 (an ETB receptor antagonist) effectively attenuated SE-induced neuronal damage as well as reduction in LIMK2 mRNA/protein expression. In addition, BQ788 alleviated up-regulation of Rho kinase 1 (ROCK1) expression and impairment of DRP1-mediated mitochondrial fission in CA1 neurons following SE. BQ788 also attenuated neuronal death and up-regulation of LIMK2 expression induced by exogenous ET-1 injection. CONCLUSION These findings suggest that ET-1 may be one of the upstream effectors for programmed neuronal necrosis through abnormal LIMK2 over-expression by ROCK1.
Collapse
Affiliation(s)
- Ah-Reum Ko
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Hye-Won Hyun
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Su-Ji Min
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Ji-Eun Kim
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea.
| | - Tae-Cheon Kang
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea.
| |
Collapse
|
214
|
Apoptosis, Necrosis, and Necroptosis in the Gut and Intestinal Homeostasis. Mediators Inflamm 2015; 2015:250762. [PMID: 26483605 PMCID: PMC4592906 DOI: 10.1155/2015/250762] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) form a physiochemical barrier that separates the intestinal lumen from the host's internal milieu and is critical for electrolyte passage, nutrient absorption, and interaction with commensal microbiota. Moreover, IECs are strongly involved in the intestinal mucosal inflammatory response as well as in mucosal innate and adaptive immune responses. Cell death in the intestinal barrier is finely controlled, since alterations may lead to severe disorders, including inflammatory diseases. The emerging picture indicates that intestinal epithelial cell death is strictly related to the maintenance of tissue homeostasis. This review is focused on previous reports on different forms of cell death in intestinal epithelium.
Collapse
|
215
|
Amaral EP, Lasunskaia EB, D'Império-Lima MR. Innate immunity in tuberculosis: how the sensing of mycobacteria and tissue damage modulates macrophage death. Microbes Infect 2015; 18:11-20. [PMID: 26369715 DOI: 10.1016/j.micinf.2015.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
The success of Mycobacterium tuberculosis as a human pathogen has been attributed to the ability of the bacillus to proliferate inside macrophages and to induce cell death. This review describes how the sensors of the innate immune system modulate the cell death pathways in infected macrophages and, consequently, the pathogenesis of tuberculosis.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Elena B Lasunskaia
- Laboratory of Recognition Biology, Center of Biosciences and Biotechnology, State University of North Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | |
Collapse
|
216
|
TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 2015; 302:2-22. [DOI: 10.1016/j.neuroscience.2015.06.038] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/17/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
|
217
|
Pastor JC, Rojas J, Pastor-Idoate S, Di Lauro S, Gonzalez-Buendia L, Delgado-Tirado S. Proliferative vitreoretinopathy: A new concept of disease pathogenesis and practical consequences. Prog Retin Eye Res 2015. [PMID: 26209346 DOI: 10.1016/j.preteyeres.2015.07.005] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During the last four decades, proliferative vitreoretinopathy (PVR) has defied the efforts of many researchers to prevent its occurrence or development. Thus, PVR is still the major complication following retinal detachment (RD) surgery and a bottle-neck for advances in cell therapy that require intraocular surgery. In this review we tried to combine basic and clinical knowledge, as an example of translational research, providing new and practical information for clinicians. PVR was defined as the proliferation of cells after RD. This idea was used for classifying PVR and also for designing experimental models used for testing many drugs, none of which were successful in humans. We summarize current information regarding the pathogenic events that follow any RD because this information may be the key for understanding and treating the earliest stages of PVR. A major focus is made on the intraretinal changes derived mainly from retinal glial cell reactivity. These responses can lead to intraretinal PVR, an entity that has not been clearly recognized. Inflammation is one of the major components of PVR, and we describe new genetic biomarkers that have the potential to predict its development. New treatment approaches are analyzed, especially those directed towards neuroprotection, which can also be useful for preventing visual loss after any RD. We also summarize the results of different surgical techniques and clinical information that is oriented toward the identification of high risk patients. Finally, we provide some recommendations for future classification of PVR and for designing comparable protocols for testing new drugs or techniques.
Collapse
Affiliation(s)
- J Carlos Pastor
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain.
| | - Jimena Rojas
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Universitario Austral, Universidad Austral, Buenos Aires, Argentina
| | - Salvador Pastor-Idoate
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Manchester Royal Eye Hospital, Manchester Vision Regeneration (MVR) Lab at NIHR/Wellcome Trust, Manchester, United Kingdom
| | - Salvatore Di Lauro
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| | - Lucia Gonzalez-Buendia
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| | - Santiago Delgado-Tirado
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| |
Collapse
|
218
|
Yashin DV, Ivanova OK, Soshnikova NV, Sheludchenkov AA, Romanova EA, Dukhanina EA, Tonevitsky AG, Gnuchev NV, Gabibov AG, Georgiev GP, Sashchenko LP. Tag7 (PGLYRP1) in Complex with Hsp70 Induces Alternative Cytotoxic Processes in Tumor Cells via TNFR1 Receptor. J Biol Chem 2015; 290:21724-31. [PMID: 26183779 DOI: 10.1074/jbc.m115.639732] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Indexed: 12/11/2022] Open
Abstract
Tag7 (also known as peptidoglycan recognition protein PGRP-S, PGLYRP1), an innate immunity protein, interacts with Hsp70 to form a stable Tag7-Hsp70 complex with cytotoxic activity against some tumor cell lines. In this study, we have analyzed the programmed cell death mechanisms that are induced when cells interact with the Tag7-Hsp70 complex, which was previously shown to be released by human lymphocytes and is cytotoxic to cancer cells. We show that this complex induces both apoptotic and necroptotic processes in the cells. Apoptosis follows the classic caspase-8 and caspase-3 activation pathway. Inhibition of apoptosis leads to a switch to the RIP1-dependent necroptosis. Both of these cytotoxic processes are initiated by the involvement of TNFR1, a receptor for TNF-α. Our results suggest that the Tag7-Hsp70 complex is a novel ligand for this receptor. One of its components, the innate immunity protein Tag7, can bind to the TNFR1 receptor, thereby inhibiting the cytotoxic actions of the Tag7-Hsp70 complex and TNF-α, an acquired immunity cytokine.
Collapse
Affiliation(s)
- Denis V Yashin
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Olga K Ivanova
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Natalia V Soshnikova
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Anton A Sheludchenkov
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Elena A Romanova
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Elena A Dukhanina
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334, the Engelhardt Institute of Molecular Biology, RAS, Moscow 119334
| | - Alexander G Tonevitsky
- the P.A. Hertsen Moscow Research Oncology Institute, 2nd Botkinskii p. 3, Moscow 125284, and
| | - Nikolai V Gnuchev
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Alexander G Gabibov
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334, the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Mikluho-Maklaya 16/10, Moscow 117997, Russia
| | - Georgii P Georgiev
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334
| | - Lidia P Sashchenko
- From the Institute of Gene Biology, Russian Academy of Sciences (RAS), Vavilova 34/5, Moscow 119334,
| |
Collapse
|
219
|
Koo MJ, Rooney KT, Choi ME, Ryter SW, Choi AMK, Moon JS. Impaired oxidative phosphorylation regulates necroptosis in human lung epithelial cells. Biochem Biophys Res Commun 2015; 464:875-80. [PMID: 26187663 DOI: 10.1016/j.bbrc.2015.07.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/09/2015] [Indexed: 12/25/2022]
Abstract
Cellular metabolism can impact cell life or death outcomes. While metabolic dysfunction has been linked to cell death, the mechanisms by which metabolic dysfunction regulates the cell death mode called necroptosis remain unclear. Our study demonstrates that mitochondrial oxidative phosphorylation (OXPHOS) activates programmed necrotic cell death (necroptosis) in human lung epithelial cells. Inhibition of mitochondrial respiration and ATP synthesis induced the phosphorylation of mixed lineage kinase domain-like protein (MLKL) and necroptotic cell death. Furthermore, we demonstrate that the activation of AMP-activated protein kinase (AMPK), resulting from impaired mitochondrial OXPHOS, regulates necroptotic cell death. These results suggest that impaired mitochondrial OXPHOS contributes to necroptosis in human lung epithelial cells.
Collapse
Affiliation(s)
- Michael Jakun Koo
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristen T Rooney
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mary E Choi
- Division of Nephrology & Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Stefan W Ryter
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jong-Seok Moon
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
220
|
Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell 2015; 59:298-308. [PMID: 26166707 DOI: 10.1016/j.molcel.2015.06.011] [Citation(s) in RCA: 1462] [Impact Index Per Article: 146.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/30/2015] [Accepted: 06/04/2015] [Indexed: 02/07/2023]
Abstract
Ferroptosis has emerged as a new form of regulated necrosis that is implicated in various human diseases. However, the mechanisms of ferroptosis are not well defined. This study reports the discovery of multiple molecular components of ferroptosis and its intimate interplay with cellular metabolism and redox machinery. Nutrient starvation often leads to sporadic apoptosis. Strikingly, we found that upon deprivation of amino acids, a more rapid and potent necrosis process can be induced in a serum-dependent manner, which was subsequently determined to be ferroptosis. Two serum factors, the iron-carrier protein transferrin and amino acid glutamine, were identified as the inducers of ferroptosis. We further found that the cell surface transferrin receptor and the glutamine-fueled intracellular metabolic pathway, glutaminolysis, played crucial roles in the death process. Inhibition of glutaminolysis, the essential component of ferroptosis, can reduce heart injury triggered by ischemia/reperfusion, suggesting a potential therapeutic approach for treating related diseases.
Collapse
Affiliation(s)
- Minghui Gao
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Prashant Monian
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Nosirudeen Quadri
- Department of Medicine, Langone Medical Center, New York University, 522 1(st) Avenue, New York, NY 10016, USA
| | - Ravichandran Ramasamy
- Department of Medicine, Langone Medical Center, New York University, 522 1(st) Avenue, New York, NY 10016, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
221
|
Lai XH, Xu Y, Chen XM, Ren Y. Macrophage cell death upon intracellular bacterial infection. ACTA ACUST UNITED AC 2015; 2:e779. [PMID: 26690967 DOI: 10.14800/macrophage.779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophage-pathogen interaction is a complex process and the outcome of this tag-of-war for both sides is to live or die. Without attempting to be comprehensive, this review will discuss the complexity and significance of the interaction outcomes between macrophages and some facultative intracellular bacterial pathogens as exemplified by Francisella, Salmonella, Shigella and Yersinia. Upon bacterial infection, macrophages can die by a variety of ways, such as apoptosis, autophagic cell death, necrosis, necroptosis, oncosis, pyronecrosis, pyroptosis etc, which is the focus of this review.
Collapse
Affiliation(s)
- Xin-He Lai
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunsheng Xu
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Dermato-venerology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Ming Chen
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Pediatric Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Ren
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA)
| |
Collapse
|
222
|
Chronic Treatment with a Water-Soluble Extract from the Culture Medium of Ganoderma lucidum Mycelia Prevents Apoptosis and Necroptosis in Hypoxia/Ischemia-Induced Injury of Type 2 Diabetic Mouse Brain. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:865986. [PMID: 25945116 PMCID: PMC4402482 DOI: 10.1155/2015/865986] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 02/03/2015] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus has been known to increase systemic oxidative stress by chronic hyperglycemia and visceral obesity and aggravate cerebral ischemic injury. On the basis of our previous study regarding a water-soluble extract from the culture medium of Ganoderma lucidum mycelia (designed as MAK), which exerts antioxidative and neuroprotective effects, the present study was conducted to evaluate the preventive effects of MAK on apoptosis and necroptosis (a programmed necrosis) induced by hypoxia/ischemia (H/I) in type 2 diabetic KKAy mice. H/I was induced by a combination of unilateral common carotid artery ligation with hypoxia (8% O2 for 20 min) and subsequent reoxygenation. Pretreatment with MAK (1 g/kg, p.o.) for a week significantly reduced H/I-induced neurological deficits and brain infarction volume assessed at 24 h of reoxygenation. Histochemical analysis showed that MAK significantly suppressed superoxide production, neuronal cell death, and vacuolation in the ischemic penumbra, which was accompanied by a decrease in the numbers of TUNEL- or cleaved caspase-3-positive cells. Furthermore, MAK decreased the expression of receptor-interacting protein kinase 3 mRNA and protein, a key molecule for necroptosis. These results suggest that MAK confers resistance to apoptotic and necroptotic cell death and relieves H/I-induced cerebral ischemic injury in type 2 diabetic mice.
Collapse
|
223
|
Omoto S, Guo H, Talekar GR, Roback L, Kaiser WJ, Mocarski ES. Suppression of RIP3-dependent necroptosis by human cytomegalovirus. J Biol Chem 2015; 290:11635-48. [PMID: 25778401 DOI: 10.1074/jbc.m115.646042] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Indexed: 12/23/2022] Open
Abstract
Necroptosis is an alternate programmed cell death pathway that is unleashed by caspase-8 compromise and mediated by receptor-interacting protein kinase 3 (RIP3). Murine cytomegalovirus (CMV) and herpes simplex virus (HSV) encode caspase-8 inhibitors that prevent apoptosis together with competitors of RIP homotypic interaction motif (RHIM)-dependent signal transduction to interrupt the necroptosis. Here, we show that pro-necrotic murine CMV M45 mutant virus drives virus-induced necroptosis during nonproductive infection of RIP3-expressing human fibroblasts, whereas WT virus does not. Thus, M45-encoded RHIM competitor, viral inhibitor of RIP activation, sustains viability of human cells like it is known to function in infected mouse cells. Importantly, human CMV is shown to block necroptosis induced by either TNF or M45 mutant murine CMV in RIP3-expressing human cells. Human CMV blocks TNF-induced necroptosis after RIP3 activation and phosphorylation of the mixed lineage kinase domain-like (MLKL) pseudokinase. An early, IE1-regulated viral gene product acts on a necroptosis step that follows MLKL phosphorylation prior to membrane leakage. This suppression strategy is distinct from RHIM signaling competition by murine CMV or HSV and interrupts an execution process that has not yet been fully elaborated.
Collapse
Affiliation(s)
- Shinya Omoto
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Hongyan Guo
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Ganesh R Talekar
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Linda Roback
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - William J Kaiser
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Edward S Mocarski
- From the Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
224
|
Oncogenic human T-cell lymphotropic virus type 1 tax suppression of primary innate immune signaling pathways. J Virol 2015; 89:4880-93. [PMID: 25694597 DOI: 10.1128/jvi.02493-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/06/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human T-cell lymphotropic virus type I (HTLV-1) is an oncogenic retrovirus considered to be the etiological agent of adult T-cell leukemia (ATL). The viral transactivator Tax is regarded as the oncoprotein responsible for contributing toward the transformation process. Here, we demonstrate that Tax potently inhibits the activity of DEx(D/H) box helicases RIG-I and MDA5 as well as Toll-dependent TIR-domain-containing adapter-inducing interferon-β (TRIF), which function as cellular sensors or mediators of viral RNA and facilitate innate immune responses, including the production of type I IFN. Tax manifested this function by binding to the RIP homotypic interaction motif (RHIM) domains of TRIF and RIP1 to disrupt interferon regulatory factor 7 (IRF7) activity, a critical type I IFN transcription factor. These data provide further mechanistic insight into HTLV-1-mediated subversion of cellular host defense responses, which may help explain HTLV-1-related pathogenesis and oncogenesis. IMPORTANCE It is predicted that up to 15% of all human cancers may involve virus infection. For example, human T-cell lymphotropic virus type 1 (HTLV-1) has been reported to infect up to 25 million people worldwide and is the causative agent of adult T-cell leukemia (ATL). We show here that HTLV-1 may be able to successfully infect the T cells and remain latent due to the virally encoded product Tax inhibiting a key host defense pathway. Understanding the mechanisms by which Tax subverts the immune system may lead to the development of a therapeutic treatment for HTLV-1-mediated disease.
Collapse
|
225
|
Bollino D, Balan I, Aurelian L. Valproic acid induces neuronal cell death through a novel calpain-dependent necroptosis pathway. J Neurochem 2015; 133:174-86. [PMID: 25581256 DOI: 10.1111/jnc.13029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/24/2014] [Accepted: 01/05/2015] [Indexed: 02/06/2023]
Abstract
A growing body of evidence indicates that valproic acid (VPA), a histone deacetylase inhibitor used to treat epilepsy and mood disorders, has histone deacetylase-related and -unrelated neurotoxic activity, the mechanism of which is still poorly understood. We report that VPA induces neuronal cell death through an atypical calpain-dependent necroptosis pathway that initiates with downstream activation of c-Jun N-terminal kinase 1 (JNK1) and increased expression of receptor-interacting protein 1 (RIP-1) and is accompanied by cleavage and mitochondrial release/nuclear translocation of apoptosis-inducing factor, mitochondrial release of Smac/DIABLO, and inhibition of the anti-apoptotic protein X-linked inhibitor of apoptosis (XIAP). Coinciding with apoptosis-inducing factor nuclear translocation, VPA induces phosphorylation of the necroptosis-associated histone H2A family member H2AX, which is known to contribute to lethal DNA degradation. These signals are inhibited in neuronal cells that express constitutively activated MEK/ERK and/or PI3-K/Akt survival pathways, allowing them to resist VPA-induced cell death. The data indicate that VPA has neurotoxic activity and identify a novel calpain-dependent necroptosis pathway that includes JNK1 activation and RIP-1 expression. A growing body of evidence indicates that valproic acid (VPA) has neurotoxic activity, the mechanism of which is still poorly understood. We report, for the first time, that VPA activates a previously unrecognized calpain-dependent necroptosis cascade that initiates with JNK1 activation and involves AIF cleavage/nuclear translocation and H2AX phosphorylation as well as an altered Smac/DIABLO to XIAP balance.
Collapse
Affiliation(s)
- Dominique Bollino
- Department of Pharmacology, University of Maryland, Baltimore, Maryland, USA
| | | | | |
Collapse
|
226
|
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015; 517:311-20. [PMID: 25592536 DOI: 10.1038/nature14191] [Citation(s) in RCA: 1578] [Impact Index Per Article: 157.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023]
Abstract
Regulated cell death has essential functions in development and in adult tissue homeostasis. Necroptosis is a newly discovered pathway of regulated necrosis that requires the proteins RIPK3 and MLKL and is induced by death receptors, interferons, toll-like receptors, intracellular RNA and DNA sensors, and probably other mediators. RIPK1 has important kinase-dependent and scaffolding functions that inhibit or trigger necroptosis and apoptosis. Mouse-model studies have revealed important functions for necroptosis in inflammation and suggested that it could be implicated in the pathogenesis of many human inflammatory diseases. We discuss the mechanisms regulating necroptosis and its potential role in inflammation and disease.
Collapse
Affiliation(s)
- Manolis Pasparakis
- Institute for Genetics, Centre for Molecular Medicine and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany
| | - Peter Vandenabeele
- 1] VIB Inflammation Research Center, Ghent University, UGhent-VIB Research Building FSVM, 9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium. [3] Methusalem program, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| |
Collapse
|
227
|
|
228
|
|
229
|
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 having 1479=1479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
230
|
|
231
|
|
232
|
|
233
|
|
234
|
|
235
|
|
236
|
|
237
|
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 order by 1-- ocnp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
238
|
|
239
|
|
240
|
|
241
|
Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 and 2810=2810-- wbae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
242
|
Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 and make_set(6705=6705,9963)-- tutl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
243
|
|
244
|
|
245
|
|
246
|
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 having 6610=1325-- ftul] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
247
|
Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 order by 1-- qnpz] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
248
|
|
249
|
Necroptosis and its role in inflammation. Nature 2015. [DOI: 10.1038/nature14191 and 9718=9916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
250
|
|