1
|
Lv Q, Han X, Ni J, Ma Q, Dai R, Liu J, Liu J, Zhai Y, Shen Q, Sun L, Liu H, Rao J, Xu H. Anti-ANGPTL3-FLD monoclonal antibody treatment ameliorates podocyte lesions through attenuating mitochondrial damage. Cell Death Dis 2022; 13:867. [PMID: 36229446 PMCID: PMC9562403 DOI: 10.1038/s41419-022-05313-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Proteinuria, an indication of kidney disease, is caused by the malfunction of podocytes, which play a key role in maintaining glomerular filtration. Angiopoietin-like 3 (ANGPTL3) has been documented to have a cell-autonomous involvement in podocytes, and deletion of Angptl3 in podocytes reduced proteinuria in adriamycin-induced nephropathy. Here, we developed a monoclonal antibody (mAb) against ANGPTL3 to investigate its effects on podocyte injury in an ADR nephropathy mouse model and puromycin (PAN) induced podocyte damage in vitro. The mAb against the human ANGPTL3-FLD sequence (5E5F6) inhibited the binding of ANGPTL3-FLD to integrin β3. Treatment with the 5E5F6 mAb in ADR nephropathy mice mitigated proteinuria and led to a significant decline in podocyte apoptosis, reactive oxygen species (ROS) generation and mitochondrial fragmentation. In PAN-induced podocyte damage in vitro, the 5E5F6 mAb blocked the ANPGPLT3-mediated activation of integrin αvβ3 and Rac1, which regulated the mitochondrial homeostasis. Altogether, anti-ANGPLT3-FLD mAb attenuates proteinuria and podocyte lesions in ADR mice models, as well as PAN-induced podocyte damage, in part through regulating mitochondrial functions. Our study provides a therapeutic approach for targeting ANGPTL3 in proteinuric kidney disease.
Collapse
Affiliation(s)
- Qianying Lv
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Xinli Han
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Jiajia Ni
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Qianqian Ma
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Rufeng Dai
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Jiaojiao Liu
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Jialu Liu
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Yihui Zhai
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Qian Shen
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Li Sun
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China ,grid.411333.70000 0004 0407 2968Department of Rheumatology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China
| | - Haimei Liu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China ,grid.411333.70000 0004 0407 2968Department of Rheumatology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China
| | - Jia Rao
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Hong Xu
- grid.411333.70000 0004 0407 2968Department of Nephrology, Children’s Hospital of Fudan University, National Pediatric Medical Center of CHINA, Shanghai, China ,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| |
Collapse
|
2
|
Cao Y, Shen M, Jiang Y, Sun SC, Liu H. Melatonin reduces oxidative damage in mouse granulosa cells via restraining JNK-dependent autophagy. Reproduction 2018; 155:307-319. [DOI: 10.1530/rep-18-0002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 01/23/2018] [Indexed: 01/02/2023]
Abstract
Oxidative stress-induced granulosa cell (GCs) injury is believed to be a common trigger for follicular atresia. Emerging evidence indicates that excessive autophagy occurs in mammalian cells with oxidative damage. N-acetyl-5-methoxytrypamine (melatonin) has been shown to prevent GCs from oxidative injury, although the exact mechanism remains to be elucidated. Here, we first demonstrated that the suppression of autophagy through the JNK/BCL-2/BECN1 signaling is engaged in melatonin-mediated GCs protection against oxidative damage. Melatonin inhibited the loss of GCs viability, formation of GFP-MAP1LC3B puncta, accumulation of MAP1LC3B-II blots, degradation of SQSTM1 and the expression of BECN1, which was correlated with impaired activation of JNK during oxidative stress. On the other hand, blocking of autophagy and/or JNK also reduced the level of H2O2-induced GCs death, but failed to further restore GCs viability in the presence of melatonin. Particularly, the suppression of autophagy provided no additional protective effects when GCs were pretreated with JNK inhibitor and/or melatonin. Importantly, we found that the enhanced interaction between BCL-2 and BECN1 might be a responsive mechanism for autophagy suppression via the melatonin/JNK pathway. Moreover, blocking the downstream antioxidant system of melatonin using specific inhibitors further confirmed a direct role of melatonin/JNK/autophagy axis in preserving GCs survival without scavenging reactive oxygen species (ROS). Taken together, our findings uncover a novel function of melatonin in preventing GCs from oxidative damage by targeting JNK-mediated autophagy, which might contribute to develop therapeutic strategies for patients with ovulation failure-related disorders.
Collapse
|
3
|
Huang H, Du W, Brekken RA. Extracellular Matrix Induction of Intracellular Reactive Oxygen Species. Antioxid Redox Signal 2017; 27:774-784. [PMID: 28791881 DOI: 10.1089/ars.2017.7305] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is the noncellular component secreted by cells and is present within all tissues and organs. The ECM provides the structural support required for tissue integrity and also contributes to diseases, including cancer. Many diseases rich in ECM are characterized by changes in reactive oxygen species (ROS) levels that have been shown to have important context-dependent functions. Recent Advances: Many studies have found that the ECM affects ROS production through integrins. The activation of integrins by ECM ligands results in stimulation of multiple pathways that can generate ROS. Furthermore, control of ECM-integrin interaction by matricellular proteins is an underappreciated pathway that functions as an ROS rheostat in remodeling tissues. CRITICAL ISSUES A better understanding of how the ECM affects the generation of intracellular ROS is required for advances in the development of therapeutic strategies that affect or exploit oxidative stress. FUTURE DIRECTIONS Targeting ROS generation can be therapeutic or can promote disease progression in a context-dependent manner. Many ECM proteins can impact ROS generation. However, given the breadth of different proteins that constitute the ECM and the cell surface receptors that interact with ECM proteins, there are likely many tissue and microenvironmental-specific ROS-generating pathways that have yet to be investigated in depth. Identifying canonical pathways of ECM-induced ROS generation should be a priority for the field. Antioxid. Redox Signal. 27, 774-784.
Collapse
Affiliation(s)
- Huocong Huang
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas
| | - Wenting Du
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas
| | - Rolf A Brekken
- 1 Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research , Dallas, Texas.,2 Department of Pharmacology, UT Southwestern, Dallas, Texas
| |
Collapse
|
4
|
Haga S, Kanno A, Ozawa T, Morita N, Asano M, Ozaki M. Detection of Necroptosis in Ligand-Mediated and Hypoxia-Induced Injury of Hepatocytes Using a Novel Optic Probe-Detecting Receptor-Interacting Protein (RIP)1/RIP3 Binding. Oncol Res 2017; 26:503-513. [PMID: 28770700 PMCID: PMC7844641 DOI: 10.3727/096504017x15005102445191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Liver injury is often observed in various pathological conditions including posthepatectomy state and cancer chemotherapy. It occurs mainly as a consequence of the combined necrotic and apoptotic types of cell death. In order to study liver/hepatocyte injury by the necrotic type of cell death, we studied signal-regulated necrosis (necroptosis) by developing a new optic probe for detecting receptor-interacting protein kinase 1 (RIP)/RIP3 binding, an essential process for necroptosis induction. In the mouse hepatocyte cell line, TIB-73 cells, TNF-α/cycloheximide (T/C) induced RIP1/3 binding only when caspase activity was suppressed by the caspase-specific inhibitor z-VAD-fmk (zVAD). T/C/zVAD-induced RIP1/3 binding was inhibited by necrostatin-1 (Nec-1), an allosteric inhibitor of RIP1. The reduced cell survival by T/C/zVAD was improved by Nec-1. These facts indicate that T/C induces necroptosis of hepatocytes when the apoptotic pathway is inhibited/unavailable. FasL also induced cell death, which was only partially inhibited by zVAD, indicating the possible involvement of necroptosis rather than apoptosis. FasL activated caspase 3 and, similarly, induced RIP1/3 binding when the caspases were inactivated. Interestingly, FasL-induced RIP1/3 binding was significantly suppressed by the antioxidants Trolox and N-acetyl cysteine (NAC), suggesting the involvement of reactive oxygen species (ROS) in FasL-induced necroptotic cellular processes. H₂O₂, by itself, induced RIP1/3 binding that was suppressed by Nec-1, but not by zVAD. Hypoxia induced RIP1/3 binding after reoxygenation, which was suppressed by Nec-1 or by the antioxidants. Cell death induced by hypoxia/reoxygenation (H/R) was also improved by Nec-1. Similar to H₂O₂, H/R did not require caspase inhibition for RIP1/3 binding, suggesting the involvement of a caspase-independent mechanism for non-ligand-induced and/or redox-mediated necroptosis. These data indicate that ROS can induce necroptosis and mediate the FasL- and hypoxia-induced necroptosis via a molecular mechanism that differs from a conventional caspase-dependent pathway. In conclusion, necroptosis is potentially involved in liver/hepatocyte injury induced by oxidative stress and FasL in the absence of apoptosis.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Akira Kanno
- Department of Environmental Applied Chemistry, Faculty of Engineering, University of ToyamaToyamaJapan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of TokyoTokyoJapan
| | - Naoki Morita
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)Sapporo, HokkaidoJapan
| | - Mami Asano
- Laboratory of Molecular and Functional Bio-Imaging, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| |
Collapse
|
5
|
Dimauro I, Mercatelli N, Caporossi D. Exercise-induced ROS in heat shock proteins response. Free Radic Biol Med 2016; 98:46-55. [PMID: 27021964 DOI: 10.1016/j.freeradbiomed.2016.03.028] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/18/2016] [Accepted: 03/24/2016] [Indexed: 11/26/2022]
Abstract
Cells have evolved multiple and sophisticated stress response mechanisms aiming to prevent macromolecular (including proteins, lipids, and nucleic acids) damage and to maintain or re-establish cellular homeostasis. Heat shock proteins (HSPs) are among the most highly conserved, ubiquitous, and abundant proteins in all organisms. Originally discovered more than 50 years ago through heat shock stress, they display multiple, remarkable roles inside and outside cells under a variety of stresses, including also oxidative stress and radiation, recognizing unfolded or misfolded proteins and facilitating their restructuring. Exercise consists in a combination of physiological stresses, such as metabolic disturbances, changes in circulating levels of hormones, increased temperature, induction of mild to severe inflammatory state, increased production of reactive oxygen and nitrogen species (ROS and RNS). As a consequence, exercise is one of the main stimuli associated with a robust increase in different HSPs in several tissues, which appears to be also fundamental in facilitating the cellular remodeling processes related to the training regime. Among all factors involved in the exercise-related modulation of HSPs level, the ROS production in the contracting muscle or in other tissues represents one of the most attracting, but still under discussion, mechanism. Following exhaustive or damaging muscle exercise, major oxidative damage to proteins and lipids is likely involved in HSP expression, together with mechanically induced damage to muscle proteins and the inflammatory response occurring several days into the recovery period. Instead, the transient and reversible oxidation of proteins by physiological concentrations of ROS seems to be involved in the activation of stress response following non-damaging muscle exercise. This review aims to provide a critical update on the role of HSPs response in exercise-induced adaptation or damage in humans, focusing on experimental results where the link between redox homeostasis and HSPs expression by exercise has been addressed. Further, with the support of in vivo and in vitro studies, we discuss the putative molecular mechanisms underlying the ROS-mediated modulation of HSP expression and/or activity during exercise.
Collapse
Affiliation(s)
- Ivan Dimauro
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy
| | - Neri Mercatelli
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy
| | - Daniela Caporossi
- Unit of Biology, Genetics and Biochemistry, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy.
| |
Collapse
|
6
|
Koriauli S, Natsvlishvili N, Barbakadze T, Mikeladze D. Knockdown of interleukin-10 induces the redistribution of sigma1-receptor and increases the glutamate-dependent NADPH-oxidase activity in mouse brain neurons. Biol Res 2015; 48:55. [PMID: 26453192 PMCID: PMC4599652 DOI: 10.1186/s40659-015-0048-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/01/2015] [Indexed: 01/30/2023] Open
Abstract
Background In the central nervous system, interleukin-10 (IL-10) provides trophic and survival effects directly on neurons, modulates neurite plasticity, and has a pivotal importance in the neuronal regeneration in neurodegenerative and neuroinflammatory conditions. This cytokine is primarily produced by glial cells and has beneficial effects on the neuronal viability. However, the mechanisms of IL-10-elicited neuroprotection are not clear. Results Membrane preparations, isolated from wild-type (Wt) and IL-10 knockout (KO) mice brain were used in this study. It has been shown that compared to wild-type mice, in IL-10 KO mice brain, the amount of immunoglobulin binding protein (BiP) is greatly increased, whereas the content of sigma receptor-1 (SigR1) is not changed significantly. Co-immunoprecipitation experiments have shown that the association of SigR1 with small GTPase Rac1 (Ras-related C3 botulinum toxin substrate 1), NR2B subunit of NMDA-receptor (NMDAR) and inositol-3-phosphate receptor (IP3R) is higher in the IL-10 KO mice brain than in the Wt mice brain. Besides, we have found that either glutamate or sigma ligands, separately or together, do not change glutamate-induced NADPH-oxidase (NOX) activity in Wt-type mice brain membrane preparations, whereas in IL-10 KO mice high concentration of glutamate markedly increases the NOX-dependent production of reactive oxygen species (ROS). Glutamate-dependent ROS production was decreased to the normal levels by the action of sigma-agonists. Conclusions It has been concluded that IL-10 deprivation, at least in part, can lead to the induction of ER-stress, which causes BiP expression and SigR1 redistribution between components of endoplasmic reticulum (ER) and plasma membrane. Moreover, IL-10 deficiency can change the specific organization of NMDAR, increasing the surface expression of SigR1-sensitive NR2B-containing NMDAR. In these conditions, glutamate-dependent ROS production is greatly increased leading to the initiation of apoptosis. In this circumstances, sigma-ligands could play a preventive role against NMDA receptor-mediated excitotoxicity.
Collapse
Affiliation(s)
- S Koriauli
- Ilia State University, 3/5 K. Cholokashvili Ave., 0162, Tbilisi, Georgia.
| | - N Natsvlishvili
- Ilia State University, 3/5 K. Cholokashvili Ave., 0162, Tbilisi, Georgia. .,I. Beritashvili Center of Experimental Biomedicine, 14 Gotua Str., Tbilisi, Georgia.
| | - T Barbakadze
- Ilia State University, 3/5 K. Cholokashvili Ave., 0162, Tbilisi, Georgia. .,I. Beritashvili Center of Experimental Biomedicine, 14 Gotua Str., Tbilisi, Georgia.
| | - D Mikeladze
- Ilia State University, 3/5 K. Cholokashvili Ave., 0162, Tbilisi, Georgia. .,I. Beritashvili Center of Experimental Biomedicine, 14 Gotua Str., Tbilisi, Georgia.
| |
Collapse
|
7
|
Osborn-Heaford HL, Murthy S, Gu L, Larson-Casey JL, Ryan AJ, Shi L, Glogauer M, Neighbors JD, Hohl R, Carter AB. Targeting the isoprenoid pathway to abrogate progression of pulmonary fibrosis. Free Radic Biol Med 2015; 86:47-56. [PMID: 25958207 PMCID: PMC4554879 DOI: 10.1016/j.freeradbiomed.2015.04.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 04/07/2015] [Accepted: 04/27/2015] [Indexed: 11/22/2022]
Abstract
Fibrotic remodeling in lung injury is a major cause of morbidity. The mechanism that mediates the ongoing fibrosis is unclear, and there is no available treatment to abate the aberrant repair. Reactive oxygen species (ROS) have a critical role in inducing fibrosis by modulating extracellular matrix deposition. Specifically, mitochondrial hydrogen peroxide (H2O2) production by alveolar macrophages is directly linked to pulmonary fibrosis as inhibition of mitochondrial H2O2 attenuates the fibrotic response in mice. Prior studies indicate that the small GTP-binding protein, Rac1, directly mediates H2O2 generation in the mitochondrial intermembrane space. Geranylgeranylation of the C-terminal cysteine residue (Cys(189)) is required for Rac1 activation and mitochondrial import. We hypothesized that impairment of geranylgeranylation would limit mitochondrial oxidative stress and, thus, abrogate progression of pulmonary fibrosis. By targeting the isoprenoid pathway with a novel agent, digeranyl bisphosphonate (DGBP), which impairs geranylgeranylation, we demonstrate that Rac1 mitochondrial import, mitochondrial oxidative stress, and progression of the fibrotic response to lung injury are significantly attenuated. These observations reveal that targeting the isoprenoid pathway to alter Rac1 geranylgeranylation halts the progression of pulmonary fibrosis after lung injury.
Collapse
Affiliation(s)
| | | | - Linlin Gu
- Deparment of Medicine, University of Alabama at Birmingham, AL
| | - Jennifer L Larson-Casey
- Free Radical and Radiation Biology Program, University of Iowa
- Deparment of Medicine, University of Alabama at Birmingham, AL
| | - Alan J Ryan
- Department of Internal Medicine, University of Iowa
| | - Lei Shi
- Human Toxicology Program, University of Iowa
| | - Michael Glogauer
- Canadian Institutes of Health Research Group in Matrix Dynamics, University of Toronto, Toronto, Ontario, Canada
| | | | - Raymond Hohl
- Department of Internal Medicine, University of Iowa
- Department of Pharmacology, University of Iowa
| | - A Brent Carter
- Department of Internal Medicine, University of Iowa
- Free Radical and Radiation Biology Program, University of Iowa
- Human Toxicology Program, University of Iowa
- Deparment of Medicine, University of Alabama at Birmingham, AL
- Iowa City VA Healthcare System, Iowa City, IA
- Birmingham VAMC, Birmingham, AL
| |
Collapse
|
8
|
Abstract
The heat shock response (HSR) is an ancient and highly conserved process that is essential for coping with environmental stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms temporarily subject themselves to thermal stress in the face of infections. We review the phylogenetically conserved mechanisms that regulate fever and discuss the effects that febrile-range temperatures have on multiple biological processes involved in host defense and cell death and survival, including the HSR and its implications for patients with severe sepsis, trauma, and other acute systemic inflammatory states. Heat shock factor-1, a heat-induced transcriptional enhancer is not only the central regulator of the HSR but also regulates expression of pivotal cytokines and early response genes. Febrile-range temperatures exert additional immunomodulatory effects by activating mitogen-activated protein kinase cascades and accelerating apoptosis in some cell types. This results in accelerated pathogen clearance, but increased collateral tissue injury, thus the net effect of exposure to febrile range temperature depends in part on the site and nature of the pathologic process and the specific treatment provided.
Collapse
Affiliation(s)
- Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine and the Baltimore V.A. Medical Center, Baltimore, Maryland
| | | | | |
Collapse
|
9
|
Shen CH, Tung SY, Huang WS, Lu CC, Lee KC, Hsieh YY, Chang PJ, Liang HF, Chen JH, Lin TH, Hsieh MC, Kuo HC. Exploring the effects of tert-butylhydroperoxide induced liver injury using proteomic approach. Toxicology 2014; 316:61-70. [PMID: 24394546 DOI: 10.1016/j.tox.2013.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/04/2013] [Accepted: 12/22/2013] [Indexed: 01/29/2023]
Abstract
Tert-butyl hydroperoxide (t-BHP), an organic lipid hydroperoxide analog, has been demonstrated to exert pro-oxidant effects to evaluate mechanisms involving oxidative stress in hepatocyte cells and rat liver. Herein, we present an investigation of the event of molecular mechanism of t-BHP related acute liver injury. A proteomic approach was used to identify proteins which are differentially expressed in liver cells following t-BHP treatment and the mechanism of its action in apoptotic and endoplasmic reticulum stress pathways. Our results demonstrate that the t-BHP treatment of liver cells increased cell cytoxicity and apoptosis. t-BHP dose-dependent induction of cell apoptosis and stained liver sections relieved the acute rat liver injury were accompanied by sustained phosphorylation of JNK1/2 and p65. In addition, there were 13 differentially displayed proteins between the t-BHP-induced and untreated were assayed and validated in vivo. Furthermore, we demonstrated that t-BHP induced human Chang liver cell viability and apoptosis properties by up-regulating the levels of ETFA (electron transfer flavoprotein subunit alpha). This study demonstrated that there was an increase in the cellular levels of ETFA in the t-BHP induction in viability and apoptosis via the activation of JNK1/2 and NFκB signaling modules. NAC administration and shRNA ETFA conferred resistance to t-BHP-increased ETFA and CHOP expression via IRE1-alpha/TRAF2 complex formation, activation of JNK1/2 and p50. We concluded that the mechanism of t-BHP-induced an apoptosis cascade and endoplasmic reticulum stress in hepatocyte cells by up-regulation of ETFA, providing a new mechanism for liver injury.
Collapse
Affiliation(s)
- Chien-Heng Shen
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Shui-Yi Tung
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Shih Huang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Chien-Chang Lu
- Department of Colorectal Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ko-Chao Lee
- Department of Colorectal Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yung-Yu Hsieh
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Hwey-Fang Liang
- Department of Nursing, Chang Gung University of Science and Technology, Taiwan; Chronic Diseases and Health Promotion Research Center, CGUST, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Jiann-Hwa Chen
- School of Medicine, Fu-Jen Catholic University, Taipei, Taiwan; Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Emergency Medicine, Cathay General Hospital, Taipei 10630, Taiwan
| | - Tseng-Hsi Lin
- Division of Hematology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Meng Chiao Hsieh
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Hsing-Chun Kuo
- Department of Nursing, Chang Gung University of Science and Technology, Taiwan; Chronic Diseases and Health Promotion Research Center, CGUST, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
10
|
Kajita K, Kuwano Y, Kitamura N, Satake Y, Nishida K, Kurokawa K, Akaike Y, Honda M, Masuda K, Rokutan K. Ets1 and heat shock factor 1 regulate transcription of the Transformer 2β gene in human colon cancer cells. J Gastroenterol 2013; 48:1222-33. [PMID: 23361474 DOI: 10.1007/s00535-012-0745-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 12/25/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND Transformer (Tra) 2β is a member of the serine/arginine-rich (SR)-like protein family that regulates alternative splicing of numerous genes in a concentration-dependent manner. Several types of cancer cells up-regulate Tra2β expression, while the regulatory mechanism of Tra2β expression remains to be elucidated. In this study, we examined the transcriptional regulation and possible functions of Tra2β in human colon cancer cells. METHODS We cloned 959 bp-upstream of the human TRA2β 5'-flank into luciferase constructs. Chromatin immunoprecipitation (ChIP) was employed to identify crucial cis element(s) and trans activator(s) of the TRA2β promoter. Tra2β expression in the human colon and colon cancer tissues was examined by immunohistochemistry. RESULTS In response to sodium arsenite, colon cancer cells (HCT116) increased levels of TRA2β1 mRNA encoding a functional, full-length Tra2β with a peak around 6 h without changing its mRNA stability. Transient expression assays using a reporter gene driven by serially truncated TRA2β promoters and Chip assay demonstrated that an Ets1-binding site present at -64 to -55 bp was crucial for basal transcription, while three heat shock elements (HSEs) located at -145 to -99 bp mediated the oxidant-induced transactivation of TRA2β. Tra2β knockdown caused apoptosis of HCT116 cells. Tra2β were preferentially expressed in proliferative compartment of normal human colonic glands and adenocarcinomas, where Ets1 and heat shock factor 1 were also highly expressed. CONCLUSIONS Our results suggest that oxidative stress-responsive Tra2β may play an important role in colon cancer growth.
Collapse
Affiliation(s)
- Keisuke Kajita
- Department of Stress Science, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Fittipaldi S, Dimauro I, Mercatelli N, Caporossi D. Role of exercise-induced reactive oxygen species in the modulation of heat shock protein response. Free Radic Res 2013; 48:52-70. [DOI: 10.3109/10715762.2013.835047] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
12
|
Talukder MAH, Elnakish MT, Yang F, Nishijima Y, Alhaj MA, Velayutham M, Hassanain HH, Zweier JL. Cardiomyocyte-specific overexpression of an active form of Rac predisposes the heart to increased myocardial stunning and ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2012; 304:H294-302. [PMID: 23161879 DOI: 10.1152/ajpheart.00367.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The GTP-binding protein Rac regulates diverse cellular functions including activation of NADPH oxidase, a major source of superoxide production (O(2)(·-)). Rac1-mediated NADPH oxidase activation is increased after myocardial infarction (MI) and heart failure both in animals and humans; however, the impact of increased myocardial Rac on impending ischemia-reperfusion (I/R) is unknown. A novel transgenic mouse model with cardiac-specific overexpression of constitutively active mutant form of Zea maize Rac D (ZmRacD) gene has been reported with increased myocardial Rac-GTPase activity and O(2)(·-) generation. The goal of the present study was to determine signaling pathways related to increased myocardial ZmRacD and to what extent hearts with increased ZmRacD proteins are susceptible to I/R injury. The effect of myocardial I/R was examined in young adult wild-type (WT) and ZmRacD transgenic (TG) mice. In vitro reversible myocardial I/R for postischemic cardiac function and in vivo regional myocardial I/R for MI were performed. Following 20-min global ischemia and 45-min reperfusion, postischemic cardiac contractile function and heart rate were significantly reduced in TG hearts compared with WT hearts. Importantly, acute regional myocardial I/R (30-min ischemia and 24-h reperfusion) caused significantly larger MI in TG mice compared with WT mice. Western blot analysis of cardiac homogenates revealed that increased myocardial ZmRacD gene expression is associated with concomitant increased levels of NADPH oxidase subunit gp91(phox), O(2)(·-), and P(21)-activated kinase. Thus these findings provide direct evidence that increased levels of active myocardial Rac renders the heart susceptible to increased postischemic contractile dysfunction and MI following acute I/R.
Collapse
Affiliation(s)
- M A Hassan Talukder
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Wu Q, Lin J, Liu JZ, Wang X, Lim W, Oh M, Park J, Rajashekar CB, Whitham SA, Cheng NH, Hirschi KD, Park S. Ectopic expression of Arabidopsis glutaredoxin AtGRXS17 enhances thermotolerance in tomato. PLANT BIOTECHNOLOGY JOURNAL 2012; 10:945-55. [PMID: 22762155 DOI: 10.1111/j.1467-7652.2012.00723.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
While various signalling networks regulate plant responses to heat stress, the mechanisms regulating and unifying these diverse biological processes are largely unknown. Our previous studies indicate that the Arabidopsis monothiol glutaredoxin, AtGRXS17, is crucial for temperature-dependent postembryonic growth in Arabidopsis. In the present study, we further demonstrate that AtGRXS17 has conserved functions in anti-oxidative stress and thermotolerance in both yeast and plants. In yeast, AtGRXS17 co-localized with yeast ScGrx3 in the nucleus and suppressed the sensitivity of yeast grx3grx4 double-mutant cells to oxidative stress and heat shock. In plants, GFP-AtGRXS17 fusion proteins initially localized in the cytoplasm and the nuclear envelope but migrated to the nucleus during heat stress. Ectopic expression of AtGRXS17 in tomato plants minimized photo-oxidation of chlorophyll and reduced oxidative damage of cell membrane systems under heat stress. This enhanced thermotolerance correlated with increased catalase (CAT) enzyme activity and reduced H₂O₂ accumulation in AtGRXS17-expressing tomatoes. Furthermore, during heat stress, expression of the heat shock transcription factor (HSF) and heat shock protein (HSP) genes was up-regulated in AtGRXS17-expressing transgenic plants compared with wild-type controls. Thus, these findings suggest a specific protective role of a redox protein against temperature stress and provide a genetic engineering strategy to improve crop thermotolerance.
Collapse
Affiliation(s)
- Qingyu Wu
- Department of Horticulture, Forestry and Recreation Resources, Kansas State University, Manhattan, KS, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Osborn-Heaford HL, Ryan AJ, Murthy S, Racila AM, He C, Sieren JC, Spitz DR, Carter AB. Mitochondrial Rac1 GTPase import and electron transfer from cytochrome c are required for pulmonary fibrosis. J Biol Chem 2012; 287:3301-12. [PMID: 22157762 PMCID: PMC3270985 DOI: 10.1074/jbc.m111.308387] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/21/2011] [Indexed: 12/22/2022] Open
Abstract
The generation of reactive oxygen species, particularly H(2)O(2), from alveolar macrophages is causally related to the development of pulmonary fibrosis. Rac1, a small GTPase, is known to increase mitochondrial H(2)O(2) generation in macrophages; however, the mechanism by which this occurs is not known. This study shows that Rac1 is localized in the mitochondria of alveolar macrophages from asbestosis patients, and mitochondrial import requires the C-terminal cysteine of Rac1 (Cys-189), which is post-translationally modified by geranylgeranylation. Furthermore, H(2)O(2) generation mediated by mitochondrial Rac1 requires electron transfer from cytochrome c to a cysteine residue on Rac1 (Cys-178). Asbestos-exposed mice harboring a conditional deletion of Rac1 in macrophages demonstrated decreased oxidative stress and were significantly protected from developing pulmonary fibrosis. These observations demonstrate that mitochondrial import and direct electron transfer from cytochrome c to Rac1 modulates mitochondrial H(2)O(2) production in alveolar macrophages pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Chao He
- Radiation Oncology, Free Radical and Radiation Biology Program, and
| | - Jessica C. Sieren
- Department of Radiology, University of Iowa Carver College of Medicine, and
| | - Douglas R. Spitz
- Radiation Oncology, Free Radical and Radiation Biology Program, and
| | - A. Brent Carter
- From the Departments of Internal Medicine and
- Radiation Oncology, Free Radical and Radiation Biology Program, and
- Human Toxicology Program, University of Iowa College of Public Health, Iowa City, Iowa 52242
| |
Collapse
|
15
|
Overmeyer JH, Maltese WA. Death pathways triggered by activated Ras in cancer cells. Front Biosci (Landmark Ed) 2011; 16:1693-713. [PMID: 21196257 DOI: 10.2741/3814] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ras GTPases are best known for their ability to serve as molecular switches regulating cell growth, differentiation and survival. Gene mutations that result in expression of constitutively active forms of Ras have been linked to oncogenesis in animal models and humans. However, over the past two decades, evidence has gradually accumulated to support a paradoxical role for Ras proteins in the initiation of cell death pathways. In this review we survey the literature pointing to the ability of activated Ras to promote cell death under conditions where cancer cells encounter apoptotic stimuli or Ras is ectopically expressed. In some of these cases Ras acts through known effectors and well defined apoptotic death pathways. However, in other cases it appears that Ras operates by triggering novel non-apoptotic death mechanisms that are just beginning to be characterized. Understanding these mechanisms and the factors that go into changing the nature of Ras signaling from pro-survival to pro-death could set the stage for development of novel therapeutic approaches aimed at manipulating pro-death Ras signaling pathways in cancer.
Collapse
Affiliation(s)
- Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine, Toledo, Ohio 43614, USA
| | | |
Collapse
|
16
|
Simão MF, Leite RB, Rocha C, Cancela ML. Changes in bioturbation of iron biogeochemistry and in molecular response of the clam Ruditapes decussates upon Perkinsus olseni infection. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2010; 59:433-443. [PMID: 20232199 DOI: 10.1007/s00244-010-9490-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 02/16/2010] [Indexed: 05/28/2023]
Abstract
A series of artificial microcosms was used to test the effect of clam density on benthic iron biogeochemistry and, subsequently, if the response of clam Ruditapes decussatus to infection with Perkinsus olseni, a common opportunistic parasite known to be iron dependent, was correlated with the dynamics of iron sediment pore waters within the chambers. Three series of benthic microcosms were used in the experiment, comparing similar densities of clams (none, one, two, three, or four individuals/chamber) between a control set (no deliberate infection) and two parallel sets of clams that were deliberately infected with the parasite after 10 days of incubation. Fifteen chambers were used simultaneously and the experiment was conducted for 35 days. In order to avoid spurious effects of differential organic loading and clam feeding efficiency on the oxidative state of the sediment, the iron balance was tentatively shifted during incubation toward decreased dissolved iron in pore water. This was done by applying a constant flow of air to all chambers and refraining from supplying extra organic matter during the experimental run, which led to the reduction of benthic oxygen demand as the experiment progressed. Results showed that microcosms bearing both higher clam densities and lower infection levels were able to exert a quantitative influence in iron biogeochemistry through bioturbation activity. This effect was significantly depressed in chambers hosting clams with high infection levels. In addition, analysis of molecular markers responsive to iron and parasite stress revealed an upper regulation of HSP70 and ferritin in infected clams, thus suggesting a role of those molecules on both host protection and response to parasite presence by limiting iron availability. Together, these findings suggest a correlation between the expression of clam molecular iron/stress markers and iron bioavailability, which can be modified by the presence or absence of Perkinsus infection. In turn, we propose that clam lethargy in response to parasite invasion might help to combat infection by reducing iron mobilization in the surrounding sediment through a decrease in bioturbation activity, thus reducing its availability to the parasite.
Collapse
|
17
|
Chudinova EM, Nadezhdina ES, Ivanov PA. Is the microtubule disruption-induced alteration of peroxide concentration a factor inhibiting the assembly of ribonucleoprotein stress granules? Biophysics (Nagoya-shi) 2010. [DOI: 10.1134/s000635091005012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
18
|
Chen M, Gu H, Ye Y, Lin B, Sun L, Deng W, Zhang J, Liu J. Protective effects of hesperidin against oxidative stress of tert-butyl hydroperoxide in human hepatocytes. Food Chem Toxicol 2010; 48:2980-7. [DOI: 10.1016/j.fct.2010.07.037] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 07/19/2010] [Accepted: 07/23/2010] [Indexed: 01/30/2023]
|
19
|
Divi UK, Rahman T, Krishna P. Brassinosteroid-mediated stress tolerance in Arabidopsis shows interactions with abscisic acid, ethylene and salicylic acid pathways. BMC PLANT BIOLOGY 2010; 10:151. [PMID: 20642851 PMCID: PMC3095295 DOI: 10.1186/1471-2229-10-151] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 07/19/2010] [Indexed: 05/18/2023]
Abstract
BACKGROUND Brassinosteroids (BRs) play crucial roles in plant development and also promote tolerance to a range of abiotic stresses. Although much has been learned about their roles in plant development, the mechanisms by which BRs control plant stress responses and regulate stress-responsive gene expression are not fully known. Since BR interacts with other plant hormones, it is likely that the stress tolerance conferring ability of BR lies in part in its interactions with other stress hormones. RESULTS Using a collection of Arabidopsis mutants that are either deficient in or insensitive to abscisic acid (ABA), ethylene (ET), jasmonic acid (JA) and salicylic acid (SA), we studied the effects of 24-epibrassinloide (EBR) on basic thermotolerance and salt tolerance of these mutants. The positive impact of EBR on thermotolerance in proportion to wild type was evident in all mutants studied, with the exception of the SA-insensitive npr1-1 mutant. EBR could rescue the ET-insensitive ein2 mutant from its hypersensitivity to salt stress-induced inhibition of seed germination, but remained ineffective in increasing the survival of eto1-1 (ET-overproducer) and npr1-1 seedlings on salt. The positive effect of EBR was significantly greater in the ABA-deficient aba1-1 mutant as compared to wild type, indicating that ABA masks BR effects in plant stress responses. Treatment with EBR increased expression of various hormone marker genes in both wild type and mutant seedlings, although to different levels. CONCLUSIONS These results together indicate that the redox-sensitive protein NPR1 (NONEXPRESSOR OF PATHOGENESIS-RELATED GENES1), a master regulator of SA-mediated defense genes, is likely a critical component of EBR-mediated increase in thermotolerance and salt tolerance, but it is not required for EBR-mediated induction of PR-1 (PATHOGENESIS-RELATED1) gene expression; that BR exerts anti-stress effects independently as well as through interactions with other hormones; that ABA inhibits BR effects during stress; and that BR shares transcriptional targets with other hormones.
Collapse
Affiliation(s)
- Uday K Divi
- Department of Biology, University of Western Ontario, London, ON, N6A 5B7, Canada
| | - Tawhidur Rahman
- Department of Biology, University of Western Ontario, London, ON, N6A 5B7, Canada
| | - Priti Krishna
- Department of Biology, University of Western Ontario, London, ON, N6A 5B7, Canada
| |
Collapse
|
20
|
Murthy S, Ryan A, He C, Mallampalli RK, Carter AB. Rac1-mediated mitochondrial H2O2 generation regulates MMP-9 gene expression in macrophages via inhibition of SP-1 and AP-1. J Biol Chem 2010; 285:25062-73. [PMID: 20529870 DOI: 10.1074/jbc.m109.099655] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aberrant matrix deposition is a hallmark of pulmonary fibrosis and is characterized by an imbalance between matrix deposition and degradation. We have previously shown that mice harboring a conditional deletion of the GTP-binding protein, Rac1, in macrophages are protected from asbestos-induced pulmonary fibrosis. To investigate the contribution of aberrant matrix degradation, we addressed the role of Rac1 in regulating expression of macrophage-specific MMP-9 (matrix metalloproteinase-9). We found that MMP-9 gene transcription was significantly greater in Rac1 null macrophages. Deletion and mutational analysis of the MMP-9 promoter revealed that both SP-1 and AP-1 are essential for MMP-9 transcription. Overexpression of constitutive active Rac1 (V12) revealed that H(2)O(2) was derived from the mitochondria. Rac1-induced H(2)O(2) generation down-regulated MMP-9 gene transcription, whereas catalase overexpression in WT cells enhanced MMP-9 expression. SP-1 interacted directly with both c-Jun and c-Fos, and H(2)O(2) decreased this binding, suggesting that SP-1 and AP-1 function cooperatively to regulate MMP-9 transcription. Rac1-mediated H(2)O(2) inhibited the ERK MAPK, which was essential for activation of SP-1 and AP-1. ERK activation and MMP-9 expression were recovered by overexpressing catalase or transfecting siRNA for the mitochondrial iron-sulfur protein, Rieske. These observations were recapitulated in vivo. MMP-9 mRNA was higher in alveolar macrophages isolated from Rac1 null mice and wild type mice given catalase. Rac1 regulates MMP-9 transcription via mitochondrial H(2)O(2) generation, providing a potential mechanism by which Rac1 null mice fail to develop pulmonary fibrosis.
Collapse
Affiliation(s)
- Shubha Murthy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
21
|
Shah NG, Tulapurkar ME, Singh IS, Shelhamer JH, Cowan MJ, Hasday JD. Prostaglandin E2 potentiates heat shock-induced heat shock protein 72 expression in A549 cells. Prostaglandins Other Lipid Mediat 2010; 93:1-7. [PMID: 20382255 DOI: 10.1016/j.prostaglandins.2010.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 03/23/2010] [Accepted: 03/31/2010] [Indexed: 12/11/2022]
Abstract
The heat shock (HS) response is an important cytoprotective response comprising the expression of heat shock proteins (HSPs) and orchestrated by the heat/stress-induced transcription factor, heat shock factor-1 (HSF-1). Previous studies suggest that the activation threshold and magnitude of the HS response may be modified by treatment with arachidonic acid (AA). We analyzed the effect of exogenous AA and its metabolites, PGE(2), LTD(4), and 15-HETE on HSF-1-dependent gene expression in A549 human respiratory epithelial-like cells. When added at 1microM, PGE(2) much more than LTD(4), but not 15-HETE increased activity of a synthetic HSF-1-dependent reporter after HS exposure (42 degrees C for 2h), but had no effect in the absence of HS. Exposing A549 cells to HS stimulated the release of PGE(2) and treatment with the cyclooxygenase inhibitor, ibuprofen, reduced HS-induced HSF-1-dependent transcription. PGE(2) increased HS-induced HSP72 mRNA and protein expression but EMSA and Western blot analysis failed to show an effect on HSF-1 DNA binding activity or post-translational modification. In summary, we showed that HS stimulates the generation of PGE(2), which augments the generation of HSPs. The clinical consequences of this pathway have yet to be determined.
Collapse
Affiliation(s)
- Nirav G Shah
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
22
|
Vedam K, Nishijima Y, Druhan LJ, Khan M, Moldovan NI, Zweier JL, Ilangovan G. Role of heat shock factor-1 activation in the doxorubicin-induced heart failure in mice. Am J Physiol Heart Circ Physiol 2010; 298:H1832-41. [PMID: 20363884 DOI: 10.1152/ajpheart.01047.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Treating cancer patients with chemotherapeutics, such as doxorubicin (Dox), cause dilated cardiomyopathy and congestive heart failure because of oxidative stress. On the other hand, heat shock factor-1 (HSF-1), a transcription factor for heat shock proteins (Hsps), is also known to be activated in response to oxidative stress. However, the possible role of HSF-1 activation and the resultant Hsp25 in chemotherapeutic-induced heart failure has not been investigated. Using HSF-1 wild-type (HSF-1(+/+)) and knock-out (HSF-1(-/-)) mice, we tested the hypothesis that activation of HSF-1 plays a role in the development of Dox-induced heart failure. Higher levels of Hsp25 and its phosphorylated forms were found in the failing hearts of Dox-treated HSF-1(+/+) mice. More than twofold increase in Hsp25 mRNA level was found in Dox-treated hearts. Proteomic analysis showed that there is accumulation and aggregation of Hsp25 in Dox-treated failing hearts. Additionally, Hsp25 was found to coimmunoprecipitate with p53 and vice versa. Further studies indicated that the Dox-induced higher levels of Hsp25 transactivated p53 leading to higher levels of the pro-apoptotic protein Bax, but other p53-related proteins remained unaltered. Moreover, HSF-1(-/-) mice showed significantly reduced Dox-induced heart failure and higher survival rate, and there was no change in Bax upon treating with Dox in HSF-1(-/-) mice. From these results we propose a novel mechanism for Dox-induced heart failure: increased expression of Hsp25 because of oxidant-induced activation of HSF-1 transactivates p53 to increase Bax levels, which leads to heart failure.
Collapse
Affiliation(s)
- Kaushik Vedam
- Division of Cardiovascular medicine, Davis Heart & Lung Research Institute, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Byun JY, Yoon CH, An S, Park IC, Kang CM, Kim MJ, Lee SJ. The Rac1/MKK7/JNK pathway signals upregulation of Atg5 and subsequent autophagic cell death in response to oncogenic Ras. Carcinogenesis 2009; 30:1880-8. [PMID: 19783847 DOI: 10.1093/carcin/bgp235] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To prevent the development of malignancies, mammalian cells activate disposal programs, such as programmed cell death, in response to deregulated oncogene expression. However, the molecular basis for regulation of cellular disposal machinery in response to activated oncogenes is unclear at present. In this study, we show that upregulation of the autophagy-related protein, Atg5, is critically required for the oncogenic H-ras-induced autophagic cell death and that Rac1/mitogen-activated kinase kinase (MKK) 7/c-Jun N-terminal kinase (JNK) signals upregulation of Atg5. Overexpression of H-ras(V12) induced marked autophagic vacuole formation and cell death in normal fibroblasts, which remained unaffected by a caspase inhibitor. Pretreatment with Bafilomycin A1, an autophagy inhibitor, completely attenuated H-ras(V12)-induced cell death as well as autophagic vacuole formation. Selective production of Atg5 was observed in cells overexpressing H-ras(V12), and small interfering RNA (siRNA) targeting of Atg5 clearly inhibited autophagic cell death. Interestingly, inhibition of JNK or c-Jun by specific siRNA suppressed Atg5 upregulation and autophagic cell death. Moreover, inhibition of MKK7, but not MKK4, effectively attenuated H-ras(V12)-induced JNK activation. In addition, ectopic expression of RacN17 or Rac1-siRNA effectively inhibited MKK7-JNK activation, Atg5 upregulation and autophagic cell death. These data support the notion that upregulation of Atg5 is required for the oncogenic H-ras-induced autophagic cell death in normal fibroblasts and that activation of Rac1/MKK7/JNK-signaling pathway leads to upregulation of Atg5 in response to oncogenic H-ras. Our findings suggest that in cells acquiring deregulated oncogene expression, oncogenic stress triggers autophagic cell death, which protects cells against malignant progression.
Collapse
Affiliation(s)
- Joo-Yun Byun
- Laboratory of Molecular Biochemistry, Department of Chemistry, Hanyang University, 17 Haengdang-Dong, Seongdong-Ku, Seoul 133-791, Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
Murthy S, Adamcakova-Dodd A, Perry SS, Tephly LA, Keller RM, Metwali N, Meyerholz DK, Wang Y, Glogauer M, Thorne PS, Carter AB. Modulation of reactive oxygen species by Rac1 or catalase prevents asbestos-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2009; 297:L846-55. [PMID: 19684199 DOI: 10.1152/ajplung.90590.2008] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The release of reactive oxygen species (ROS) and cytokines by alveolar macrophages has been demonstrated in asbestos-induced pulmonary fibrosis, but the mechanism linking alveolar macrophages to the pathogenesis is not known. The GTPase Rac1 is a second messenger that plays an important role in host defense. In this study, we demonstrate that Rac1 null mice are protected from asbestos-induced pulmonary fibrosis, as determined by histological and biochemical analysis. We hypothesized that Rac1 induced pulmonary fibrosis via generation of ROS. Asbestos increased TNF-alpha and ROS in a Rac1-dependent manner. TNF-alpha was elevated only 1 day after exposure, whereas ROS generation progressively increased in bronchoalveolar lavage cells obtained from wild-type (WT) mice. To determine whether ROS generation contributed to pulmonary fibrosis, we overexpressed catalase in WT monocytes and observed a decrease in ROS generation in vitro. More importantly, administration of catalase to WT mice attenuated the development of fibrosis in vivo. For the first time, these results demonstrate that Rac1 plays a crucial role in asbestos-induced pulmonary fibrosis. Moreover, it suggests that a simple intervention may be useful to prevent progression of the disease.
Collapse
Affiliation(s)
- Shubha Murthy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang PL, Niidome T, Akaike A, Kihara T, Sugimoto H. Rac1 inhibition negatively regulates transcriptional activity of the amyloid precursor protein gene. J Neurosci Res 2009; 87:2105-14. [DOI: 10.1002/jnr.22039] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
26
|
Gieni RS, Hendzel MJ. Actin dynamics and functions in the interphase nucleus: moving toward an understanding of nuclear polymeric actin. Biochem Cell Biol 2009; 87:283-306. [PMID: 19234542 DOI: 10.1139/o08-133] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Actin exists as a dynamic equilibrium of monomers and polymers within the nucleus of living cells. It is utilized by the cell for many aspects of gene regulation, including mRNA processing, chromatin remodelling, and global gene expression. Polymeric actin is now specifically linked to transcription by RNA polymerase I, II, and III. An active process, requiring both actin polymers and myosin, appears to drive RNA polymerase I transcription, and is also implicated in long-range chromatin movement. This type of mechanism brings activated genes from separate chromosomal territories together, and then participates in their compartmentalization near nuclear speckles. Nuclear speckle formation requires polymeric actin, and factors promoting polymerization, such as profilin and PIP2, are concentrated there. A review of the literature shows that a functional population of G-actin cycles between the cytoplasm and the nucleoplasm. Its nuclear concentration is dependent on the cytoplasmic G-actin pool, as well as on the activity of import and export mechanisms and the availability of interactions that sequester it within the nucleus. The N-WASP-Arp2/3 actin polymer-nucleating mechanism functions in the nucleus, and its mediators, including NCK, PIP2, and Rac1, can be found in the nucleoplasm, where they likely influence the kinetics of polymer formation. The actin polymer species produced are tightly regulated, and may take on conformations not easily recognized by phalloidin. Many of the factors that cleave F-actin in the cytoplasm are present at high levels in the nucleoplasm, and are also likely to affect actin dynamics there. The absolute and relative G-actin content in the nucleoplasm and the cytoplasm of a cell contains information about the homeostatic state of that cell. We propose that the cycling of G-actin between the nucleus and cytoplasm represents a signal transduction mechanism that can function through both extremes of global cellular G-actin content. MAL signalling within the serum response factor pathway, when G-actin levels are low, represents a well-studied example of actin functioning in signal transduction. The translocation of NCK into the nucleus, along with G-actin, during dissolution of the cytoskeleton in response to DNA damage represents another instance of a unique signalling mechanism operating when G-actin levels are high.
Collapse
Affiliation(s)
- Randall S Gieni
- Cross Cancer Institute and Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, ABT6G1Z2, Canada
| | | |
Collapse
|
27
|
Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal 2009; 11:777-90. [PMID: 18828708 DOI: 10.1089/ars.2008.2270] [Citation(s) in RCA: 573] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Reactive oxygen species (ROS) have been identified as signaling molecules in various pathways regulating both cell survival and cell death. Autophagy, a self-digestion process that degrades intracellular structures in response to stress, such as nutrient starvation, is also involved in both cell survival and cell death. Alterations in both ROS and autophagy regulation contribute to cancer initiation and progression, and both are targets for developing therapies to induce cell death selectively in cancer cells. Many stimuli that induce ROS generation also induce autophagy, including nutrient starvation, mitochondrial toxins, hypoxia, and oxidative stress. Some of these stimuli are under clinical investigation as cancer treatments, such as 2-methoxyestrodial and arsenic trioxide. Recently, it was demonstrated that ROS can induce autophagy through several distinct mechanisms involving Atg4, catalase, and the mitochondrial electron transport chain (mETC). This leads to both cell-survival and cell-death responses and could be selective toward cancer cells. In this review, we give an overview of the roles ROS and autophagy play in cell survival and cell death, and their importance to cancer. Furthermore, we describe how autophagy is mediated by ROS and the implications of this regulation to cancer treatments.
Collapse
Affiliation(s)
- Meghan B Azad
- Manitoba Institute of Cell Biology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
28
|
Zhang P, Zhang X, Hao X, Wang Y, Hui Y, Wang H, Hu D, Zhou J. Rac1 activates HIF-1 in retinal pigment epithelium cells under hypoxia. Graefes Arch Clin Exp Ophthalmol 2009; 247:633-9. [PMID: 19165496 DOI: 10.1007/s00417-008-1031-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 12/10/2008] [Accepted: 12/22/2008] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Upregulation of vascular endothelial growth factor (VEGF) in hypoxic retinal pigment epithelium (RPE) cells, mediated by hypoxia-inducible factor-1 (HIF-1) is responsible for choroidal neovascularization (CNV). HIF-1alpha is the inducible subunit of HIF-1, but the underlying mechanisms by which RPE cells sense a decrease in oxygen concentration and transduce this signal to HIF-1alpha are largely unknown. Rho family small GTPase Rac1, as a potential intermediate, possibly plays a pivotal role in activating HIF-1alpha in RPE cells under hypoxia. AIMS To further define Rac1 playing an essential role in the induction of HIF-1alpha expression in RPE cells under hypoxia. METHODS In this study, we examined the expression of HIF-1alpha and Rac1 in human RPE cells under hypoxia for 0, 1, 2, 4, 8, 12 and 24 h by RT-PCR and Western blot. To elucidate whether Rac1 is responsible for activating the expression of hypoxia-induced HIF-1alpha, human RPE cells were treated with Rac1 inhibitor NSC23766 under hypoxia for 0, 1, 2, 4, 8, 12 and 24 h, and expression of HIF-1alpha and Rac1 measured by RT-PCR and Western blot. RESULTS The mRNA expression of HIF-1alpha and Rac1 in RPE cells significantly increased in a time-dependent manner, reaching the maximum at 4 h, and thereafter slowly declined. HIF-1alpha protein induction in human RPE cells was found after 1 h of hypoxia, reaching the maximum at 8 h, and then slowly declined. In response to hypoxia, the levels of Rac1 protein significantly increased, reaching the maximum at 4 h, and then slowly declined. After treatment with NSC23766, both HIF-1alpha and Rac1 expression were significantly inhibited in hypoxic RPE cells. CONCLUSIONS Rac1 is crucial to activate HIF-1 in RPE cells under hypoxia, which may be a novel target other than VEGF and HIF-1 in developing CNV inhibitors.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Singh IS, Shah NG, Almutairy E, Hasday JD. Role of HSF1 in Infectious Disease. HEAT SHOCK PROTEINS 2009. [DOI: 10.1007/978-90-481-2976-8_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Haga S, Terui K, Fukai M, Oikawa Y, Irani K, Furukawa H, Todo S, Ozaki M. Preventing hypoxia/reoxygenation damage to hepatocytes by p66(shc) ablation: up-regulation of anti-oxidant and anti-apoptotic proteins. J Hepatol 2008; 48:422-32. [PMID: 18191273 DOI: 10.1016/j.jhep.2007.11.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 11/07/2007] [Accepted: 11/07/2007] [Indexed: 12/30/2022]
Abstract
BACKGROUND/AIMS Ischemia/reperfusion damage to the liver remains a serious concern in many clinical situations. Major mechanisms for this certainly include oxidative stress. METHODS The effects of ablating the p66 isoform of ShcA (p66(shc)) on hypoxia/reoxygenation (H/R)-induced oxidative stress and cell injury in hepatocytes were investigated. RESULTS Immediately after reoxygenation, AML12 cells were clearly under oxidative stress; many cells underwent apoptosis. However, knockdown of p66(shc) by specific RNAi markedly decreased cellular oxidative stress and H/R-induced apoptosis, as well as conferring resistance to H(2)O(2) insult. These data suggest that prevention of apoptosis conferred by ablation of p66(shc) results from changed ROS-scavenging, but not inhibition of ROS generation. These data were also confirmed in fibroblasts from p66(shc) knockout mice. Anti-oxidant molecules, such as MnSOD and Ref-1 and the anti-apoptotic molecule Bcl-xL were up-regulated, and pro-apoptotic FLICE was down-regulated, by ablation of p66(shc). Interestingly, catalase expression was not affected in p66(shc)-knockdown-AML12 cells although it is a major target in other cell types. CONCLUSIONS Our findings suggest that in hepatocytes, ablation of p66(shc) is cytoprotective against H/R-induced oxidative stress, with MnSOD and Ref-1 playing critical roles, and with up-regulation of Bcl-xL and down-regulation of FLICE contributing jointly to preventing cells from undergoing oxidant-induced apoptosis.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Surgery, Hokkaido University School of Medicine, Sapporo, Hokkaido, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ouyang W, Luo W, Zhang D, Jian J, Ma Q, Li J, Shi X, Chen J, Gao J, Huang C. PI-3K/Akt pathway-dependent cyclin D1 expression is responsible for arsenite-induced human keratinocyte transformation. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1-6. [PMID: 18197291 PMCID: PMC2199295 DOI: 10.1289/ehp.10403] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 10/04/2007] [Indexed: 05/21/2023]
Abstract
BACKGROUND Long-term exposure of arsenite leads to human skin cancer. However, the exact mechanisms of arsenite-induced human skin carcinogenesis remain to be defined. OBJECTIVES In this study, we investigated the potential role of PI-3K/Akt/cyclin D1in the transformation of human keratinocytic cells upon arsenite exposure. METHODS We used the soft agar assay to evaluate the cell transformation activity of arsenite exposure and the nude mice xenograft model to determine the tumorigenesis of arsenite-induced transformed cells. We used the dominant negative mutant and gene knockdown approaches to elucidate the signaling pathway involved in this process. RESULTS Our results showed that repeated long-term exposure of HaCat cells to arsenite caused cell transformation, as indicated by anchorage-independent growth in soft agar. The tumorigenicity of these transformed cells was confirmed in nude mice. Treatment of cells with arsenite also induced significant activation of PI-3K and Akt, which was responsible for the anchorage-independent cell growth induced by arsenite exposure. Furthermore, our data also indicated that cyclin D1 is an important downstream molecule involved in PI-3K/Akt-mediated cell transformation upon arsenite exposure based on the facts that inhibition of cyclin D1 expression by dominant negative mutants of PI-3K, and Akt, or the knockdown of the cyclin D1 expression by its specific siRNA in the HaCat cells resulted in impairing of anchorage-independent growth of HaCat cells induced by arsenite. CONCLUSION Our results demonstrate that PI-3K/Akt-mediated cyclin D1 expression is at least one key event implicated in the arsenite human skin carcinogenic effect.
Collapse
Affiliation(s)
- Weiming Ouyang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Wenjing Luo
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
- Department of Occupational and Environmental Health Sciences, Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Jinlong Jian
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Qian Ma
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Xianglin Shi
- Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky, USA
| | - Jingyuan Chen
- Department of Occupational and Environmental Health Sciences, Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical College, Wenzhou, Zhejiang, People’s Republic of China
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
- Address correspondence to C. Huang, Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Rd., Tuxedo, NY 10987 USA. Telephone: (845) 731-3519. Fax: (845) 351-2320. E-mail:
| |
Collapse
|
32
|
Ozaki M, Todo S. Surgical stress and tumor behavior: impact of ischemia-reperfusion and hepatic resection on tumor progression. Liver Transpl 2007; 13:1623-6. [PMID: 18044752 DOI: 10.1002/lt.21230] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE. Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 2007; 110:2641-9. [PMID: 17525289 DOI: 10.1182/blood-2006-11-053728] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Plasma cells producing high levels of paraprotein are dependent on the unfolded protein response (UPR) and chaperone proteins to ensure correct protein folding and cell survival. We hypothesized that disrupting client-chaperone interactions using heat shock protein 90 (Hsp90) inhibitors would result in an inability to handle immunoglobulin production with the induction of the UPR and myeloma cell death. To study this, myeloma cells were treated with Hsp90 inhibitors as well as known endoplasmic reticulum stress inducers and proteasome inhibitors. Treatment with thapsigargin and tunicamycin led to the activation of all 3 branches of the UPR, with early splicing of XBP1 indicative of IRE1 activation, upregulation of CHOP consistent with ER resident kinase (PERK) activation, and activating transcription factor 6 (ATF6) splicing. 17-AAG and radicicol also induced splicing of XBP1, with the induction of CHOP and activation of ATF6, whereas bortezomib resulted in the induction of CHOP and activation of ATF6 with minimal effects on XBP1. After treatment with all drugs, expression levels of the molecular chaperones BiP and GRP94 were increased. All drugs inhibited proliferation and induced cell death with activation of JNK and caspase cleavage. In conclusion, Hsp90 inhibitors induce myeloma cell death at least in part via endoplasmic reticulum stress and the UPR death pathway.
Collapse
Affiliation(s)
- Emma L Davenport
- Section of Haemato-Oncology, Cancer Research UK, The Institute of Cancer Research, Sutton, Surrey, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Moraitis C, Curran BPG. Can the different heat shock response thresholds found in fermenting and respiring yeast cells be attributed to their differential redox states? Yeast 2007; 24:653-66. [PMID: 17533621 DOI: 10.1002/yea.1498] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In this study we used a heat-shock (HS) reporter gene to demonstrate that respiring cells are intrinsically less sensitive (by 5 degrees C) than their fermenting counterparts to a sublethal heat shock. We also used an oxidant-sensitive fluorescent probe to demonstrate that this correlates with lower levels of sublethal reactive oxygen species (ROS) accumulation in heat-stressed respiring cells. Moreover, this relationship between HS induction of the reporter gene and ROS accumulation extends to respiring cells that have had their ROS levels modified by treatment with the anti-oxidant ascorbic acid and the pro-oxidant H(2)O(2). Thus, by demonstrating that the ROS/HSR correlation previously demonstrated in fermenting cells also holds for respiring cells (despite their greater HS insensitivity and higher level of intrinsic thermotolerance), we provide evidence that the intracellular redox state may influence both the sensitivity of the heat-shock response (HSR) and stress tolerance in the yeast Saccharomyces cerevisiae.
Collapse
Affiliation(s)
- Christos Moraitis
- School of Biological and Chemical Sciences, Queen Mary College, University of London, Mile End Road, London E1 4NS, U.K
| | | |
Collapse
|
35
|
Wheeler DS, Wong HR. Heat shock response and acute lung injury. Free Radic Biol Med 2007; 42:1-14. [PMID: 17157189 PMCID: PMC1790871 DOI: 10.1016/j.freeradbiomed.2006.08.028] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 08/23/2006] [Accepted: 08/29/2006] [Indexed: 11/19/2022]
Abstract
All cells respond to stress through the activation of primitive, evolutionarily conserved genetic programs that maintain homeostasis and assure cell survival. Stress adaptation, which is known in the literature by a myriad of terms, including tolerance, desensitization, conditioning, and reprogramming, is a common paradigm found throughout nature, in which a primary exposure of a cell or organism to a stressful stimulus (e.g., heat) results in an adaptive response by which a second exposure to the same stimulus produces a minimal response. More interesting is the phenomenon of cross-tolerance, by which a primary exposure to a stressful stimulus results in an adaptive response whereby the cell or organism is resistant to a subsequent stress that is different from the initial stress (i.e., exposure to heat stress leading to resistance to oxidant stress). The heat shock response is one of the more commonly described examples of stress adaptation and is characterized by the rapid expression of a unique group of proteins collectively known as heat shock proteins (also commonly referred to as stress proteins). The expression of heat shock proteins is well described in both whole lungs and in specific lung cells from a variety of species and in response to a variety of stressors. More importantly, in vitro data, as well as data from various animal models of acute lung injury, demonstrate that heat shock proteins, especially Hsp27, Hsp32, Hsp60, and Hsp70 have an important cytoprotective role during lung inflammation and injury.
Collapse
Affiliation(s)
- Derek S. Wheeler
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center; Kindervelt Laboratory for Critical Care Medicine Research, Children’s Hospital Research Foundation;]Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Hector R. Wong
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center; Kindervelt Laboratory for Critical Care Medicine Research, Children’s Hospital Research Foundation;]Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
36
|
Szymczyk KH, Kerr BAE, Freeman TA, Adams CS, Steinbeck MJ. Involvement of hydrogen peroxide in the differentiation and apoptosis of preosteoclastic cells exposed to arsenite. Biochem Pharmacol 2006; 72:761-9. [PMID: 16876763 DOI: 10.1016/j.bcp.2006.06.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 06/08/2006] [Accepted: 06/08/2006] [Indexed: 11/21/2022]
Abstract
Long-term exposure to sodium arsenite (AsO(2)) promotes the development of various cancers. Paradoxically, arsenic also induces pro-myelomonocytic leukemia cell differentiation, and at higher concentrations, apoptosis. The present study investigated the effects of AsO(2) on preosteoclasts. When treated with 2.5-5microM AsO(2), RAW264.7 cells underwent osteoclast differentiation as evidenced by an increase in the number of multinucleate cells expressing tartrate resistant acid phosphatase (TRAP). The appearance of these phenotypic markers was preceded by a low level increase in extracellular production of H(2)O(2) and was prevented by the addition of catalase (4.5microg/ml), an enzyme that removes H(2)O(2). Only at high concentrations (10-25microM) of AsO(2) was a significant loss of cell viability and a high level increase in H(2)O(2) production (1.5microM) observed. Apoptosis was blocked by pretreatment with diphenylene iodonium chloride (2microM), a NAD(P)H-flavoprotein inhibitor, suggesting the involvement of NADPH-oxidase. The data show that AsO(2), dose-dependently, stimulates increasing amounts of H(2)O(2) production. Moreover, at concentrations found in tissues of individuals exposed to geochemical AsO(2), osteoclasts underwent an H(2)O(2)-dependent differentiation. Therefore, chronic exposure to low-level amounts of AsO(2) could result in increased bone resorption and contribute to bone related pathologies.
Collapse
Affiliation(s)
- K H Szymczyk
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street, Suite 501 Curtis, Philadelphia, PA, USA.
| | | | | | | | | |
Collapse
|
37
|
Abstract
Although reactive oxidants have long been stigmatized as unwanted metabolic byproducts, the expression of oxidases specifically functioning to produce these same molecules in a regulated fashion is surprisingly pervasive throughout metazoan and plant evolution. Although the involvement of oxidants in many signaling pathways is well documented, the cellular strategies for conferring pathway specificity to such reactive molecules have remained more recondite. Recent studies now suggest that cells may spatially restrict oxidant production to allow microdomain-specific signaling.
Collapse
Affiliation(s)
- Lance S Terada
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
38
|
Abstract
BACKGROUND Reactive oxygen species are produced in a highly localized and specific pattern in biological stress responses. The present review examines the redox regulatory aspects of a number of molecular stress response mechanisms in both prokaryotes and eukaryotes. SCOPE The present review provides examples representing both the cytoplasmic stress response, often studied as the heat shock response, as well as the stress response of the endoplasmic reticulum, known as the unfolded protein response. The examples have been selected to illustrate the variety of ways that redox signals mediate and affect stress responses. CONCLUSIONS Redox regulatory mechanisms are intricately embedded in both the cytoplasmic and endoplasmic reticulum stress responses at multiple levels. Many different stimuli, both internal and external, activate endogenous production of reactive oxygen species as a necessary part of the intracellular communication system that activates stress responses.
Collapse
Affiliation(s)
- Nina Fedoroff
- Huck Institutes of the Life Sciences and Biology Department, Penn State University, University Park, PA 16803, USA.
| |
Collapse
|
39
|
Abstract
Many cellular signaling molecules exist in different conformations corresponding to active and inactive states. Transition between these states is regulated by reversible modifications, such as phosphorylation, or by binding of nucleotide triphosphates, their regulated hydrolysis to diphosphates, and their exchange against fresh triphosphates. Specificity and efficiency of cellular signaling is further maintained by regulated subcellular localization of signaling molecules as well as regulated protein-protein interaction. Hence, it is not surprising that molecular chaperones--proteins that are able to specifically interact with distinct conformations of other proteins--could per se interfere with cellular signaling. Hence, it is not surprising that chaperones have co-evolved as integral components of signaling networks where they can function in the maturation as well as in regulating the transition between active and inactive state of signaling molecules, such as receptors, transcriptional regulators and protein kinases. Furthermore, new classes of specific chaperones are emerging and their role in histone-mediated chromatin remodeling and RNA folding are under investigation.
Collapse
Affiliation(s)
- M Gaestel
- Institute of Biochemistry, Medical School Hannover, Germany.
| |
Collapse
|
40
|
Khanday FA, Santhanam L, Kasuno K, Yamamori T, Naqvi A, Dericco J, Bugayenko A, Mattagajasingh I, Disanza A, Scita G, Irani K. Sos-mediated activation of rac1 by p66shc. ACTA ACUST UNITED AC 2006; 172:817-22. [PMID: 16520382 PMCID: PMC2063726 DOI: 10.1083/jcb.200506001] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The Son of Sevenless 1 protein (sos1) is a guanine nucleotide exchange factor (GEF) for either the ras or rac1 GTPase. We show that p66shc, an adaptor protein that promotes oxidative stress, increases the rac1-specific GEF activity of sos1, resulting in rac1 activation. P66shc decreases sos1 bound to the growth factor receptor bound protein (grb2) and increases the formation of the sos1–eps8–e3b1 tricomplex. The NH2-terminal proline-rich collagen homology 2 (CH2) domain of p66shc associates with full-length grb2 in vitro via the COOH-terminal src homology 3 (C-SH3) domain of grb2. A proline-rich motif (PPLP) in the CH2 domain mediates this association. The CH2 domain competes with the proline-rich COOH-terminal region of sos1 for the C-SH3 domain of grb2. P66shc-induced dissociation of sos1 from grb2, formation of the sos1–eps8–e3b1 complex, rac1-specific GEF activity of sos1, rac1 activation, and oxidative stress are also mediated by the PPLP motif in the CH2 domain. This relationship between p66shc, grb2, and sos1 provides a novel mechanism for the activation of rac1.
Collapse
Affiliation(s)
- Firdous A Khanday
- Cardiovascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wu RF, Xu YC, Ma Z, Nwariaku FE, Sarosi GA, Terada LS. Subcellular targeting of oxidants during endothelial cell migration. ACTA ACUST UNITED AC 2006; 171:893-904. [PMID: 16330715 PMCID: PMC2171295 DOI: 10.1083/jcb.200507004] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endogenous oxidants participate in endothelial cell migration, suggesting that the enzymatic source of oxidants, like other proteins controlling cell migration, requires precise subcellular localization for spatial confinement of signaling effects. We found that the nicotinamide adenine dinucleotide phosphate reduced (NADPH) oxidase adaptor p47phox and its binding partner TRAF4 were sequestered within nascent, focal complexlike structures in the lamellae of motile endothelial cells. TRAF4 directly associated with the focal contact scaffold Hic-5, and the knockdown of either protein, disruption of the complex, or oxidant scavenging blocked cell migration. An active mutant of TRAF4 activated the NADPH oxidase downstream of the Rho GTPases and p21-activated kinase 1 (PAK1) and oxidatively modified the focal contact phosphatase PTP-PEST. The oxidase also functioned upstream of Rac1 activation, suggesting its participation in a positive feedback loop. Active TRAF4 initiated robust membrane ruffling through Rac1, PAK1, and the oxidase, whereas the knockdown of PTP-PEST increased ruffling independent of oxidase activation. Our data suggest that TRAF4 specifies a molecular address within focal complexes that is targeted for oxidative modification during cell migration.
Collapse
Affiliation(s)
- Ru Feng Wu
- University of Texas Southwestern, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
42
|
Wang J, He H, Yu L, Xia HHX, Lin MCM, Gu Q, Li M, Zou B, An X, Jiang B, Kung HF, Wong BCY. HSF1 down-regulates XAF1 through transcriptional regulation. J Biol Chem 2006; 281:2451-9. [PMID: 16303760 DOI: 10.1074/jbc.m505890200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Studies have indicated the role of HSF1 (heat-shock transcription factor 1) in repressing the transcription of some nonheat shock genes. XAF1 (XIAP-associated factor 1) was an inhibitor of apoptosis-interacting protein with the effect of antagonizing the cytoprotective role of XIAP. XAF1 expression was lower in gastrointestinal cancers than in normal tissues with the mechanism unclear. Here we showed that gastrointestinal cancer tissues expressed higher levels of HSF1 than matched normal tissues. The expression of XAF1 and HSF1 was negatively correlated in gastrointestinal cancer cell lines. Stress stimuli, including heat, hypo-osmolarity, and H2O2, significantly suppressed the expression of XAF1, whereas the alteration of HSF1 expression negatively correlated with XAF1 expression. We cloned varying lengths of the 5'-flanking region of the XAF1 gene into luciferase reporter vectors, and we evaluated their promoter activities. A transcription silencer was found between the -592- and -1414-nucleotide region that was rich in nGAAn/nT-TCn elements (where n indicates G, A, T, or C). A high affinity and functional HSF1-binding element within the -862/-821-nucleotide region was determined by electrophoretic mobility shift assay and chromatin immunoprecipitation assay. Inactivation of this "heat-shock element" by either site-directed mutation or an HSF1 inhibitor, pifithrin-alpha, restored the promoter activity of the silencer structure. Moreover, pretreatment with antioxidants suppressed HSF1 binding activity and increased the transcriptional activity and expression of XAF1. These findings suggested that endogenous stress pressure in cancer cells sustained the high level expression of HSF1 and subsequently suppressed XAF1 expression, implicating the synergized effect of two anti-apoptotic protein families, HSP and inhibitors of apoptosis, in cytoprotection under stress circumstances.
Collapse
Affiliation(s)
- Jide Wang
- Institute for Digestive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Chou YH, Ho FM, Liu DZ, Lin SY, Tsai LH, Chen CH, Ho YS, Hung LF, Liang YC. The possible role of heat shock factor-1 in the negative regulation of heme oxygenase-1. Int J Biochem Cell Biol 2005; 37:604-15. [PMID: 15618017 DOI: 10.1016/j.biocel.2004.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Revised: 08/10/2004] [Accepted: 08/20/2004] [Indexed: 10/26/2022]
Abstract
We examined a possible role for heat shock factor-1 (HSF-1) in the negative regulation of HO-1 gene expression in human Hep3B hepatoma cells responding to stimulation with 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) and arsenite. Overexpression of HSF-1 and heat-shock experiments indicated that HSF-1 repressed the 15d-PGJ2-and arsenite-induced HO-1 gene expression through directly binding to the consensus heat shock element (HSE) of the HO-1 gene promoter. In addition, point mutations at specific HSE sequences of the HO-1 promoter-driven luciferase plasmid (pGL2/hHO3.2-Luc) abolished the heat shock- and HSF-1-mediated repression of reporter activity. Overall, it is possible that HSF-1 negatively regulates HO-1 gene expression, and that the HSE present in the -389 to -362 region mediates HSF-1-induced repression of human HO-1 gene expression.
Collapse
Affiliation(s)
- Yenn-Hwei Chou
- Department of Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Caron A, Desrosiers RR, Béliveau R. Kidney ischemia–reperfusion regulates expression and distribution of tubulin subunits, β-actin and rho GTPases in proximal tubules. Arch Biochem Biophys 2004; 431:31-46. [PMID: 15464724 DOI: 10.1016/j.abb.2004.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 07/12/2004] [Indexed: 10/26/2022]
Abstract
Ischemic injury is characterized by a loss of cell polarity and a release of proximal tubule epithelial cells resulting from cytoskeletal reorganization. This study used a reversible unilateral renal ischemia-reperfusion model to investigate the expression and distribution of cytoskeletal components and Rho GTPases at protein and mRNA levels in proximal tubule fractions. Ischemia strongly increased beta-actin and alpha-tubulin expressions that were predominantly found in nuclear fractions. Rho GTPases and caveolin-1 expression were upregulated by ischemia and were enriched mainly in Triton-soluble membranes. Rac1 expression was stimulated in the soluble fractions during reperfusion. Rho GTPases mRNA levels were similarly regulated by ischemia-reperfusion suggesting that changes in their expressions could occur at gene or mRNA levels. ERM protein expression and distribution were unaffected by ischemia-reperfusion. Together, these data show that renal ischemia-reperfusion induced expression and redistribution of actin and microtubule cytoskeleton components in addition to Rho GTPases in proximal tubules, suggesting that they participate in an adaptive response to cellular lesions.
Collapse
Affiliation(s)
- Annick Caron
- Laboratoire de médecine moléculaire, Centre de cancérologie Charles Bruneau, Hôpital Ste-Justine, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal, Que., Canada, H3C 3P8
| | | | | |
Collapse
|
45
|
Terui K, Haga S, Enosawa S, Ohnuma N, Ozaki M. Hypoxia/re-oxygenation-induced, redox-dependent activation of STAT1 (signal transducer and activator of transcription 1) confers resistance to apoptotic cell death via hsp70 induction. Biochem J 2004; 380:203-9. [PMID: 14984365 PMCID: PMC1224164 DOI: 10.1042/bj20031891] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Revised: 02/24/2004] [Accepted: 02/25/2004] [Indexed: 11/17/2022]
Abstract
STAT1 (signal transducer and activator of transcription 1) is potentially involved in cell survival, as well as cell death, in different types of cells. The present study was designed to examine the effects of STAT1 on hypoxia/re-oxygenation (H/R)-induced cell death and/or survival, and the underlying mechanisms of any such effects. H/R was shown to induce apoptotic cell death of rat hepatocytes. The addition of a STAT1-specific inhibitor, fludarabine, significantly increased the fraction of apoptotic cells after H/R. Following H/R, STAT1 was activated and sequential phosphorylation of Tyr701 and Ser727 was observed, which could be inhibited by the antioxidant N-acetyl-L-cysteine. Tyrosine and serine phosphorylation of STAT1 was mediated by Janus kinase 2 and phosphoinositide 3-kinase/Akt kinase respectively in a redox-dependent manner following H/R. STAT1-induced HSP70 (heat-shock protein 70) expression and the suppression of apoptosis occurred concomitantly. In conclusion, STAT1 activation, in a redox-dependent manner, following H/R may play crucial roles in cell survival, at least partly via HSP70 induction.
Collapse
Affiliation(s)
- Keita Terui
- Bioengineering Laboratory, Department of Innovative Surgery National Research Institute for Child Health and Development 3-35-31, Taishi-do, Setagaya, Tokyo, 154-8567, Japan
| | | | | | | | | |
Collapse
|
46
|
Shin HK, Kim YK, Kim KY, Lee JH, Hong KW. Remnant Lipoprotein Particles Induce Apoptosis in Endothelial Cells by NAD(P)H Oxidase–Mediated Production of Superoxide and Cytokines via Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 Activation. Circulation 2004; 109:1022-8. [PMID: 14967724 DOI: 10.1161/01.cir.0000117403.64398.53] [Citation(s) in RCA: 210] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Remnant lipoprotein particles (RLPs), products of lipolytic degradation of triglyceride-rich lipoprotein derived from VLDL, exert atherogenesis. In this study, we observed how RLPs induced cytotoxicity in human umbilical vein endothelial cells (HUVECs) and cilostazol prevented cell death.
Methods and Results—
RLPs were isolated from the plasma of hyperlipidemic patients by use of an immunoaffinity gel mixture of anti–apolipoprotein A-1 and anti–apolipoprotein B-100 monoclonal antibodies. RLPs (50 μg/mL) significantly increased superoxide formation in HUVECs associated with elevated gp91phox mRNA and protein expression and Rac1 translocation, accompanied by increased production of tumor necrosis factor (TNF)-α and interleukin-1β, DNA fragmentation, and cell death. Cilostazol (1 to 100 μmol/L) significantly suppressed not only NAD(P)H oxidase–dependent superoxide production but also TNF-α and interleukin-1β release and restored viability. RLPs activated a lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), which was not inhibited by cilostazol. Treatment of HUVECs with monoclonal antibody for LOX-1 attenuated RLP-mediated production of superoxide, TNF-α, and interleukin-1β and DNA fragmentation.
Conclusions—
RLPs stimulated NAD(P)H oxidase–dependent superoxide formation and induction of cytokines in HUVECs via activation of LOX-1, consequently leading to reduction in cell viability with DNA fragmentation, and cilostazol exerts a cell-protective effect by suppressing these variables.
Collapse
Affiliation(s)
- Hwa Kyoung Shin
- Department of Pharmacology, College of Medicine, Pusan National University, Pusan, Korea
| | | | | | | | | |
Collapse
|
47
|
Fiorenza MT, Bevilacqua A, Canterini S, Torcia S, Pontecorvi M, Mangia F. Early transcriptional activation of the hsp70.1 gene by osmotic stress in one-cell embryos of the mouse. Biol Reprod 2004; 70:1606-13. [PMID: 14766729 DOI: 10.1095/biolreprod.103.024877] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In fertilized mouse eggs, de novo transcription of embryonic genes is first observed during the S phase of the one-cell stage. This transcription, however, is mostly limited to the male pronucleus and possibly uncoupled from translation, making the functional meaning obscure. We found that one-cell mouse embryos respond to the osmotic shock of in vitro isolation with migration of HSF1, the canonical stress activator of mammalian heat shock genes, to pronuclei and by transient transcription of the hsp70.1, but not hsp70.3 and hsp90, heat shock genes. Isolated growing dictyate oocytes also display a nuclear HSF1 localization, but, in contrast with embryos, they transcribe both hsp70.1 and hsp70.3 genes only after heat shock. Intranuclear injection of double-stranded oligodeoxyribonucleotides containing HSE, GAGA box or GC box consensus sequences, and antibodies raised to transcription factors HSF1, HSF2, Drosophila melanogaster GAGA factor, or Sp1 demonstrated that hsp70.1 transcription depends on HSF1 in both oocytes and embryos and that Sp1 is dispensable in oocytes and inhibitory in the embryos. Hsp70.1 thus represents the first endogenous gene so far identified to be physiologically activated and tightly regulated after fertilization in mammals.
Collapse
Affiliation(s)
- Maria Teresa Fiorenza
- Istituto Pasteur-Fondazione Cenci Bolognetti and Department of Psychology, Section of Neuroscience, UniversityLa Sapienza of Rome, 00185 Rome, Italy
| | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- Michitaka Ozaki
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
49
|
Jung DK, Bae GU, Kim YK, Han SH, Choi WS, Kang H, Seo DW, Lee HY, Cho EJ, Lee HW, Han JW. Hydrogen peroxide mediates arsenite activation of p70(s6k) and extracellular signal-regulated kinase. Exp Cell Res 2003; 290:144-54. [PMID: 14516795 DOI: 10.1016/s0014-4827(03)00320-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
To define the mechanism of arsenite-induced tumor promotion, we examined the role of reactive oxygen species (ROS) in the signaling pathways of cells exposed to arsenite. Arsenite treatment resulted in the persistent activation of p70(s6k) and extracellular signal-regulated kinase 1/2 (ERK1/2) which was accompanied by an increase in intracellular ROS production. The predominant produced appeared to be H(2)O(2), because the arsenite-induced increase in dichlorofluorescein (DCF) fluorescence was completely abolished by pretreatment with catalase but not with heat-inactivated catalase. Elimination of H(2)O(2) by catalase or N-acetyl-L-cysteine inhibited the arsenite-induced activation of p70(s6k) and ERK1/2, indicating the possible role of H(2)O(2) in the arsenite activation of the p70(s6k) and the ERK1/2 signaling pathways. A specific inhibitor of p70(s6k), rapamycin, and calcium chelators significantly blocked the activation of p70(s6k) induced by arsenite. While the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and LY294002 completely abrogated arsenite activation of p70(s6k), ERK1/2 activation by arsenite was not affected by these inhibitors, indicating that H(2)O(2) might act as an upstream molecule of PI3K as well as ERK1/2. Consistent with these results, none of the inhibitors impaired H(2)O(2) production by arsenite. DNA binding activity of AP-1, downstream of ERK1/2, was also inhibited by catalase, N-acetyl-L-cysteine, and the MEK inhibitor PD98059, which significantly blocked arsenite activation of ERK1/2. Taken together, these studies provide insight into mechanisms of arsenite-induced tumor promotion and suggest that H(2)O(2) plays a critical role in tumor promotion by arsenite through activation of the ERK1/2 and p70(s6k) signaling pathways.
Collapse
Affiliation(s)
- Dong Keun Jung
- Department of Biochemistry and Molecular Biology, College of Pharmacy, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bar-Or D. Proposed mechanism for induction of heat shock protein 70 by geranylgeranyl acetone by prenylation of Ras proteins *. Crit Care Med 2003; 31:2700-1. [PMID: 14605546 DOI: 10.1097/01.ccm.0000092457.89851.eb] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|