51
|
Mizuma A, You JS, Yenari MA. Targeting Reperfusion Injury in the Age of Mechanical Thrombectomy. Stroke 2018; 49:1796-1802. [PMID: 29760275 DOI: 10.1161/strokeaha.117.017286] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Atsushi Mizuma
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.).,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.).,Department of Neurology, Tokai University School of Medicine, Isehara, Japan (A.M.)
| | - Je Sung You
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.).,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.).,Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, South Korea (J.S.Y.)
| | - Midori A Yenari
- From the Department of Neurology, University of California, San Francisco (A.M., J.S.Y., M.A.Y.) .,San Francisco Veterans Affairs Medical Center, CA (A.M., J.S.Y., M.A.Y.)
| |
Collapse
|
52
|
Kumfu S, Charununtakorn ST, Jaiwongkam T, Chattipakorn N, Chattipakorn SC. Humanin Exerts Neuroprotection During Cardiac Ischemia-Reperfusion Injury. J Alzheimers Dis 2018; 61:1343-1353. [DOI: 10.3233/jad-170708] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sirinart Kumfu
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Savitree T. Charununtakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
53
|
Bernhart E, Kogelnik N, Prasch J, Gottschalk B, Goeritzer M, Depaoli MR, Reicher H, Nusshold C, Plastira I, Hammer A, Fauler G, Malli R, Graier WF, Malle E, Sattler W. 2-Chlorohexadecanoic acid induces ER stress and mitochondrial dysfunction in brain microvascular endothelial cells. Redox Biol 2018; 15:441-451. [PMID: 29413957 PMCID: PMC5975063 DOI: 10.1016/j.redox.2018.01.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 02/06/2023] Open
Abstract
Peripheral leukocytes induce blood-brain barrier (BBB) dysfunction through the release of cytotoxic mediators. These include hypochlorous acid (HOCl) that is formed via the myeloperoxidase-H2O2-chloride system of activated phagocytes. HOCl targets the endogenous pool of ether phospholipids (plasmalogens) generating chlorinated inflammatory mediators like e.g. 2-chlorohexadecanal and its conversion product 2-chlorohexadecanoic acid (2-ClHA). In the cerebrovasculature these compounds inflict damage to brain microvascular endothelial cells (BMVEC) that form the morphological basis of the BBB. To follow subcellular trafficking of 2-ClHA we synthesized a ‘clickable’ alkyne derivative (2-ClHyA) that phenocopied the biological activity of the parent compound. Confocal and superresolution structured illumination microscopy revealed accumulation of 2-ClHyA in the endoplasmic reticulum (ER) and mitochondria of human BMVEC (hCMEC/D3 cell line). 2-ClHA and its alkyne analogue interfered with protein palmitoylation, induced ER-stress markers, reduced the ER ATP content, and activated transcription and secretion of interleukin (IL)−6 as well as IL-8. 2-ClHA disrupted the mitochondrial membrane potential and induced procaspase-3 and PARP cleavage. The protein kinase R-like ER kinase (PERK) inhibitor GSK2606414 suppressed 2-ClHA-mediated activating transcription factor 4 synthesis and IL-6/8 secretion, but showed no effect on endothelial barrier dysfunction and cleavage of procaspase-3. Our data indicate that 2-ClHA induces potent lipotoxic responses in brain endothelial cells and could have implications in inflammation-induced BBB dysfunction.
Collapse
Affiliation(s)
- Eva Bernhart
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Nora Kogelnik
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Jürgen Prasch
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Madeleine Goeritzer
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Maria Rosa Depaoli
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Helga Reicher
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Christoph Nusshold
- Institute of Physiological Chemistry, Medical University of Graz, Austria.
| | - Ioanna Plastira
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Astrid Hammer
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Cell Biology, Histology and Embryology, Medical University of Graz, Austria.
| | - Günter Fauler
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria.
| | - Roland Malli
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Ernst Malle
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Wolfgang Sattler
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| |
Collapse
|
54
|
Li X, Guo H, Zhao L, Wang B, Liu H, Yue L, Bai H, Jiang H, Gao L, Feng D, Qu Y. Adiponectin attenuates NADPH oxidase-mediated oxidative stress and neuronal damage induced by cerebral ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3265-3276. [DOI: 10.1016/j.bbadis.2017.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/12/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
|
55
|
Keilhoff G, Esser T, Titze M, Ebmeyer U, Schild L. High-potential defense mechanisms of neocortex in a rat model of transient asphyxia induced cardiac arrest. Brain Res 2017; 1674:42-54. [DOI: 10.1016/j.brainres.2017.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/24/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023]
|
56
|
Zhang B, Yang N, Mo ZM, Lin SP, Zhang F. IL-17A Enhances Microglial Response to OGD by Regulating p53 and PI3K/Akt Pathways with Involvement of ROS/HMGB1. Front Mol Neurosci 2017; 10:271. [PMID: 28912678 PMCID: PMC5583146 DOI: 10.3389/fnmol.2017.00271] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/11/2017] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia-reperfusion injury (IRI) has a complex pathogenesis, and interleukin-17 (IL-17) is a newly identified class of the cytokine family that plays an important role in ischemic inflammation. An oxygen-glucose deprivation (OGD) model showed that IL-17A expression was significantly up-regulated in microglial cells. After IL-17A siRNA transfection, the inhibition of proliferation, and the increased apoptosis in microglial cells, induced by OGD/reperfusion, was improved, and the elevation of Caspase-3, Caspase-8, Caspase-9, and poly ADP ribose polymerase (PARP) activities was inhibited. Mass spectrometry demonstrated that IL-17A functioned through a series of factors associated with oxidative stress and apoptosis and regulated Caspase-3 activity and apoptosis in microglial cells via the p53 and PI3K/Akt signaling pathways. IL-17A, HMGB1, and ROS were regulated mutually to exhibit a synergistic effect in the OGD model of microglial cells, but the down-regulation of IL-17A or HMGB1 expression did not completely inhibit the production of ROS. These findings demonstrated that ROS might be located upstream of IL-17A and HMGB1 so that ROS can regulate HMGB1/IL-17A expression to affect the p53 and PI3K/Akt signaling pathways and therefore promote the occurrence of apoptosis in microglial cells. These findings provide a novel evidence for the role of IL-17A in ischemic cerebral diseases.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Ning Yang
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Zhi-Ming Mo
- Department of Neurology, the Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Shao-Peng Lin
- Department of Emergency, the Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Feng Zhang
- Department of Neurosurgery, the Fourth Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| |
Collapse
|
57
|
Zhang B, Yang N, Lin SP, Zhang F. Suitable Concentrations of Uric Acid Can Reduce Cell Death in Models of OGD and Cerebral Ischemia-Reperfusion Injury. Cell Mol Neurobiol 2017; 37:931-939. [PMID: 27709309 PMCID: PMC11482132 DOI: 10.1007/s10571-016-0430-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022]
Abstract
Cerebral infarction (CI) is a common clinical cerebrovascular disease, and to explore the pathophysiological mechanisms and seek effective treatment means are the hotspot and difficult point in medical research nowadays. Numerous studies have confirmed that uric acid plays an important role in CI, but the mechanism has not yet been clarified. When treating HT22 and BV-2 cells with different concentrations of uric acid, uric acid below 450 μM does not have significant effect on cell viability, but uric acid more than 500 μM can significantly inhibit cell viability. After establishing models of OGD (oxygen-glucose deprivation) with HT22 and BV-2 cells, uric acid at a low concentration (50 μM) cannot improve cell viability and apoptosis, and Reactive oxygen species (ROS) levels during OGD/reoxygenation; a suitable concentration (300 μM) of uric acid can significantly improve cell viability and apoptosis, and reduce ROS production during OGD/reoxygenation; but a high concentration (1000 μM) of uric acid can further reduce cell viability and enhance ROS production. After establishing middle cerebral artery occlusion of male rats with suture method, damage and increase of ROS production in brain tissue could be seen, and after adding suitable concentration of uric acid, the degree of brain damage and ROS production was reduced. Therefore, different concentrations of uric acid should have different effect, and suitable concentrations of uric acid have neuroprotective effect, and this finding may provide guidance for study on the clinical curative effect of uric acid.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology, The Fifth Affiliated Hospital of GuangZhou Medical University, 621# Harbour Road, Whampoa District, Guangzhou, 510700, Guangdong Province, China
| | - Ning Yang
- Department of Neurology, The Fifth Affiliated Hospital of GuangZhou Medical University, 621# Harbour Road, Whampoa District, Guangzhou, 510700, Guangdong Province, China.
| | - Shao-Peng Lin
- Department of Emergency, The Second Affiliated Hospital of GuangZhou Medical University, Guangzhou, 510260, Guangdong Province, China
| | - Feng Zhang
- Department of Neurosurgery, The Fourth Affiliated Hospital of GuangZhou Medical University, Guangzhou, 511447, Guangdong Province, China
| |
Collapse
|
58
|
Li Z, Chen H, Lv J, Zhao R. The application and neuroprotective mechanisms of cerebral ischemic post-conditioning: A review. Brain Res Bull 2017; 131:39-46. [DOI: 10.1016/j.brainresbull.2017.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/06/2017] [Indexed: 01/17/2023]
|
59
|
Chen BH, Park JH, Ahn JH, Cho JH, Kim IH, Lee JC, Won MH, Lee CH, Hwang IK, Kim JD, Kang IJ, Cho JH, Shin BN, Kim YH, Lee YL, Park SM. Pretreated quercetin protects gerbil hippocampal CA1 pyramidal neurons from transient cerebral ischemic injury by increasing the expression of antioxidant enzymes. Neural Regen Res 2017; 12:220-227. [PMID: 28400803 PMCID: PMC5361505 DOI: 10.4103/1673-5374.200805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Quercetin (QE; 3,5,7,3′,4′-pentahydroxyflavone), a well-known flavonoid, has been shown to prevent against neurodegenerative disorders and ischemic insults. However, few studies are reported regarding the neuroprotective mechanisms of QE after ischemic insults. Therefore, in this study, we investigated the effects of QE on ischemic injury and the expression of antioxidant enzymes in the hippocampal CA1 region of gerbils subjected to 5 minutes of transient cerebral ischemia. QE was pre-treated once daily for 15 days before ischemia. Pretreatment with QE protected hippocampal CA1 pyramidal neurons from ischemic injury, which was confirmed by neuronal nuclear antigen immunohistochemistry and Fluoro-Jade B histofluorescence staining. In addition, pretreatment with QE significantly increased the expression levels of endogenous antioxidant enzymes Cu/Zn superoxide dismutase, Mn superoxide dismutase, catalase and glutathione peroxidase in the hippocampal CA1 pyramidal neurons of animals with ischemic injury. These findings demonstrate that pretreated QE displayed strong neuroprotective effects against transient cerebral ischemia by increasing the expression of antioxidant enzymes.
Collapse
Affiliation(s)
- Bai Hui Chen
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Bich Na Shin
- Department of Physiology, College of Medicine, and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon, South Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yun Lyul Lee
- Department of Physiology, College of Medicine, and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon, South Korea
| | - Seung Min Park
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea
| |
Collapse
|
60
|
Li W, Yang S. Targeting oxidative stress for the treatment of ischemic stroke: Upstream and downstream therapeutic strategies. Brain Circ 2016; 2:153-163. [PMID: 30276293 PMCID: PMC6126224 DOI: 10.4103/2394-8108.195279] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022] Open
Abstract
Excessive oxygen and its chemical derivatives, namely reactive oxygen species (ROS), produce oxidative stress that has been known to lead to cell injury in ischemic stroke. ROS can damage macromolecules such as proteins and lipids and leads to cell autophagy, apoptosis, and necrosis to the cells. This review describes studies on the generation of ROS, its role in the pathogenesis of ischemic stroke, and recent development in therapeutic strategies in reducing oxidative stress after ischemic stroke.
Collapse
Affiliation(s)
- Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shaohua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
61
|
Yao Y, Miao W, Liu Z, Han W, Shi K, Shen Y, Li H, Liu Q, Fu Y, Huang D, Shi FD. Dimethyl Fumarate and Monomethyl Fumarate Promote Post-Ischemic Recovery in Mice. Transl Stroke Res 2016; 7:535-547. [PMID: 27614618 PMCID: PMC5065588 DOI: 10.1007/s12975-016-0496-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 01/21/2023]
Abstract
Oxidative stress plays an important role in cerebral ischemia-reperfusion injury. Dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) are antioxidant agents that can activate the nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway and induce the expression of antioxidant proteins. Here, we evaluated the impact of DMF and MMF on ischemia-induced brain injury and whether the Nrf2 pathway mediates the effects provided by DMF and MMF in cerebral ischemia-reperfusion injury. Using a mouse model of transient focal brain ischemia, we show that DMF and MMF significantly reduce neurological deficits, infarct volume, brain edema, and cell death. Further, DMF and MMF suppress glial activation following brain ischemia. Importantly, the protection of DMF and MMF was mostly evident during the subacute stage and was abolished in Nrf2-/- mice, indicating that the Nrf2 pathway is required for the beneficial effects of DMF and MMF. Together, our data indicate that DMF and MMF have therapeutic potential in cerebral ischemia-reperfusion injury and their protective role is likely mediated by the Nrf2 pathway.
Collapse
Affiliation(s)
- Yang Yao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Weimin Miao
- The State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Zhijia Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wei Han
- Department of Radiology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Kaibin Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yi Shen
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Handong Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Ying Fu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - DeRen Huang
- Neurology and Neuroscience Associates, Unity Health Network, Akron, OH, USA
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA.
| |
Collapse
|
62
|
Simões D, Riva P, Peliciari-Garcia RA, Cruzat VF, Graciano MF, Munhoz AC, Taneda M, Cipolla-Neto J, Carpinelli AR. Melatonin modifies basal and stimulated insulin secretion via NADPH oxidase. J Endocrinol 2016; 231:235-244. [PMID: 27803236 DOI: 10.1530/joe-16-0259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/04/2016] [Indexed: 01/10/2023]
Abstract
Melatonin is a hormone synthesized in the pineal gland, which modulates several functions within the organism, including the synchronization of glucose metabolism and glucose-stimulated insulin secretion (GSIS). Melatonin can mediate different signaling pathways in pancreatic islets through two membrane receptors and via antioxidant or pro-oxidant enzymes modulation. NADPH oxidase (NOX) is a pro-oxidant enzyme responsible for the production of the reactive oxygen specie (ROS) superoxide, generated from molecular oxygen. In pancreatic islets, NOX-derived ROS can modulate glucose metabolism and regulate insulin secretion. Considering the roles of both melatonin and NOX in islets, the aim of this study was to evaluate the association of NOX and ROS production on glucose metabolism, basal and GSIS in pinealectomized rats (PINX) and in melatonin-treated isolated pancreatic islets. Our results showed that ROS content derived from NOX activity was increased in PINX at baseline (2.8 mM glucose), which was followed by a reduction in glucose metabolism and basal insulin secretion in this group. Under 16.7 mM glucose, an increase in both glucose metabolism and GSIS was observed in PINX islets, without changes in ROS content. In isolated pancreatic islets from control animals incubated with 2.8 mM glucose, melatonin treatment reduced ROS content, whereas in 16.7 mM glucose, melatonin reduced ROS and GSIS. In conclusion, our results demonstrate that both basal and stimulated insulin secretion can be regulated by melatonin through the maintenance of ROS homeostasis in pancreatic islets.
Collapse
Affiliation(s)
- Daniel Simões
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Patrícia Riva
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Antonio Peliciari-Garcia
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
- Department of Biological SciencesLaboratory of Biosystems, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Vinicius Fernandes Cruzat
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Maria Fernanda Graciano
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Munhoz
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Marco Taneda
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - José Cipolla-Neto
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
63
|
Zhang R, Xu M, Wang Y, Xie F, Zhang G, Qin X. Nrf2—a Promising Therapeutic Target for Defensing Against Oxidative Stress in Stroke. Mol Neurobiol 2016; 54:6006-6017. [DOI: 10.1007/s12035-016-0111-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 09/06/2016] [Indexed: 12/30/2022]
|
64
|
Coucha M, Abdelsaid M, Li W, Johnson MH, Orfi L, El-Remessy AB, Fagan SC, Ergul A. Nox4 contributes to the hypoxia-mediated regulation of actin cytoskeleton in cerebrovascular smooth muscle. Life Sci 2016; 163:46-54. [PMID: 27558234 DOI: 10.1016/j.lfs.2016.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/12/2016] [Accepted: 08/19/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Ischemia/reperfusion and the resulting oxidative/nitrative stress impair cerebral myogenic tone via actin depolymerization. While it is known that NADPH oxidase (Nox) family is a major source of vascular oxidative stress; the extent and mechanisms by which Nox activation contributes to actin depolymerization, and equally important, the relative role of Nox isoforms in this response is not clear. AIM To determine the role of Nox4 in hypoxia-mediated actin depolymerization and myogenic-tone impairment in cerebral vascular smooth muscle. MAIN METHODS Control and Nox4 deficient (siRNA knock-down) human brain vascular smooth muscle cells (HBVSMC) were exposed to 30-min hypoxia/45-min reoxygenation. Nox2, Nox4, inducible and neuronal nitric oxide synthase (iNOS and nNOS) and nitrotyrosine levels as well as F:G actin were determined. Myogenic-tone was measured using pressurized arteriography in middle cerebral artery isolated from rats subjected to sham, 30-min ischemia/45-min reperfusion or ex-vivo oxygen glucose deprivation in the presence and absence of Nox inhibitors. RESULTS Nox4 and iNOS expression were significantly upregulated following hypoxia or ischemia/reperfusion. Hypoxia augmented nitrotyrosine levels while reducing F actin. These effects were nullified by inhibiting nitration with epicatechin or pharmacological or molecular inhibition of Nox4. Ischemia/reperfusion impaired myogenic-tone, which was restored by the selective inhibition of Nox4. CONCLUSION Nox4 activation in VSMCs contributes to actin depolymerization after hypoxia, which could be the underlying mechanism for myogenic-tone impairment following ischemia/reperfusion.
Collapse
Affiliation(s)
- Maha Coucha
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Mohammed Abdelsaid
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | - Weiguo Li
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | | | | | - Azza B El-Remessy
- Charlie Norwood VA Medical Center, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Susan C Fagan
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Neurology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States.
| |
Collapse
|
65
|
Brennan GP, Dey D, Chen Y, Patterson KP, Magnetta EJ, Hall AM, Dube CM, Mei YT, Baram TZ. Dual and Opposing Roles of MicroRNA-124 in Epilepsy Are Mediated through Inflammatory and NRSF-Dependent Gene Networks. Cell Rep 2016; 14:2402-12. [PMID: 26947066 DOI: 10.1016/j.celrep.2016.02.042] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 12/22/2015] [Accepted: 02/04/2016] [Indexed: 12/25/2022] Open
Abstract
Insult-provoked transformation of neuronal networks into epileptic ones involves multiple mechanisms. Intervention studies have identified both dysregulated inflammatory pathways and NRSF-mediated repression of crucial neuronal genes as contributors to epileptogenesis. However, it remains unclear how epilepsy-provoking insults (e.g., prolonged seizures) induce both inflammation and NRSF and whether common mechanisms exist. We examined miR-124 as a candidate dual regulator of NRSF and inflammatory pathways. Status epilepticus (SE) led to reduced miR-124 expression via SIRT1--and, in turn, miR-124 repression--via C/EBPα upregulated NRSF. We tested whether augmenting miR-124 after SE would abort epileptogenesis by preventing inflammation and NRSF upregulation. SE-sustaining animals developed epilepsy, but supplementing miR-124 did not modify epileptogenesis. Examining this result further, we found that synthetic miR-124 not only effectively blocked NRSF upregulation and rescued NRSF target genes, but also augmented microglia activation and inflammatory cytokines. Thus, miR-124 attenuates epileptogenesis via NRSF while promoting epilepsy via inflammation.
Collapse
Affiliation(s)
- Gary P Brennan
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Deblina Dey
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Katelin P Patterson
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Eric J Magnetta
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Alicia M Hall
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Celine M Dube
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yu-Tang Mei
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
66
|
Fuentes E, Palomo I. Role of oxidative stress on platelet hyperreactivity during aging. Life Sci 2016; 148:17-23. [PMID: 26872977 DOI: 10.1016/j.lfs.2016.02.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 12/13/2022]
Abstract
Thrombotic events are common causes of morbidity and mortality in the elderly. Age-accelerated vascular injury is commonly considered to result from increased oxidative stress. There is abundant evidence that oxidative stress regulate several components of thrombotic processes, including platelet activation. Thus oxidative stress can trigger platelet hyperreactivity by decreasing nitric oxide bioavailability. Therefore oxidative stress measurement may help in the early identification of asymptomatic subjects at risk of thrombosis. In addition, oxidative stress inhibitors and platelet-derived nitric oxide may represent a novel anti-aggregation/-activation approach. In this article the relative contribution of oxidative stress and platelet activation in aging is explored.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Chile.
| |
Collapse
|
67
|
Lushchak VI. Time-course and intensity-based classifications of oxidative stresses and their potential application in biomedical, comparative and environmental research. Redox Rep 2016; 21:262-70. [PMID: 26828292 DOI: 10.1080/13510002.2015.1126940] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE We propose some clues for classification of oxidative stresses based on their intensity and time-course. BACKGROUND Oxidative stress is studied for more than three decades and it is clear that it may differ on the parameters of interest. But up to now there is no any system for formal discrimination between different types of the stress. Such approach can provide important benefits at description of experimental data. METHOD We briefly review information on oxidative stresses and show that the theoretical concept is actually poorly developed since introduction of the first definition in 1985 by H. Sies. We argue that the stresses can differ on their intensities and time-curses, but there was no theoretical basis for discrimination between them. RESULTS On the basis of these analyses, we propose two systems of classifications of oxidative stresses enabling their description taking into account their intensity and time-course. We analyze essential biomarkers of oxidative stress to be used for classification such as levels of modified by reactive oxygen species proteins, lipids, nucleic acids, and low molecular mass compounds. Finally, we describe potential applications of the proposed classifications to biomedical, comparative and environmental research. CONCLUSION The proposed classifications of oxidative stress may facilitate description of experimental data and their comparison between different organisms and methods of induction of oxidative stresses. Additionally this work may provide some clues to develop quantitative approaches for formal categorization of oxidative stresses. APPLICATION Most applications of the classifications proposed are theoretical and applied studies where oxidative stress takes place.
Collapse
Affiliation(s)
- Volodymyr I Lushchak
- a Department of Biochemistry and Biotechnology , Vasyl Stefanyk Precarpathian National University , 57 Shevchenko Str., Ivano-Frankivsk 76018 , Ukraine
| |
Collapse
|
68
|
Suzuki Y, Nagai N, Umemura K. A Review of the Mechanisms of Blood-Brain Barrier Permeability by Tissue-Type Plasminogen Activator Treatment for Cerebral Ischemia. Front Cell Neurosci 2016; 10:2. [PMID: 26834557 PMCID: PMC4724711 DOI: 10.3389/fncel.2016.00002] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/04/2016] [Indexed: 11/13/2022] Open
Abstract
Cerebrovascular homeostasis is maintained by the blood-brain barrier (BBB), which forms a mechanical and functional barrier between systemic circulation and the central nervous system (CNS). In patients with ischemic stroke, the recombinant tissue-type plasminogen activator (rt-PA) is used to accelerate recanalization of the occluded vessels. However, rt-PA is associated with a risk of increasing intracranial bleeding (ICB). This effect is thought to be caused by the increase in cerebrovascular permeability though various factors such as ischemic reperfusion injury and the activation of matrix metalloproteinases (MMPs), but the detailed mechanisms are unknown. It was recently found that rt-PA treatment enhances BBB permeability not by disrupting the BBB, but by activating the vascular endothelial growth factor (VEGF) system. The VEGF regulates both the dissociation of endothelial cell (EC) junctions and endothelial endocytosis, and causes a subsequent increase in vessel permeability through the VEGF receptor-2 (VEGFR-2) activation in ECs. Here, we review the possibility that rt-PA increases the penetration of toxic molecules derived from the bloodstream including rt-PA itself, without disrupting the BBB, and contributes to these detrimental processes in the cerebral parenchyma.
Collapse
Affiliation(s)
- Yasuhiro Suzuki
- Department of Pharmacology, Hamamatsu University School of MedicineHamamatsu, Japan; School of Pharmaceutical Sciences, Ohu UniversityKoriyama, Japan
| | - Nobuo Nagai
- Faculty of Bioscience, Department of Animal Bioscience, Nagahama Institute of Bio-Science and Technology Nagahama, Japan
| | - Kazuo Umemura
- Department of Pharmacology, Hamamatsu University School of Medicine Hamamatsu, Japan
| |
Collapse
|
69
|
Nakka VP, Prakash-Babu P, Vemuganti R. Crosstalk Between Endoplasmic Reticulum Stress, Oxidative Stress, and Autophagy: Potential Therapeutic Targets for Acute CNS Injuries. Mol Neurobiol 2016; 53:532-544. [PMID: 25482050 PMCID: PMC4461562 DOI: 10.1007/s12035-014-9029-6] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/30/2014] [Indexed: 01/06/2023]
Abstract
Endoplasmic reticulum (ER) stress induces a variety of neuronal cell death pathways that play a critical role in the pathophysiology of stroke. ER stress occurs when unfolded/misfolded proteins accumulate and the folding capacity of ER chaperones exceeds the capacity of ER lumen to facilitate their disposal. As a consequence, a complex set of signaling pathways will be induced that transmit from ER to cytosol and nucleus to compensate damage and to restore the normal cellular homeostasis, collectively known as unfolded protein response (UPR). However, failure of UPR due to severe or prolonged stress leads to cell death. Following acute CNS injuries, chronic disturbances in protein folding and oxidative stress prolong ER stress leading to sustained ER dysfunction and neuronal cell death. While ER stress responses have been well studied after stroke, there is an emerging need to study the association of ER stress with other cell pathways that exacerbate neuronal death after an injury. In this review, we summarize the current understanding of the role for ER stress in acute brain injuries, highlighting the diverse molecular mechanisms associated with ER stress and its relation to oxidative stress and autophagy. We also discussed the existing and developing therapeutic options aimed to reduce ER stress to protect the CNS after acute injuries.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
- Department of Biotechnology & Bioinformatics, School of Life sciences, University of Hyderabad, Hyderabad, India
| | - Phanithi Prakash-Babu
- Department of Biotechnology & Bioinformatics, School of Life sciences, University of Hyderabad, Hyderabad, India
| | - Raghu Vemuganti
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
70
|
Almutairi MMA, Gong C, Xu YG, Chang Y, Shi H. Factors controlling permeability of the blood-brain barrier. Cell Mol Life Sci 2016; 73:57-77. [PMID: 26403789 PMCID: PMC11108286 DOI: 10.1007/s00018-015-2050-8] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 12/27/2022]
Abstract
As the primary protective barrier for neurons in the brain, the blood-brain barrier (BBB) exists between the blood microcirculation system and the brain parenchyma. The normal BBB integrity is essential in protecting the brain from systemic toxins and maintaining the necessary level of nutrients and ions for neuronal function. This integrity is mediated by structural BBB components, such as tight junction proteins, integrins, annexins, and agrin, of a multicellular system including endothelial cells, astrocytes, pericytes, etc. BBB dysfunction is a significant contributor to the pathogeneses of a variety of brain disorders. Many signaling factors have been identified to be able to control BBB permeability through regulating the structural components. Among those signaling factors are inflammatory mediators, free radicals, vascular endothelial growth factor, matrix metalloproteinases, microRNAs, etc. In this review, we provide a summary of recent progress regarding these structural components and signaling factors, relating to their roles in various brain disorders. Attention is also devoted to recent research regarding impact of pharmacological agents such as isoflurane on BBB permeability and how iron ion passes across BBB. Hopefully, a better understanding of the factors controlling BBB permeability helps develop novel pharmacological interventions of BBB hyperpermeability under pathological conditions.
Collapse
Affiliation(s)
- Mohammed M A Almutairi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS, 66045, USA
| | - Chen Gong
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS, 66045, USA
| | - Yuexian G Xu
- Department of Anesthesiology, School of Medicine, University of Kansas, Kansas City, KS, 66160, USA
| | - Yanzhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, 050016, China
| | - Honglian Shi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS, 66045, USA.
| |
Collapse
|
71
|
Cardiovascular and Hepatic Toxicity of Cocaine: Potential Beneficial Effects of Modulators of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:8408479. [PMID: 26823954 PMCID: PMC4707355 DOI: 10.1155/2016/8408479] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/19/2015] [Accepted: 11/01/2015] [Indexed: 12/20/2022]
Abstract
Oxidative stress (OS) is thought to play an important role in the pharmacological and toxic effects of various drugs of abuse. Herein we review the literature on the mechanisms responsible for the cardiovascular and hepatic toxicity of cocaine with special focus on OS-related mechanisms. We also review the preclinical and clinical literature concerning the putative therapeutic effects of OS modulators (such as N-acetylcysteine, superoxide dismutase mimetics, nitroxides and nitrones, NADPH oxidase inhibitors, xanthine oxidase inhibitors, and mitochondriotropic antioxidants) for the treatment of cocaine toxicity. We conclude that available OS modulators do not appear to have clinical efficacy.
Collapse
|
72
|
Shi GX, Wang XR, Yan CQ, He T, Yang JW, Zeng XH, Xu Q, Zhu W, Du SQ, Liu CZ. Acupuncture elicits neuroprotective effect by inhibiting NAPDH oxidase-mediated reactive oxygen species production in cerebral ischaemia. Sci Rep 2015; 5:17981. [PMID: 26656460 PMCID: PMC4674709 DOI: 10.1038/srep17981] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 11/10/2015] [Indexed: 11/09/2022] Open
Abstract
In the current study, we aimed to investigate whether NADPH oxidase, a major ROS-producing enzyme, was involved in the antioxidant effect of acupuncture on cognitive impairment after cerebral ischaemia. The cognitive function, infract size, neuron cell loss, level of superoxide anion and expression of NADPH oxidase subunit in hippocampus of two-vessel occlusion (2VO) rats were determined after 2-week acupuncture. Furthermore, the cognitive function and production of O2(-) were determined in the presence and absence of NADPH oxidase agonist (TBCA) and antagonist (Apocynin). The effect of acupuncture on cognitive function after cerebral ischaemia in gp91phox-KO mice was evaluated by Morris water maze. Acupuncture reduced infarct size, attenuated overproduction of O2(-), and reversed consequential cognitive impairment and neuron cell loss in 2VO rats. The elevations of gp91phox and p47phox after 2VO were significantly decreased after acupuncture treatment. However, no differences of gp91phox mRNA were found among any experimental groups. Furthermore, these beneficial effects were reversed by TBCA, whereas apocynin mimicked the effect of acupuncture by improving cognitive function and decreasing O2(-) generation. Acupuncture failed to improve the memory impairment in gp91phox KO mice. Full function of the NADPH oxidase enzyme plays an important role in neuroprotective effects against cognitive impairment via inhibition of NAPDH oxidase-mediated oxidative stress.
Collapse
Affiliation(s)
- Guang-Xia Shi
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Chao-Qun Yan
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Jing-Wen Yang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Xiang-Hong Zeng
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Qian Xu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Wen Zhu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| |
Collapse
|
73
|
Carbone F, Teixeira PC, Braunersreuther V, Mach F, Vuilleumier N, Montecucco F. Pathophysiology and Treatments of Oxidative Injury in Ischemic Stroke: Focus on the Phagocytic NADPH Oxidase 2. Antioxid Redox Signal 2015; 23:460-489. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/05/2014] [Accepted: 03/16/2014] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Phagocytes play a key role in promoting the oxidative stress after ischemic stroke occurrence. The phagocytic NADPH oxidase (NOX) 2 is a membrane-bound enzyme complex involved in the antimicrobial respiratory burst and free radical production in these cells. RECENT ADVANCES Different oxidants have been shown to induce opposite effects on neuronal homeostasis after a stroke. However, several experimental models support the detrimental effects of NOX activity (especially the phagocytic isoform) on brain recovery after stroke. Therapeutic strategies selectively targeting the neurotoxic ROS and increasing neuroprotective oxidants have recently produced promising results. CRITICAL ISSUES NOX2 might promote carotid plaque rupture and stroke occurrence. In addition, NOX2-derived reactive oxygen species (ROS) released by resident and recruited phagocytes enhance cerebral ischemic injury, activating the inflammatory apoptotic pathways. The aim of this review is to update evidence on phagocyte-related oxidative stress, focusing on the role of NOX2 as a potential therapeutic target to reduce ROS-related cerebral injury after stroke. FUTURE DIRECTIONS Radical scavenger compounds (such as Ebselen and Edaravone) are under clinical investigation as a therapeutic approach against stroke. On the other hand, NOX inhibition might represent a promising strategy to prevent the stroke-related injury. Although selective NOX inhibitors are not yet available, nonselective compounds (such as apocynin and fasudil) provided encouraging results in preclinical studies. Whereas additional studies are needed to better evaluate this therapeutic potential in human beings, the development of specific NOX inhibitors (such as monoclonal antibodies, small-molecule inhibitors, or aptamers) might further improve brain recovery after stroke.
Collapse
Affiliation(s)
- Federico Carbone
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Priscila Camillo Teixeira
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
74
|
Lu XY, Wang HD, Xu JG, Ding K, Li T. Deletion of Nrf2 Exacerbates Oxidative Stress After Traumatic Brain Injury in Mice. Cell Mol Neurobiol 2015; 35:713-21. [PMID: 25732597 PMCID: PMC11486234 DOI: 10.1007/s10571-015-0167-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is a worldwide public health and medical problem. Oxidative stress is recognized as an important contributing factor in the pathogenesis of TBI. The present study was designed to explore the anti-oxidative effect of Nuclear factor erythroid 2-related factor 2 (Nrf2) on brain damage induced by traumatic injury in a mouse model. Moderate weight-drop impact head injury was induced in adult male mice. The mice were randomly divided into four groups: Nrf2(+/+) sham-operation, Nrf2(-/-) sham-operation, Nrf2(+/+) TBI, and Nrf2(-/-) TBI group. Neurological scores were evaluated 24 h after TBI, followed by collection of the brain specimens. Brain edema was detected by the wet-dry ratio method. The expression of NOX2 protein in the brain specimen was investigated using Western Blot analysis and immunohistochemical staining. In addition, malondialdehyde (MDA) level and superoxide dismutase (SOD) activity were evaluated in the brain tissues. Twenty-four hours after TBI, our results showed Nrf2(+/+) TBI mice have more severe neurological deficits and brain edema than Nrf2(+/+) sham group. On the other hand, the Nrf2(-/-) TBI mice were found to have significantly increased neurological deficits and brain edema, compared to Nrf2(+/+) TBI mice (P < 0.05). At the same time, we found that the expression of NOX2 protein, MDA level were significantly increased in Nrf2(-/-) mice, while SOD activity was considerably decreased after TBI compared to Nrf2(+/+) mice (P < 0.05). We demonstrated that deletion of Nrf2 exacerbates brain injury after TBI in mice, suggesting that Nrf2 may play an important role in protecting brain injury after TBI, possibly by modulating oxidative stress.
Collapse
Affiliation(s)
- Xin-Yu Lu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Han-Dong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Jian-Guo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| |
Collapse
|
75
|
Wu JS, Tsai HD, Cheung WM, Hsu CY, Lin TN. PPAR-γ Ameliorates Neuronal Apoptosis and Ischemic Brain Injury via Suppressing NF-κB-Driven p22phox Transcription. Mol Neurobiol 2015; 53:3626-3645. [PMID: 26108185 DOI: 10.1007/s12035-015-9294-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-γ), a stress-induced transcription factor, protects neurons against ischemic stroke insult by reducing oxidative stress. NADPH oxidase (NOX) activation, a major driving force in ROS generation in the setting of reoxygenation/reperfusion, constitutes an important pathogenetic mechanism of ischemic brain damage. In the present study, both transient in vitro oxygen-glucose deprivation and in vivo middle cerebral artery (MCA) occlusion-reperfusion experimental paradigms of ischemic neuronal death were used to investigate the interaction between PPAR-γ and NOX. With pharmacological (PPAR-γ antagonist GW9662), loss-of-function (PPAR-γ siRNA), and gain-of-function (Ad-PPAR-γ) approaches, we first demonstrated that 15-deoxy-∆(12,14)-PGJ2 (15d-PGJ2), via selectively attenuating p22phox expression, inhibited NOX activation and the subsequent ROS generation and neuronal death in a PPAR-γ-dependent manner. Secondly, results of promoter analyses and subcellular localization studies further revealed that PPAR-γ, via inhibiting hypoxia-induced NF-κB nuclear translocation, indirectly suppressed NF-κB-driven p22phox transcription. Noteworthily, postischemic p22phox siRNA treatment not only reduced infarct volumes but also improved functional outcome. In summary, we report a novel transrepression mechanism involving PPAR-γ downregulation of p22phox expression to suppress the subsequent NOX activation, ischemic neuronal death, and brain infarct. Identification of a PPAR-γ → NF-κB → p22phox neuroprotective signaling cascade opens a new avenue for protecting the brain against ischemic insult.
Collapse
Affiliation(s)
- Jui-Sheng Wu
- Institute of Biomedical Sciences, Academia Sinica, Rm 404, Taipei, 11529, Taiwan, Republic of China
| | - Hsin-Da Tsai
- Institute of Biomedical Sciences, Academia Sinica, Rm 404, Taipei, 11529, Taiwan, Republic of China
| | - Wai-Mui Cheung
- Institute of Biomedical Sciences, Academia Sinica, Rm 404, Taipei, 11529, Taiwan, Republic of China
| | - Chung Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Rm 404, Taipei, 11529, Taiwan, Republic of China. .,Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan. .,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
76
|
Tang H, Pan CS, Mao XW, Liu YY, Yan L, Zhou CM, Fan JY, Zhang SY, Han JY. Role of NADPH oxidase in total salvianolic acid injection attenuating ischemia-reperfusion impaired cerebral microcirculation and neurons: implication of AMPK/Akt/PKC. Microcirculation 2015; 21:615-27. [PMID: 24702968 DOI: 10.1111/micc.12140] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/02/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE TSI is a new drug derived from Chinese medicine for treatment of ischemic stroke in China. The aim of this study was to verify the therapeutic effect of TSI in a rat model of MCAO, and further explore the mechanism for its effect. METHODS Male Sprague-Dawley rats were subjected to right MCAO for 60 minutes followed by reperfusion. TSI (1.67 mg/kg) was administrated before reperfusion via femoral vein injection. Twenty-four hours after reperfusion, the fluorescence intensity of DHR 123 in, leukocyte adhesion to and albumin leakage from the cerebral venules were observed. Neurological scores, TTC staining, brain water content, Nissl staining, TUNEL staining, and MDA content were assessed. Bcl-2/Bax, cleaved caspase-3, NADPH oxidase subunits p47(phox)/p67(phox)/gp91(phox), and AMPK/Akt/PKC were analyzed by Western blot. RESULTS TSI attenuated I/R-induced microcirculatory disturbance and neuron damage, activated AMPK, inhibited NADPH oxidase subunits membrane translocation, suppressed Akt phosphorylation, and PKC translocation. CONCLUSIONS TSI attenuates I/R-induced brain injury in rats, supporting its clinic use for treatment of acute ischemic stroke. The role of TSI may benefit from its antioxidant activity, which is most likely implemented via inactivation of NADPH oxidase through a signaling pathway implicating AMPK/Akt/PKC.
Collapse
Affiliation(s)
- Hao Tang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Harrison IP, Selemidis S. Understanding the biology of reactive oxygen species and their link to cancer: NADPH oxidases as novel pharmacological targets. Clin Exp Pharmacol Physiol 2015; 41:533-42. [PMID: 24738947 DOI: 10.1111/1440-1681.12238] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS), the cellular products of myriad physiological processes, have long been understood to lead to cellular damage if produced in excess and to be a causative factor in cancer through the oxidation and nitration of various macromolecules. Reactive oxygen species influence various hallmarks of cancer, such as cellular proliferation and angiogenesis, through the promotion of cell signalling pathways intrinsic to these processes and can also regulate the function of key immune cells, such as macrophages and regulatory T cells, which promote angiogenesis in the tumour environment. Herein we emphasize the family of NADPH oxidase enzymes as the most likely source of ROS, which promote angiogenesis and tumourigenesis through signalling pathways within endothelial, immune and tumour cells. In this review we focus on the pharmacological inhibitors of NADPH oxidases and suggest that, compared with traditional anti-oxidants, they are likely to offer better alternatives for suppression of tumour angiogenesis. Despite the emerging enthusiasm towards the use of NADPH oxidase inhibitors for cancer therapy, this field is still in its infancy; in particular, there is a glaring lack of knowledge of the roles of NADPH oxidases in in vivo animal models and in human cancers. Certainly a clearer understanding of the relevant signalling pathways influenced by NADPH oxidases during angiogenesis in cancer is likely to yield novel therapeutic approaches.
Collapse
Affiliation(s)
- Ian P Harrison
- Department of Pharmacology, Monash University, Melbourne, Vic., Australia
| | | |
Collapse
|
78
|
Yu J, Zhu H, Gattoni-Celli S, Taheri S, Kindy MS. Dietary supplementation of GrandFusion(®) mitigates cerebral ischemia-induced neuronal damage and attenuates inflammation. Nutr Neurosci 2015; 19:290-300. [PMID: 25879584 DOI: 10.1179/1476830515y.0000000021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Dietary supplementation of fruits and vegetables has been the main stay for nutritional benefit and overall well-being. GrandFusion(®) is a nutritional supplement that contains the natural nutrients from whole fruits and vegetables that include complex nutrients and phytonutrients that contain anti-oxidant, anti-inflammatory, and neuroprotective properties. METHODS In this study, C57BL/6 mice were fed a diet supplemented with GrandFusion(®) for 2 months prior to 1 hour of ischemia induced by occlusion of the middle cerebral artery (MCAo) followed by various times of reperfusion. Mice were subjected to MCAo for 1 hour and then at various times following reperfusion, animals were assessed for behavioral outcomes (open field testing, rotarod, and adhesive test removal), and infarct volumes (cresyl violet and triphenyltetrazolium chloride). In addition, to determine the potential mechanisms associated with treatment, the brain tissue was examined for changes in oxidative stress and inflammatory markers. RESULTS The GrandFusion(®) diet was able to show a significant protection from infarct damage in the brain and an improvement in neurological outcomes. The diet did not alter heart rate, blood pressure, pO2, pCO2, or pH. In addition, the diet mitigated inflammation by reducing microglial and astrocytic activation following ischemia and reperfusion and limiting oxidative stress. DISCUSSION The study demonstrates the neuroprotective effect of a diet rich in fruits and vegetables that contain anti-oxidant and anti-inflammatory against the impact of cerebral ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Jin Yu
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Hong Zhu
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Sebastiano Gattoni-Celli
- b Department of Radiation Oncology , Medical University of South Carolina , Charleston , SC , USA.,c Ralph H. Johnson VA Medical Center , Charleston , SC , USA
| | - Saeid Taheri
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Mark Stephen Kindy
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA.,c Ralph H. Johnson VA Medical Center , Charleston , SC , USA
| |
Collapse
|
79
|
Abstract
Understanding innate immune responses and their correlation to alloimmunity after solid organ transplantation is key to optimizing long term graft outcome. While Ischemia/Reperfusion injury (IRI) has been well studied, new insight into central mechanisms of innate immune activation, i.e. chemokine mediated cell trafficking and the role of Toll-like receptors have evolved recently. The mechanistic implications of Neutrophils, Macrophages/Monocytes, NK-cells, Dendritic cells in renal IRI has been proven by selective depletion of these cell types, thereby offering novel therapeutic interventions. At the same time, the multi-faceted role of different T-cell subsets in IRI has gained interest, highlighting the dichotomous effects of differentiated T-cells and suggesting more selective therapeutic approaches. Targeting innate immune cells and their activation and migration pathways, respectively, has been promising in experimental models holding translational potential. This review will summarize the effects of innate immune activation and potential strategies to interfere with the immunological cascade following renal IRI.
Collapse
|
80
|
Forghani R, Kim HJ, Wojtkiewicz GR, Bure L, Wu Y, Hayase M, Wei Y, Zheng Y, Moskowitz MA, Chen JW. Myeloperoxidase propagates damage and is a potential therapeutic target for subacute stroke. J Cereb Blood Flow Metab 2015; 35:485-93. [PMID: 25515211 PMCID: PMC4348390 DOI: 10.1038/jcbfm.2014.222] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 11/05/2014] [Indexed: 11/09/2022]
Abstract
Few effective treatment options exist for stroke beyond the hyperacute period. Radical generation and myeloperoxidase (MPO) have been implicated in stroke. We investigated whether pharmacologic reduction or gene deletion of this highly oxidative enzyme reduces infarct propagation and improves outcome in the transient middle cerebral artery occlusion mouse model (MCAO). Mice were treated with 4-aminobenzoic acid hydrazide (ABAH), a specific irreversible MPO inhibitor. Three treatment regimens were used: (1) daily throughout the 21-day observational period, (2) during the acute stage (first 24 hours), or (3) during the subacute stage (daily starting on day 2). We found elevated MPO activity in ipsilateral brain 3 to 21 days after ischemia. 4-Aminobenzoic acid hydrazide reduced enzyme activity by 30% to 40% and final lesion volume by 60% (P<0.01). The MPO-knockout (KO) mice subjected to MCAO also showed a similar reduction in the final lesion volume (P<0.01). The ABAH treatment or MPO-KO mice also improved neurobehavioral outcome (P<0.001) and survival (P=0.01), but ABAH had no additional beneficial effects in MPO-KO mice, confirming specificity of ABAH. Interestingly, inhibiting MPO activity during the subacute stage recapitulated most of the therapeutic benefit of continuous MPO inhibition, suggesting that MPO-targeted therapies could be useful when given after 24 hours of stroke onset.
Collapse
Affiliation(s)
- Reza Forghani
- 1] Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA [2] Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hyeon Ju Kim
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory R Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lionel Bure
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yue Wu
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Makoto Hayase
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ying Wei
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Zheng
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael A Moskowitz
- 1] Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA [2] Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John W Chen
- 1] Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA [2] Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
81
|
Manea A, Manea SA, Todirita A, Albulescu IC, Raicu M, Sasson S, Simionescu M. High-glucose-increased expression and activation of NADPH oxidase in human vascular smooth muscle cells is mediated by 4-hydroxynonenal-activated PPARα and PPARβ/δ. Cell Tissue Res 2015; 361:593-604. [PMID: 25722086 DOI: 10.1007/s00441-015-2120-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 12/29/2014] [Indexed: 01/04/2023]
Abstract
High glucose induces vascular smooth muscle cell (SMC) dysfunction by generating oxidative stress attributable, in part, to the up-regulated NADPH oxidases (Nox). We have attempted to elucidate the high-glucose-generated molecular signals that mediate this effect and hypothesize that products of high-glucose-induced lipid peroxidation regulate Nox by activating peroxisome proliferator-activated receptors (PPARs). Human aortic SMCs were exposed to glucose (5.5-25 mM) or 4-hydroxynonenal (1-25 μM, 4-HNE). Lucigenin assay, real-time polymerase chain reaction, western blot, and promoter analyses were employed to investigate Nox. We found that high glucose generated an increase in Nox activity and expression. It also promoted oxidative stress that consequently induced lipid peroxidation, which resulted in the production of 4-HNE. Pharmacological inhibition of Nox activity significantly reduced the formation of high-glucose-induced 4-HNE. Exposure of SMCs to non-cytotoxic concentrations (1-10 μM) of 4-HNE alone mimicked the effect of high glucose incubation, whereas scavenging of 4-HNE by N-acetyl L-cysteine completely abolished both the effects of high glucose and 4-HNE. The latter exerted its effect by activating PPARα and PPARβ/δ, but not PPARγ, as assessed pharmacologically by the inhibitory effect of selective antagonists and following the silencing of the expression of these receptors. These new data indicate that 4-HNE, generated following Nox activation, functions as an endogenous activator of PPARα and PPARβ/δ. The newly discovered "lipid peroxidation products-PPARs-Nox axis" represents a novel mechanism of Nox regulation and an additional therapeutic target for oxidative stress in diabetes.
Collapse
Affiliation(s)
- Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8 B.P. Hasdeu Street, 050568, Bucharest, Romania,
| | | | | | | | | | | | | |
Collapse
|
82
|
Neuroprotection for ischaemic stroke: Current status and challenges. Pharmacol Ther 2015; 146:23-34. [DOI: 10.1016/j.pharmthera.2014.09.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
|
83
|
Bai J, Lyden PD. Revisiting Cerebral Postischemic Reperfusion Injury: New Insights in Understanding Reperfusion Failure, Hemorrhage, and Edema. Int J Stroke 2015; 10:143-52. [DOI: 10.1111/ijs.12434] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023]
Abstract
Cerebral postischemic reperfusion injury is defined as deterioration of ischemic brain tissue that parallels and antagonizes the benefits of restoring cerebral circulation after therapeutic thrombolysis for acute ischemic stroke. To understand the paradox of injury caused by treatment, we first emphasize the phenomenon in which recanalization of an occluded artery does not lead to tissue reperfusion. Additionally, no-reflow after recanalization may be due to injury of the neurovascular unit, distal microthrombosis, or both, and certainly worsens outcome. We examine the mechanism of molecular and sub-cellular damage in the neurovascular unit, notably oxidative stress, mitochondrial dysfunction, and apoptosis. At the level of the neurovascular unit, which mediates crosstalk between the damaged brain and systemic responses in blood, we summarize emerging evidence demonstrating that individual cell components play unique and cumulative roles that lead to damage of the blood–brain barrier and neurons. Furthermore, we review the latest developments in establishing a link between the immune system and microvascular dysfunction during ischemic reperfusion. Progress in assessing reperfusion injury has also been made, and we review imaging studies using various magnetic resonance imaging modalities. Lastly, we explore potential treatment approaches, including ischemic preconditioning, postconditioning, pharmacologic agents, and hypothermia.
Collapse
Affiliation(s)
- Jilin Bai
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Patrick D. Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
84
|
Brennan-Minnella AM, Won SJ, Swanson RA. NADPH oxidase-2: linking glucose, acidosis, and excitotoxicity in stroke. Antioxid Redox Signal 2015; 22:161-74. [PMID: 24628477 PMCID: PMC4281853 DOI: 10.1089/ars.2013.5767] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Neuronal superoxide production contributes to cell death in both glutamate excitotoxicity and brain ischemia (stroke). NADPH oxidase-2 (NOX2) is the major source of neuronal superoxide production in these settings, and regulation of NOX2 activity can thereby influence outcome in stroke. RECENT ADVANCES Reduced NOX2 activity can rescue cells from oxidative stress and cell death that otherwise occur in excitotoxicity and ischemia. NOX2 activity is regulated by several factors previously shown to affect outcome in stroke, including glucose availability, intracellular pH, protein kinase ζ/δ, casein kinase 2, phosphoinositide-3-kinase, Rac1/2, and phospholipase A2. The newly identified functions of these factors as regulators of NOX2 activity suggest alternative mechanisms for their effects on ischemic brain injury. CRITICAL ISSUES Key aspects of these regulatory influences remain unresolved, including the mechanisms by which rac1 and phospholipase activities are coupled to N-methyl-D-aspartate (NMDA) receptors, and whether superoxide production by NOX2 triggers subsequent superoxide production by mitochondria. FUTURE DIRECTIONS It will be important to establish whether interventions targeting the signaling pathways linking NMDA receptors to NOX2 in brain ischemia can provide a greater neuroprotective efficacy or a longer time window to treatment than provided by NMDA receptor blockade alone. It will likewise be important to determine whether dissociating superoxide production from the other signaling events initiated by NMDA receptors can mitigate the deleterious effects of NMDA receptor blockade.
Collapse
|
85
|
Üllen A, Nusshold C, Glasnov T, Saf R, Cantillo D, Eibinger G, Reicher H, Fauler G, Bernhart E, Hallstrom S, Kogelnik N, Zangger K, Oliver Kappe C, Malle E, Sattler W. Covalent adduct formation between the plasmalogen-derived modification product 2-chlorohexadecanal and phloretin. Biochem Pharmacol 2015; 93:470-81. [PMID: 25576489 PMCID: PMC4321883 DOI: 10.1016/j.bcp.2014.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 12/02/2022]
Abstract
Hypochlorous acid added as reagent or generated by the myeloperoxidase (MPO)-H2O2-Cl− system oxidatively modifies brain ether-phospholipids (plasmalogens). This reaction generates a sn2-acyl-lysophospholipid and chlorinated fatty aldehydes. 2-Chlorohexadecanal (2-ClHDA), a prototypic member of chlorinated long-chain fatty aldehydes, has potent neurotoxic potential by inflicting blood–brain barrier (BBB) damage. During earlier studies we could show that the dihydrochalcone-type polyphenol phloretin attenuated 2-ClHDA-induced BBB dysfunction. To clarify the underlying mechanism(s) we now investigated the possibility of covalent adduct formation between 2-ClHDA and phloretin. Coincubation of 2-ClHDA and phloretin in phosphatidylcholine liposomes revealed a half-life of 2-ClHDA of approx. 120 min, decaying at a rate of 5.9 × 10−3 min−1. NMR studies and enthalpy calculations suggested that 2-ClHDA-phloretin adduct formation occurs via electrophilic aromatic substitution followed by hemiacetal formation on the A-ring of phloretin. Adduct characterization by high-resolution mass spectroscopy confirmed these results. In contrast to 2-ClHDA, the covalent 2-ClHDA-phloretin adduct was without adverse effects on MTT reduction (an indicator for metabolic activity), cellular adenine nucleotide content, and barrier function of brain microvascular endothelial cells (BMVEC). Of note, 2-ClHDA-phloretin adduct formation was also observed in BMVEC cultures. Intraperitoneal application and subsequent GC–MS analysis of brain lipid extracts revealed that phloretin is able to penetrate the BBB of C57BL/6J mice. Data of the present study indicate that phloretin scavenges 2-ClHDA, thereby attenuating 2-ClHDA-mediated brain endothelial cell dysfunction. We here identify a detoxification pathway for a prototypic chlorinated fatty aldehyde (generated via the MPO axis) that compromises BBB function in vitro and in vivo.
Collapse
Affiliation(s)
- Andreas Üllen
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Christoph Nusshold
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Toma Glasnov
- Christian Doppler Laboratory for Flow Chemistry, Institute of Chemistry, University of Graz, Graz, Austria
| | - Robert Saf
- Institute of Chemistry and Technology of Materials, Graz University of Technology, Graz, Austria
| | - David Cantillo
- Institute of Chemistry, University of Graz, Graz, Austria
| | - Gerald Eibinger
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Helga Reicher
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Günter Fauler
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Eva Bernhart
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Seth Hallstrom
- Institute of Physiological Chemistry, Medical University of Graz, Graz, Austria
| | - Nora Kogelnik
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Klaus Zangger
- Institute of Chemistry, University of Graz, Graz, Austria
| | - C Oliver Kappe
- Institute of Chemistry, University of Graz, Graz, Austria
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria.
| |
Collapse
|
86
|
Liu Z, Jing Y, Yin J, Mu J, Yao T, Gao L. Downregulation of thioredoxin reductase 1 expression in the substantia nigra pars compacta of Parkinson's disease mice. Neural Regen Res 2014; 8:3275-83. [PMID: 25206649 PMCID: PMC4145943 DOI: 10.3969/j.issn.1673-5374.2013.35.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/21/2013] [Indexed: 11/18/2022] Open
Abstract
Because neurons are susceptible to oxidative damage and thioredoxin reductase 1 is extensively distributed in the central nervous system and has antioxidant properties, we speculated that the enzyme may be involved in the pathogenesis of Parkinson's disease. A Parkinson's disease model was produced by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine into C57BL/6 mice. Real-time reverse transcription-PCR, western blot analysis and colorimetric assay showed that the levels of thioredoxin reductase 1 mRNA and protein were decreased, along with a significant reduction in thioredoxin reductase activity, in the midbrain of Parkinson's disease mice compared with normal mice. Immunohistochemical staining revealed that the number of thioredoxin reductase 1-positive neurons in the substantia nigra pars compacta of Parkinson's disease mice was significantly decreased compared with normal mice. These experimental findings suggest that the expression of thioredoxin reductase 1 in the substantia nigra pars compacta of Parkinson's disease mice is significantly decreased, and that the enzyme may be associated with disease onset.
Collapse
Affiliation(s)
- Zihua Liu
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Yuhong Jing
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China ; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou 730000, Gansu Province, China
| | - Jie Yin
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Jiying Mu
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Tingting Yao
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Liping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China ; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
87
|
Nour M, Scalzo F, Liebeskind DS. Ischemia-reperfusion injury in stroke. INTERVENTIONAL NEUROLOGY 2014; 1:185-99. [PMID: 25187778 DOI: 10.1159/000353125] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite ongoing advances in stroke imaging and treatment, ischemic and hemorrhagic stroke continue to debilitate patients with devastating outcomes at both the personal and societal levels. While the ultimate goal of therapy in ischemic stroke is geared towards restoration of blood flow, even when mitigation of initial tissue hypoxia is successful, exacerbation of tissue injury may occur in the form of cell death, or alternatively, hemorrhagic transformation of reperfused tissue. Animal models have extensively demonstrated the concept of reperfusion injury at the molecular and cellular levels, yet no study has quantified this effect in stroke patients. These preclinical models have also demonstrated the success of a wide array of neuroprotective strategies at lessening the deleterious effects of reperfusion injury. Serial multimodal imaging may provide a framework for developing therapies for reperfusion injury.
Collapse
Affiliation(s)
- May Nour
- Departments of Neurology and Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Calif., USA
| | - Fabien Scalzo
- Departments of Neurology and Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Calif., USA
| | - David S Liebeskind
- Departments of Neurology and Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Calif., USA
| |
Collapse
|
88
|
Han Z, Shen F, He Y, Degos V, Camus M, Maze M, Young WL, Su H. Activation of α-7 nicotinic acetylcholine receptor reduces ischemic stroke injury through reduction of pro-inflammatory macrophages and oxidative stress. PLoS One 2014; 9:e105711. [PMID: 25157794 PMCID: PMC4144901 DOI: 10.1371/journal.pone.0105711] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022] Open
Abstract
Activation of α-7 nicotinic acetylcholine receptor (α-7 nAchR) has a neuro-protective effect on ischemic and hemorrhagic stroke. However, the underlying mechanism is not completely understood. We hypothesized that α-7 nAchR agonist protects brain injury after ischemic stroke through reduction of pro-inflammatory macrophages (M1) and oxidative stress. C57BL/6 mice were treated with PHA568487 (PHA, α-7 nAchR agonist), methyllycaconitine (MLA, nAchR antagonist), or saline immediately and 24 hours after permanent occlusion of the distal middle cerebral artery (pMCAO). Behavior test, lesion volume, CD68+, M1 (CD11b+/Iba1+) and M2 (CD206/Iba1+) microglia/macrophages, and phosphorylated p65 component of NF-kB in microglia/macrophages were quantified using histological stained sections. The expression of M1 and M2 marker genes, anti-oxidant genes and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase were quantified using real-time RT-PCR. Compared to the saline-treated mice, PHA mice had fewer behavior deficits 3 and 7 days after pMCAO, and smaller lesion volume, fewer CD68+ and M1 macrophages, and more M2 macrophages 3 and 14 days after pMCAO, whereas MLA's effects were mostly the opposite in several analyses. PHA increased anti-oxidant genes and NADPH oxidase expression associated with decreased phosphorylation of NF-kB p65 in microglia/macrophages. Thus, reduction of inflammatory response and oxidative stress play roles in α-7 nAchR neuro-protective effect.
Collapse
Affiliation(s)
- Zhenying Han
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Tianjin Medical University General Hospital, Tianjin, China
| | - Fanxia Shen
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Yue He
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Vincent Degos
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Hôpital Pitié Salpetrière, Assistance Publique-Hopitaux de Paris (APHP), Université Pierre et Marie Curie-Paris VI and UMR INSERM 1141, Paris, France
| | - Marine Camus
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Mervyn Maze
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - William L. Young
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
89
|
Zhao H, Wang R, Tao Z, Yan F, Gao L, Liu X, Wang N, Min L, Jia Y, Zhao Y, Ji X, Luo Y. Activation of T-LAK-cell-originated protein kinase-mediated antioxidation protects against focal cerebral ischemia-reperfusion injury. FEBS J 2014; 281:4411-20. [PMID: 25065601 DOI: 10.1111/febs.12948] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/13/2014] [Accepted: 07/23/2014] [Indexed: 12/16/2022]
Abstract
T-LAK-cell-originated protein kinase (TOPK), a MAPKK-like kinase, is crucial for neural progenitor cell proliferation; however, the function of TOPK and the molecular mechanism underlying cerebral ischemia-reperfusion injury remains unknown. Therefore, we investigated the role of TOPK in experimental stroke. Sprague-Dawley rats underwent transient middle cerebral artery occlusion (tMCAO) and reperfusion, and TOPK small interfering RNA (siRNA) was delivered by intracerebroventricular injection at the beginning of MCAO. After TOPK overexpression and H2O2 stimulation in PC12 neuronal cells, antioxidative proteins, apoptosis-related proteins and signal pathways were detected by western blot analysis, the levels of the peroxidation products (malondialdehyde and 3-nitrotyrosine) were measured with ELISA. Phosphorylation of TOPK was increased in rat cortical neurons following tMCAO. TOPK overexpression in PC12 cells augmented levels of antioxidative proteins (peroxiredoxin 1 and 2, heme oxygenase 1 and manganese superoxide dismutase), as well as total superoxide dismutase activity, along with inhibition of malondialdehyde and 3-nitrotyrosine upon H2O2 stimulation. TOPK overexpression increased cell viability and reduced expression of caspase 3 and caspase 12 in PC12 cells in response to H2O2 . The p-ERK level was increased by TOPK overexpression, and antioxidative protection afforded by TOPK was abolished by blocking the extracellular signal-regulated kinase pathway in PC12 cells. TOPK siRNA increased the infarct volume and reduced total superoxide dismutase activity in the cortex in vivo after MCAO. These data reveal that activating TOPK confers neuroprotection against focal cerebral ischemia-reperfusion injury by antioxidative function, in part through activation of the extracellular signal-regulated kinase pathway.
Collapse
Affiliation(s)
- Haiping Zhao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Li H, Wang Y, Feng D, Liu Y, Xu M, Gao A, Tian F, Zhang L, Cui Y, Wang Z, Chen G. Alterations in the time course of expression of the Nox family in the brain in a rat experimental cerebral ischemia and reperfusion model: effects of melatonin. J Pineal Res 2014; 57:110-9. [PMID: 24867613 DOI: 10.1111/jpi.12148] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 05/23/2014] [Indexed: 12/01/2022]
Abstract
Ischemia-reperfusion (I/R) injury induces the generation of reactive oxygen species (ROS), which results in a poor prognosis for ischemic stroke patients. This study was designed to evaluate the time course of expression of the Nox family, a major source of ROS, and whether melatonin, a potent scavenger of ROS, influences these parameters in a rat model of cerebral I/R caused by middle cerebral artery occlusion (MCAO). After 2-hr occlusion, the filament was withdrawn to allow reperfusion. At 0, 3, 6, 12, 24, and 48 hr after reperfusion, brain tissue samples were obtained for assays. Among the Nox family, the mRNA and protein levels of Nox2 and Nox4 were increased both in the ischemic hemisphere and contralateral counterpart in the experimental I/R rats at 0 hr after reperfusion, peaked at 3 hr, and then returned to the basal level at 24 hr. Double-immunofluorescence staining further confirmed the expressions of Nox2 and Nox4 in three major types of brain cells, including neurons, astrocytes, and endothelial cells. In addition, melatonin (5 mg/kg) or its vehicle was injected intraperitoneally at 0.5 hr before MCAO. Compared with I/R + vehicle group, melatonin pretreatment diminished the increased expression of Nox2 and Nox4, reduced ROS levels, and inhibited cell apoptosis. Our findings suggested that the inhibition of Nox2 and Nox4 expressions by melatonin may essentially contribute to its antioxidant and anti-apoptotic effects during brain I/R.
Collapse
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Zhao H, Wang R, Tao Z, Gao L, Yan F, Gao Z, Liu X, Ji X, Luo Y. Ischemic postconditioning relieves cerebral ischemia and reperfusion injury through activating T-LAK cell-originated protein kinase/protein kinase B pathway in rats. Stroke 2014; 45:2417-24. [PMID: 25013016 DOI: 10.1161/strokeaha.114.006135] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Ischemic postconditioning (IPostC) protects against ischemic brain injury. To date, no study has examined the role of T-LAK-cell-originated protein kinase (TOPK) in IPostC-afforded neuroprotection. We explored the molecular mechanism related with TOPK in antioxidant effect of IPostC against ischemia/reperfusion. METHODS Focal ischemia was induced in rats by transient middle cerebral artery occlusion. Reactive oxygen species production in the peri-infarct cortex was detected using dihydroethidium. Malondialdehyde, as a marker of lipid peroxidation, and 3-nitrotyrosine, as a marker of protein oxidation, were detected by ELISA. The expression or location of antioxidant proteins and signal molecules TOPK, phosphatase, and tensin homolog, and Akt was analyzed by Western blotting and immunofluorescence. RESULTS Our results revealed that IPostC relieved transient middle cerebral artery occlusion-induced oxidative damage by reducing reactive oxygen species, malondialdehyde, and 3-nitrotyrosine accumulation in the peri-infarct cortex and raised levels of antioxidants perioxiredoxin-1, peroxiredoxin-2, and thioredoxin-1. In addition, IPostC increased p-AKT and p-TOPK levels, which colocalized in neural cells. In vitro TOPK knockdown by small interfering RNA decreased the levels of antioxidants peroxiredoxin-1, thioredoxin, and manganese superoxide dismutase activity in PC12 cells. In vivo intracerebroventricular injection of TOPK small interfering RNA reversed IPostC-induced neuroprotection by increasing infarct volume and nitric oxide content and reducing manganese superoxide dismutase activity. Moreover, IPostC-evoked Akt activation was blocked by TOPK small interfering RNA in vivo, but the decreased phosphorylated phosphatase and tensin homolog level in ischemia/reperfusion was not influenced by IPostC or by TOPK small interfering RNA treatment. CONCLUSIONS Our results suggest that the antioxidative effects of TOPK/Akt might contribute to the neuroprotection of IPostC treatment against transient middle cerebral artery occlusion.
Collapse
Affiliation(s)
- Haiping Zhao
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Rongliang Wang
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Zhen Tao
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Li Gao
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Feng Yan
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Zhi Gao
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Xiangrong Liu
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.)
| | - Xunming Ji
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.).
| | - Yumin Luo
- From the Cerebrovascular Diseases Research Institute (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.) and Department of Neurology (L.G.), Xuanwu Hospital of Capital Medical University, Beijing, China; Cerebralvascular Diseases Research Laboratory, Beijing Geriatric Medical Research Center, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.); and Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China (H.Z., R.W., Z.T., F.Y., Z.G., X.L., X.J., Y.L.).
| |
Collapse
|
92
|
Oxidative Stress and the Use of Antioxidants in Stroke. Antioxidants (Basel) 2014; 3:472-501. [PMID: 26785066 PMCID: PMC4665418 DOI: 10.3390/antiox3030472] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/08/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022] Open
Abstract
Transient or permanent interruption of cerebral blood flow by occlusion of a cerebral artery gives rise to an ischaemic stroke leading to irreversible damage or dysfunction to the cells within the affected tissue along with permanent or reversible neurological deficit. Extensive research has identified excitotoxicity, oxidative stress, inflammation and cell death as key contributory pathways underlying lesion progression. The cornerstone of treatment for acute ischaemic stroke remains reperfusion therapy with recombinant tissue plasminogen activator (rt-PA). The downstream sequelae of events resulting from spontaneous or pharmacological reperfusion lead to an imbalance in the production of harmful reactive oxygen species (ROS) over endogenous anti-oxidant protection strategies. As such, anti-oxidant therapy has long been investigated as a means to reduce the extent of injury resulting from ischaemic stroke with varying degrees of success. Here we discuss the production and source of these ROS and the various strategies employed to modulate levels. These strategies broadly attempt to inhibit ROS production or increase scavenging or degradation of ROS. While early clinical studies have failed to translate success from bench to bedside, the combination of anti-oxidants with existing thrombolytics or novel neuroprotectants may represent an avenue worthy of clinical investigation. Clearly, there is a pressing need to identify new therapeutic alternatives for the vast majority of patients who are not eligible to receive rt-PA for this debilitating and devastating disease.
Collapse
|
93
|
Moran E, Ding L, Wang Z, Cheng R, Chen Q, Moore R, Takahashi Y, Ma JX. Protective and antioxidant effects of PPARα in the ischemic retina. Invest Ophthalmol Vis Sci 2014; 55:4568-76. [PMID: 24825105 DOI: 10.1167/iovs.13-13127] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Previous studies have demonstrated that peroxisome proliferator-activated receptor-alpha (PPARα) agonists have therapeutic effects in diabetic retinopathy, although the mechanism of action remains incompletely understood. The purpose of this study was to evaluate PPARα's protective effects in the ischemic retina, and to delineate its molecular mechanism of action. METHODS For the oxygen-induced retinopathy (OIR) model, wild-type (WT), and PPARα knockout (PPARα(-/-)) mice were exposed to 75% O₂ from postnatal day 7 (P7) to P12 and treated with the PPARα agonist fenofibric acid (Feno-FA) from P12 to P16. At P17, the effects of Feno-FA on retinal glial fibrillary acidic protein (GFAP) expression, apoptotic DNA cleavage, and TUNEL labeling were analyzed. Cultured retinal cells were exposed to CoCl₂ to induce hypoxia, and TUNEL staining and 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein dye were used to measure apoptosis and reactive oxygen species (ROS) generation. Western blotting was used to measure GFAP levels and cell signaling. RESULTS Feno-FA decreased retinal apoptosis and oxidative stress in WT but not PPARα(-/-) OIR mice. Peroxisome proliferator-activated receptor-alpha knockout OIR mice showed increased retinal cell death and glial activation in comparison to WT OIR mice. Feno-FA treatment and PPARα overexpression protected cultured retinal cells from hypoxic cell death and decreased ROS levels. Nuclear hypoxia-inducible factor-α (HIF-1α) and nicotine adenine dinucleotide phosphate oxidase-4 (Nox 4) were increased in OIR retinas and downregulated by Feno-FA in WT but not in PPARα(-/-) mice. CONCLUSIONS Peroxisome proliferator-activated receptor-alpha has a potent antiapoptotic effect in the ischemic retina. This protective effect may be mediated in part through downregulation of HIF-1α/Nox 4 and consequently alleviation of oxidative stress.
Collapse
Affiliation(s)
- Elizabeth Moran
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Lexi Ding
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongxiao Wang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rui Cheng
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Qian Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Robert Moore
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Yusuke Takahashi
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Jian-xing Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
94
|
Kim HR, Lee A, Choi EJ, Kie JH, Lim W, Lee HK, Moon BI, Seoh JY. Attenuation of experimental colitis in glutathione peroxidase 1 and catalase double knockout mice through enhancing regulatory T cell function. PLoS One 2014; 9:e95332. [PMID: 24743300 PMCID: PMC3990669 DOI: 10.1371/journal.pone.0095332] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 03/25/2014] [Indexed: 12/02/2022] Open
Abstract
Reactive oxygen species (ROS) have been implicated in the progression of inflammatory diseases including inflammatory bowel diseases (IBD). Meanwhile, several studies suggested the protective role of ROS in immune-mediated inflammatory diseases, and it was recently reported that dextran sodium sulfate (DSS)-induced colitis was attenuated in mice with an elevated level of ROS due to deficiency of peroxiredoxin II. Regulatory T cells (Tregs) are critical in the prevention of IBD and Treg function was reported to be closely associated with ROS level, but it has been investigated only in lowered levels of ROS so far. In the present study, in order to clarify the relationship between ROS level and Treg function, and their role in the pathogenesis of IBD, we investigated mice with an elevated level of ROS due to deficiency of both glutathione peroxidase (GPx)-1 and catalase (Cat) for the susceptibility of DSS-induced colitis in association with Treg function. The results showed that DSS-induced colitis was attenuated and Tregs were hyperfunctional in GPx1−/− × Cat−/− mice. In vivo administration of N-acetylcysteine (NAC) aggravated DSS-induced colitis and decreased Treg function to the level comparable to WT mice. Attenuated Th17 cell differentiation from naïve CD4+ cells as well as impaired production of IL-6 and IL-17A by splenocytes upon stimulation suggested anti-inflammatory tendency of GPx1−/− × Cat−/− mice. Suppression of Stat3 activation in association with enhancement of indoleamine 2,3-dioxygenase and FoxP3 expression might be involved in the immunosuppressive mechanism of GPx1−/− × Cat−/− mice. Taken together, it is implied that ROS level is critical in the regulation of Treg function, and IBD may be attenuated in appropriately elevated levels of ROS.
Collapse
Affiliation(s)
- Hyung-Ran Kim
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Anbok Lee
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Korea
| | - Eun-Jeong Choi
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Jeong-Hae Kie
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Woosung Lim
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Korea
| | - Hyeon Kook Lee
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Korea
| | - Byung-In Moon
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Korea
- * E-mail: (JYS); (BIM)
| | - Ju-Young Seoh
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
- * E-mail: (JYS); (BIM)
| |
Collapse
|
95
|
Kim HR, Lee A, Choi EJ, Hong MP, Kie JH, Lim W, Lee HK, Moon BI, Seoh JY. Reactive oxygen species prevent imiquimod-induced psoriatic dermatitis through enhancing regulatory T cell function. PLoS One 2014; 9:e91146. [PMID: 24608112 PMCID: PMC3946742 DOI: 10.1371/journal.pone.0091146] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/07/2014] [Indexed: 11/29/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease resulting from immune dysregulation. Regulatory T cells (Tregs) are important in the prevention of psoriasis. Traditionally, reactive oxygen species (ROS) are known to be implicated in the progression of inflammatory diseases, including psoriasis, but many recent studies suggested the protective role of ROS in immune-mediated diseases. In particular, severe cases of psoriasis vulgaris have been reported to be successfully treated by hyperbaric oxygen therapy (HBOT), which raises tissue level of ROS. Also it was reported that Treg function was closely associated with ROS level. However, it has been only investigated in lowered levels of ROS so far. Thus, in this study, to clarify the relationship between ROS level and Treg function, as well as their role in the pathogenesis of psoriasis, we investigated imiquimod-induced psoriatic dermatitis (PD) in association with Treg function both in elevated and lowered levels of ROS by using knockout mice, such as glutathione peroxidase-1−/− and neutrophil cytosolic factor-1−/− mice, as well as by using HBOT or chemicals, such as 2,3-dimethoxy-1,4-naphthoquinone and N-acetylcysteine. The results consistently showed Tregs were hyperfunctional in elevated levels of ROS, whereas hypofunctional in lowered levels of ROS. In addition, imiquimod-induced PD was attenuated in elevated levels of ROS, whereas aggravated in lowered levels of ROS. For the molecular mechanism that may link ROS level and Treg function, we investigated the expression of an immunoregulatory enzyme, indoleamine 2,3-dioxygenase (IDO) which is induced by ROS, in PD lesions. Taken together, it was implied that appropriately elevated levels of ROS might prevent psoriasis through enhancing IDO expression and Treg function.
Collapse
Affiliation(s)
- Hyung-Ran Kim
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Anbok Lee
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Eun-Jeong Choi
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Min-Pyo Hong
- College of Arts and Sciences, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Jeong-Hae Kie
- Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Hyeon Kook Lee
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Seoul, Korea
| | - Ju-Young Seoh
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
96
|
Uekawa K, Hasegawa Y, Ma M, Nakagawa T, Katayama T, Sueta D, Toyama K, Kataoka K, Koibuchi N, Kawano T, Kuratsu JI, Kim-Mitsuyama S. Rosuvastatin ameliorates early brain injury after subarachnoid hemorrhage via suppression of superoxide formation and nuclear factor-kappa B activation in rats. J Stroke Cerebrovasc Dis 2014; 23:1429-39. [PMID: 24529602 DOI: 10.1016/j.jstrokecerebrovasdis.2013.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/29/2013] [Accepted: 12/03/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Statins, or 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors, have been suggested to possess pleiotropic effects, including antioxidant and anti-inflammatory properties. We investigated the protective effects of pretreatment with rosuvastatin, a relatively hydrophilic statin, on early brain injury (EBI) after a subarachnoid hemorrhage (SAH), using the endovascular perforation SAH model. METHODS Eighty-six male Sprague-Dawley rats were randomly divided into 3 groups: (1) sham operation, (2) SAH+vehicle, and (3) SAH+10 mg/kg rosuvastatin. Rosuvastatin or vehicle was orally administered to rats once daily from 7 days before to 1 day after the SAH operation. After SAH, we examined the effects of rosuvastatin on the neurologic score, brain water content, neuronal cell death estimated by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate nick end labeling staining, blood-brain barrier disruption by immunoglobulin G (IgG) extravasation, oxidative stress, and proinflammatory molecules. RESULTS Compared with the vehicle group, rosuvastatin significantly improved the neurologic score and reduced the brain water content, neuronal cell death, and IgG extravasation. Rosuvastatin inhibited brain superoxide production, nuclear factor-kappa B (NF-κB) activation, and the increase in activated microglial cells after SAH. The increased expressions of tumor necrosis factor-alpha, endothelial matrix metalloproteinase-9, and neuronal cyclooxygenase-2 induced by SAH were prevented by rosuvastatin pretreatment. CONCLUSIONS The present study demonstrates that rosuvastatin pretreatment ameliorates EBI after SAH through the attenuation of oxidative stress and NF-κB-mediated inflammation.
Collapse
Affiliation(s)
- Ken Uekawa
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yu Hasegawa
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mingjie Ma
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Nakagawa
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tetsuji Katayama
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensuke Toyama
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keiichiro Kataoka
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobutaka Koibuchi
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takayuki Kawano
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun-ichi Kuratsu
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shokei Kim-Mitsuyama
- Department of Pharmacology and Molecular Therapeutics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
97
|
Pretreatment with tert-butylhydroquinone attenuates cerebral oxidative stress in mice after traumatic brain injury. J Surg Res 2013; 188:206-12. [PMID: 24387843 DOI: 10.1016/j.jss.2013.11.1106] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 10/21/2013] [Accepted: 11/21/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a worldwide health problem, identified as a major cause of death and disability. Increasing evidence has shown that oxidative stress plays an important role in TBI pathogenesis. The antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), is a known mediator in protection against TBI-induced brain damage. The objective of this study was to test whether tert-butylhydroquinone (tBHQ), a novel Nrf2 activator, can protect against TBI-induced oxidative stress. METHODS Adult male imprinting control region mice were randomly divided into three groups: (1) sham + vehicle group; (2) TBI + vehicle group; and (3) TBI + tBHQ group. Closed-head brain injury was applied using the Feeney weight-drop method. We accessed the neurologic outcome of mice at 24 h after TBI, and subsequently measured protein levels of Nrf2 and the NOX2 subunit of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), the concentration of malondialdehyde, superoxide dismutase activity, and brain edema. RESULT The NOX2 protein level was increased fivefold in the TBI + vehicle group, whereas pretreatment with tBHQ markedly attenuated the NOX2 protein expression relative to that in the TBI + vehicle group. TBI increased Nrf2 formation by 5% compared with the sham group, whereas treatment with tBHQ further upregulated the Nrf2 protein level by 12% compared with the sham group. The level of the oxidative damage marker malondialdehyde was reduced by 29% in the TBI + tBHQ group compared with the TBI + vehicle group, Moreover, pretreatment with tBHQ significantly increased the antioxidant enzyme superoxide dismutase activity. Administration of tBHQ also significantly decreased TBI-induced brain edema and neurologic deficits. CONCLUSIONS Pretreatment with tBHQ effectively attenuated markers of cerebral oxidative stress after TBI, thus supporting the testing of tBHQ as a potential neuroprotectant and adjunct therapy for TBI patients.
Collapse
|
98
|
Wang Z, Wei X, Liu K, Zhang X, Yang F, Zhang H, He Y, Zhu T, Li F, Shi W, Zhang Y, Xu H, Liu J, Yi F. NOX2 deficiency ameliorates cerebral injury through reduction of complexin II-mediated glutamate excitotoxicity in experimental stroke. Free Radic Biol Med 2013; 65:942-951. [PMID: 23982049 DOI: 10.1016/j.freeradbiomed.2013.08.166] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 08/12/2013] [Accepted: 08/15/2013] [Indexed: 10/26/2022]
Abstract
Although NADPH oxidase (NOX)-mediated oxidative stress is considered one of the major mechanisms triggering the pathogenic actions of ischemic stroke and very recent studies have indicated that NADPH oxidase is a major source of reactive oxygen species (ROS) production controlling glutamate release, how neuronal NADPH oxidase activation is coupled to glutamate release is not well understood. Therefore, in this study, we used an in vivo transient middle cerebral artery occlusion model and in vitro primary cell cultures to test whether complexins, the regulators of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes necessary for vesicle fusion, are associated with NOX2-derived ROS and contribute to glutamate-mediated excitotoxicity in ischemic stroke. In this study, we first identified the upregulation of complexin II in the ischemic brain and evaluated its potential role in ischemic stroke showing that gene silencing of complexin II ameliorated cerebral injury as evidenced by reduced infarction volume, neurological deficit, and neuron necrosis accompanied by decreased glutamate levels, consistent with the results from NOX2(-/-) mice with ischemic stroke. We further demonstrated that complexin II expression was mediated by NOX2 in primary cultured neurons subjected to oxygen-glucose deprivation (OGD) and contributed to OGD-induced glutamate release and neuron necrosis via SNARE signaling. Taken together, these findings for the first time provide evidence that complexin II is a central target molecule that links NADPH oxidase-derived ROS to glutamate-mediated neuronal excitotoxicity in ischemic stroke.
Collapse
Affiliation(s)
- Ziying Wang
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xinbing Wei
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Kang Liu
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiumei Zhang
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fan Yang
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Hongyu Zhang
- Department of Geriatrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Yeteng He
- Department of Orthopedics, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Tianfeng Zhu
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fengli Li
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Weichen Shi
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yan Zhang
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Huiyan Xu
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiang Liu
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fan Yi
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
99
|
NADPH oxidase and the degeneration of dopaminergic neurons in parkinsonian mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:157857. [PMID: 24379900 PMCID: PMC3863573 DOI: 10.1155/2013/157857] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/08/2013] [Accepted: 10/23/2013] [Indexed: 11/18/2022]
Abstract
Several lines of investigation have implicated oxidative stress in Parkinson's disease (PD) pathogenesis, but the mechanisms involved are still unclear. In this study, we characterized the involvement of NADPH oxidase (Nox), a multisubunit enzyme that catalyzes the reduction of oxygen, in the 6-hydroxydopamine- (6-OHDA-) induced PD mice model and compared for the first time the effects of this neurotoxin in mice lacking gp91phox−/−, the catalytic subunit of Nox2, and pharmacological inhibition of Nox with apocynin. Six-OHDA induced increased protein expression of p47phox, a Nox subunit, in striatum. gp91phox−/− mice appear to be completely protected from dopaminergic cell loss, whereas the apocynin treatment conferred only a limited neuroprotection. Wt mice treated with apocynin and gp91phox−/− mice both exhibited ameliorated apomorphine-induced rotational behavior. The microglial activation observed within the striatum and the substantia nigra pars compacta (SNpc) of 6-OHDA-injected Wt mice was prevented by apocynin treatment and was not detected in gp91phox−/− mice. Apocynin was not able to attenuate astrocyte activation in SN. The results support a role for Nox2 in the 6-OHDA-induced degeneration of dopaminergic neurons and glial cell activation in the nigrostriatal pathway and reveal that no comparable 6-OHDA effects were observed between apocynin-treated and gp91phox−/− mice groups.
Collapse
|
100
|
Intracellular pH reduction prevents excitotoxic and ischemic neuronal death by inhibiting NADPH oxidase. Proc Natl Acad Sci U S A 2013; 110:E4362-8. [PMID: 24163350 DOI: 10.1073/pnas.1313029110] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sustained activation of N-methyl-d-aspartate (NMDA) -type glutamate receptors leads to excitotoxic neuronal death in stroke, brain trauma, and neurodegenerative disorders. Superoxide production by NADPH oxidase is a requisite event in the process leading from NMDA receptor activation to excitotoxic death. NADPH oxidase generates intracellular H(+) along with extracellular superoxide, and the intracellular H(+) must be released or neutralized to permit continued NADPH oxidase function. In cultured neurons, NMDA-induced superoxide production and neuronal death were prevented by intracellular acidification by as little as 0.2 pH units, induced by either lowered medium pH or by inhibiting Na(+)/H(+) exchange. In mouse brain, superoxide production induced by NMDA injections or ischemia-reperfusion was likewise prevented by inhibiting Na(+)/H(+) exchange and by reduced expression of the Na(+)/H(+) exchanger-1 (NHE1). Neuronal intracellular pH and neuronal Na(+)/H(+) exchange are thus potent regulators of excitotoxic superoxide production. These findings identify a mechanism by which cell metabolism can influence coupling between NMDA receptor activation and superoxide production.
Collapse
|