1
|
Ahmed S, Tripathy RK, Pande AH, Sharma SS. Neuroprotective potential of ApoE-mimetic peptide (ApoEFrag) in stroke models: Neurobehavioural and mechanistic study. Int J Biol Macromol 2025; 304:140790. [PMID: 39924027 DOI: 10.1016/j.ijbiomac.2025.140790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Stroke is the second leading cause of death worldwide, and currently, there is no effective neuroprotective agent available for its treatment. Though apolipoprotein E (ApoE) showed potential as a neuroprotective agent for CNS disorders, however, its large size limits its clinical application. To overcome this issue, smaller ApoE-mimetic peptides that mimic the biological functions of ApoE have been developed. In this study, we designed and characterized a novel ApoE-mimetic peptide, ApoEFrag, and explored its neuroprotective potential in experimental stroke models. ApoEFrag was evaluated for its ability to interact with oxidized lipids and lack of self-aggregation potential. In the in vitro cerebral ischemia model, ApoEFrag demonstrated neuroprotection against glutamate-induced neuronal damage in SH-SY5Y cells by maintaining mitochondrial health and reducing reactive oxygen species (ROS) levels. To confirm ApoEFrag's neuroprotective potential, ApoEFrag was investigated in middle cerebral artery occlusion (MCAO) induced ischemic stroke in rats. ApoEFrag administration significantly reduced infarct size, improved neurological function, and lowered mortality. ApoEFrag exhibited anti-inflammatory effects, reduced astrocyte activation and apoptosis, and promoted neurogenesis. Overall, ApoEFrag shows promising neuroprotective effects and could be a potential therapeutic approach for the treatment of stroke.
Collapse
Affiliation(s)
- Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
2
|
Zaric BL, Macvanin MT, Isenovic ER. Free radicals: Relationship to Human Diseases and Potential Therapeutic applications. Int J Biochem Cell Biol 2023; 154:106346. [PMID: 36538984 DOI: 10.1016/j.biocel.2022.106346] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Reactive species are highly-reactive enzymatically, or non-enzymatically produced compounds with important roles in physiological and pathophysiological cellular processes. Although reactive species represent an extensively researched topic in biomedical sciences, many aspects of their roles and functions remain unclear. This review aims to systematically summarize findings regarding the biochemical characteristics of various types of reactive species and specify the localization and mechanisms of their production in cells. In addition, we discuss the specific roles of free radicals in cellular physiology, focusing on the current lines of research that aim to identify the reactive oxygen species-initiated cascades of reactions resulting in adaptive or pathological cellular responses. Finally, we present recent findings regarding the therapeutic modulations of intracellular levels of reactive oxygen species, which may have substantial significance in developing novel agents for treating several diseases.
Collapse
Affiliation(s)
- Bozidarka L Zaric
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Yilmaz S, Alkan T, Ballar Kirmizibayrak P. A new underlying mechanism for the neuroprotective effect of bosutinib: Reverting toxicity-induced PARylation in SIN1-mediated neurotoxicity. J Biochem Mol Toxicol 2021; 35:e22915. [PMID: 34519134 DOI: 10.1002/jbt.22915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 11/09/2022]
Abstract
Increased levels of reactive oxygen and nitrogen species play an important role in the development and progression of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. The overproduction of these highly reactive chemical species leads to DNA damage and subsequent activation of the poly(ADP-ribose)polymerase (PARP) enzyme. Several studies have demonstrated the potential use of PARP inhibitors for neuroprotection. We previously reported that the dual Src/Abl kinase inhibitor bosutinib (BOS) decreases PARP activity and acts as a chemosensitizer in cancer cells. In this study, we evaluated the neuroprotective potential of BOS with respect to its inhibitory effect on cellular poly(ADP-ribos)ylation (PARylation) using a 3-morpholinosydnonimine (SIN1)-mediated cellular toxicity model. Our data suggest that pretreatment with BOS, especially at lower doses, significantly decreased the level of SIN1-induced cellular PARylation. This regulation pattern of PARylation was found to be associated with the protective effect of BOS against SIN1 on the viability of retinoic acid-differentiated SH-SY5Y cells. Furthermore, while PARP-1 expression was decreased, phosphorylation of SAPK/JNK was not reverted at the observed neuroprotective doses of BOS. In conclusion, we suggest a novel mechanism for the neuroprotective effect of BOS involving the inhibition of cellular PARylation.
Collapse
Affiliation(s)
- Sinem Yilmaz
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey.,Department of Bioengineering, Faculty of Engineering, University of Alanya Aladdin Keykubat, Antalya, Turkey
| | - Tolgaç Alkan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Petek Ballar Kirmizibayrak
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Turkey.,Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
4
|
Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood–brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res 2018; 52:1220-1239. [PMID: 30468092 DOI: 10.1080/10715762.2018.1521519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| | - Xi Chen
- Department of Core Facility, the People’s Hospital of Bao-an Shenzhen, Shenzhen, PR China
- The 8th People’s Hospital of Shenzhen, the Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, PR China
| | - Yunhao Luo
- School of Chinese Medicine, the University of Hong Kong, PR China
| | - Jiangang Shen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
5
|
Slosky LM, Vanderah TW. Therapeutic potential of peroxynitrite decomposition catalysts: a patent review. Expert Opin Ther Pat 2015; 25:443-66. [PMID: 25576197 DOI: 10.1517/13543776.2014.1000862] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Peroxynitrite is a cytotoxic oxidant species implicated in a host of pathologies, including inflammatory and neurodegenerative diseases, cancer, radiation injury and chronic pain. With the recognition of the role of peroxynitrite in disease, numerous experimental and therapeutic tools have arisen to probe peroxyntirite's pathophysiological contribution and attenuate its oxidative damage. Peroxynitrite decomposition catalysts (PNDCs) are redox-active compounds that detoxify peroxynitrite by catalyzing its isomerization or reduction to nitrate or nitrite. AREAS COVERED This review discusses recent research articles and patents published 1995 - 2014 on the development and therapeutic use of PNDCs. Iron and manganese metalloporphyrin PNDCs attenuate the toxic effects of peroxynitrite and are currently being developed for clinical applications. Additionally, some Mn porphyrin-based PNDCs have optimized pharmaceutical properties such that they exhibit greater peroxynitrite selectivity. Other classes of PNDC agents, including bis(hydroxyphenyl)dipyrromethenes and metallocorroles, have demonstrated preclinical efficacy, oral availability and reduced toxicity risk. EXPERT OPINION Interest in the drug-like properties of peroxynitrite-neutralizing agents has grown with the realization that PNDCs will be powerful tools in the treatment of disease. The design of compounds with enhanced oral availability and peroxynitrite selectivity is a critical step toward the availability of safe, effective and selective redox modulators for the treatment of peroxynitrite-associated pathologies.
Collapse
Affiliation(s)
- Lauren M Slosky
- University of Arizona, Department of Pharmacology , Life Science North Rm 621, 1501 North Campbell Ave., Tucson, AZ 85721 , USA
| | | |
Collapse
|
6
|
Free radical scavenging activity and neuroprotective potentials of D138, one Cu(II)/Zn(II) Schiff-base complex derived from N,N'-bis(2-hydroxynaphthylmethylidene)-1,3-propanediamine. Neurochem Res 2014; 39:1834-44. [PMID: 25069642 DOI: 10.1007/s11064-014-1392-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/08/2014] [Accepted: 07/16/2014] [Indexed: 10/25/2022]
Abstract
There is increasing evidence that free radicals play an important role in neuronal damages induced by diabetes mellitus or cerebral ischemia insults. Antioxidants with free radical scavenging activities have been shown to be beneficial and neuroprotective for these pathological conditions. Here, we report free radical scavenging activity and neuroprotective potential of D138, one copper(II)/zinc(II) Schiff-base complex derived from N,N'-2(2-hydroxynaphthylmethylidene)-1,3-propanediamine. The data from three in vitro assays, 2,2-diphenyl-1-picrylhydrazyl assay, nitro blue tetrazolium assay and hydroxyl radical scavenging assay, indicated that D138 presented a potent free radical scavenging activity. The neuroprotective and antioxidative effects of D138 were further evaluated in vivo using bilateral common carotid artery occlusion (BCCAO) mouse model and streptozotocin (STZ) diabetic mouse model. Our results indicated that treatment of D138 significantly ameliorated the hippocampal neuronal damage and the oxidative stress levels in these animal models. Moreover, D138 also reversed the behavioral deficiencies induced by BCCAO or STZ, as assessed by Y-maze test and fear conditioning test. In conclusion, all these findings support that D138 exerts free radical scavenging and neuroprotective activities and has the potentials to be a potent therapeutic candidate for brain oxidative damage induced by cerebral ischemia or diabetes mellitus.
Collapse
|
7
|
Sheng H, Chaparro RE, Sasaki T, Izutsu M, Pearlstein RD, Tovmasyan A, Warner DS. Metalloporphyrins as therapeutic catalytic oxidoreductants in central nervous system disorders. Antioxid Redox Signal 2014; 20:2437-64. [PMID: 23706004 DOI: 10.1089/ars.2013.5413] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Metalloporphyrins, characterized by a redox-active transitional metal (Mn or Fe) coordinated to a cyclic porphyrin core ligand, mitigate oxidative/nitrosative stress in biological systems. Side-chain substitutions tune redox properties of metalloporphyrins to act as potent superoxide dismutase mimics, peroxynitrite decomposition catalysts, and redox regulators of transcription factor function. With oxidative/nitrosative stress central to pathogenesis of CNS injury, metalloporphyrins offer unique pharmacologic activity to improve the course of disease. RECENT ADVANCES Metalloporphyrins are efficacious in models of amyotrophic lateral sclerosis, Alzheimer's disease, epilepsy, neuropathic pain, opioid tolerance, Parkinson's disease, spinal cord injury, and stroke and have proved to be useful tools in defining roles of superoxide, nitric oxide, and peroxynitrite in disease progression. The most substantive recent advance has been the synthesis of lipophilic metalloporphyrins offering improved blood-brain barrier penetration to allow intravenous, subcutaneous, or oral treatment. CRITICAL ISSUES Insufficient preclinical data have accumulated to enable clinical development of metalloporphyrins for any single indication. An improved definition of mechanisms of action will facilitate preclinical modeling to define and validate optimal dosing strategies to enable appropriate clinical trial design. Due to previous failures of "antioxidants" in clinical trials, with most having markedly less biologic activity and bioavailability than current-generation metalloporphyrins, a stigma against antioxidants has discouraged the development of metalloporphyrins as CNS therapeutics, despite the consistent definition of efficacy in a wide array of CNS disorders. FUTURE DIRECTIONS Further definition of the metalloporphyrin mechanism of action, side-by-side comparison with "failed" antioxidants, and intense effort to optimize therapeutic dosing strategies are required to inform and encourage clinical trial design.
Collapse
Affiliation(s)
- Huaxin Sheng
- 1 Department of Anesthesiology, Duke University Medical Center (DUMC) , Durham, North Carolina
| | | | | | | | | | | | | |
Collapse
|
8
|
Jangra A, Datusalia AK, Sharma SS. Reversal of neurobehavioral and neurochemical alterations in STZ-induced diabetic rats by FeTMPyP, a peroxynitrite decomposition catalyst and 1,5-Isoquinolinediol a poly(ADP-ribose) polymerase inhibitor. Neurol Res 2013; 36:619-26. [PMID: 24620961 DOI: 10.1179/1743132813y.0000000301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In this study, we have evaluated the involvement of nitrosative stress and poly-ADP ribosyl polymerase (PARP) in diabetes induced neurobehavioral and neurochemical changes using pharmacological agents peroxynitrite decomposition catalyst (FeTMPyP) and a PARP inhibitor (1,5-Isoquinolinediol) in diabetic brains. METHODS The extent of neurobehavioral changes was assessed by functional observation battery, motor coordination activity (rota rod performance) and passive avoidance test. Neurochemical changes were assessed by measuring nicotinamide adenine dinucleotide (NAD), malondialdehyde, acetylcholinesterase, neurotransmitters (GABA and glutamate) levels in the hippocampus. GABA and glutamate were measured by high-performance liquid chromatography with electrochemical detection method. RESULTS Two weeks' treatment with FeTMPyP (3 mg/kg, i.p.) and 1,5-Isoquinolinediol (3 mg/kg, i.p.) improved the cognitive deficits in diabetic rats as observed in passive avoidance test. Both the agents inhibited lipid peroxidation and improves the acetylcholinesterase level in the hippocampus. 1,5-Isoquinolinediol treatment also improves the NAD, neurotransmitter level in the hippocampus. DISCUSSION These results suggest that peroxynitrite decomposition catalyst and PARP inhibitor have beneficial effects in neurobehavioral alterations induced by diabetes. Improvement in neurobehavioral alteration may be attributed to reversal of neurotransmitter homeostasis deficits.
Collapse
|
9
|
Sharma SS, Kaundal RK. Neuroprotective effects of 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox), an antioxidant in middle cerebral artery occlusion induced focal cerebral ischemia in rats. Neurol Res 2013; 29:304-9. [PMID: 17509231 DOI: 10.1179/016164107x158983] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES In the present study, we have investigated the neuroprotective potential of 6hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox), in middle cerebral artery occlusion (MCAO) induced focal cerebral ischemia. METHODS Sprague-Dawley rats were subjected to 2 hours of MCAO followed by 22 or 70 hours of reperfusion. After reperfusion, rats were evaluated for neurological deficits and cerebral infarction. Brain malondialdehyde (MDA) level and in situ terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling (TUNEL) were also estimated. RESULTS Focal cerebral ischemia produced a significant infarct volume and neurological scores as compared with sham-operated animals. Cerebral ischemia reperfusion injury was associated with an increase in lipid peroxidation in ipsilateral and contralateral hemisphere of brain along with an increase in TUNEL positive cells in ipsilateral hemisphere of brain sections indicating oxidative stress and DNA fragmentation, respectively. Trolox (10 and 30 mg/kg, i.p.) treatment significantly decreased neurological damage which was evident from the reduction in infarct volume and neurological score. Trolox (30 mg/kg) also attenuated oxidative stress and DNA fragmentation. DISCUSSION Oxidative stress-induced neuronal damage is implicated in the pathophysiology of cerebral ischemia. Our study suggests that Trolox is a potent neuroprotective agent in focal cerebral ischemia and its neuroprotective effects may be attributed to the reduction of lipid peroxidation and DNA fragmentation.
Collapse
Affiliation(s)
- Shyam S Sharma
- Molecular Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India.
| | | |
Collapse
|
10
|
Pujari RR, Vyawahare NS, Thakurdesai PA. Protective effects of Phoenix dactylifera against oxidative stress and neuronal damage induced by global cerebral ischemia in rats. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.biomag.2013.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
11
|
Kelly-Cobbs AI, Prakash R, Li W, Pillai B, Hafez S, Coucha M, Johnson MH, Ogbi SN, Fagan SC, Ergul A. Targets of vascular protection in acute ischemic stroke differ in type 2 diabetes. Am J Physiol Heart Circ Physiol 2013; 304:H806-15. [PMID: 23335797 DOI: 10.1152/ajpheart.00720.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemorrhagic transformation is an important complication of acute ischemic stroke, particularly in diabetic patients receiving thrombolytic treatment with tissue plasminogen activator, the only approved drug for the treatment of acute ischemic stroke. The objective of the present study was to determine the effects of acute manipulation of potential targets for vascular protection [i.e., NF-κB, peroxynitrite, and matrix metalloproteinases (MMPs)] on vascular injury and functional outcome in a diabetic model of cerebral ischemia. Ischemia was induced by middle cerebral artery occlusion in control and type 2 diabetic Goto-Kakizaki rats. Treatment groups received a single dose of the peroxynitrite decomposition catalyst 5,10,15,20-tetrakis(4-sulfonatophenyl)prophyrinato iron (III), the nonspecific NF-κB inhibitor curcumin, or the broad-spectrum MMP inhibitor minocycline at reperfusion. Poststroke infarct volume, edema, hemorrhage, neurological deficits, and MMP-9 activity were evaluated. All acute treatments reduced MMP-9 and hemorrhagic transformation in diabetic groups. In addition, acute curcumin and minocycline therapy reduced edema in these animals. Improved neurological function was observed in varying degrees with treatment, as indicated by beam-walk performance, modified Bederson scores, and grip strength; however, infarct size was similar to untreated diabetic animals. In control animals, all treatments reduced MMP-9 activity, yet bleeding was not improved. Neuroprotection was only conferred by curcumin and minocycline. Uncovering the underlying mechanisms contributing to the success of acute therapy in diabetes will advance tailored stroke therapies.
Collapse
Affiliation(s)
- Aisha I Kelly-Cobbs
- Department of Physiology, Georgia Regents University Augusta, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Chen XM, Chen HS, Xu MJ, Shen JG. Targeting reactive nitrogen species: a promising therapeutic strategy for cerebral ischemia-reperfusion injury. Acta Pharmacol Sin 2013; 34:67-77. [PMID: 22842734 DOI: 10.1038/aps.2012.82] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ischemic stroke accounts for nearly 80% of stroke cases. Recanalization with thrombolysis is a currently crucial therapeutic strategy for re-building blood supply, but the thrombolytic therapy often companies with cerebral ischemia-reperfusion injury, which are mediated by free radicals. As an important component of free radicals, reactive nitrogen species (RNS), including nitric oxide (NO) and peroxynitrite (ONOO(-)), play important roles in the process of cerebral ischemia-reperfusion injury. Ischemia-reperfusion results in the production of nitric oxide (NO) and peroxynitrite (ONOO(-)) in ischemic brain, which trigger numerous molecular cascades and lead to disruption of the blood brain barrier and exacerbate brain damage. There are few therapeutic strategies available for saving ischemic brains and preventing the subsequent brain damage. Recent evidence suggests that RNS could be a therapeutic target for the treatment of cerebral ischemia-reperfusion injury. Herein, we reviewed the recent progress regarding the roles of RNS in the process of cerebral ischemic-reperfusion injury and discussed the potentials of drug development that target NO and ONOO(-) to treat ischemic stroke. We conclude that modulation for RNS level could be an important therapeutic strategy for preventing cerebral ischemia-reperfusion injury.
Collapse
|
13
|
Khan M, Dhammu TS, Sakakima H, Shunmugavel A, Gilg AG, Singh AK, Singh I. The inhibitory effect of S-nitrosoglutathione on blood-brain barrier disruption and peroxynitrite formation in a rat model of experimental stroke. J Neurochem 2012; 123 Suppl 2:86-97. [PMID: 23050646 PMCID: PMC3481195 DOI: 10.1111/j.1471-4159.2012.07947.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The hallmark of stroke injury is endothelial dysfunction leading to blood-brain barrier (BBB) leakage and edema. Among the causative factors of BBB disruption are accelerating peroxynitrite formation and the resultant decreased bioavailability of nitric oxide (NO). S-nitrosoglutathione (GSNO), an S-nitrosylating agent, was found not only to reduce the levels of peroxynitrite but also to protect the integrity of BBB in a rat model of cerebral ischemia and reperfusion (IR). A treatment with GSNO (3 μmol/kg) after IR reduced 3-nitrotyrosine levels in and around vessels and maintained NO levels in brain. This mechanism protected endothelial function by reducing BBB leakage, increasing the expression of Zonula occludens-1 (ZO-1), decreasing edema, and reducing the expression of matrix metalloproteinase-9 and E-selectin in the neurovascular unit. An administration of the peroxynitrite-forming agent 3-morpholino sydnonimine (3 μmol/kg) at reperfusion increased BBB leakage and decreased the expression of ZO-1, supporting the involvement of peroxynitrite in BBB disruption and edema. Mechanistically, the endothelium-protecting action of GSNO was invoked by reducing the activity of nuclear factor kappa B and increasing the expression of S-nitrosylated proteins. Taken together, the results support the ability of GSNO to improve endothelial function by reducing nitroxidative stress in stroke.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Harutoshi Sakakima
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | | | - Anne G Gilg
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Avtar K. Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson VA Medical Center, Charleston, SC
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
14
|
Singh N, Sharma G, Singh N, Hanif K. A Comparative Study of Neuroprotective Effect of Single and Combined Blockade of AT1 Receptor and PARP-1 in Focal Cerebral Ischaemia in Rat. Int J Stroke 2012; 9:560-8. [DOI: 10.1111/j.1747-4949.2012.00916.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/01/2012] [Indexed: 11/30/2022]
Abstract
Background Cerebral ischaemia results in enhanced expression of type 1 angiotensin receptor and oxidative stress. Free radicals due to oxidative stress lead to excessive DNA damage causing overactivation of poly (ADP-ribose) polymerase-1 resulting in neuronal death. Activation of both type 1 angiotensin receptors and poly (ADP-ribose) polymerase-1 following cerebral ischaemia takes place simultaneously, but until now, no study has explored the effect of combined blockade of both angiotensin type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 in cerebral ischaemia. Aim Our purpose was to compare the effect of single and combined treatment with angiotensin type 1 angiotensin receptor blocker, candesartan, and the poly (ADP-ribose) polymerase-1 inhibitor, 1, 5 isoquinolinediol, on brain damage and oxidative stress in transient focal cerebral ischaemia in rats. Method Transient focal cerebral ischaemia was induced in Sprague-Dawley rats by an intraluminal technique for two-hours following 48 h of reperfusion. Candesartan (0·05 mg/kg) was administered just after initiation of ischaemia followed by a repeat administration at 24 h while 1, 5 isoquinolinediol (0·1 mg/kg) was given one-hour after of ischaemia. After 24 h of reperfusion, neurological deficit was evaluated in the different treatment groups. After 48 h of reperfusion, the rats were sacrificed and the brain was isolated. Ischaemic brain damage by 2,3,5 triphenyl tetrazolium chloride staining, oxidative stress markers, and levels of reactive oxygen species were determined biochemically. Result Single treatment with candesartan and 1, 5 isoquinolinediol significantly reduced neurological deficit, infarct, and oedema volume as compared to ischaemic control and different vehicle groups for each of the drugs. However, treatment with candesartan + 1, 5 isoquinolinediol offered greater reduction in neurological deficit, cerebral infarct volume, and oedema as compared to single-drug treatments. Furthermore, treatment with candesartan + 1, 5 isoquinolinediol significantly decreased oxidative stress as compared to single treatments with each drug. Conclusion The study suggests that blockade of either type 1 angiotensin receptor or poly (ADP-ribose) polymerase-1 alone provides neuroprotection, but the better result was achieved when both type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 were blocked together by the combined use of their pharmacological inhibitor in transient cerebral ischaemia in rat.
Collapse
|
15
|
Prajapati KD, Sharma SS, Roy N. Hepatocyte nuclear factor-1alpha mediated upregulation of albumin expression in focal ischemic rat brain. Neurol Res 2012; 34:25-31. [PMID: 22196858 DOI: 10.1179/1743132811y.0000000052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Exogenous human albumin has been shown to be neuroprotective in experimental ischemic stroke and it is currently investigated in clinical trials. However, the role of endogenous expression of albumin and its transcriptional regulation in the ischemic brain is not known. We have previously reported the upregulation of de novo synthesis of albumin in the ischemic rat brain (at 0 and 22 hours of reperfusion after 2 hours of ischemia). In this study, we analyzed the role of transcription factors in albumin expression in ischemic rat brain. METHODS The putative transcription factor binding sites for the albumin promoter was analyzed using transcription factor search computational tool and validated in rat middle cerebral artery occlusion model of transient cerebral ischemia. RESULTS Computational analysis predicted approximately 20 transcription factor binding sites including hepatocyte nuclear factor-1alpha (HNF-1alpha). We found for the first time mRNA and protein expression of HNF-1alpha in the control and ischemic rat brain. There was no significant difference in mRNA and protein expression of HNF-1alpha between control and ischemic (0, 2 and 22 hours of reperfusion) group but there was increased interaction of HNF-1alpha with p300 (known interacting partner for HNF-1alpha, a histone acetyl-transferase) in 0- and 22-hour reperfusion groups. Also albumin promoter binding activity of HNF-1alpha in ischemic animals of 0- and 22-hour reperfusion groups significantly increased compared to respective control group animals. DISCUSSION Although, HNF-1alpha is mainly expressed in the rat liver and involved in hepatic expression of albumin, our study conclusively shows for the first time de novo synthesis of HNF-1alpha in rat brain. Moreover, an increased interaction of HNF-1alpha with p300 and albumin promoter seems to be responsible for overexpression of albumin in ischemic conditions.
Collapse
Affiliation(s)
- Kanaiyalal D Prajapati
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, India
| | | | | |
Collapse
|
16
|
Peroxynitrite and Nitroxidative Stress: Detection Probes and Micro-Sensors. A Case of a Nanostructured Catalytic Film. ACTA ACUST UNITED AC 2011. [DOI: 10.1021/bk-2011-1083.ch011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
17
|
Khan M, Sakakima H, Dhammu TS, Shunmugavel A, Im YB, Gilg AG, Singh AK, Singh I. S-nitrosoglutathione reduces oxidative injury and promotes mechanisms of neurorepair following traumatic brain injury in rats. J Neuroinflammation 2011; 8:78. [PMID: 21733162 PMCID: PMC3158546 DOI: 10.1186/1742-2094-8-78] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Accepted: 07/06/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) induces primary and secondary damage in both the endothelium and the brain parenchyma, collectively termed the neurovascular unit. While neurons die quickly by necrosis, a vicious cycle of secondary injury in endothelial cells exacerbates the initial injury in the neurovascular unit following TBI. In activated endothelial cells, excessive superoxide reacts with nitric oxide (NO) to form peroxynitrite. Peroxynitrite has been implicated in blood brain barrier (BBB) leakage, altered metabolic function, and neurobehavioral impairment. S-nitrosoglutathione (GSNO), a nitrosylation-based signaling molecule, was reported not only to reduce brain levels of peroxynitrite and oxidative metabolites but also to improve neurological function in TBI, stroke, and spinal cord injury. Therefore, we investigated whether GSNO promotes the neurorepair process by reducing the levels of peroxynitrite and the degree of oxidative injury. METHODS TBI was induced by controlled cortical impact (CCI) in adult male rats. GSNO or 3-Morpholino-sydnonimine (SIN-1) (50 μg/kg body weight) was administered orally two hours following CCI. The same dose was repeated daily until endpoints. GSNO-treated (GSNO group) or SIN-1-treated (SIN-1 group) injured animals were compared with vehicle-treated injured animals (TBI group) and vehicle-treated sham-operated animals (Sham group) in terms of peroxynitrite, NO, glutathione (GSH), lipid peroxidation, blood brain barrier (BBB) leakage, edema, inflammation, tissue structure, axon/myelin integrity, and neurotrophic factors. RESULTS SIN-1 treatment of TBI increased whereas GSNO treatment decreased peroxynitrite, lipid peroxides/aldehydes, BBB leakage, inflammation and edema in a short-term treatment (4-48 hours). GSNO also reduced brain infarctions and enhanced the levels of NO and GSH. In a long-term treatment (14 days), GSNO protected axonal integrity, maintained myelin levels, promoted synaptic plasticity, and enhanced the expression of neurotrophic factors. CONCLUSION Our findings indicate the participation of peroxynitrite in the pathobiology of TBI. GSNO treatment of TBI not only reduces peroxynitrite but also protects the integrity of the neurovascular unit, indicating that GSNO blunts the deleterious effects of peroxynitrite. A long-term treatment of TBI with the same low dose of GSNO promotes synaptic plasticity and enhances the expression of neurotrophic factors. These results support that GSNO reduces the levels of oxidative metabolites, protects the neurovascular unit, and promotes neurorepair mechanisms in TBI.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang C, Li S, Shang DJ, Wang XL, You ZL, Li HB. Antihyperglycemic and neuroprotective effects of one novel Cu-Zn SOD mimetic. Bioorg Med Chem Lett 2011; 21:4320-4. [PMID: 21669524 DOI: 10.1016/j.bmcl.2011.05.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 05/05/2011] [Accepted: 05/17/2011] [Indexed: 11/24/2022]
Abstract
Increasing evidence supports that OS plays important roles in diabetes mellitus and cerebral ischemia. This suggests that recovering an impaired endogenous superoxide dismutase (SOD) enzyme system induced by OS with a mimetic would be beneficial and protective for these diseases. In present study, one nonpeptidyl small molecular weight compound (D34) was synthesized. Its SOD mimetic activity and the potential therapeutic actions were also evaluated both in vivo and in vitro. The in vitro nitro blue tetrazolium (NBT) assay indicated that D34 presents an SOD mimetic activity. D34 (20μmol/kg) exhibited significant antihyperglycemic activity in alloxan-diabetic mice. D34 could also ameliorate the cerebral neuronal death in hippocampus of global cerebral ischemia mice. Furthermore, the D34 treatment significantly decreased malondialdehyde (MDA) contents and increased SOD activities in brains or livers of diabetes mice or cerebral ischemic mice. In conclusion, these preliminary findings support that D34 exhibits SOD mimetic activity and possesses significant antihyperglycemic and neuroprotective effects.
Collapse
Affiliation(s)
- Che Wang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | | | | | | | | | | |
Collapse
|
19
|
Prajapati KD, Sharma SS, Roy N. Upregulation of albumin expression in focal ischemic rat brain. Brain Res 2010; 1327:118-24. [PMID: 20193666 DOI: 10.1016/j.brainres.2010.02.063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 02/19/2010] [Accepted: 02/22/2010] [Indexed: 01/24/2023]
Abstract
To gain insight into the early and late changes in protein expression following focal transient cerebral ischemia in rat, proteomic approach was undertaken. Proteomic profiling using two-dimensional gel electrophoresis indicated upregulation of albumin protein after 2h of ischemia and 22h of reperfusion among the other altered proteins. Further, the albumin overexpression was verified by quantitative real time PCR at mRNA level, validated by western blotting and immunohistochemistry. Although, exogenous human albumin therapy is already under clinical trials for cerebral ischemia, its endogenous expression in ischemic rat brain at mRNA and protein levels has not been investigated as yet. Here we report for the first time de novo synthesis of albumin in the ischemic rat brain. This study paves the way for further investigation of signaling mechanisms leading to albumin overexpression, so that it can be exploited as a therapeutic intervention.
Collapse
Affiliation(s)
- Kanaiyalal D Prajapati
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Punjab-160 062, India
| | | | | |
Collapse
|
20
|
Guven A, Uysal B, Akgul O, Cermik H, Gundogdu G, Surer I, Ozturk H, Korkmaz A. Scavenging of Peroxynitrite Reduces Renal Ischemia/Reperfusion Injury. Ren Fail 2009; 30:747-54. [DOI: 10.1080/08860220802213039] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
21
|
Arora M, Kumar A, Kaundal RK, Sharma SS. Amelioration of neurological and biochemical deficits by peroxynitrite decomposition catalysts in experimental diabetic neuropathy. Eur J Pharmacol 2008; 596:77-83. [PMID: 18768138 DOI: 10.1016/j.ejphar.2008.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 07/31/2008] [Accepted: 08/07/2008] [Indexed: 12/11/2022]
Abstract
Diabetic neuropathy, a major complication of diabetes, affects more than 60% of diabetic patients. Recently, involvement of peroxynitrite has been postulated in diabetic neuropathy. In the present study, we have studied the effects of peroxynitrite decomposition catalysts (PDC's)-5,10,15,20-tetrakis(4-sulfonatophenyl) porphyrinato iron(III) [FeTPPS] and 5,10,15,20-tetrakis(N-methyl-4-pyridyl)porphyrinato iron(III) [FeTMPyP]-in experimental diabetic neuropathy. Male Sprague-Dawley rats, with six weeks of untreated diabetes were treated for two weeks with peroxynitrite decomposition catalysts. Diabetic animals showed a significant decrease in motor nerve conduction velocity and nerve blood flow, nociception as evident from decreased tail flick latency (hyperalgesia) and increased paw withdrawal pressure (mechanical allodynia) along with elevation in peroxynitrite and reduction in nerve glutathione levels. Two weeks treatment with PDC's significantly improved all the above stated functional and biochemical deficits. Aftermath of this study advocates the beneficial effects of peroxynitrite decomposition catalysts in experimental diabetic neuropathy.
Collapse
Affiliation(s)
- Manish Arora
- Molecular Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, India
| | | | | | | |
Collapse
|
22
|
Rabkin SW, Klassen SS. Metalloporphyrins as a therapeutic drug class against peroxynitrite in cardiovascular diseases involving ischemic reperfusion injury. Eur J Pharmacol 2008; 586:1-8. [DOI: 10.1016/j.ejphar.2008.02.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 01/18/2008] [Accepted: 02/06/2008] [Indexed: 10/22/2022]
|
23
|
Protective effects of 4-amino1,8-napthalimide, a poly (ADP-ribose) polymerase inhibitor in experimental diabetic neuropathy. Life Sci 2007; 82:570-6. [PMID: 18262571 DOI: 10.1016/j.lfs.2007.11.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 11/16/2007] [Accepted: 11/27/2007] [Indexed: 02/07/2023]
Abstract
Peripheral diabetic neuropathy is a heterogeneous group of disorders, and is known to affect 50-60% of diabetic patients. Poly (ADP-ribose) polymerase (PARP) activation has been identified as one of the key components in the pathogenesis of diabetic neuropathy. In the present study we have targeted PARP overactivation in diabetic neuropathy using a known PARP inhibitor, 4 amino 1, 8-napthalimide (4-ANI). Streptozotocin induced diabetic rats developed neuropathy within 6 weeks, which was evident from significant reduction in motor nerve conduction velocity (MNCV), nerve blood flow (NBF) along with neuropathic pain and abnormal sensory perception. Six weeks after diabetes induction Sprague Dawley rats were treated with 4-ANI (3 and 10 mg/kg, p.o.) for a period of two weeks (seventh and eighth weeks). Two week treatment with 4-ANI showed improvement in nerve conduction, nerve blood flow and reduction in tail flick responses and mechanical allodynia in diabetic animals. 4-ANI also attenuated PAR immunoreactivity and NAD depletion in nerves of diabetic animals. Results of present study suggest the potential of PARP inhibitors like 4-ANI in the treatment of diabetic neuropathy.
Collapse
|
24
|
Szabó C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov 2007; 6:662-80. [PMID: 17667957 DOI: 10.1038/nrd2222] [Citation(s) in RCA: 1679] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peroxynitrite--the product of the diffusion-controlled reaction of nitric oxide with superoxide radical--is a short-lived oxidant species that is a potent inducer of cell death. Conditions in which the reaction products of peroxynitrite have been detected and in which pharmacological inhibition of its formation or its decomposition have been shown to be of benefit include vascular diseases, ischaemia-reperfusion injury, circulatory shock, inflammation, pain and neurodegeneration. In this Review, we first discuss the biochemistry and pathophysiology of peroxynitrite and then focus on pharmacological strategies to attenuate the toxic effects of peroxynitrite. These include its catalytic reduction to nitrite and its isomerization to nitrate by metalloporphyrins, which have led to potential candidates for drug development for cardiovascular, inflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Csaba Szabó
- Department of Surgery, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, University Heights, Newark, New Jersey 07103-2714, USA.
| | | | | |
Collapse
|
25
|
Maier C, Scheuerle A, Hauser B, Schelzig H, Szabó C, Radermacher P, Kick J. The selective poly(ADP)ribose-polymerase 1 inhibitor INO1001 reduces spinal cord injury during porcine aortic cross-clamping-induced ischemia/reperfusion injury. Intensive Care Med 2007; 33:845-850. [PMID: 17361386 DOI: 10.1007/s00134-007-0585-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 02/12/2007] [Indexed: 11/29/2022]
Abstract
OBJECTIVE It is well-established that poly(ADP)ribose-polymerase (PARP) assumes major importance during ischemic brain damage, and the selective PARP-1 inhibitor PJ34 reduced spinal cord damage in murine aortic occlusion-induced ischemia/reperfusion injury. We investigated the effect of the PARP-1 inhibitor INO1001 on aortic-occlusion-related porcine spinal cord injury. DESIGN AND SETTING Prospective, randomized, controlled experimental study in an animal laboratory. PATIENTS AND PARTICIPANTS Ten anesthetized, mechanically ventilated, and instrumented pigs. INTERVENTIONS Animals underwent 45 min of thoracic aortic cross-clamping after receiving vehicle (n=5) or intravenous INO1001 (n=5, total dose 4 mg/kg administered both before clamping and during reperfusion). During reperfusion continuous intravenous norepinephrine was incrementally adjusted to maintain blood pressure at or above 80% of the preclamping level. Plasma INO1001 levels were analyzed by HPLC. After 4[Symbol: see text]h of reperfusion spinal cord biopsy samples were analyzed for neuronal damage (hematoxyline-eosine and Nissl staining), expression of the cyclin-dependent kinase inhibitor genes p21 and p27 (immunohistochemistry), and apoptosis (terminal deoxynucleotidyl transferase mediated nick end labeling assay). MEASUREMENTS AND RESULTS Plasma INO1001 levels were 0.8-2.3 and 0.30-0.76 mM before and after clamping, respectively. While 3-5% of the spinal cord neurons were irreversibly damaged in the INO1001 animals, the neuronal cell injury was three times higher in the control group. Neither p21 and p27 expression nor apoptosis showed any intergroup difference. CONCLUSIONS The selective PARP-1 inhibitor INO1001 markedly reduced aortic occlusion-induced spinal cord injury. Given the close correlation reported in the literature between morphological damage and impaired spinal cord function, INO1001 may improve spinal cord recovery after thoracic aortic cross-clamping.
Collapse
Affiliation(s)
- Christian Maier
- Sektion Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, 89073, Ulm, Germany
- Abteilung Thorax- und Gefäßchirurgie, Universitätsklinikum, 89070, Ulm, Germany
| | | | - Balázs Hauser
- Sektion Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, 89073, Ulm, Germany
- Aneszteziológiai és Intenzív Terápiás Klinika, Semmelweis Egyetem, 1125, Budapest, Hungary
| | - Hubert Schelzig
- Abteilung Thorax- und Gefäßchirurgie, Universitätsklinikum, 89070, Ulm, Germany
| | - Csaba Szabó
- Department of Surgery, University of Medicine and Dentistry, Newark, N.J., USA
| | - Peter Radermacher
- Sektion Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, 89073, Ulm, Germany.
| | - Jochen Kick
- Abteilung Thorax- und Gefäßchirurgie, Universitätsklinikum, 89070, Ulm, Germany
| |
Collapse
|
26
|
Sharma SS, Gupta S. Neuroprotective effect of MnTMPyP, a superoxide dismutase/catalase mimetic in global cerebral ischemia is mediated through reduction of oxidative stress and DNA fragmentation. Eur J Pharmacol 2007; 561:72-9. [PMID: 17320858 DOI: 10.1016/j.ejphar.2006.12.039] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 12/18/2006] [Accepted: 12/21/2006] [Indexed: 10/23/2022]
Abstract
Excessive generation of free radicals and decreased levels of the antioxidant enzymes such as superoxide dismutase (SOD) and catalase have been observed after brain ischemic reperfusion injury. In the present study, we have investigated the neuroprotective potential of MnTMPyP (Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin pentachloride), a SOD/Catalase mimetic in bilateral carotid artery occlusion model of global cerebral ischemia in Mongolian gerbils. Five minutes of bilateral carotid artery occlusion produced global cerebral ischemia, which was evident from the neurological deficits, spontaneous motor activity and the decrease in the number of viable hippocampal CA1 neurons. Global ischemia was also associated with increased levels of malondialdehyde, decreased levels of SOD and catalase, and increased TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) positive cells, indicating oxidative stress and DNA fragmentation. Administration of a single dose of MnTMPyP, 1 mg/kg i.p. (30 min before occlusion), produced no significant neuroprotection; however, 3 mg/kg i.p. (30 min before to occlusion) produced significant reduction in neurological score, spontaneous motor activity and CA1 pyramidal neuronal damage. MnTMPyP also attenuated the increased levels of malondialdehyde and improved the levels of SOD and catalase, and inhibited DNA fragmentation in the ischemic animals. Multiple administration of MnTMPyP, 3 mg/kg i.p. (three times: 30 min before, 1 h and 3 h after occlusion), produced better neuroprotection as compared to single dose administration. This study demonstrates that the neuroprotective effect of MnTMPyP in global ischemia is mediated through reduction in oxidative stress and DNA fragmentation.
Collapse
Affiliation(s)
- Shyam S Sharma
- Molecular Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar (Mohali), Punjab-160062, India.
| | | |
Collapse
|
27
|
Sharma SS, Dhar A, Kaundal RK. FeTPPS protects against global cerebral ischemic-reperfusion injury in gerbils. Pharmacol Res 2007; 55:335-42. [PMID: 17292620 DOI: 10.1016/j.phrs.2007.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 12/13/2006] [Accepted: 01/08/2007] [Indexed: 11/18/2022]
Abstract
Neuronal damage following cerebral ischemia is mediated by various mechanisms, among which nitrosative stress plays an important role. Peroxynitrite, a powerful oxidant, contributes heavily to the neuronal damage in cerebral ischemic-reperfusion (IR) injury. In the present study, we have investigated the neuroprotective effects of a peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis(4-sulfonatophenyl) porphyrinato iron(III) [FeTPPS] in global cerebral IR injury in gerbils. Neurological damage was significantly attenuated by FeTPPS treatment (1 and 3mgkg(-1), i.p.) as evident from reduction in neurological symptoms, hyperlocomotion, memory impairment and CA1 hippocampal neuronal damage in IR challenged gerbils. FeTPPS treatment also attenuated the increased malondialdehyde (MDA) levels and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) positive cells after cerebral IR injury. Results of this study demonstrates the neuroprotective activity of FeTPPS in global cerebral IR injury and its neuroprotective effects may be attributed to reduction in oxidative stress and DNA fragmentation.
Collapse
Affiliation(s)
- Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India.
| | | | | |
Collapse
|
28
|
Tauskela JS, Brunette E, Kiedrowski L, Lortie K, Hewitt M, Morley P. Unconventional neuroprotection against Ca2+ -dependent insults by metalloporphyrin catalytic antioxidants. J Neurochem 2006; 98:1324-42. [PMID: 16895586 DOI: 10.1111/j.1471-4159.2006.03973.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We evaluated whether both inert and catalytically active metalloporphyrin antioxidants, meso-substituted with either phenyl-based or N-alkylpyridinium-based groups, suppress Ca(2+)-dependent neurotoxicity in cell culture models of relevance to cerebral ischemia. Representatives from both metalloporphyrin classes, regardless of antioxidant strength, protected cultured cortical neurons or PC-12 cultures against the Ca(2+) ionophores ionomycin or A23187, by suppressing neurotoxic Ca(2+) influx. Some metalloporphyrins suppressed excitotoxic Ca(2+) influx indirectly induced by the Ca(2+) ionophores in cortical neurons. Metalloporphyrins did not quench intracellular fluorescence, suggesting localization to the plasma membrane interface and/or interference with Ca(2+) ionophores. Metalloporphyrins suppressed ionomycin-induced Mn(2+) influx, but did not protect cortical neurons against pyrithione, a Zn(2+) ionophore. In other Ca(2+)-dependent paradigms, Ca(2+) influx via plasma membrane depolarization, but not through reversal of plasmalemmal Na(+)/Ca(2+) exchangers, was modestly suppressed by Mn(III)meso-tetrakis(4-benzoic acid)porphyrin (Mn(III)TBAP) or by an inert analog, Zn(II)TBAP. Mn(III)TBAP and Zn(II)TBAP potently protected cortical neurons against long-duration oxygen-glucose deprivation (OGD), performed in the presence of antagonists of NMDA, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate and L-type voltage-gated Ca(2+) channels, raising the possibility of an unconventional mode of blockade of transient receptor protein melastatin 7 channels by a metalloTBAP family of metalloporphyrins. The present study extends the range of Ca(2+)-dependent insults for which metalloporphyrins demonstrate unconventional neuroprotection. MetalloTBAPs appear capable of targeting an OGD temporal continuum.
Collapse
Affiliation(s)
- Joseph S Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Pathophysiology Group, Ottawa, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
29
|
Kaundal RK, Shah KK, Sharma SS. Neuroprotective effects of NU1025, a PARP inhibitor in cerebral ischemia are mediated through reduction in NAD depletion and DNA fragmentation. Life Sci 2006; 79:2293-302. [PMID: 16935310 DOI: 10.1016/j.lfs.2006.07.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/24/2006] [Accepted: 07/27/2006] [Indexed: 10/24/2022]
Abstract
Oxidative stress induced cell injury is reported to contribute to the pathogenesis of cerebral ischemia. Reactive oxygen species such as hydrogen peroxide (H2O2) and superoxide radical along with nitric oxide and peroxynitrite generated during ischemia-reperfusion injury, causes the overactivation of poly (ADP-ribose) polymerase (PARP) leading to neuronal cell death. In the present study we have evaluated the effects of PARP inhibitor, 8-hydroxy-2 methyl-quinazolin-4-[3H]one (NU1025) in H2O2 and 3-morphilinosyndonimine (SIN-1) induced cytotoxicity in PC12 cells as well as in middle cerebral artery occlusion (MCAO) induced focal cerebral ischemia in rats. Exposure of PC12 cells to H2O2 (0.4 mM) and SIN-1 (0.8 mM) resulted in a significant decrease in cell viability after 6 h. Pretreatment with NU1025 (0.2 mM) restored cell viability to approximately 73 and 82% in H2O2 and SIN-1 injured cells, respectively. In MCAO studies, NU1025 was administered at different time points (1 h before reperfusion, immediately before reperfusion, 3 h after reperfusion and 6 h after reperfusion). NU1025 at 1 and 3 mg/kg reduced total infarct volume to 25% and 45%, respectively, when administered 1 h before reperfusion. NU1025 also produced significant improvement in neurological deficits. Neuroprotection with NU1025 was associated with reduction in PAR accumulation, reversal of brain NAD depletion and reduction in DNA fragmentation. Results of this study demonstrate the neuroprotective activity of NU1025 and suggest its potential in cerebral ischemia.
Collapse
Affiliation(s)
- Ravinder K Kaundal
- Molecular Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar (Mohali), Punjab-160062, India
| | | | | |
Collapse
|
30
|
Gupta S, Sharma SS. Neuroprotective effects of trolox in global cerebral ischemia in gerbils. Biol Pharm Bull 2006; 29:957-61. [PMID: 16651726 DOI: 10.1248/bpb.29.957] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stroke is a third leading cause of death and oxygen free radicals have been shown to be involved in its pathophysiology. In the present study, we have investigated neuroprotective potential of trolox, a free radical scavenger in bilateral carotid arteries occlusion (5 min) model of global cerebral ischemia in Mongolian gerbils. Gerbils were treated with trolox (3, 10 or 30 mg/kg, i.p.) 30 min prior to occlusion. There was a significant increase in neurological symptoms and locomotor activity in ischemic animals as compared with the sham-operated animals. These effects were attenuated by trolox 30 mg/kg, i.p. Significant increase in the number of the surviving neurons in the hippocampal CA1 pyramidal region was observed in ischemic animals treated with trolox 30 mg/kg, i.p. There was significant increase in the level of malondialdehyde (MDA) in ischemic animals indicating oxidative stress. Elevated levels of MDA in ischemic animals (25.79+/-3.34 microM/mg of protein) were reduced (16.43+/-3.32 microM/mg of protein) and (8.98+/-0.89 microM/mg of protein) by trolox 10 and 30 mg/kg, i.p., respectively. This study demonstrates the neuroprotective potential of trolox in global cerebral ischemia in gerbils.
Collapse
Affiliation(s)
- Sangeetha Gupta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, India
| | | |
Collapse
|
31
|
Tauskela JS, Brunette E, Hewitt M, Mealing G, Morley P. Competing approaches to excitotoxic neuroprotection by inert and catalytic antioxidant porphyrins. Neurosci Lett 2006; 401:236-41. [PMID: 16631306 DOI: 10.1016/j.neulet.2006.03.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 03/03/2006] [Accepted: 03/15/2006] [Indexed: 10/24/2022]
Abstract
The goal of this study was to determine if novel porphyrins protect cultured cortical neurons from excitotoxic NMDA exposure or oxygen-glucose deprivation (OGD), which model key aspects of cerebral ischemia. Porphyrins were chosen based on conventional and unconventional criteria. Metalloporphyrin catalytic antioxidants possessing a redox-sensitive metal core can exhibit potent and wide-ranging catalytic antioxidant abilities, which are conventionally believed to underlie neuroprotection. We report here that a recent-generation potent peroxynitrite decomposition catalyst, FP-15, protected a majority of neurons against OGD and NMDA toxicity, without suppressing NMDA-mediated intracellular Ca2+ (Cai2+) elevations or whole-cell currents. We have previously shown that neuroprotection against OGD and NMDA toxicity correlated with an ability to suppress neurotoxic Cai2+ elevations and not antioxidant ability. We now evaluate if this unconventional mechanism extends to inert metal-free porphyrins. Neuron cultures were completely protected against OGD and NMDA toxicity by H2-meso-tetrakis(3-benzoic acid)porphyrin (H2-TBAP(3)) or H2-meso-tetrakis(4-sulfonatophenyl)porphyrin (H2-TPPS(4)), although only H2-TPPS(4) suppressed (completely) NMDA-induced Cai2+ rises. H2-meso-tetrakis(3,3'-benzoic acid)porphyrin (H2-TBAP(3,3')) or H2-meso-tetrakis(N-methylpyridynium-4-yl)porphyrin (H2-TM-PyP(4)) provided at least partial protection against OGD and NMDA toxicity and partially suppressed NMDA-induced Cai2+ elevations. Despite the complexity of Ca2+-independent and -dependent based mechanisms, the inventory of porphyrins demonstrating neuroprotection in ischemia-relevant insults is now expanded to include FP-15 and inert metal-free compounds, although with no apparent advantage gained by using FP-15.
Collapse
Affiliation(s)
- Joseph Stephen Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Pathophysiology Group, Montreal Road Campus, Building M-54, Ottawa, Ontario, Canada K1A 0R6.
| | | | | | | | | |
Collapse
|
32
|
Richards EM, Rosenthal RE, Kristian T, Fiskum G. Postischemic hyperoxia reduces hippocampal pyruvate dehydrogenase activity. Free Radic Biol Med 2006; 40:1960-70. [PMID: 16716897 PMCID: PMC2570699 DOI: 10.1016/j.freeradbiomed.2006.01.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 01/20/2006] [Accepted: 01/23/2006] [Indexed: 01/04/2023]
Abstract
The pyruvate dehydrogenase complex (PDHC) is a mitochondrial matrix enzyme that catalyzes the oxidative decarboxylation of pyruvate and represents the sole bridge between anaerobic and aerobic cerebral energy metabolism. Previous studies demonstrating loss of PDHC enzyme activity and immunoreactivity during reperfusion after cerebral ischemia suggest that oxidative modifications are involved. This study tested the hypothesis that hyperoxic reperfusion exacerbates loss of PDHC enzyme activity, possibly due to tyrosine nitration or S-nitrosation. We used a clinically relevant canine ventricular fibrillation cardiac arrest model in which, after resuscitation and ventilation on either 100% O2 (hyperoxic) or 21-30% O2 (normoxic), animals were sacrificed at 2 h reperfusion and the brains removed for enzyme activity and immunoreactivity measurements. Animals resuscitated under hyperoxic conditions exhibited decreased PDHC activity and elevated 3-nitrotyrosine immunoreactivity in the hippocampus but not the cortex, compared to nonischemic controls. These measures were unchanged in normoxic animals. In vitro exposure of purified PDHC to peroxynitrite resulted in a dose-dependent loss of activity and increased nitrotyrosine immunoreactivity. These results support the hypothesis that oxidative stress contributes to loss of hippocampal PDHC activity during cerebral ischemia and reperfusion and suggest that PDHC is a target of peroxynitrite.
Collapse
Affiliation(s)
- Erica M. Richards
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Robert E. Rosenthal
- Program in Trauma, Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
| | - Gary Fiskum
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
- Corresponding author. Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA. Fax: +1 410 706 2550. E-mail address: (G. Fiskum)
| |
Collapse
|
33
|
Ding W, Hudson LG, Liu KJ. Inorganic arsenic compounds cause oxidative damage to DNA and protein by inducing ROS and RNS generation in human keratinocytes. Mol Cell Biochem 2005; 279:105-12. [PMID: 16283519 DOI: 10.1007/s11010-005-8227-y] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Arsenic is a naturally occurring element that is present in food, soil, and water. Inorganic arsenic can accumulate in human skin and is associated with increased risk of skin cancer. Oxidative stress due to arsenic exposure is proposed as one potential mode of carcinogenic action. The purpose of this study is to investigate the specific reactive oxygen and nitrogen species that are responsible for the arsenic-induced oxidative damage to DNA and protein. Our results demonstrated that exposure of human keratinocytes to trivalent arsenite caused the generation of 8-hydroxyl-2'-deoxyguanine (8-OHdG) and 3-nitrotyrosine (3-NT) in a concentration- and time-dependent manner. Pentavalent arsenate had similar effects, but to a significantly less extent. The observed oxidative damage can be suppressed by pre-treating cells with specific antioxidants. Furthermore, we found that pre-treating cells with Nomega-nitro-L-arginine methyl ester (L-NAME), an inhibitor of nitric oxide synthase (NOS), or with 5,10,15,20-tetrakis (N-methyl-4'-pyridyl) porphinato iron (III) chloride (FeTMPyP), a decomposition catalyst of peroxynitrite, suppressed the generation of both 8-OHdG and 3-NT, which indicated that peroxynitrite, a product of the reaction of nitric oxide and superoxide, played an important role in arsenic-induced oxidative damage to both DNA and protein. These findings highlight the involvement of peroxynitrite in the molecular mechanism underlying arsenic-induced human skin carcinogenesis.
Collapse
Affiliation(s)
- Wei Ding
- Program of Toxicology, College of Pharmacy, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
34
|
Tauskela JS, Brunette E, O'Reilly N, Mealing G, Comas T, Gendron TF, Monette R, Morley P. An alternative Ca2+‐dependent mechanism of neuroprotection by the metalloporphyrin class of superoxide dismutase mimetics. FASEB J 2005; 19:1734-6. [PMID: 16081500 DOI: 10.1096/fj.05-3795fje] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study challenges the conventional view that metalloporphyrins protect cultured cortical neurons in models of cerebral ischemia by acting as intracellular catalytic antioxidants [superoxide dismutase (SOD) mimetics]. High SOD-active Mn(III)porphyrins meso-substituted with N,N'-dimethylimidazolium or N-alkylpyridinium groups did not protect neurons against oxygen-glucose deprivation (OGD), although lower SOD-active and -inactive para isomers protected against N-methyl-D-aspartate (NMDA) exposure. Mn(III)meso-tetrakis(4-benzoic acid)porphyrin (Mn(III)TBAP), as well as SOD-inactive metalloTBAPs and other phenyl ring- or beta-substituted metalloporphyrins that contained redox-insensitive metals, protected cultures against OGD and NMDA neurotoxicity. Crucially, neuroprotective metalloporphyrins suppressed OGD- or NMDA-induced rises in intracellular Ca2+ concentration in the same general rank order as observed for neuroprotection. Results from paraquat toxicity, intracellular fluorescence quenching, electrophysiology, mitochondrial Ca2+, and spontaneous synaptic activity experiments suggest a model in which metalloporphyrins, acting at the plasma membrane, protect neurons against OGD by suppressing postsynaptic NMDA receptor-mediated Ca2+ rises, thereby indirectly preventing accumulation of neurotoxic mitochondrial Ca2+ levels. Though neuroprotective in a manner not originally intended, SOD-inactive metalloporphyrins may represent promising therapeutic agents in diseases such as cerebral ischemia, in which Ca2+ toxicity is implicated. Conventional syntheses aimed at improving the catalytic antioxidant capability and/or intracellular access of metalloporphyrins may not yield improved efficacy in some disease models.
Collapse
Affiliation(s)
- Joseph S Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Pathophysiology Group, Ottawa, ON, Canada.
| | | | | | | | | | | | | | | |
Collapse
|