1
|
Chen H, He Y, Chen S, Qi S, Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res 2020; 158:104877. [PMID: 32407958 DOI: 10.1016/j.phrs.2020.104877] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Oxidative/nitrosative stress and neuroinflammation are critical pathological processes in cerebral ischemia-reperfusion injury, and their intimate interactions mediate neuronal damage, blood-brain barrier (BBB) damage and hemorrhagic transformation (HT) during ischemic stroke. We review current progress towards understanding the interactions of oxidative/nitrosative stress and inflammatory responses in ischemic brain injury. The interactions between reactive oxygen species (ROS)/reactive nitrogen species (RNS) and innate immune receptors such as TLR2/4, NOD-like receptor, RAGE, and scavenger receptors are crucial pathological mechanisms that amplify brain damage during cerebral ischemic injury. Furthermore, we review the current progress of omics and systematic biology approaches for studying complex network regulations related to oxidative/nitrosative stress and inflammation in the pathology of ischemic stroke. Targeting oxidative/nitrosative stress and neuroinflammation could be a promising therapeutic strategy for ischemic stroke treatment. We then review recent advances in discovering compounds from medicinal herbs with the bioactivities of simultaneously regulating oxidative/nitrosative stress and pro-inflammatory molecules for minimizing ischemic brain injury. These compounds include sesamin, baicalin, salvianolic acid A, 6-paradol, silymarin, apocynin, 3H-1,2-Dithiole-3-thione, (-)-epicatechin, rutin, Dl-3-N-butylphthalide, and naringin. We finally summarize recent developments of the omics and systematic biology approaches for exploring the molecular mechanisms and active compounds of Traditional Chinese Medicine (TCM) formulae with the properties of antioxidant and anti-inflammation for neuroprotection. The comprehensive omics and systematic biology approaches provide powerful tools for exploring therapeutic principles of TCM formulae and developing precision medicine for stroke treatment.
Collapse
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Yacong He
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China; School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
2
|
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
|
3
|
Sriram CS, Jangra A, Kasala ER, Bodduluru LN, Bezbaruah BK. Targeting poly(ADP-ribose)polymerase1 in neurological diseases: A promising trove for new pharmacological interventions to enter clinical translation. Neurochem Int 2014; 76:70-81. [PMID: 25049175 DOI: 10.1016/j.neuint.2014.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022]
Abstract
The highly conserved abundant nuclear protein poly(ADP-ribose)polymerase1 (PARP1) functions at the center of cellular stress response and is mainly implied in DNA damage repair mechanism. Apart from its involvement in DNA damage repair, it does sway multiple vital cellular processes such as cell death pathways, cell aging, insulator function, chromatin modification, transcription and mitotic apparatus function. Since brain is the principal organ vulnerable to oxidative stress and inflammatory responses, upon stress encounters robust DNA damage can occur and intense PARP1 activation may result that will lead to various CNS diseases. In the context of soaring interest towards PARP1 as a therapeutic target for newer pharmacological interventions, here in the present review, we are attempting to give a silhouette of the role of PARP1 in the neurological diseases and the potential of its inhibitors to enter clinical translation, along with its structural and functional aspects.
Collapse
Affiliation(s)
- Chandra Shekhar Sriram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India.
| | - Ashok Jangra
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Eshvendar Reddy Kasala
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Lakshmi Narendra Bodduluru
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Babul Kumar Bezbaruah
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India; Department of Pharmacology, III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| |
Collapse
|
4
|
Curtin N, Szabo C. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol Aspects Med 2013; 34:1217-56. [PMID: 23370117 PMCID: PMC3657315 DOI: 10.1016/j.mam.2013.01.006] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/12/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
The aim of this article is to describe the current and potential clinical translation of pharmacological inhibitors of poly(ADP-ribose) polymerase (PARP) for the therapy of various diseases. The first section of the present review summarizes the available preclinical and clinical data with PARP inhibitors in various forms of cancer. In this context, the role of PARP in single-strand DNA break repair is relevant, leading to replication-associated lesions that cannot be repaired if homologous recombination repair (HRR) is defective, and the synthetic lethality of PARP inhibitors in HRR-defective cancer. HRR defects are classically associated with BRCA1 and 2 mutations associated with familial breast and ovarian cancer, but there may be many other causes of HRR defects. Thus, PARP inhibitors may be the drugs of choice for BRCA mutant breast and ovarian cancers, and extend beyond these tumors if appropriate biomarkers can be developed to identify HRR defects. Multiple lines of preclinical data demonstrate that PARP inhibition increases cytotoxicity and tumor growth delay in combination with temozolomide, topoisomerase inhibitors and ionizing radiation. Both single agent and combination clinical trials are underway. The final part of the first section of the present review summarizes the current status of the various PARP inhibitors that are in various stages of clinical development. The second section of the present review summarizes the role of PARP in selected non-oncologic indications. In a number of severe, acute diseases (such as stroke, neurotrauma, circulatory shock and acute myocardial infarction) the clinical translatability of PARP inhibition is supported by multiple lines of preclinical data, as well as observational data demonstrating PARP activation in human tissue samples. In these disease indications, PARP overactivation due to oxidative and nitrative stress drives cell necrosis and pro-inflammatory gene expression, which contributes to disease pathology. Accordingly, multiple lines of preclinical data indicate the efficacy of PARP inhibitors to preserve viable tissue and to down-regulate inflammatory responses. As the clinical trials with PARP inhibitors in various forms of cancer progress, it is hoped that a second line of clinical investigations, aimed at testing of PARP inhibitors for various non-oncologic indications, will be initiated, as well.
Collapse
Affiliation(s)
- Nicola Curtin
- Department of Experimental Cancer Therapy, Northern Institute for Cancer Research, Newcastle University, University of Newcastle Upon Tyne, UK
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
5
|
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
|
6
|
Egi Y, Matsuura S, Maruyama T, Fujio M, Yuki S, Akira T. Neuroprotective effects of a novel water-soluble poly(ADP-ribose) polymerase-1 inhibitor, MP-124, in in vitro and in vivo models of cerebral ischemia. Brain Res 2011; 1389:169-76. [PMID: 21420942 DOI: 10.1016/j.brainres.2011.03.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/10/2011] [Accepted: 03/13/2011] [Indexed: 11/28/2022]
Abstract
Cerebral ischemia induces excessive activation of poly(ADP-ribose) polymerase-1 (PARP-1), leading to neuronal cell death and the development of post-ischemic dysfunction. Blockade of PARP-related signals during cerebral ischemia has become a focus of interest as a new therapeutic approach for acute stroke treatment. The purpose of the present study was to examine the pharmacological profiles of MP-124, a novel water-soluble PARP-1 inhibitor, and its neuroprotective effects on ischemic injury in vitro and in vivo. MP-124 demonstrated competitive inhibition of the PARP-1 activity of human recombinant PARP-1 enzyme (Ki=16.5nmol/L). In P388D(1) cells, MP-124 inhibited the LDH leakage induced by H(2)O(2) in a concentration-dependent manner. (IC(50)=20.8nmol/L). In rat primary cortical neurons, MP-124 also inhibited the NAD depletion and polymerized ADP-ribose formation induced by H(2)O(2) exposure. Moreover, we investigated the neuroprotective effects of MP-124 in rat permanent and transient stroke models. In the rat permanent middle cerebral artery occlusion (MCAO) model, MP-124 was administered intravenously for 24h from 5min after the onset of MCAO. MP-124 (1, 3 and 10mg/kg/h) significantly inhibited the cerebral infarction in a dose-dependent manner (18, 42 and 48%). In rat transient MCAO model, MP-124 was administered intravenously from 30min after the onset of MCAO. MP-124 (3 and 10mg/kg/h) significantly reduced the infarct volume (53% and 50%). The present findings suggest that MP-124 acts as a potent neuroprotective agent in focal ischemia and its actions can be attributed to a reduction in NAD depletion and PAR formation.
Collapse
Affiliation(s)
- Yasuhiro Egi
- Department II, Central Nervous System, Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan.
| | | | | | | | | | | |
Collapse
|
7
|
Ferraris DV. Evolution of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. From concept to clinic. J Med Chem 2010; 53:4561-84. [PMID: 20364863 DOI: 10.1021/jm100012m] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Dana V Ferraris
- Johns Hopkins University Brain Science Institute, 855 N. Wolfe Street, Baltimore, Maryland 21205, USA.
| |
Collapse
|
8
|
Szabadfi K, Mester L, Reglodi D, Kiss P, Babai N, Racz B, Kovacs K, Szabo A, Tamas A, Gabriel R, Atlasz T. Novel neuroprotective strategies in ischemic retinal lesions. Int J Mol Sci 2010; 11:544-561. [PMID: 20386654 PMCID: PMC2852854 DOI: 10.3390/ijms11020544] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 01/27/2010] [Accepted: 01/27/2010] [Indexed: 02/04/2023] Open
Abstract
Retinal ischemia can be effectively modeled by permanent bilateral common carotid artery occlusion, which leads to chronic hypoperfusion-induced degeneration in the entire rat retina. The complex pathways leading to retinal cell death offer a complex approach of neuroprotective strategies. In the present review we summarize recent findings with different neuroprotective candidate molecules. We describe the protective effects of intravitreal treatment with: (i) urocortin 2; (ii) a mitochondrial ATP-sensitive K+ channel opener, diazoxide; (iii) a neurotrophic factor, pituitary adenylate cyclase activating polypeptide; and (iv) a novel poly(ADP-ribose) polymerase inhibitor (HO3089). The retinoprotective effects are demonstrated with morphological description and effects on apoptotic pathways using molecular biological techniques.
Collapse
Affiliation(s)
- Krisztina Szabadfi
- Department of Experimental Zoology and Neurobiology, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(K.S.);
(N.B.);
(R.G.)
| | - Laszlo Mester
- Department of Biochemistry and Medical Chemistry, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(L.M.);
(B.R.);
(K.K.);
(A.S.)
| | - Dora Reglodi
- Department of Anatomy, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(D.R.);
(P.K.);
(A.T.)
| | - Peter Kiss
- Department of Anatomy, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(D.R.);
(P.K.);
(A.T.)
| | - Norbert Babai
- Department of Experimental Zoology and Neurobiology, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(K.S.);
(N.B.);
(R.G.)
| | - Boglarka Racz
- Department of Biochemistry and Medical Chemistry, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(L.M.);
(B.R.);
(K.K.);
(A.S.)
| | - Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(L.M.);
(B.R.);
(K.K.);
(A.S.)
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(L.M.);
(B.R.);
(K.K.);
(A.S.)
| | - Andrea Tamas
- Department of Anatomy, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(D.R.);
(P.K.);
(A.T.)
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(K.S.);
(N.B.);
(R.G.)
| | - Tamas Atlasz
- Department of Experimental Zoology and Neurobiology, University of Pecs, H-7624 Pecs, Hungary; E-Mails:
(K.S.);
(N.B.);
(R.G.)
- Department of Sportbiology, University of Pecs, H-7624 Pecs, Hungary
- Author to whom correspondence should be addressed; E-Mail:
; Tel.: +36-72-503-600/4613; Fax: +36-72-501-517
| |
Collapse
|
9
|
Lubbers LS, Rowe BA, Hodge LM, Browne SE, Gundersdorf R, Jones P, Hess FJ, Reynolds IJ. PISA, a novel pharmacodynamic assay for assessing poly(ADP-ribose) polymerase (PARP) activity in situ. J Pharmacol Toxicol Methods 2010; 61:319-28. [PMID: 20132901 DOI: 10.1016/j.vascn.2010.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 01/22/2010] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Poly ADP-ribose polymerase (PARP) maintains genomic integrity by repairing DNA strand breaks, however over-activation of PARP following neural tissue injury is hypothesized to cause neuronal death. Therefore, PARP inhibitors have potential for limiting neural injury under certain conditions. A reliable method for assessing PARP activity in brain is critical for development of novel inhibitors with CNS activity. We developed the PARP In Situ Activity (PISA) assay to provide a direct, quantitative assessment of CNS PARP activity in vitro or in vivo. METHODS The assay utilized brain sections from rats with striatal kainic acid (KA) lesions and 3H- or biotinylated NAD+ as the substrate to assess PARP activity. Following optimization of the assay, it was used to assess in vitro and in vivo efficacy of known and novel PARP inhibitors. The assay also was used to assess PARP activity in male and female gonad-intact and ovariectomized rats. RESULTS Using 3H-NAD+ as the substrate, PARP activity was greater (p<0.01) in tissue from KA-lesioned vs. non-lesioned rats. Using biotinylated NAD+ it was revealed that PARP activity was present ipsilateral to the KA injection site, and labeling was blocked by incubation with excess unlabeled NAD+ or PARP inhibitors. The PARP inhibitor, 3-aminobenzamide and several novel inhibitors reduced (p<0.01) polymerase activity in vitro. Furthermore, the inhibitor MRLSD303 reduced (p<0.001) PARP activity in vivo in both male and female rats. Finally, administration of the novel PARP inhibitor MRLIT115 dose-dependently reduced (p<0.001) polymerase activity in vivo. DISCUSSION The PISA assay provides a direct, quantitative method for assessing PARP activity in vitro and provides critical information on factors underlying in vivo efficacy of chemical inhibitors including brain penetration and target engagement. These findings support use of the PISA assay as a screening tool for testing efficacy of PARP inhibitors in brain.
Collapse
Affiliation(s)
- Laura S Lubbers
- Department of Stroke and Neurodegeneration, Merck Research Laboratories, 770 Sumneytown Pike, PO Box 4, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Chaitanya GV, Babu PP. Differential PARP cleavage: an indication of heterogeneous forms of cell death and involvement of multiple proteases in the infarct of focal cerebral ischemia in rat. Cell Mol Neurobiol 2009; 29:563-73. [PMID: 19225880 PMCID: PMC11506027 DOI: 10.1007/s10571-009-9348-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 01/07/2009] [Indexed: 11/28/2022]
Abstract
AIM Poly (ADP-ribose) polymerase (PARP) is a nuclear repair enzyme whose role is widely depicted in various physiological and pathological processes. In the present study, we wanted to check the status of PARP and the role of various cell death proteases involved in apoptotic and non-apoptotic forms of cell death during transient focal cerebral ischemia in rat model. The activation of these proteases can result in the production of PARP fragments which can be treated as specific signature fragments to the particular protease involved in the pathology and hence the type of cell death. RESULTS In the ischemic samples, we observed activation of calpain, cathepsin-b, caspase-3, and granzyme-b which were known to act on and cleave PARP to produce specific signature fragments by Western blot and immunohistochemical analysis. Cresyl violet staining showed the presence of apoptotic and necrotic cell deaths. Further we observed interaction of AIF and gra-b with PARP in double immunofluorescence and co-immunoprecipitation experiments. CONCLUSION Activation of calpains, cathepsin-b, caspase-3, and granzyme-b correlated with either apoptotic or necrotic cell deaths in cresyl violet staining. The appearance of PARP signature fragments gives a clear idea on the involvement of particular protease in the pathology. Appearance of signature fragments like 89- and 50-kDa indicates the involvement of apoptotic and necrotic cell death in the pathology. Further interaction of AIF and gra-b with PARP also indicates the involvement of non-apoptotic modes of cell death during the pathology of focal cerebral ischemia.
Collapse
Affiliation(s)
- Ganta Vijay Chaitanya
- Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 Andhra Pradesh India
| | - Phanithi Prakash Babu
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 Andhra Pradesh India
| |
Collapse
|
11
|
Protection against chronic hypoperfusion-induced retinal neurodegeneration by PARP inhibition via activation of PI-3-kinase Akt pathway and suppression of JNK and p38 MAP kinases. Neurotox Res 2009; 16:68-76. [PMID: 19526300 DOI: 10.1007/s12640-009-9049-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/25/2009] [Accepted: 03/27/2009] [Indexed: 12/11/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) activation is considered as a major regulator of cell death in various pathophysiological conditions, however, no direct information is available about its role in chronic hypoperfusion-induced neuronal death. Here, we provide evidence for the protective effect of PARP inhibition on degenerative retinal damage induced by bilateral common carotid artery occlusion (BCCAO), an adequate chronic hypoperfusion murine model. We found that BCCAO in adult male Wistar rats led to severe degeneration of all retinal layers that was attenuated by a carboxaminobenzimidazol-derivative PARP inhibitor (HO3089) administered unilaterally into the vitreous body immediately following carotid occlusion and then 4 times in a 2-week-period. Normal morphological structure of the retina was preserved and the thickness of the retinal layers was increased in HO3089-treated eyes compared to the BCCAO eyes. For Western blot studies, HO3089 was administered immediately after BCCAO and retinas were removed 4 h later. According to Western blot analysis utilizing phosphorylation-specific primary antibodies, besides activating poly-ADP-ribose (PAR) synthesis, BCCAO induced phosphorylation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). HO3089 inhibited PAR synthesis, and decreased the phosphorylation of these proapoptotic MAPKs. In addition, HO3089 treatment induced phosphorylation, that is activation, of the protective Akt/glycogen synthase kinase (GSK)-3beta and extracellular signal-regulated kinase (ERK1/2) signaling pathways. These data indicate that PARP activation has a major role in mediating chronic hypoperfusion-induced neuronal death, and inhibition of the enzyme prevents the pathological changes both in the morphology and the kinase signaling cascades involved. These results identify PARP inhibition as a possible molecular target in the clinical management of chronic hypoperfusion-induced neurodegenerative diseases including ocular ischemic syndrome.
Collapse
|
12
|
Ischemic tolerance as an active and intrinsic neuroprotective mechanism. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:171-95. [PMID: 18790275 DOI: 10.1016/s0072-9752(08)01909-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
13
|
Pan X, Sun L, Ma W, Tang Y, Long C, Tian L, Liu N, Feng Z, Zheng J. Overactivation of poly(adenosine phosphate-ribose) polymerase 1 and molecular events in neuronal injury after deep hypothermic circulatory arrest: study in a rabbit model. J Thorac Cardiovasc Surg 2007; 134:1227-33. [PMID: 17976454 DOI: 10.1016/j.jtcvs.2007.05.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/18/2007] [Accepted: 05/09/2007] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Although deep hypothermic circulatory arrest has been known to induce neuronal injury, the molecular mechanism of this damage has not been identified. We studied the key molecular mediators through cellular energy failure, excitotoxicity, and overactivation of poly(adenosine diphosphate-ribose) polymerase 1 in brain tissues of a rabbit model of deep hypothermic circulatory arrest similar to clinical settings. METHODS We established 2 models of cardiopulmonary bypass (n = 15) and deep hypothermic circulatory arrest (n = 15) associated with cerebral microdialysis in rabbits. Deep hypothermic circulatory arrest lasted for 60 minutes. The measurements of glucose, lactate, pyruvate, and glutamate collected by means of microdialysis were quantified by using a microdialysis analyzer and high-performance liquid chromatography. The overactivation of poly(adenosine diphosphate-ribose) polymerase 1 was assessed by detecting immunostaining of poly(adenosine diphosphate-ribose). Histologic studies were used to identify neuronal morphologic changes and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling staining and poly(adenosine diphosphate-ribose) polymerase 1 Western blotting were used to identify apoptotic cells and early apoptotic signals. RESULTS Deep hypothermic circulatory arrest significantly increased the lactate/pyruvate and lactate/glucose ratios and the glutamate value, whereas cardiopulmonary bypass did not (P < .05). Deep hypothermic circulatory arrest significantly increased the numbers of poly(adenosine diphosphate-ribose)-positive and apoptotic neurons compared with cardiopulmonary bypass (P < .05). The cleavage of poly(adenosine diphosphate-ribose) polymerase 1 was only found in the deep hypothermic circulatory arrest group. More injured neurons were found in the deep hypothermic circulatory arrest group (histologic scores, P < .05). CONCLUSIONS This study demonstrated that deep hypothermic circulatory arrest results in an overactivation of poly(adenosine diphosphate-ribose) polymerase 1, and that there were molecular events consisting of cellular energy failure, excitotoxicity, overactivation of poly(adenosine diphosphate-ribose) polymerase 1, and necrosis and/or apoptosis in neuronal injury.
Collapse
Affiliation(s)
- Xudong Pan
- Department of Cardiovascular Surgery and Aortic Surgery Center, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Traumatic brain injury (TBI) is a devastating occurrence that may result in short- and long-term complications. Oxidative stress (an imbalance between oxidants and antioxidants) plays a critical role in the development of secondary injuries following TBI and, consequently, in patient outcomes. Secondary injuries resulting from oxidative stress produce DNA strand breaks that activate poly(adenosine diphosphate [ADP]-ribose) polymerase-1 (PARP-1) and produce another level of injury. PARP-1 functions as a DNA-damage sensor and signaling molecule. In response to the severe DNA damage after brain injury, PARP-1 becomes overactivated and depletes the cells' energy sources, which leads to cellular and neuronal death. Recently, PARP-1 inhibition has been studied in various animal models of brain injury with promising results. TBI treatments based on PARP-1 inhibition in humans are far from the clinical arena, although descriptive studies of PARP-1 activation in humans are beginning to emerge. Nurses should become involved in this area of collaborative research in human response to brain injury by helping design and implement safe and meaningful clinical trials.
Collapse
|
15
|
Diaz-Hernandez JI, Moncada S, Bolaños JP, Almeida A. Poly(ADP-ribose) polymerase-1 protects neurons against apoptosis induced by oxidative stress. Cell Death Differ 2007; 14:1211-21. [PMID: 17347665 DOI: 10.1038/sj.cdd.4402117] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In neurons, DNA is prone to free radical damage, although repair mechanisms preserve the genomic integrity. However, activation of the DNA repair system, poly(ADP-ribose) polymerase (PARP-1), is thought to cause neuronal death through NAD+ depletion and mitochondrial membrane potential (delta psi(m)) depolarization. Here, we show that abolishing PARP-1 activity in primary cortical neurons can either enhance or prevent apoptotic death, depending on the intensity of an oxidative stress. Only in severe oxidative stress does PARP-1 activation result in NAD+ and ATP depletion and neuronal death. To investigate the role of PARP-1 in an endogenous model of oxidative stress, we used an RNA interference (RNAi) strategy to specifically knock down glutamate-cysteine ligase (GCL), the rate-limiting enzyme of glutathione biosynthesis. GCL RNAi spontaneously elicited a mild type of oxidative stress that was enough to stimulate PARP-1 in a Ca2+-calmodulin kinase II-dependent manner. GCL RNAi-mediated PARP-1 activation facilitated DNA repair, although neurons underwent delta psi(m) loss followed by some apoptotic death. PARP-1 inhibition did not prevent delta psi(m) loss, but enhanced the vulnerability of neurons to apoptosis upon GCL silencing. Conversely, mild expression of PARP-1 partially prevented to GCL RNAi-dependent apoptosis. Thus, in the mild progressive damage likely occur in neurodegenerative diseases, PARP-1 activation plays a neuroprotective role that should be taken into account when considering therapeutic strategies.
Collapse
Affiliation(s)
- J I Diaz-Hernandez
- Unidad de Investigación, Hospital Universitario de Salamanca-Instituto de Estudios Ciencias de la Salud de Castilla y León, Salamanca, Spain
| | | | | | | |
Collapse
|
16
|
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
|