1
|
Peng B, Mohammed FS, Tang X, Liu J, Sheth KN, Zhou J. Nanotechnology approaches to drug delivery for the treatment of ischemic stroke. Bioact Mater 2025; 43:145-161. [PMID: 39386225 PMCID: PMC11462157 DOI: 10.1016/j.bioactmat.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Ischemic stroke is a major global public health concern that lacks effective treatment options. A significant challenge lies in delivering therapeutic agents to the brain due to the restrictive nature of the blood-brain barrier (BBB). The BBB's selectivity hampers the delivery of therapeutically relevant quantities of agents to the brain, resulting in a lack of FDA-approved pharmacotherapies for stroke. In this article, we review therapeutic agents that have been evaluated in clinical trials or are currently undergoing clinical trials. Subsequently, we survey strategies for synthesizing and engineering nanoparticles (NPs) for drug delivery to the ischemic brain. We then provide insights into the potential clinical translation of nanomedicine, offering a perspective on its transformative role in advancing stroke treatment strategies. In summary, existing literature suggests that drug delivery represents a major barrier for clinical translation of stroke pharmacotherapies. While nanotechnology has shown significant promise in addressing this challenge, further advancements aimed at improving delivery efficiency and simplifying formulations are necessary for successful clinical translation.
Collapse
Affiliation(s)
- Bin Peng
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Farrah S. Mohammed
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| | - Xiangjun Tang
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurosurgery, Taihe Hospital, Hubei, 442000, PR China
| | - Jia Liu
- Department of Neurosurgery, New Haven, CT, 06510, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Yale University, New Haven, CT, 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| |
Collapse
|
2
|
Choi DH, Choi IA, Lee J. Role of NADPH Oxidases in Stroke Recovery. Antioxidants (Basel) 2024; 13:1065. [PMID: 39334724 PMCID: PMC11428334 DOI: 10.3390/antiox13091065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke is one of the most significant causes of death and long-term disability globally. Overproduction of reactive oxygen species by NADPH oxidase (NOX) plays an important role in exacerbating oxidative stress and causing neuronal damage after a stroke. There is growing evidence that NOX inhibition prevents ischemic injury and that the role of NOX in brain damage or recovery depends on specific post-stroke phases. In addition to studies on post-stroke neuroprotection by NOX inhibition, recent reports have also demonstrated the role of NOX in stroke recovery, a critical process for brain adaptation and functional reorganization after a stroke. Therefore, in this review, we investigated the role of NOX in stroke recovery with the aim of integrating preclinical findings into potential therapeutic strategies to improve stroke recovery.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
- Department of Medical Science, Konkuk University School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - In-Ae Choi
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
- Department of Occupational Therapy, Division of Health, Baekseok University, Cheonan-si 31065, Republic of Korea
| | - Jongmin Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
3
|
Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int J Mol Sci 2022; 23:ijms23137292. [PMID: 35806303 PMCID: PMC9266317 DOI: 10.3390/ijms23137292] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Differential evolution of apoptosis, programmed necrosis, and autophagy, parthanatos is a form of cell death mediated by poly(ADP-ribose) polymerase 1 (PARP1), which is caused by DNA damage. PARP1 hyper-activation stimulates apoptosis-inducing factor (AIF) nucleus translocation, and accelerates nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP) depletion, leading to DNA fragmentation. The mechanisms of parthanatos mainly include DNA damage, PARP1 hyper-activation, PAR accumulation, NAD+ and ATP depletion, and AIF nucleus translocation. Now, it is reported that parthanatos widely exists in different diseases (tumors, retinal diseases, neurological diseases, diabetes, renal diseases, cardiovascular diseases, ischemia-reperfusion injury...). Excessive or defective parthanatos contributes to pathological cell damage; therefore, parthanatos is critical in the therapy and prevention of many diseases. In this work, the hallmarks and molecular mechanisms of parthanatos and its related disorders are summarized. The questions raised by the recent findings are also presented. Further understanding of parthanatos will provide a new treatment option for associated conditions.
Collapse
|
4
|
Liu S, Luo W, Wang Y. Emerging role of PARP-1 and PARthanatos in ischemic stroke. J Neurochem 2021; 160:74-87. [PMID: 34241907 DOI: 10.1111/jnc.15464] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/10/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
Cell death is a key feature of neurological diseases, including stroke and neurodegenerative disorders. Studies in a variety of ischemic/hypoxic mouse models demonstrate that poly(ADP-ribose) polymerase 1 (PARP-1)-dependent cell death, also named PARthanatos, plays a pivotal role in ischemic neuronal cell death and disease progress. PARthanatos has its unique triggers, processors, and executors that convey a highly orchestrated and programmed signaling cascade. In addition to its role in gene transcription, DNA damage repair, and energy homeostasis through PARylation of its various targets, PARP-1 activation in neuron and glia attributes to brain damage following ischemia/reperfusion. Pharmacological inhibition or genetic deletion of PARP-1 reduces infarct volume, eliminates inflammation, and improves recovery of neurological functions in stroke. Here, we reviewed the role of PARP-1 and PARthanatos in stroke and their therapeutic potential.
Collapse
Affiliation(s)
- Shuiqiao Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Weibo Luo
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yingfei Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
Koehler RC, Dawson VL, Dawson TM. Targeting Parthanatos in Ischemic Stroke. Front Neurol 2021; 12:662034. [PMID: 34025565 PMCID: PMC8131834 DOI: 10.3389/fneur.2021.662034] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parthanatos is a cell death signaling pathway in which excessive oxidative damage to DNA leads to over-activation of poly(ADP-ribose) polymerase (PARP). PARP then generates the formation of large poly(ADP-ribose) polymers that induce the release of apoptosis-inducing factor from the outer mitochondrial membrane. In the cytosol, apoptosis-inducing factor forms a complex with macrophage migration inhibitory factor that translocates into the nucleus where it degrades DNA and produces cell death. In a review of the literature, we identified 24 publications from 13 laboratories that support a role for parthanatos in young male mice and rats subjected to transient and permanent middle cerebral artery occlusion (MCAO). Investigators base their conclusions on the use of nine different PARP inhibitors (19 studies) or PARP1-null mice (7 studies). Several studies indicate a therapeutic window of 4-6 h after MCAO. In young female rats, two studies using two different PARP inhibitors from two labs support a role for parthanatos, whereas two studies from one lab do not support a role in young female PARP1-null mice. In addition to parthanatos, a body of literature indicates that PARP inhibitors can reduce neuroinflammation by interfering with NF-κB transcription, suppressing matrix metaloproteinase-9 release, and limiting blood-brain barrier damage and hemorrhagic transformation. Overall, most of the literature strongly supports the scientific premise that a PARP inhibitor is neuroprotective, even when most did not report behavior outcomes or address the issue of randomization and treatment concealment. Several third-generation PARP inhibitors entered clinical oncology trials without major adverse effects and could be repurposed for stroke. Evaluation in aged animals or animals with comorbidities will be important before moving into clinical stroke trials.
Collapse
Affiliation(s)
- Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, United States
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Physiology, The Johns Hopkins University, Baltimore, MD, United States
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
6
|
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: 103] [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
|
7
|
Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, Fang W, Xiong X. The Involvement and Therapy Target of Immune Cells After Ischemic Stroke. Front Immunol 2019; 10:2167. [PMID: 31572378 PMCID: PMC6749156 DOI: 10.3389/fimmu.2019.02167] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
After ischemic stroke, the integrity of the blood-brain barrier is compromised. Peripheral immune cells, including neutrophils, T cells, B cells, dendritic cells, and macrophages, infiltrate into the ischemic brain tissue and play an important role in regulating the progression of ischemic brain injury. In this review, we will discuss the role of different immune cells after stroke in the secondary inflammatory reaction and focus on the phenotypes and functions of macrophages in ischemic stroke, as well as briefly introduce the anti-ischemic stroke therapy targeting macrophages.
Collapse
Affiliation(s)
- Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, China
| | - Xiqun Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daniel Smerin
- Department of Neurosurgery, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Huang C, Mazdeyasna S, Chen L, Abu Jawdeh EG, Bada HS, Saatman KE, Chen L, Yu G. Noninvasive noncontact speckle contrast diffuse correlation tomography of cerebral blood flow in rats. Neuroimage 2019; 198:160-169. [DOI: 10.1016/j.neuroimage.2019.05.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/05/2023] Open
|
9
|
Kotagale NR, Taksande BG, Inamdar NN. Neuroprotective offerings by agmatine. Neurotoxicology 2019; 73:228-245. [DOI: 10.1016/j.neuro.2019.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022]
|
10
|
McGurk L, Rifai OM, Bonini NM. Poly(ADP-Ribosylation) in Age-Related Neurological Disease. Trends Genet 2019; 35:601-613. [PMID: 31182245 DOI: 10.1016/j.tig.2019.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022]
Abstract
A central and causative feature of age-related neurodegenerative disease is the deposition of misfolded proteins in the brain. To devise novel approaches to treatment, regulatory pathways that modulate these aggregation-prone proteins must be defined. One such pathway is post-translational modification by the addition of poly(ADP-ribose) (PAR), which promotes protein recruitment and localization in several cellular contexts. Mounting evidence implicates PAR in seeding the abnormal localization and accumulation of proteins that are causative of neurodegenerative disease. Inhibitors of PAR polymerase (PARP) activity have been developed as cancer therapeutics, raising the possibility that they could be used to treat neurodegenerative disease. We focus on pathways regulated by PAR in neurodegenerative disease, with emphasis on amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD).
Collapse
Affiliation(s)
- Leeanne McGurk
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Olivia M Rifai
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
McGurk L, Mojsilovic-Petrovic J, Van Deerlin VM, Shorter J, Kalb RG, Lee VM, Trojanowski JQ, Lee EB, Bonini NM. Nuclear poly(ADP-ribose) activity is a therapeutic target in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2018; 6:84. [PMID: 30157956 PMCID: PMC6114235 DOI: 10.1186/s40478-018-0586-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and fatal motor neuron disease. Diagnosis typically occurs in the fifth decade of life and the disease progresses rapidly leading to death within ~ 2–5 years of symptomatic onset. There is no cure, and the few available treatments offer only a modest extension in patient survival. A protein central to ALS is the nuclear RNA/DNA-binding protein, TDP-43. In > 95% of ALS patients, TDP-43 is cleared from the nucleus and forms phosphorylated protein aggregates in the cytoplasm of affected neurons and glia. We recently defined that poly(ADP-ribose) (PAR) activity regulates TDP-43-associated toxicity. PAR is a posttranslational modification that is attached to target proteins by PAR polymerases (PARPs). PARP-1 and PARP-2 are the major enzymes that are active in the nucleus. Here, we uncovered that the motor neurons of the ALS spinal cord were associated with elevated nuclear PAR, suggesting elevated PARP activity. Veliparib, a small-molecule inhibitor of nuclear PARP-1/2, mitigated the formation of cytoplasmic TDP-43 aggregates in mammalian cells. In primary spinal-cord cultures from rat, Veliparib also inhibited TDP-43-associated neuronal death. These studies uncover that PAR activity is misregulated in the ALS spinal cord, and a small-molecular inhibitor of PARP-1/2 activity may have therapeutic potential in the treatment of ALS and related disorders associated with abnormal TDP-43 homeostasis.
Collapse
|
12
|
Shekhar S, Cunningham MW, Pabbidi MR, Wang S, Booz GW, Fan F. Targeting vascular inflammation in ischemic stroke: Recent developments on novel immunomodulatory approaches. Eur J Pharmacol 2018; 833:531-544. [PMID: 29935175 DOI: 10.1016/j.ejphar.2018.06.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
Abstract
Ischemic stroke is a devastating and debilitating medical condition with limited therapeutic options. However, accumulating evidence indicates a central role of inflammation in all aspects of stroke including its initiation, the progression of injury, and recovery or wound healing. A central target of inflammation is disruption of the blood brain barrier or neurovascular unit. Here we discuss recent developments in identifying potential molecular targets and immunomodulatory approaches to preserve or protect barrier function and limit infarct damage and functional impairment. These include blocking harmful inflammatory signaling in endothelial cells, microglia/macrophages, or Th17/γδ T cells with biologics, third generation epoxyeicosatrienoic acid (EET) analogs with extended half-life, and miRNA antagomirs. Complementary beneficial pathways may be enhanced by miRNA mimetics or hyperbaric oxygenation. These immunomodulatory approaches could be used to greatly expand the therapeutic window for thrombolytic treatment with tissue plasminogen activator (t-PA). Moreover, nanoparticle technology allows for the selective targeting of endothelial cells for delivery of DNA/RNA oligonucleotides and neuroprotective drugs. In addition, although likely detrimental to the progression of ischemic stroke by inducing inflammation, oxidative stress, and neuronal cell death, 20-HETE may also reduce susceptibility of onset of ischemic stroke by maintaining autoregulation of cerebral blood flow. Although the interaction between inflammation and stroke is multifaceted, a better understanding of the mechanisms behind the pro-inflammatory state at all stages will hopefully help in developing novel immunomodulatory approaches to improve mortality and functional outcome of those inflicted with ischemic stroke.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA; Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
13
|
Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations. Pharmacol Res 2016; 110:242-264. [DOI: 10.1016/j.phrs.2015.12.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
|
14
|
Kwiatkowski D, Czarny P, Galecki P, Bachurska A, Talarowska M, Orzechowska A, Bobińska K, Bielecka-Kowalska A, Pietras T, Szemraj J, Maes M, Sliwinski T. Variants of Base Excision Repair Genes MUTYH , PARP1 and XRCC1 in Alzheimer's Disease Risk. Neuropsychobiology 2016; 71:176-86. [PMID: 25998844 DOI: 10.1159/000381985] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/30/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Many clinical studies have shown that oxidative stress pathways and the efficiency of the oxidative DNA damage base excision repair (BER) system are associated with the pathogenesis of Alzheimer's disease (AD). Reduced BER efficiency may result from polymorphisms of BER-related genes. In the present study, we examine whether single nucleotide polymorphisms (SNPs) of BER genes are associated with increased risk of AD. METHODS SNP genotyping was carried out on DNA isolated from peripheral blood mononuclear cells obtained from 120 patients with AD and 110 healthy volunteers. Samples were genotyped for the presence of BER-related SNPs, i.e. XRCC1-rs1799782, rs25487; MUTYH-rs3219489, and PARP1-rs1136410. RESULTS We found a positive association between AD risk and the presence of G/A genotype variant of the XRCC1 rs25487 polymorphism [odds ratio (OR) = 3.762, 95% CI: 1.793-7.891]. The presence of the A/A genotype of this polymorphism reduced the risk of AD (OR = 0.485, 95% CI: 0.271-0.870). In cases of the PARP1 gene rs1136410 polymorphism, we observed that the T/C variant increases (OR = 4.159, 95% CI: 1.978-8.745) while the T/T variant reduces risk (OR = 0.240, 95% CI: 0.114-0.556) of AD. CONCLUSIONS We conclude that BER gene polymorphisms may play an important role in the etiology of AD. Diagnosing the presence or absence of particular genetic variants may be an important marker of AD. Further research on a larger population is needed. There is also a need to examine polymorphisms of other BER in the context of AD risk.
Collapse
|
15
|
Sun Y, Zhang L, Chen Y, Zhan L, Gao Z. Therapeutic Targets for Cerebral Ischemia Based on the Signaling Pathways of the GluN2B C Terminus. Stroke 2015; 46:2347-53. [DOI: 10.1161/strokeaha.115.009314] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/09/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Yongjun Sun
- From the Department of Pharmacy (Y.S., Y.C., L. Zhan, Z.G.), Hebei Research Center of Pharmaceutical and Chemical Engineering (Y.S., Z.G.), and State Key Laboratory Breeding Base—Hebei Province Key Laboratory of Molecular Chemistry for Drug (Z.G.), Hebei University of Science and Technology, Shijiazhuang, China; and Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China (L. Zhang)
| | - Linan Zhang
- From the Department of Pharmacy (Y.S., Y.C., L. Zhan, Z.G.), Hebei Research Center of Pharmaceutical and Chemical Engineering (Y.S., Z.G.), and State Key Laboratory Breeding Base—Hebei Province Key Laboratory of Molecular Chemistry for Drug (Z.G.), Hebei University of Science and Technology, Shijiazhuang, China; and Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China (L. Zhang)
| | - You Chen
- From the Department of Pharmacy (Y.S., Y.C., L. Zhan, Z.G.), Hebei Research Center of Pharmaceutical and Chemical Engineering (Y.S., Z.G.), and State Key Laboratory Breeding Base—Hebei Province Key Laboratory of Molecular Chemistry for Drug (Z.G.), Hebei University of Science and Technology, Shijiazhuang, China; and Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China (L. Zhang)
| | - Liying Zhan
- From the Department of Pharmacy (Y.S., Y.C., L. Zhan, Z.G.), Hebei Research Center of Pharmaceutical and Chemical Engineering (Y.S., Z.G.), and State Key Laboratory Breeding Base—Hebei Province Key Laboratory of Molecular Chemistry for Drug (Z.G.), Hebei University of Science and Technology, Shijiazhuang, China; and Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China (L. Zhang)
| | - Zibin Gao
- From the Department of Pharmacy (Y.S., Y.C., L. Zhan, Z.G.), Hebei Research Center of Pharmaceutical and Chemical Engineering (Y.S., Z.G.), and State Key Laboratory Breeding Base—Hebei Province Key Laboratory of Molecular Chemistry for Drug (Z.G.), Hebei University of Science and Technology, Shijiazhuang, China; and Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China (L. Zhang)
| |
Collapse
|
16
|
Fatokun AA, Dawson VL, Dawson TM. Parthanatos: mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 2014; 171:2000-16. [PMID: 24684389 PMCID: PMC3976618 DOI: 10.1111/bph.12416] [Citation(s) in RCA: 399] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/27/2013] [Accepted: 09/02/2013] [Indexed: 12/12/2022] Open
Abstract
Cells die by a variety of mechanisms. Terminally differentiated cells such as neurones die in a variety of disorders, in part, via parthanatos, a process dependent on the activity of poly (ADP-ribose)-polymerase (PARP). Parthanatos does not require the mediation of caspases for its execution, but is clearly mechanistically dependent on the nuclear translocation of the mitochondrial-associated apoptosis-inducing factor (AIF). The nuclear translocation of this otherwise beneficial mitochondrial protein, occasioned by poly (ADP-ribose) (PAR) produced through PARP overactivation, causes large-scale DNA fragmentation and chromatin condensation, leading to cell death. This review describes the multistep course of parthanatos and its dependence on PAR signalling and nuclear AIF translocation. The review also discusses potential targets in the parthanatos cascade as promising avenues for the development of novel, disease-modifying, therapeutic agents.
Collapse
Affiliation(s)
- Amos A Fatokun
- Institute of Cell Signalling, School of Biomedical Sciences, University of NottinghamNottingham, UK
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
17
|
Chen Y, Won SJ, Xu Y, Swanson RA. Targeting microglial activation in stroke therapy: pharmacological tools and gender effects. Curr Med Chem 2014; 21:2146-55. [PMID: 24372213 PMCID: PMC4076056 DOI: 10.2174/0929867321666131228203906] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/02/2013] [Accepted: 06/03/2013] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is caused by critical reductions in blood flow to brain or spinal cord. Microglia are the resident immune cells of the central nervous system, and they respond to stroke by assuming an activated phenotype that releases cytotoxic cytokines, reactive oxygen species, proteases, and other factors. This acute, innate immune response may be teleologically adapted to limit infection, but in stroke this response can exacerbate injury by further damaging or killing nearby neurons and other cell types, and by recruiting infiltration of circulating cytotoxic immune cells. The microglial response requires hours to days to fully develop, and this time interval presents a clinically accessible time window for initiating therapy. Because of redundancy in cytotoxic microglial responses, the most effective therapeutic approach may be to target the global gene expression changes involved in microglial activation. Several classes of drugs can do this, including histone deacetylase inhibitors, minocycline and other PARP inhibitors, corticosteroids, and inhibitors of TNFα and scavenger receptor signaling. Here we review the pre-clinical studies in which these drugs have been used to suppress microglial activation after stroke. We also review recent advances in the understanding of sex differences in the CNS inflammatory response, as these differences are likely to influence the efficacy of drugs targeting post-stroke brain inflammation.
Collapse
Affiliation(s)
| | | | | | - R A Swanson
- Dept. of Neurology, University of California San Francisco; and Neurology Service, San Francisco Veterans Affairs Medical Center, 4150 Clement St, San Francisco, CA 94121, USA.
| |
Collapse
|
18
|
Feng X, Koh DW. Inhibition of poly(ADP-ribose) polymerase-1 or poly(ADP‑ribose) glycohydrolase individually, but not in combination, leads to improved chemotherapeutic efficacy in HeLa cells. Int J Oncol 2012; 42:749-56. [PMID: 23254695 PMCID: PMC3583631 DOI: 10.3892/ijo.2012.1740] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/19/2012] [Indexed: 01/19/2023] Open
Abstract
The genome-protecting role of poly(ADP-ribose) (PAR) has identified PAR polymerase-1 (PARP-1) and PAR glycohydrolase (PARG), two enzymes responsible for the synthesis and hydrolysis of PAR, as chemotherapeutic targets. Each has been previously individually evaluated in chemotherapy, but the effects of combination PARP-1 and PARG inhibition in cancer cells are not known. Here we determined the effects of the inhibition of PARP-1 and the absence or RNAi knockdown of PARG on PAR synthesis, cell death after chemotherapy and long-term viability. Using three experimental/clinical PARP-1 inhibitors in PARG-null cells, we show decreased levels of PAR and increased short-term and long-term viability with each inhibitor, with the exception of DPQ. Treatment with the experimental chemotherapeutic agent, N-methyl-N’-nitro-N-nitrosoguanidine (MNNG), led to increased cell death in PARG-null cells, but decreased cell death when pretreated with each PARP-1 inhibitor. Similar results were observed in MNNG-treated HeLa cells, where RNAi knockdown of PARG or pretreatment with ABT-888 led to increased HeLa cell death, whereas combination PARG RNAi knockdown + ABT-888 failed to produce increased cell death. The results demonstrate the ability of the PARP-1 inhibitors to decrease PAR levels, maintain viability and decrease PAR-mediated cell death after chemotherapeutic treatment in the absence of PARG. Further, the results demonstrate that the combination of PARP-1 and PARG inhibition in chemotherapy does not produce increased HeLa cell death. Thus, the results indicate that inhibiting both PARP-1 and PARG, which both are chemotherapeutic targets that increase cancer cell death, does not lead to synergistic cell death in HeLa cells. Therefore, strategies that target PAR metabolism for the improved treatment of cancer may be required to target PARP-1 and PARG individually in order to optimize cancer cell death.
Collapse
Affiliation(s)
- Xiaoxing Feng
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Pullman, WA 99164-6534, USA
| | | |
Collapse
|
19
|
Natural Inhibitors of Poly(ADP-ribose) Polymerase-1. Mol Neurobiol 2012; 46:55-63. [DOI: 10.1007/s12035-012-8257-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 03/01/2012] [Indexed: 01/08/2023]
|
20
|
Molderings GJ, Haenisch B. Agmatine (decarboxylated l-arginine): Physiological role and therapeutic potential. Pharmacol Ther 2012; 133:351-65. [DOI: 10.1016/j.pharmthera.2011.12.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 12/05/2011] [Indexed: 01/14/2023]
|
21
|
The pharmacological importance of agmatine in the brain. Neurosci Biobehav Rev 2012; 36:502-19. [DOI: 10.1016/j.neubiorev.2011.08.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 06/23/2011] [Accepted: 08/18/2011] [Indexed: 01/28/2023]
|
22
|
Maynard KI. Hormesis pervasiveness and its potential implications for pharmaceutical research and development. Dose Response 2011; 9:377-86. [PMID: 22013400 DOI: 10.2203/dose-response.11-026.maynard] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
This mini-review illustrates that hormesis is not only confined to the areas of biochemistry, radiation biology and toxicology, where it is traditionally known, but illustrates, by citing published scientific literature, that it is found across a wide range of biomedical science and clinical medicine such as neuroscience, cardiology and oncology. The use of techniques and technology, including high through-put screening, micro-dosing or phase 0 studies, pharmacometrics and adaptive trial design in the clinic, are proposed to illustrate how acknowledging the potential impact of hormesis throughout different stages of drug discovery and development, including hurdles related to efficacy and safety, could help the pharmaceutical industry address some of its major and frequently mentioned challenges.
Collapse
|
23
|
Experimental Unilateral Spermatic Cord Torsion: The Effect of Polypolymerase Enzyme Inhibitor on Histopathological and Biochemical Changes in the Early and Late Periods in the Ipsilateral and Contralateral Testicles. Urology 2010; 76:507.e1-5. [DOI: 10.1016/j.urology.2010.03.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 03/03/2010] [Accepted: 03/26/2010] [Indexed: 11/19/2022]
|
24
|
Li X, Klaus JA, Zhang J, Xu Z, Kibler KK, Andrabi SA, Rao K, Yang ZJ, Dawson TM, Dawson VL, Koehler RC. Contributions of poly(ADP-ribose) polymerase-1 and -2 to nuclear translocation of apoptosis-inducing factor and injury from focal cerebral ischemia. J Neurochem 2010; 113:1012-22. [PMID: 20236222 PMCID: PMC2860677 DOI: 10.1111/j.1471-4159.2010.06667.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Excessive oxidative damage to DNA leads to activation of poly(ADP-ribose) polymerase-1 (PARP-1), accumulation of PAR polymers, translocation of apoptosis-inducing factor (AIF) from mitochondria to the nucleus, and cell death. In this study, we compared the effect of gene deletion of PARP-1 and PARP-2, enzymes activated by DNA oxidative damage, in male mice subjected to 2 h of focal cerebral ischemia. Infarct volume at 3 days of reperfusion was markedly decreased to a similar extent in PARP-1- and PARP-2-null mice. The ischemia-induced increase in nuclear AIF accumulation was largely suppressed in both knockout genotypes. The transient increase in PAR during early reperfusion was nearly blocked in PARP-1-null mice, but only moderately decreased at 1-h reperfusion in PARP-2-null mice. Differences in the tissue volume at risk, as assessed by arterial casts and autoradiographic analysis of regional blood flow, did not fully account for the large reductions in AIF translocation and infarct volume in both PARP null mice. Cell death was attenuated in PARP-2-null neurons exposed to a submaximal concentration of 100 microM NMDA for 5 min, but not in those exposed to a near-maximal toxic concentration of 500 microM NMDA. We conclude that PARP-2 contributes substantially to nuclear translocation of AIF and infarct size after transient focal cerebral ischemia in male mice, but that protection is disproportionate to the attenuation of overall PARP activity.
Collapse
Affiliation(s)
- Xiaoling Li
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Judith A. Klaus
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Jian Zhang
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Zhenfeng Xu
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Kathleen K. Kibler
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Shaida A. Andrabi
- Department of Neurology, The Johns Hopkins University, Baltimore, Maryland, USA
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Karthik Rao
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Zeng-Jin Yang
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Ted M. Dawson
- Department of Neurology, The Johns Hopkins University, Baltimore, Maryland, USA
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Valina L. Dawson
- Department of Neurology, The Johns Hopkins University, Baltimore, Maryland, USA
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Raymond C. Koehler
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Xu Z, Zhang J, David KK, Yang ZJ, Li X, Dawson TM, Dawson VL, Koehler RC. Endonuclease G does not play an obligatory role in poly(ADP-ribose) polymerase-dependent cell death after transient focal cerebral ischemia. Am J Physiol Regul Integr Comp Physiol 2010; 299:R215-21. [PMID: 20427721 DOI: 10.1152/ajpregu.00747.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of poly(ADP-ribose) polymerase (PARP) and subsequent translocation of apoptosis-inducing factor contribute to caspase-independent neuronal injury from N-methyl-d-aspartate, oxygen-glucose deprivation, and ischemic stroke. Some studies have implicated endonuclease G in the DNA fragmentation associated with caspase-independent cell death. Here, we compared wild-type and endonuclease G null mice to investigate whether endonuclease G plays a role in the PARP-dependent injury that results from transient focal cerebral ischemia. Latex casts did not reveal differences in the cerebral arterial distribution territory or posterior communicating arterial diameter, and the decrease in laser-Doppler flux during middle cerebral artery occlusion was similar in wild-type and endonuclease G null mice. After 90 min of occlusion and 1 day of reperfusion, similar degrees of nuclear translocation of apoptosis-inducing factor and DNA degradation were evident in male wild-type and null mice. At 3 days of reperfusion, infarct volume and neurological deficit scores were not different between male wild-type and endonuclease G null mice or between female wild-type and endonuclease G null mice. These data demonstrate that endonuclease G is not required for the pathogenesis of transient focal ischemia in either male or female mice. Treatment with a PARP inhibitor decreased infarct volume and deficit scores equivalently in male wild-type and endonuclease G null mice, indicating that the injury in endonuclease G null mice remains dependent on PARP. Thus endonuclease G is not obligatory for executing PARP-dependent injury during ischemic stroke.
Collapse
Affiliation(s)
- Zhenfeng Xu
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Strosznajder RP, Czubowicz K, Jesko H, Strosznajder JB. Poly(ADP-ribose) metabolism in brain and its role in ischemia pathology. Mol Neurobiol 2010; 41:187-96. [PMID: 20411356 DOI: 10.1007/s12035-010-8124-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 03/23/2010] [Indexed: 11/24/2022]
Abstract
The biological roles of poly(ADP-ribose) polymers (PAR) and poly(ADP-ribosyl)ation of proteins in the central nervous system are diverse. The homeostasis of PAR orchestrated by poly(ADP-ribose) polymerase-1 (PARP-1) and poly(ADP-ribose) glycohydrolase (PARG) is crucial for cell physiology and pathology. Both enzymes are ubiquitously distributed in neurons and glia; however, they are segregated at the subcellular level. PARP-1 serves as a "nick sensor" for single- or double-stranded breaks in DNA and is involved in long and short patch base-excision repair, while PARG breaks down PAR. The stimulation of PARP-1 and PAR formation can activate proinflammatory transcription factors, including nuclear factor kappa B. However, hyperactivation of PARP-1 can result in depletion of NAD/ATP, and in PAR-dependent mitochondrial pore formation leading to release of apoptosis inducing factor and cell death. The role of PAR as a death signaling molecule in brain ischemia-reperfusion and inflammation as well as the effect of gender and aging is presented in this review. Modulating the PAR level through pharmacological or genetic intervention on PARP-1/PARG activity and gene expression should be a valuable way for neuroprotective strategy.
Collapse
Affiliation(s)
- Robert Piotr Strosznajder
- Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego St., 02-106, Warsaw, Poland.
| | | | | | | |
Collapse
|
27
|
Kövesdi E, Bukovics P, Besson V, Nyirádi J, Lückl J, Pál J, Sümegi B, Dóczi T, Hernádi I, Büki A. A novel PARP inhibitor L-2286 in a rat model of impact acceleration head injury: an immunohistochemical and behavioral study. Int J Mol Sci 2010; 11:1253-68. [PMID: 20480019 PMCID: PMC2871115 DOI: 10.3390/ijms11041253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 03/11/2010] [Accepted: 03/22/2010] [Indexed: 11/17/2022] Open
Abstract
We examined the neuro/axono-protective potential of a novel poly (ADP-ribose) polymerase (PARP) inhibitor L-2286 in a rat impact acceleration brain injury model. Male Wistar rats (n = 70) weighing 300–350 grams were used to determine the most effective intracerebroventricular (i.c.v.) dose of L-2286 administered 30 min after injury, and to test the neuroprotective effect at two time points (immediately, and 30 min after injury). The neuroprotective effect of L-2286 was tested using immunohistochemical (amyloid precursor protein and mid-sized mouse anti-neurofilament clone RMO-14.9 antibody) and behavioral tests (beam-balance, open-field and elevated plus maze). At both time-points, a 100 μg/rat dose of i.c.v. L-2286 significantly (p < 0.05) reduced the density of damaged axons in the corticospinal tract and medial longitudinal fascicle compared to controls. In the behavioral tests, treatment 30 min post-injury improved motor function, while the level of anxiety was reduced in both treatment protocols.
Collapse
Affiliation(s)
- Erzsébet Kövesdi
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - Péter Bukovics
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - Valérie Besson
- Laboratoire de Pharmacologie de la Circulation Cérébrale, UPRES EA 2510, Université René Descartes, Paris, France; E-Mail:
(V.B.)
| | - József Nyirádi
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - János Lückl
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - József Pál
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - Balázs Sümegi
- Department of BioChemistry, University of Pécs, 7624 Pécs, Hungary; E-Mail:
(B.S.)
| | - Tamás Dóczi
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
| | - István Hernádi
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624, Hungary; E-Mail:
(I.H.)
| | - András Büki
- Department of Neurosurgery, Medical Faculty, University of Pécs, 7623 Pécs, Hungary; E-Mails:
(E.K.);
(P.B.);
(J.N.);
(J.L.);
(J.P.);
(T.D.)
- Author to whom correspondence should be addressed; E-Mail:
; Tel.: +36-72-535-900; Fax: +36-72-535-931
| |
Collapse
|
28
|
Peralta-Leal A, Rodríguez-Vargas JM, Aguilar-Quesada R, Rodríguez MI, Linares JL, de Almodóvar MR, Oliver FJ. PARP inhibitors: new partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med 2009; 47:13-26. [PMID: 19362586 DOI: 10.1016/j.freeradbiomed.2009.04.008] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 04/07/2009] [Accepted: 04/08/2009] [Indexed: 12/21/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are defined as cell signaling enzymes that catalyze the transfer of ADP-ribose units from NAD(+) to a number of acceptor proteins. PARP-1, the best characterized member of the PARP family, which currently comprises 18 members, is an abundant nuclear enzyme implicated in cellular responses to DNA injury provoked by genotoxic stress. PARP is involved in DNA repair and transcriptional regulation and is now recognized as a key regulator of cell survival and cell death as well as a master component of a number of transcription factors involved in tumor development and inflammation. PARP-1 is essential to the repair of DNA single-strand breaks via the base excision repair pathway. Inhibitors of PARP-1 have been shown to enhance the cytotoxic effects of ionizing radiation and DNA-damaging chemotherapy agents, such as the methylating agents and topoisomerase I inhibitors. There are currently at least five PARP inhibitors in clinical trial development. Recent in vitro and in vivo evidence suggests that PARP inhibitors could be used not only as chemo/radiotherapy sensitizers, but also as single agents to selectively kill cancers defective in DNA repair, specifically cancers with mutations in the breast cancer-associated genes (BRCA1 and BRCA2). PARP becomes activated in response to oxidative DNA damage and depletes cellular energy pools, thus leading to cellular dysfunction in various tissues. The activation of PARP may also induce various cell death processes and promotes an inflammatory response associated with multiple organ failure. Inhibition of PARP activity is protective in a wide range of inflammatory and ischemia-reperfusion-associated diseases, including cardiovascular diseases, diabetes, rheumatoid arthritis, endotoxic shock, and stroke. The aim of this review is to overview the emerging data in the literature showing the role of PARP in the pathogenesis of cancer and inflammatory diseases and unravel the solid body of literature that supports the view that PARP is an important target for therapeutic intervention in critical illness.
Collapse
Affiliation(s)
- Andreína Peralta-Leal
- Instituto de Parasitología y Biomedicina López Neyra, Consejo Superior de Investigaciones Cientificas (CSIC), Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Besson VC. Drug targets for traumatic brain injury from poly(ADP-ribose)polymerase pathway modulation. Br J Pharmacol 2009; 157:695-704. [PMID: 19371326 DOI: 10.1111/j.1476-5381.2009.00229.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The deleterious pathophysiological cascade induced after traumatic brain injury (TBI) is initiated by an excitotoxic process triggered by excessive glutamate release. Activation of the glutamatergic N-methyl-D-aspartate receptor, by increasing calcium influx, activates nitric oxide (NO) synthases leading to a toxic production of NO. Moreover, after TBI, free radicals are highly produced and participate to a deleterious oxidative stress. Evidence has showed that the major toxic effect of NO comes from its combination with superoxide anion leading to peroxynitrite formation, a highly reactive and oxidant compound. Indeed, peroxynitrite mediates nitrosative stress and is a potent inducer of cell death through its reaction with lipids, proteins and DNA. Particularly DNA damage, caused by both oxidative and nitrosative stresses, results in activation of poly(ADP-ribose) polymerase (PARP), a nuclear enzyme implicated in DNA repair. In response to excessive DNA damage, massive PARP activation leads to energetic depletion and finally to cell death. Since 10 years, accumulating data have showed that inactivation of PARP, either pharmacologically or using PARP null mice, induces neuroprotection in experimental models of TBI. Thus TBI generating NO, oxidative and nitrosative stresses promotes PARP activation contributing in post-traumatic motor, cognitive and histological sequelae. The mechanisms by which PARP inhibitors provide protection might not entirely be related to the preservation of cellular energy stores, but might also include other PARP-mediated mechanisms that needed to be explored in a TBI context. Ten years of experimental research provided rational basis for the development of PARP inhibitors as treatment for TBI.
Collapse
Affiliation(s)
- Valerie C Besson
- Equipe de Recherche 'Pharmacologie de la Circulation Cérébrale' (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France.
| |
Collapse
|
30
|
Calabrese EJ. Drug therapies for stroke and traumatic brain injury often display U-shaped dose responses: occurrence, mechanisms, and clinical implications. Crit Rev Toxicol 2008; 38:557-77. [PMID: 18615310 DOI: 10.1080/10408440802014287] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This article explores the occurrence of U-shaped dose responses induced by neuroprotective agents in animal stroke and traumatic brain injury (TBI) screening/preclinical studies. The assessment was stimulated by suggestions that U-shaped dose responses may be common for neuroprotective agents in stroke and TBI models, and its lack of both recognition and understanding may be a factor contributing to the failure of many promising drugs to be protective in clinical trials. Over 30 agents with neuroprotective properties in animal stroke/TBI models were identified that act via U-shaped dose responses in a broad range of experimental protocols. These findings suggest that U-shaped dose responses in animal stroke/TBI models may be a general occurrence and have significant implications for drug discovery, drug development, and clinical practice.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Public Health, School of Public Health and Health Sciences, Environmental Health Sciences Division, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
31
|
Beneke S. Poly(ADP-ribose) polymerase activity in different pathologies--the link to inflammation and infarction. Exp Gerontol 2008; 43:605-614. [PMID: 18511226 DOI: 10.1016/j.exger.2008.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 03/25/2008] [Accepted: 04/14/2008] [Indexed: 12/27/2022]
Abstract
DNA repair and aging are two phenomena closely connected to each other. The poly(ADP-ribosyl)ation reaction has been implicated in both of them. Poly(ADP-ribose) was originally discovered as an enzymatic reaction product after DNA damage. Soon it became evident that it is necessary for regulation of different repair pathways. Also, evidence accumulated that poly(ADP-ribose) formation capacity is at least correlated with the life span of mammalian species. As a NAD(+)-consuming process, poly(ADP-ribosyl)ation can lead to cell death by energy depletion. This finding opened the area for investigation of poly(ADP-ribose) polymerase activity and polymer formation in pathologies. This review provides an introduction into the wide and complex field of poly(ADP-ribosyl)ation in different pathologies with regards of cell death regulation, inflammation and resulting tissue damage.
Collapse
Affiliation(s)
- Sascha Beneke
- University of Konstanz, Molecular Toxicology Group, Universiteatsstr. 10, Box X911, 78457 Konstanz, Germany
| |
Collapse
|
32
|
Chatterjee PK. Novel pharmacological approaches to the treatment of renal ischemia-reperfusion injury: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2007; 376:1-43. [PMID: 18038125 DOI: 10.1007/s00210-007-0183-5] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 08/01/2007] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion (I-R) contributes to the development of ischemic acute renal failure (ARF). Multi-factorial processes are involved in the development and progression of renal I-R injury with the generation of reactive oxygen species, nitric oxide and peroxynitrite, and the decline of antioxidant protection playing major roles, leading to dysfunction, injury, and death of the cells of the kidney. Renal inflammation, involving cytokine/adhesion molecule cascades with recruitment, activation, and diapedesis of circulating leukocytes is also implicated. Clinically, renal I-R occurs in a variety of medical and surgical settings and is responsible for the development of acute tubular necrosis (a characteristic feature of ischemic ARF), e.g., in renal transplantation where I-R of the kidney directly influences graft and patient survival. The cellular mechanisms involved in the development of renal I-R injury have been targeted by several pharmacological interventions. However, although showing promise in experimental models of renal I-R injury and ischemic ARF, they have not proved successful in the clinical setting (e.g., atrial natriuretic peptide, low-dose dopamine). This review highlights recent pharmacological developments, which have shown particular promise against experimental renal I-R injury and ischemic ARF, including novel antioxidants and antioxidant enzyme mimetics, nitric oxide and nitric oxide synthase inhibitors, erythropoietin, peroxisome-proliferator-activated receptor agonists, inhibitors of poly(ADP-ribose) polymerase, carbon monoxide-releasing molecules, statins, and adenosine. Novel approaches such as recent research involving combination therapies and the potential of non-pharmacological strategies are also considered.
Collapse
Affiliation(s)
- Prabal K Chatterjee
- Division of Pharmacology and Therapeutics, School of Pharmacy and Biomolecular Sciences, University of Brighton, Cockcroft Building, Lewes Road, Moulsecoomb, Brighton BN2 4GJ, UK.
| |
Collapse
|
33
|
Park SY, Lee JH, Kim CD, Rhim BY, Hong KW, Lee WS. Beneficial synergistic effects of concurrent treatment with cilostazol and probucol against focal cerebral ischemic injury in rats. Brain Res 2007; 1157:112-20. [PMID: 17521620 DOI: 10.1016/j.brainres.2007.04.051] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/12/2007] [Accepted: 04/13/2007] [Indexed: 11/28/2022]
Abstract
In the present study, we assessed the beneficial synergistic effects of concurrent treatment with low doses of cilostazol and probucol against focal cerebral ischemic infarct in rats. The ischemic infarct induced by 2-h occlusion of middle cerebral artery (MCA) and 22-h reperfusion was significantly reduced in rat brain that received cilostazol (20 mg/kg) and probucol (30 mg/kg) twice together with prominent improvement of neurological function compared to the effect of cilostazol or probucol monotherapy. Increased myeloperoxidase activity, a marker of neutrophil infiltration, observed in the penumbral zone of vehicle-treated brain was more significantly reduced by cilostazol plus probucol in combination. Increased superoxide-, nitrotyrosine (a marker of peroxynitrite)-, poly(ADP-ribose) [a marker for poly(ADP-ribose) polymerase activity]-, and cleaved caspase-3-positive cells (a proapoptotic marker) in the vehicle sample were significantly attenuated by the combination therapy, while individual treatment with low dose of cilostazol or probucol showed a marginal effect. Taken together, it is suggested that the neuroprotective potentials of combination therapy with low doses of cilostazol plus probucol may provide beneficial therapeutic intervention in reducing the focal cerebral ischemic infarct in rats.
Collapse
Affiliation(s)
- So Youn Park
- Department of Pharmacology, College of Medicine, Pusan National University, Busan 602-739, Republic of Korea
| | | | | | | | | | | |
Collapse
|
34
|
Lee JH, Park SY, Shin HK, Kim CD, Lee WS, Hong KW. Poly(ADP-ribose) polymerase inhibition by cilostazol is implicated in the neuroprotective effect against focal cerebral ischemic infarct in rat. Brain Res 2007; 1152:182-90. [PMID: 17433265 DOI: 10.1016/j.brainres.2007.03.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/08/2007] [Accepted: 03/09/2007] [Indexed: 11/28/2022]
Abstract
This study shows that cilostazol displayed a potent inhibition of PARP with IC(50) of 883+/-41 nM in the enzyme assay, and also significantly reversed H(2)O(2)-evoked elevated PARP activity and reduced NAD(+) levels in the PC12 cells with improvement of cell viability. In in vivo study, inhibition of PARP activity by cilostazol prevented cerebral ischemic injury induced by 2-h middle cerebral artery occlusion (MCAO) and 24-h reperfusion. The ischemic infarct was significantly reduced in the rats that received cilostazol (30 mg/kg, twice orally) with improvement of neurological function. Moreover, cilostazol treatment significantly decreased the number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL)- and poly(ADP-ribose)-positive cells associated with apoptosis-inducing factor (AIF) translocation to the nucleus in the penumbral region. Further, cilostazol significantly reduced myeloperoxidase activity, a marker of neutrophil infiltration. In line with these findings, the OX-42- (a marker of microglia) and TNF-alpha-positive cells (a marker of proapoptotic protein) were markedly increased in the vehicle samples, both of which were significantly attenuated by treatment with cilostazol. Taken together, these results suggest that neuroprotective potentials of cilostazol against focal cerebral ischemic injury are, at least in part, ascribed to its anti-inflammatory effects and PARP inhibitory activity.
Collapse
Affiliation(s)
- Jeong Hyun Lee
- Department of Pharmacology, College of Medicine, Pusan National University, Busan 602-739, Korea
| | | | | | | | | | | |
Collapse
|
35
|
Fatokun AA, Stone TW, Smith RA. Cell death in rat cerebellar granule neurons induced by hydrogen peroxide in vitro: mechanisms and protection by adenosine receptor ligands. Brain Res 2006; 1132:193-202. [PMID: 17188658 DOI: 10.1016/j.brainres.2006.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 11/02/2006] [Accepted: 11/03/2006] [Indexed: 11/23/2022]
Abstract
Oxidative stress, resulting from excessive production of reactive oxygen species (ROS), is a pathological state that causes profound cellular damage and eventual death resulting from the overactivation of glutamate receptors, and the generation of nitric oxide, superoxide and hydrogen peroxide (H(2)O(2)). As such, H(2)O(2) represents an important model for studying the neuropathology of oxidative stress in a variety of CNS disorders. The effects of H(2)O(2) on the viability of post-natal cerebellar granule neurons (CGNs), the nature of the cell death involved and the potential protection by adenosine receptors against the damage were examined in the current study. Hydrogen peroxide (10-400 microM) reduced CGN viability in a concentration- and time-dependent manner. The addition of catalase (100 U/ml) prevented this effect, and the non-specific COX inhibitor aspirin (1 mM) also alleviated the damage. A combination of H(2)O(2) (5 microM) and Cu(2+) (0.5 mM) resulted in a significant damage that was not prevented by the hydroxyl radical scavenger mannitol (50 mM). The permeability transition pore blocker cyclosporin A, the caspase-3 inhibitor Z-DEVD-fmk (40 microM) and the PARP-1 inhibitor DPQ (10 microM) each significantly protected against peroxide damage. While the A(1) adenosine receptor agonist CPA and the A(2A) receptor antagonist ZM241385 (each at 100 nM) elicited protection, the A(1) adenosine receptor blocker DPCPX and the A(2A) receptor agonist CGS21680 (each at 100 nM) showed no effect. The data demonstrate that H(2)O(2) induced oxidative stress in CGNs, involving both apoptotic and necrotic death, and this can be ameliorated by A(1) receptor activation or A(2A) receptor blockade.
Collapse
Affiliation(s)
- Amos A Fatokun
- Division of Neuroscience and Biomedical Systems, Institute of Biomedical and Life Sciences, West Medical Building, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | | | | |
Collapse
|
36
|
Goebel DJ, Winkler BS. Blockade of PARP activity attenuates poly(ADP-ribosyl)ation but offers only partial neuroprotection against NMDA-induced cell death in the rat retina. J Neurochem 2006; 98:1732-45. [PMID: 16903875 PMCID: PMC1766941 DOI: 10.1111/j.1471-4159.2006.04065.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent reports have linked neuronal cell death by necrosis to poly(ADP-ribose) polymerase-1 (PARP-1) hyperactivation. It is believed that under stress, the activity of this enzyme is up-regulated, resulting in extensive poly(ADP-ribosyl)ation of nuclear proteins, using NAD(+) as its substrate, which, in turn, leads to the depletion of NAD(+). In efforts to restore the level of NAD(+), depletion of ATP occurs, resulting in the shutdown of ATP-dependent ionic pumps. This results in cell swelling and eventual loss of membrane selectivity, hallmarks of necrosis. Reports from in vitro and in vivo studies in the brain have shown that NMDA receptor activation stimulates PARP activity and that blockade of the enzyme provides substantial neuroprotection. The present study was undertaken to determine whether PARP activity is regulated by NMDA in the rat retina, and whether blockade of PARP activity provides protection against toxic effects of NMDA. Rat retinas exposed to intravitreal injections containing NMDA, with or without the PARP inhibitor N-(6-oxo-5, 6-dihydrophenanthridin-2-yl)-(N,-dimethylamino) acetamide hydrochloride (PJ-34), were assessed for changes in PARP-1 activity as evidenced by poly(ADP-ribosyl)ation (PAR), loss of membrane integrity, morphological indicators of apoptosis and necrosis, and ganglion cell loss. Results showed that: NMDA increased PAR formation in a concentration-dependent manner and caused a decline in retinal ATP levels; PJ-34 blockade attenuated the NMDA-induced formation of PAR and decline in ATP; NMDA induced the loss of membrane selectivity to ethidium bromide (EtBr) in inner retinal neurons, but loss of membrane selectivity was not prevented by blocking PARP activity; cells stained with EtBr, or reacted for TUNEL-labeling, displayed features characteristic of both apoptosis and necrosis. In the presence of PJ-34, greater numbers of cells exhibited apoptotic features; PJ-34 provided partial neuroprotection against NMDA-induced ganglion cell loss. These findings suggest that although blockade of PARP activity fully attenuates NMDA-induced PAR formation and loss of retinal ATP content, and improves the survival of select populations of ganglion cells, this approach does not provide full neuroprotection. In contrast, blockade of PARP activity promotes apoptotic-like cell death in the majority of cells undergoing cell death. Furthermore, these studies show that the loss of membrane selectivity is not dependent upon PAR formation or the resulting decline of ATP, and suggests that an alternative pathway, other than PARP activation, exists to mediate this event.
Collapse
Affiliation(s)
- Dennis J. Goebel
- Department of Anatomy & Cell Biology, Wayne State University, Detroit, Michigan, USA
- Address correspondence and reprint requests to Dennis J. Goebel, Associate Professor, Department of Anatomy & Cell Biology, Wayne State University, School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA. E-mail:
| | - Barry S. Winkler
- Department of Anatomy & Cell Biology, Wayne State University, Detroit, Michigan, USA
- Eye Research Institute, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
37
|
Abstract
Perinatal hypoxic-ischaemic injury (HII) is a significant cause of neurodevelopmental impairment and disability. Studies employing 31P magnetic resonance spectroscopy to measure phosphorus metabolites in situ in the brains of newborn infants and animals have demonstrated that transient hypoxia-ischaemia leads to a delayed disruption in cerebral energy metabolism, the magnitude of which correlates with the subsequent neurodevelopmental impairment. Prominent among the biochemical features of HII is the loss of cellular ATP, resulting in increased intracellular Na+ and Ca2+, and decreased intracellular K+. These ionic imbalances, together with a breakdown in cellular defence systems following HII, can contribute to oxidative stress with a net increase in reactive oxygen species. Subsequent damage to lipids, proteins, and DNA and inactivation of key cellular enzymes leads ultimately to cell death. Although the precise mechanisms of neuronal loss are unclear, it is now clear both apoptosis and necrosis are the significant components of cell death following HII. A number of different factors influence whether a cell will undergo apoptosis or necrosis, including the stage of development, cell type, severity of mitochondrial injury and the availability of ATP for apoptotic execution. This review will focus on some pathological mechanisms of cell death in which there is a disruption to oxidative metabolism. The first sections will discuss the process of damage to oxidative metabolism, covering the data collected both from human infants and from animal models. Following sections will deal with the molecular mechanisms that may underlie cerebral energy failure and cell death in this form of brain injury, with a particular emphasis on the role of apoptosis and mitochondria.
Collapse
Affiliation(s)
- Deanna L. Taylor
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - A. David Edwards
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Huseyin Mehmet
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| |
Collapse
|
38
|
Pellicciari R, Camaioni E, Costantino G. 3. Life or death decisions: the cast of poly(ADP-ribose)polymerase (PARP) as a therapeutic target for brain ischaemia. PROGRESS IN MEDICINAL CHEMISTRY 2005; 42:125-69. [PMID: 15003720 DOI: 10.1016/s0079-6468(04)42003-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Affiliation(s)
- Roberto Pellicciari
- Dipartimento di Chimica e Tecnologia del Farmaco, Via del Liceo 1, 06123 Perugia, Italy
| | | | | |
Collapse
|
39
|
Abstract
Over the past decade, poly(ADP-ribosyl)ation has emerged as a crucial event in the pathogenesis of ischemic stroke. A large body of evidence unambiguously demonstrates that activity of poly(ADP-ribose) polymerase-1 (PARP-1) significantly increases during brain ischemia, and that inhibition of this enzymatic activity affords substantial neuroprotection from ischemic brain injury. This review strictly focuses on literature on poly(ADP-ribosyl)ation and ischemic stroke, highlighting the pathogenetic role of poly(ADP-ribose) in ischemic neuronal death, and the therapeutic relevance of drugs modulating its metabolism to pharmacological treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Alberto Chiarugi
- Department of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
40
|
Devalaraja-Narashimha K, Singaravelu K, Padanilam BJ. Poly(ADP-ribose) polymerase-mediated cell injury in acute renal failure. Pharmacol Res 2005; 52:44-59. [PMID: 15911333 DOI: 10.1016/j.phrs.2005.02.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Accepted: 02/01/2005] [Indexed: 01/12/2023]
Abstract
Acute Renal Failure (ARF) is the most costly kidney disease in hospitalized patients and remains as a serious problem in clinical medicine. The mortality rate among ARF patients remains around 50% and no pharmaceutical agents are currently available to improve its clinical outcome. Although several successful therapeutic approaches have been developed in animal models of the disease, translation of the results to clinical ARF remains elusive. Understanding the cellular and molecular mechanisms of vascular and tubular dysfunction in ARF is important for developing acceptable therapeutic interventions. Following an ischemic episode, cells of the affected nephron undergo necrotic and/or apoptotic cell death. Necrotic cell death is widely considered to be a futile process that cannot be modulated by pharmacological means as opposed to apoptosis. However, recent reports from various laboratories including ours indicate that inhibition or absence of poly(ADP)-ribose polymerase (PARP), one of the molecules involved in cell death, provides remarkable protection in disease models such as stroke, myocardial infarction and renal ischemia which are characterized predominantly by necrotic type of cell death. Overactivation of PARP in conditions such as ischemic renal injury leads to cellular depletion of its substrate NAD+ and consequently ATP. The severely compromised cellular energetic state induces acute cell injury and diminishes renal functions. PARP activation also enhances the expression of proinflammatory agents and adhesion molecules in ischemic kidneys. Pharmacological inhibition and gene ablation of PARP-1 decreased energy depletion, inflammatory response and improved renal functions in the setting renal ischemia/reperfusion injury. The biochemical pathways and the cellular and molecular mechanisms mediated by PARP-1 activation in eliciting the energy depletion and inflammatory responses in ischemic kidney are not fully elucidated. Dissecting the molecular mechanisms by which PARP activation contributes to oxidant-induced cell death will provide new strategies to interfere in those pathways to modulate cell death in renal ischemia. The current review evaluates the experimental evidences in animal and cell culture models implicating PARP as a pathophysiological modulator of acute renal failure with particular emphasis on ischemic renal injury.
Collapse
|
41
|
Cosi C. New inhibitors of poly(ADP-ribose) polymerase and their potential therapeutic targets. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.12.7.1047] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
42
|
Zhang J. PARP inhibition: a novel approach to treat ischaemia/reperfusion and inflammation-related injuries. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728214.4.1.209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Patel NSA, Cortes U, Di Poala R, Mazzon E, Mota-Filipe H, Cuzzocrea S, Wang ZQ, Thiemermann C. Mice Lacking the 110-kD Isoform of Poly(ADP-Ribose) Glycohydrolase Are Protected against Renal Ischemia/Reperfusion Injury. J Am Soc Nephrol 2005; 16:712-9. [PMID: 15677308 DOI: 10.1681/asn.2004080677] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The role of poly(ADP-ribose) (PAR) glycohydrolase (PARG) in the pathophysiology of renal ischemia/reperfusion (I/R) injury is not known. Poly(ADP-ribosyl)ation is rapidly stimulated in cells after DNA damage caused by the generation of reactive oxygen and nitrogen species during I/R. Continuous or excessive activation of poly(ADP-ribose) polymerase-1 produces extended chains of ADP-ribose on nuclear proteins and results in a substantial depletion of intracellular NAD(+) and subsequently, ATP, leading to cellular dysfunction and, ultimately, cell death. The key enzyme involved in polymer turnover is PARG, which possesses mainly exoglycosidase activity but can remove olig(ADP-ribose) fragments via endoglycosidic cleavage. Thus, the aim of this study was to investigate whether the absence of PARG(110) reduced the renal dysfunction, injury, and inflammation caused by I/R of the mouse kidney. Here, the renal dysfunction and injury caused by I/R (bilateral renal artery occlusion [30 min] followed by reperfusion [24 h]) in mice lacking PARG(110), the major nuclear isoform of PARG, was investigated. The following markers of renal dysfunction and injury were measured: Plasma urea, creatinine, aspartate aminotransferase, and histology. The following markers of inflammation were also measured: Myeloperoxidase activity, malondialdehyde levels, and plasma nitrite/nitrate. The degree of renal injury and dysfunction caused by I/R was significantly reduced in PARG(110)-deficient mice when compared with their wild-type littermates, and there were no differences in any of the biochemical parameters measured between sham-operated PARG(110)(-/-) mice and sham-operated wild-type littermates. Thus, it is proposed that endogenous PARG(110) plays a pivotal role in the pathophysiology of I/R injury of the kidney.
Collapse
Affiliation(s)
- Nimesh S A Patel
- Centre for Experimental Medicine, Nephrology & Critical Care, William Harvey Research Institute, Queen Mary, University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Koh SH, Park Y, Song CW, Kim JG, Kim K, Kim J, Kim MH, Lee SR, Kim DW, Yu HJ, Chang DI, Hwang SJ, Kim SH. The effect of PARP inhibitor on ischaemic cell death, its related inflammation and survival signals. Eur J Neurosci 2004; 20:1461-72. [PMID: 15355313 DOI: 10.1111/j.1460-9568.2004.03632.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) plays an important role in ischaemic cell death, and 3-aminobenzamide (3-AB), one of the PARP inhibitors, has a protective effect on ischaemic stroke. We investigated the neuroprotective mechanisms of 3-AB in ischaemic stroke. The occlusion of middle cerebral artery (MCA) was made in 170 Sprague-Dawley rats, and reperfusion was performed 2 h after the occlusion. Another 10 Sprague-Dawley rats were used for sham operation. 3-AB was administered to 85 rats 10 min before the occlusion [3-AB group (n = 85) vs. control group without 3-AB (n = 85)]. Infarct volume and water content were measured, brain magnetic resonance imaging, terminal deoxynucleotidyltransferase (TdT)-mediated dUTP-biotin nick end-labelling (TUNEL) and Cresyl violet staining were performed, and immunoreactivities (IRs) of poly(ADP-ribose) polymer (PAR), cleaved caspase-3, CD11b, intercellular adhesion molecule-1 (ICAM-1), cyclooxygenase-2 (COX-2), phospho-Akt (pAkt) and phospho-glycogen synthase kinase-3 (pGSK-3) were compared in the peri-infarcted region of the 3-AB group and its corresponding ischaemic region of the control group at 2, 8, 24 and 72 h after the occlusion. In the 3-AB group, the infarct volume and the water content were decreased (about 45% and 3.6%, respectively, at 24 h), the number of TUNEL-positive cells was decreased (about 36% at 24 h), and the IRs of PAR, cleaved caspase-3, CD11b, ICAM-1 and COX-2 were significantly reduced, while the IRs of pAkt and pGSK-3 were increased. These results suggest that 3-AB treatment could reduce the infarct volume by reducing ischaemic cell death, its related inflammation and increasing survival signals. The inhibition of PARP could be another potential neuroprotective strategy in ischaemic stroke.
Collapse
Affiliation(s)
- Seong-Ho Koh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Park EM, Cho S, Frys K, Racchumi G, Zhou P, Anrather J, Iadecola C. Interaction between inducible nitric oxide synthase and poly(ADP-ribose) polymerase in focal ischemic brain injury. Stroke 2004; 35:2896-901. [PMID: 15514191 DOI: 10.1161/01.str.0000147042.53659.6c] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND PURPOSE Overactivation of the DNA repair enzyme poly(ADP-ribose) polymerase (PARP) contributes to ischemic brain injury. Because PARP upregulates proinflammatory genes, we investigated whether inducible nitric oxide synthase (iNOS), a gene involved in the deleterious effects of postischemic inflammation, participates in the mechanisms by which PARP activation contributes to cerebral ischemic injury. METHODS The middle cerebral artery (MCA) was occluded in mice for 20 minutes using an intravascular filament, and injury volume was measured 72 hours later in Nissl-stained brain sections. mRNA expression was assessed in the postischemic brain by the quantitative "real-time" polymerase chain reaction. RESULTS The PARP inhibitor PJ34 reduced infarct volume and attenuated postischemic iNOS mRNA upregulation by 72%. To determine whether iNOS contributes to the toxicity of PARP, the iNOS inhibitor aminoguanidine was co-administered with PARP inhibitors. Unexpectedly, co-administration of PARP and iNOS inhibitors, or treatment of iNOS-null mice with PARP inhibitors, abrogated the protective effect afforded by iNOS or PARP inhibition alone. The loss of neuroprotection was associated with upregulation of the inflammatory genes iNOS, intercellular adhesion molecule-1, and gp91(phox). CONCLUSIONS The results suggest that iNOS expression contributes to the deleterious effects exerted by PARP activation in cerebral ischemia. However, iNOS activity is required for the protective effect of PARP inhibition and, conversely, PARP activity must be present for iNOS inhibition to be effective. The findings unveil a previously unrecognized deleterious interaction between iNOS and PARP that is relevant to the development of combination therapies for ischemic stroke.
Collapse
Affiliation(s)
- Eun-Mi Park
- Department of Pharmacology, College of Medicine, Ewha Women's University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
46
|
Zheng J, Devalaraja-Narashimha K, Singaravelu K, Padanilam BJ. Poly(ADP-ribose) polymerase-1 gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 2004; 288:F387-98. [PMID: 15494543 DOI: 10.1152/ajprenal.00436.2003] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased generation of reactive oxygen species (ROS) and the subsequent DNA damage and excessive activation of poly(ADP-ribose) polymerase-1 (PARP-1) have been implicated in the pathogenesis of ischemic injury. We previously demonstrated that pharmacological inhibition of PARP protects against ischemic renal injury (IRI) in rats (Martin DR, Lewington AJ, Hammerman MR, and Padanilam BJ. Am J Physiol Regul Integr Comp Physiol 279: R1834-R1840, 2000). To further define the role of PARP-1 in IRI, we tested whether genetic ablation of PARP-1 attenuates tissue injury after renal ischemia. Twenty-four hours after reperfusion following 37 min of bilateral renal pedicle occlusion, the effects of the injury on renal functions in PARP-/- and PARP+/+ mice were assessed by determining glomerular filtration rate (GFR) and the plasma levels of creatinine. The levels of plasma creatinine were decreased and GFR was augmented in PARP-/- mice. Morphological evaluation of the kidney tissues showed that the extent of damage due to the injury in PARP-/- mice was less compared with their wild-type counterparts. The levels of ROS and DNA damage were comparable in the injured kidneys of PARP+/+ and PARP-/- mice. PARP activity was induced in ischemic kidneys of PARP+/+ mice at 6-24 h postinjury. At 6, 12, and 24 h after injury, ATP levels in the PARP+/+ mice kidney declined to 28, 26, and 43%, respectively, whereas it was preserved close to normal levels in PARP-/- mice. The inflammatory cascade was attenuated in PARP-/- mice as evidenced by decreased neutrophil infiltration and attenuated expression of inflammatory molecules such as TNF-alpha, IL-1beta, and intercellular adhesion molecule-1. At 12 h postinjury, no apoptotic cell death was observed in PARP-/- mice kidneys. However, by 24 h postinjury, a comparable number of cells underwent apoptosis in both PARP-/- and PARP+/+ mice kidneys. Thus activation of PARP post-IRI contributes to cell death most likely by ATP depletion and augmentation of the inflammatory cascade in the mouse model. PARP ablation preserved ATP levels, renal functions, and attenuated inflammatory response in the setting of IRI in the mouse model. PARP inhibition may have clinical efficacy in preventing the progression of acute renal failure complications.
Collapse
Affiliation(s)
- Jianfeng Zheng
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | | | | | | |
Collapse
|
47
|
Nakajima H, Kakui N, Ohkuma K, Ishikawa M, Hasegawa T. A newly synthesized poly(ADP-ribose) polymerase inhibitor, DR2313 [2-methyl-3,5,7,8-tetrahydrothiopyrano[4,3-d]-pyrimidine-4-one]: pharmacological profiles, neuroprotective effects, and therapeutic time window in cerebral ischemia in rats. J Pharmacol Exp Ther 2004; 312:472-81. [PMID: 15466246 DOI: 10.1124/jpet.104.075465] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the pharmacological profiles of DR2313 [2-methyl-3,5,7,8-tetrahydrothiopyrano[4,3-d]pyrimidine-4-one], a newly synthesized poly(ADP-ribose) polymerase (PARP) inhibitor, and its neuroprotective effects on ischemic injuries in vitro and in vivo. DR2313 competitively inhibited poly(ADP-ribosyl)ation in nuclear extracts of rat brain in vitro (K(i) = 0.23 microM). Among several NAD(+)-utilizing enzymes, DR2313 was specific for PARP but not selective between PARP-1 and PARP-2. DR2313 also showed excellent profiles in water solubility and rat brain penetrability. In in vitro models of cerebral ischemia, exposure to hydrogen peroxide or glutamate induced cell death with overactivation of PARP, and treatment with DR2313 reduced excessive formation of poly(ADP-ribose) and cell death. In both permanent and transient focal ischemia models in rats, pretreatment with DR2313 (10 mg/kg i.v. bolus and 10 mg/kg/h i.v. infusion for 6 h) significantly reduced the cortical infarct volume. To determine the therapeutic time window of neuroprotection by DR2313, the effect of post-treatment was examined in transient focal ischemia model and compared with that of a free radical scavenger, MCI-186 (3-methyl-1-phenyl-2-pyrazolone-5-one). Pretreatment with MCI-186 (3 mg/kg i.v. bolus and 3 mg/kg/h i.v. infusion for 6 h) significantly reduced the infarct volume, whereas the post-treatment failed to show any effects. In contrast, post-treatment with DR2313 (same regimen) delaying for 2 h after ischemia still prevented the progression of infarction. These results indicate that DR2313 exerts neuroprotective effects via its potent PARP inhibition, even when the treatment is initiated after ischemia. Thus, a PARP inhibitor like DR2313 may be more useful in treating acute stroke than a free radical scavenger.
Collapse
Affiliation(s)
- Hidemitsu Nakajima
- Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd., 760 Moro-oka-cho, Kohoku-ku, Yokohama 222-8567, Japan.
| | | | | | | | | |
Collapse
|
48
|
Besson VC, Margaill I, Plotkine M, Marchand-Verrecchia C. Deleterious activation of poly(ADP-ribose)polymerase-1 in brain after in vivo oxidative stress. Free Radic Res 2004; 37:1201-8. [PMID: 14703732 DOI: 10.1080/10715760310001612568] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Oxidative stress has been shown to be implicated in the pathogenesis of central nervous system injuries such as cerebral ischemia and trauma, and chronic neurodegenerative diseases. In vitro studies show that oxidative stress, particularly peroxynitrite, could trigger DNA strand breaks, which lead to the activation of repairing enzymes including Poly(ADP-ribose) Polymerase-1 (PARP-1). As excessive activation of this enzyme induces cell death, we examined whether such a cascade also occurs in vivo in a model of oxidative stress in rat brain. For this purpose, the mitochondrial toxin malonate, which promotes free radical production, was infused into the left striatum of rats. Immunohistochemistry showed that 3-nitrotyrosine, an indicator of nitrosative stress, and poly(ADP-ribose), a marker of poly(ADP-ribose)polymerase-1 activation, were present as early as 1 h after malonate, and that they persisted for 24 h. The PARP inhibitor, 3-aminobenzamide, significantly reduced the lesion and inhibited PARP-1 activation induced by malonate. These results demonstrate that oxidative stress induced in vivo in the central nervous system leads to the activation of poly(ADP-ribose)polymerase-1, which contributes to neuronal cell death.
Collapse
Affiliation(s)
- Valérie C Besson
- Laboratoire de Pharmacologie de la Circulation Cérébrale, UPRES EA 2510, Université René Descartes, 4 avenue de l'Observatoire, F-75006 Paris, France
| | | | | | | |
Collapse
|
49
|
Fonfria E, Marshall ICB, Benham CD, Boyfield I, Brown JD, Hill K, Hughes JP, Skaper SD, McNulty S. TRPM2 channel opening in response to oxidative stress is dependent on activation of poly(ADP-ribose) polymerase. Br J Pharmacol 2004; 143:186-92. [PMID: 15302683 PMCID: PMC1575275 DOI: 10.1038/sj.bjp.0705914] [Citation(s) in RCA: 224] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. TRPM2 (melastatin-like transient receptor potential 2 channel) is a nonselective cation channel that is activated under conditions of oxidative stress leading to an increase in intracellular free Ca(2+) concentration ([Ca(2+)](i)) and cell death. We investigated the role of the DNA repair enzyme poly(ADP-ribose) polymerase (PARP) on hydrogen peroxide (H(2)O(2))-mediated TRPM2 activation using a tetracycline-inducible TRPM2-expressing cell line. 2. In whole-cell patch-clamp recordings, intracellular adenine 5'-diphosphoribose (ADP-ribose) triggered an inward current in tetracycline-induced TRPM2-human embryonic kidney (HEK293) cells, but not in uninduced cells. Similarly, H(2)O(2) stimulated an increase in [Ca(2+)](i) (pEC(50) 4.54+/-0.02) in Fluo-4-loaded TRPM2-expressing HEK293 cells, but not in uninduced cells. Induction of TRPM2 expression caused an increase in susceptibility to plasma membrane damage and mitochondrial dysfunction in response to H(2)O(2). These data demonstrate functional expression of TRPM2 following tetracycline induction in TRPM2-HEK293 cells. 3. PARP inhibitors SB750139-B (patent number DE10039610-A1 (Lubisch et al., 2001)), PJ34 (N-(6-oxo-5,6-dihydro-phenanthridin-2-yl)-N,N-dimethylacetamide) and DPQ (3, 4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone) inhibited H(2)O(2)-mediated increases in [Ca(2+)](i) (pIC(50) vs 100 microm H(2)O(2): 7.64+/-0.38; 6.68+/-0.28; 4.78+/-0.05, respectively), increases in mitochondrial dysfunction (pIC(50) vs 300 microm H(2)O(2): 7.32+/-0.23; 6.69+/-0.22; 5.44+/-0.09, respectively) and decreases in plasma membrane integrity (pIC(50) vs 300 microm H(2)O(2): 7.45+/-0.27; 6.35+/-0.18; 5.29+/-0.12, respectively). The order of potency of the PARP inhibitors in these assays (SB750139>PJ34>DPQ) was the same as for inhibition of isolated PARP enzyme. 4. SB750139-B, PJ34 and DPQ had no effect on inward currents elicited by intracellular ADP-ribose in tetracycline-induced TRPM2-HEK293 cells, suggesting that PARP inhibitors are not interacting directly with the channel. 5. SB750139-B, PJ34 and DPQ inhibited increases in [Ca(2+)](i) in a rat insulinoma cell line (CRI-G1 cells) endogenously expressing TRPM2 (pIC(50) vs 100 microm H(2)O(2): 7.64+/-0.38; 6.68+/-0.28; 4.78+/-0.05, respectively). 6. These data suggest that oxidative stress causes TRPM2 channel opening in both recombinant and endogenously expressing cell systems via activation of PARP enzymes.
Collapse
Affiliation(s)
- Elena Fonfria
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
- Author for correspondence:
| | - Ian C B Marshall
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Christopher D Benham
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Izzy Boyfield
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Jason D Brown
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Kerstin Hill
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Jane P Hughes
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Stephen D Skaper
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| | - Shaun McNulty
- Neurology and GI Centre of Excellence for Drug Discovery, GlaxoSmithKline Research and Development Limited, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW
| |
Collapse
|
50
|
Shen CC, Huang HM, Ou HC, Chen HL, Chen WC, Jeng KC. Protective effect of nicotinamide on neuronal cells under oxygen and glucose deprivation and hypoxia/reoxygenation. J Biomed Sci 2004; 11:472-81. [PMID: 15153782 DOI: 10.1007/bf02256096] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2003] [Accepted: 02/03/2004] [Indexed: 01/13/2023] Open
Abstract
Nicotinamide (vitamin B(3)) reduces the infarct volume following focal cerebral ischemia in rats; however, its mechanism of action has not been reported. After cerebral ischemia and/or reperfusion, reactive oxygen species (ROS) and reactive nitrogen species may be generated by inflammatory cells through several cellular pathways, which can lead to intracellular calcium influx and cell damage. Therefore, we investigated the mechanisms of action of nicotinamide in neuroprotection under conditions of hypoxia/reoxygenation. Results showed that nicotinamide significantly protected rat primary cortical cells from hypoxia by reducing lactate dehydrogenase release with 1 h of oxygen-glucose deprivation (OGD) stress. ROS production and calcium influx in neuronal cells during OGD were dose-dependently diminished by up to 10 mM nicotinamide (p < 0.01). This effect was further examined with OGD/reoxygenation (H/R). Cells were stained with the fluorescent dye 4,6-diamidino-2-phenylindole (DAPI) or antibodies against anti-microtubule-associated protein-2 and cleaved caspase-3. Apoptotic cells were studied using Western blotting of cytochrome c and cleaved caspase-3. Results showed that vitamin B(3) reduced cell injury, caspase-3 cleavage and nuclear condensation (DAPI staining) in neuronal cells under H/R. In addition, nicotinamide diminished c-fos and zif268 immediate-early gene expressions following OGD. Taken together, these results indicate that the neuroprotective effect of nicotinamide might occur through these mechanisms in this in vitro ischemia/reperfusion model.
Collapse
Affiliation(s)
- Chiung-Chyi Shen
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|