1
|
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: 64] [Impact Index Per Article: 21.3] [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
|
2
|
Tuo QZ, Zhang ST, Lei P. Mechanisms of neuronal cell death in ischemic stroke and their therapeutic implications. Med Res Rev 2021; 42:259-305. [PMID: 33957000 DOI: 10.1002/med.21817] [Citation(s) in RCA: 348] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 03/31/2021] [Accepted: 04/23/2021] [Indexed: 02/05/2023]
Abstract
Ischemic stroke caused by arterial occlusion is the most common type of stroke, which is among the most frequent causes of disability and death worldwide. Current treatment approaches involve achieving rapid reperfusion either pharmacologically or surgically, both of which are time-sensitive; moreover, blood flow recanalization often causes ischemia/reperfusion injury. However, even though neuroprotective intervention is urgently needed in the event of stroke, the exact mechanisms of neuronal death during ischemic stroke are still unclear, and consequently, the capacity for drug development has remained limited. Multiple cell death pathways are implicated in the pathogenesis of ischemic stroke. Here, we have reviewed these potential neuronal death pathways, including intrinsic and extrinsic apoptosis, necroptosis, autophagy, ferroptosis, parthanatos, phagoptosis, and pyroptosis. We have also reviewed the latest results of pharmacological studies on ischemic stroke and summarized emerging drug targets with a focus on clinical trials. These observations may help to further understand the pathological events in ischemic stroke and bridge the gap between basic and translational research to reveal novel neuroprotective interventions.
Collapse
Affiliation(s)
- Qing-Zhang Tuo
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shu-Ting Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
3
|
Thapa K, Khan H, Sharma U, Grewal AK, Singh TG. Poly (ADP-ribose) polymerase-1 as a promising drug target for neurodegenerative diseases. Life Sci 2020; 267:118975. [PMID: 33387580 DOI: 10.1016/j.lfs.2020.118975] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/07/2020] [Accepted: 12/19/2020] [Indexed: 02/07/2023]
Abstract
AIMS Poly (ADP-ribose) polymerase- (PARP)-1 is predominantly triggered by DNA damage. Overexpression of PARP-1 is known for its association with the pathogenesis of several CNS disorders, such as Stroke, Parkinson's disease (PD), Alzheimer's disease (AD), Huntington (HD) and Amyotrophic lateral sclerosis (ALS). NAD+ depletion resulted PARP related cell death only happened when the trial used extreme high oxidization treatment. Inhibition of PARP1/2 may induce replication related cell death due to un-repaired DNA damage. This review has discussed PARP-1 modulated downstream pathways in neurodegeneration and various FDA approved PARP-1 inhibitors. MATERIALS AND METHODS A systematic literature review of PubMed, Medline, Bentham, Scopus and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on mechanistic role of Poly (ADP-ribose) polymerase and its inhibition in Neurodegenerative diseases. KEY FINDINGS Several researchers have put forward number of potential treatments, of which PARP-1 enzyme has been regarded as a potent target intended for the handling of neurodegenerative ailments. Targeting PARP using its chemical inhibitors in various neurodegenerative may have therapeutic outcomes by reducing neuronal death mediated by PARPi. Numerous PARP-1 inhibitors have been studied in neurodegenerative diseases but they haven't been clinically evaluated. SIGNIFICANCE In this review, the pathological role of PARP-1 in various neurodegenerative diseases has been discussed along with the therapeutic role of PARP-1 inhibitors in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Uma Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | | |
Collapse
|
4
|
Sriwastva MK, Kunjunni R, Andrabi M, Prasad K, Saxena R, Subbiah V. Neuroprotective Effects of Activated Protein C Involve the PARP/AIF Pathway against Oxygen-Glucose Deprivation in SH-SY5Y Cells. Brain Sci 2020; 10:brainsci10120959. [PMID: 33321687 PMCID: PMC7764138 DOI: 10.3390/brainsci10120959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Protein C, a member of the zymogen family of serine proteases in plasma, is one of the several vitamin K dependent glycoproteins known to induce anti-apoptotic activity. However, the target molecule involved in the mechanism needs to be investigated. We sought to investigate the pathways involved in the anti-apoptotic role of activated protein C (APC) on oxygen-glucose deprivation (OGD) induced ischemic conditions in in-vitro SH-SY5Y cells. SH-SY5Y cells were exposed to OGD in an airtight chamber containing 95% N2 and 5% CO2 and media deprived of glucose for 4 h following 24 h of reoxygenation. The cell toxicity, viability, expression of receptors such as endothelial cell protein C receptor (EPCR), protease-activated receptor (PAR)1, PAR3, and apoptosis-related proteins B-cell lymphoma 2 (BCL-2), BCL-2-like protein 4 (Bax), Poly [ADP-ribose] polymerase-1 (PARP-1) were assessed. Administration of APC decreased the cellular injury when compared to the OGD exposed group in a dose-dependent manner and displayed increased expression of PAR-1, PAR-3, and EPCR. The APC treatment leads to a reduction in PARP-1 expression and cleavage and apoptosis-inducing factor (AIF) expression. The reduction of caspase-3 activity and PARP-1 and AIF expression following APC administration results in restoring mitochondrial function with decreased cellular injury and apoptosis. Our results suggested that APC has potent protective effects against in-vitro ischemia in SH-SY5Y cells by modulating mitochondrial function.
Collapse
Affiliation(s)
- Mukesh Kumar Sriwastva
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
- Correspondence: ; Tel.: +91-112659-4488
| | - Remesh Kunjunni
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| | - Mutahar Andrabi
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| | - Kameshwar Prasad
- Department of Neurology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Renu Saxena
- Department of Hematology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Vivekanandhan Subbiah
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| |
Collapse
|
5
|
Hong W, Mo F, Zhang Z, Huang M, Wei X. Nicotinamide Mononucleotide: A Promising Molecule for Therapy of Diverse Diseases by Targeting NAD+ Metabolism. Front Cell Dev Biol 2020; 8:246. [PMID: 32411700 PMCID: PMC7198709 DOI: 10.3389/fcell.2020.00246] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/24/2020] [Indexed: 02/05/2023] Open
Abstract
NAD+, a co-enzyme involved in a great deal of biochemical reactions, has been found to be a network node of diverse biological processes. In mammalian cells, NAD+ is synthetized, predominantly through NMN, to replenish the consumption by NADase participating in physiologic processes including DNA repair, metabolism, and cell death. Correspondingly, aberrant NAD+ metabolism is observed in many diseases. In this review, we discuss how the homeostasis of NAD+ is maintained in healthy condition and provide several age-related pathological examples related with NAD+ unbalance. The sirtuins family, whose functions are NAD-dependent, is also reviewed. Administration of NMN surprisingly demonstrated amelioration of the pathological conditions in some age-related disease mouse models. Further clinical trials have been launched to investigate the safety and benefits of NMN. The NAD+ production and consumption pathways including NMN are essential for more precise understanding and therapy of age-related pathological processes such as diabetes, ischemia–reperfusion injury, heart failure, Alzheimer’s disease, and retinal degeneration.
Collapse
Affiliation(s)
- Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Mo
- West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Department of Biotherapy, Chengdu, China
| | - Ziqi Zhang
- West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Department of Biotherapy, Chengdu, China
| | - Mengyuan Huang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Klimova N, Fearnow A, Long A, Kristian T. NAD + precursor modulates post-ischemic mitochondrial fragmentation and reactive oxygen species generation via SIRT3 dependent mechanisms. Exp Neurol 2019; 325:113144. [PMID: 31837320 DOI: 10.1016/j.expneurol.2019.113144] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022]
Abstract
Global cerebral ischemia depletes brain tissue NAD+, an essential cofactor for mitochondrial and cellular metabolism, leading to bioenergetics failure and cell death. The post-ischemic NAD+ levels can be replenished by the administration of nicotinamide mononucleotide (NMN), which serves as a precursor for NAD+ synthesis. We have shown that NMN administration shows dramatic protection against ischemic brain damage and inhibits post-ischemic hippocampal mitochondrial fragmentation. To understand the mechanism of NMN-induced modulation of mitochondrial dynamics and neuroprotection we used our transgenic mouse models that express mitochondria targeted yellow fluorescent protein in neurons (mito-eYFP) and mice that carry knockout of mitochondrial NAD+-dependent deacetylase sirt3 gene (SIRT3KO). Following ischemic insult, the mitochondrial NAD+ levels were depleted leading to an increase in mitochondrial protein acetylation, high reactive oxygen species (ROS) production, and excessive mitochondrial fragmentation. Administration of a single dose of NMN normalized hippocampal mitochondria NAD+ pools, protein acetylation, and ROS levels. These changes were dependent on SIRT3 activity, which was confirmed using SIRT3KO mice. Ischemia induced increase in acetylation of the key mitochondrial antioxidant enzyme, superoxide dismutase 2 (SOD2) that resulted in inhibition of its activity. This was reversed after NMN treatment followed by reduction of ROS generation and suppression of mitochondrial fragmentation. Specifically, we found that the interaction of mitochondrial fission protein, pDrp1(S616), with neuronal mitochondria was inhibited in NMN treated ischemic mice. Our data thus provide a novel link between mitochondrial NAD+ metabolism, ROS production, and mitochondrial fragmentation. Using NMN to target these mechanisms could represent a new therapeutic approach for treatment of acute brain injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Nina Klimova
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Adam Fearnow
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| | - Aaron Long
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA; Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
7
|
Hosseini L, Vafaee MS, Mahmoudi J, Badalzadeh R. Nicotinamide adenine dinucleotide emerges as a therapeutic target in aging and ischemic conditions. Biogerontology 2019; 20:381-395. [PMID: 30838484 DOI: 10.1007/s10522-019-09805-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/27/2019] [Indexed: 02/06/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) has been described as central coenzyme of redox reactions and is a key regulator of stress resistance and longevity. Aging is a multifactorial and irreversible process that is characterized by a gradual diminution in physiological functions in an organism over time, leading to development of age-associated pathologies and eventually increasing the probability of death. Ischemia is the lack of nutritive blood flow that causes damage and mortality that mostly occurs in various organs during aging. During the process of aging and related ischemic conditions, NAD+ levels decline and lead to nuclear and mitochondrial dysfunctions, resulting in age-related pathologies. The majority of studies have shown that restoring of NAD+ using supplementation with intermediates such as nicotinamide mononucleotide and nicotinamide riboside can be a valuable strategy for recovery of ischemic injury and age-associated defects. This review summarizes the molecular mechanisms responsible for the reduction in NAD+ levels during ischemic disorders and aging, as well as a particular focus is given to the recent progress in the understanding of NAD+ precursor's effects on aging and ischemia.
Collapse
Affiliation(s)
- Leila Hosseini
- Drug Applied Research Center, Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr S Vafaee
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.,Neuroscience Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neuroscience Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran. .,Molecular Medicine Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Song SH, Jee YS, Ko IG, Lee SW, Sim YJ, Kim DY, Lee SJ, Cho YS. Treadmill exercise and wheel exercise improve motor function by suppressing apoptotic neuronal cell death in brain inflammation rats. J Exerc Rehabil 2018; 14:911-919. [PMID: 30656148 PMCID: PMC6323344 DOI: 10.12965/jer.1836508.254] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 11/22/2022] Open
Abstract
Brain inflammation is involved in many brain disorders, such as brain ischemic injury, Alzheimer diseases, and Parkinson disease. Physical exercise has been recommended for the prevention and treatment of many brain inflammatory diseases. In the present study, the effects of exercise on motor function in relation with apoptotic neuronal cell death following neuroinflammation were investigated. Moreover, we compared the effect of forced exercise with voluntary exercise on neuroinflammation-induced motor malfunction. For this study, rota-rod test, vertical pole test, foot fault test, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay, immunohistochemistry for caspase-3, and western blot for Bcl-2 and Bax were performed. Lipopolysaccharide was intraventricular infused for induction of brain inflammation. Treadmill exercise and wheel exercise were conducted during 6 weeks. In the present results, Treadmill exercise and wheel exercise alleviated brain inflammation-induced motor impairments by suppressing apoptotic neuronal cell death in the motor cortex. These effects of treadmill exercise and wheel exercise were similarly appeared.
Collapse
Affiliation(s)
- Sang-Hyuk Song
- Department of Physical Activity Design, Hanseo University, Seosan, Korea
| | - Yong-Seok Jee
- Department of Physical Activity Design, Hanseo University, Seosan, Korea
| | - Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Won Lee
- Department of Physical Education, Korea Military Academy, Seoul, Korea
| | - Young-Je Sim
- Department of Physical Education, Kunsan National University, Gunsan, Korea
| | - Dae-Young Kim
- Department of Sports Healthcare, College of Humanities & Social Sciences, Inje University, Gimhae, Korea
| | - Sam-Jun Lee
- Department of Physical Education, College of Health, Welfare, and Education, Tong Myong University, Busan, Korea
| | - Young Sam Cho
- Department of Urology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Interplay between NAD + and acetyl‑CoA metabolism in ischemia-induced mitochondrial pathophysiology. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2060-2067. [PMID: 30261291 DOI: 10.1016/j.bbadis.2018.09.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022]
Abstract
Brain injury caused by ischemic insult due to significant reduction or interruption in cerebral blood flow leads to disruption of practically all cellular metabolic pathways. This triggers a complex stress response followed by overstimulation of downstream enzymatic pathways due to massive activation of post-translational modifications (PTM). Mitochondria are one of the most sensitive organelle to ischemic conditions. They become dysfunctional due to extensive fragmentation, inhibition of acetyl‑CoA production, and increased activity of NAD+ consuming enzymes. These pathologic conditions ultimately lead to inhibition of oxidative phosphorylation and mitochondrial ATP production. Both acetyl‑CoA and NAD+ are essential intermediates in cellular bioenergetics metabolism and also serve as substrates for post-translational modifications such as acetylation and ADP‑ribosylation. In this review we discuss ischemia/reperfusion-induced changes in NAD+ and acetyl‑CoA metabolism, how these affect relevant PTMs, and therapeutic approaches that restore the physiological levels of these metabolites leading to promising neuroprotection.
Collapse
|
10
|
Noh MY, Lee WM, Lee SJ, Kim HY, Kim SH, Kim YS. Regulatory T cells increase after treatment with poly (ADP-ribose) polymerase-1 inhibitor in ischemic stroke patients. Int Immunopharmacol 2018; 60:104-110. [PMID: 29709770 DOI: 10.1016/j.intimp.2018.04.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/04/2018] [Accepted: 04/24/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) are thought to play a modulatory role in immune responses and to improve outcomes after ischemic stroke. Thus, various strategies for increasing Tregs in animal models of ischemic stroke have yielded successful results. The aim of this study was to examine the potential effect of poly (ADP-ribose) polymerase-1 (PARP-1) inhibitor on Treg proportion in stroke patients. METHODS Peripheral blood samples were collected from 12 ischemic stroke patients (within 72 h of stroke onset) and 5 healthy control subjects. Flow cytometry analyses and quantitative reverse transcription polymerase chain reactions (qRT-PCR) were performed on peripheral blood mononuclear cells (PBMCs) before and after treating them with PARP-1 inhibitor (3-AB; JPI-289 1 μm, JPI-289 10 μm) for 24 h. RESULTS Treg proportions were significantly higher in healthy controls (median 2.8%, IQR 2.6-5.0%) than ischemic stroke patients (median 1.6%, IQR 1.25-2.2%) (p < 0.001). In the latter, Treg proportions were positively correlated with age (r = 0.595, p = 0.041), but not with infarct volume (r = 0.367, p = 0.241). After PARP-1 inhibitor treatment, Treg proportions among PBMCs increased in response to high dose (10 μm) JPI-289 (median 2.3%, IQR 2.0-2.9%) as did Treg-associated transcription factors such as FoxP3 and CTLA-4 mRNA. PARP-1 inhibitor treatment also decreased pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-17) and increased anti-inflammatory cytokines (IL-4, IL-10, and TGF-β1). CONCLUSION Treg proportions are reduced in ischemic stroke patients and increased by treatment with high-dose PARP-1 inhibitor JPI-289. The PARP-1 inhibitor also had a possible anti-inflammatory effect on cytokine levels, and may ameliorate the outcome of ischemic stroke by up-regulating Tregs.
Collapse
Affiliation(s)
- Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Won Moo Lee
- Department of Obstetrics and Gynecology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Su-Jung Lee
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Young Seo Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC. Neuronal Cell Death. Physiol Rev 2018; 98:813-880. [PMID: 29488822 PMCID: PMC5966715 DOI: 10.1152/physrev.00011.2017] [Citation(s) in RCA: 746] [Impact Index Per Article: 106.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Neuronal cell death occurs extensively during development and pathology, where it is especially important because of the limited capacity of adult neurons to proliferate or be replaced. The concept of cell death used to be simple as there were just two or three types, so we just had to work out which type was involved in our particular pathology and then block it. However, we now know that there are at least a dozen ways for neurons to die, that blocking a particular mechanism of cell death may not prevent the cell from dying, and that non-neuronal cells also contribute to neuronal death. We review here the mechanisms of neuronal death by intrinsic and extrinsic apoptosis, oncosis, necroptosis, parthanatos, ferroptosis, sarmoptosis, autophagic cell death, autosis, autolysis, paraptosis, pyroptosis, phagoptosis, and mitochondrial permeability transition. We next explore the mechanisms of neuronal death during development, and those induced by axotomy, aberrant cell-cycle reentry, glutamate (excitoxicity and oxytosis), loss of connected neurons, aggregated proteins and the unfolded protein response, oxidants, inflammation, and microglia. We then reassess which forms of cell death occur in stroke and Alzheimer's disease, two of the most important pathologies involving neuronal cell death. We also discuss why it has been so difficult to pinpoint the type of neuronal death involved, if and why the mechanism of neuronal death matters, the molecular overlap and interplay between death subroutines, and the therapeutic implications of these multiple overlapping forms of neuronal death.
Collapse
Affiliation(s)
- Michael Fricker
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Aviva M Tolkovsky
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Vilmante Borutaite
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Michael Coleman
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Guy C Brown
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
12
|
Czapski GA, Cieślik M, Wencel PL, Wójtowicz S, Strosznajder RP, Strosznajder JB. Inhibition of poly(ADP-ribose) polymerase-1 alters expression of mitochondria-related genes in PC12 cells: relevance to mitochondrial homeostasis in neurodegenerative disorders. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:281-288. [PMID: 29128369 DOI: 10.1016/j.bbamcr.2017.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is characterized by the release of amyloid beta peptides (Aβ) in the form of monomers/oligomers which may lead to oxidative stress, mitochondria dysfunction, synaptic loss, neuroinflammation and, in consequence, to overactivation of poly(ADP-ribose) polymerase-1 (PARP-1). However, Aβ peptides are also released in the brain ischemia, traumatic injury and in inflammatory response. PARP-1 is suggested to be a promising target in therapy of neurodegenerative disorders. We investigated the impact of PARP-1 inhibition on transcription of mitochondria-related genes in PC12 cells. Moreover, the effect of PARP-1 inhibitor (PJ34) on cells subjected to Aβ oligomers (AβO) - evoked stress was analyzed. Our data demonstrated that inhibition of PARP-1 in PC12 cells enhanced the transcription of genes for antioxidative enzymes (Sod1, Gpx1, Gpx4), activated genes regulating mitochondrial fission/fusion (Mfn1, Mfn2, Dnm1l, Opa1, Fis1), subunits of ETC complexes (mt-Nd1, Sdha, mt-Cytb) and modulated expression of several TFs, enhanced Foxo1 and decreased Nrf1, Stat6, Nfkb1. AβO elevated free radicals concentration, decreased mitochondria membrane potential (MMP) and cell viability after 24h. Gene transcription was not affected by AβO after 24h, but was significantly downregulated after 96h. In AβO stress, PJ34 exerted stimulatory effect on expression of several genes (Gpx1, Gpx4, Opa1, Mfn2, Fis1 and Sdha), decreased transcription of numerous TFs (Nrf1, Tfam, Stat3, Stat6, Trp53, Nfkb1) and prevented oxidative stress. Our results indicated that PARP-1 inhibition significantly enhanced transcription of genes involved in antioxidative defense and in regulation of mitochondria function, but was not able to ameliorate cells viability affected by Aβ.
Collapse
Affiliation(s)
- Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Przemysław L Wencel
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Sylwia Wójtowicz
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Joanna B Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| |
Collapse
|
13
|
Wencel PL, Lukiw WJ, Strosznajder JB, Strosznajder RP. Inhibition of Poly(ADP-ribose) Polymerase-1 Enhances Gene Expression of Selected Sirtuins and APP Cleaving Enzymes in Amyloid Beta Cytotoxicity. Mol Neurobiol 2017; 55:4612-4623. [PMID: 28698968 PMCID: PMC5948241 DOI: 10.1007/s12035-017-0646-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) and sirtuins (SIRTs) are involved in the regulation of cell metabolism, transcription, and DNA repair. Alterations of these enzymes may play a crucial role in Alzheimer's disease (AD). Our previous results indicated that amyloid beta (Aβ) peptides and inflammation led to activation of PARP1 and cell death. This study focused on a role of PARP1 in the regulation of gene expression for SIRTs and beta-amyloid precursor protein (βAPP) cleaving enzymes under Aβ42 oligomers (AβO) toxicity in pheochromocytoma cells (PC12) in culture. Moreover, the effect of endogenously liberated Aβ peptides in PC12 cells stably transfected with human gene for APP wild-type (APPwt) was analyzed. Our results demonstrated that AβO enhanced transcription of presenilins (Psen1 and Psen2), the crucial subunits of γ-secretase. Aβ peptides in APPwt cells activated expression of β-secretase (Bace1), Psen1, Psen2, and Parp1. The inhibitor of PARP1, PJ-34 in the presence of AβO upregulated transcription of α-secretase (Adam10), Psen1, and Psen2, but also Bace1. Concomitantly, PJ-34 enhanced mRNA level of nuclear Sirt1, Sirt6, mitochondrial Sirt4, and Parp3 in PC12 cells subjected to AβOs toxicity. Our data indicated that Aβ peptides through modulation of APP secretases may lead to a vicious metabolic circle, which could be responsible for maintaining Aβ at high level. PARP1 inhibition, besides activation of nuclear SIRTs and mitochondrial Sirt4 expression, enhanced transcription of enzyme(s) involved in βAPP metabolism, and this effect should be considered in its application against Aβ peptide toxicity.
Collapse
Affiliation(s)
- Przemysław L Wencel
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA
| | - Joanna B Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Robert Piotr Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
14
|
Park JH, Park JA, Ahn JH, Kim YH, Kang IJ, Won MH, Lee CH. Transient cerebral ischemia induces albumin expression in microglia only in the CA1 region of the gerbil hippocampus. Mol Med Rep 2017; 16:661-665. [PMID: 28586018 PMCID: PMC5482121 DOI: 10.3892/mmr.2017.6671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 02/09/2017] [Indexed: 11/30/2022] Open
Abstract
Albumin, the most abundant plasma protein, is known to exhibit a neuroprotective effect in animal models of focal and global cerebral ischemia. In the present study, the expression and immunoreactivity of albumin was examined in the hippocampus following 5 min of transient cerebral ischemia in gerbils. Albumin immunoreactivity was observed in microglia of the CA1 hippocampal region 2 days post-ischemic insult, and it was significantly increased at 4 days following ischemia-reperfusion. In addition, at 4 days post-ischemic insult, albumin-immunoreactive microglia were abundant in the stratum pyramidale of the CA1 region. The present results demonstrated that albumin was newly expressed post-injury in microglia in the CA1 region, suggesting ischemia-induced neuronal loss. Albumin expression may therefore be associated with ischemia-induced delayed neuronal death in the CA1 region following transient cerebral ischemia.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jin-A Park
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Chuncheon, Gangwon 24252, Republic of Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| |
Collapse
|
15
|
Nicotinamide mononucleotide inhibits post-ischemic NAD(+) degradation and dramatically ameliorates brain damage following global cerebral ischemia. Neurobiol Dis 2016; 95:102-10. [PMID: 27425894 DOI: 10.1016/j.nbd.2016.07.018] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/17/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is an essential cofactor for multiple cellular metabolic reactions and has a central role in energy production. Brain ischemia depletes NAD(+) pools leading to bioenergetics failure and cell death. Nicotinamide mononucleotide (NMN) is utilized by the NAD(+) salvage pathway enzyme, nicotinamide adenylyltransferase (Nmnat) to generate NAD(+). Therefore, we examined whether NMN could protect against ischemic brain damage. Mice were subjected to transient forebrain ischemia and treated with NMN or vehicle at the start of reperfusion or 30min after the ischemic insult. At 2, 4, and 24h of recovery, the proteins poly-ADP-ribosylation (PAR), hippocampal NAD(+) levels, and expression levels of NAD(+) salvage pathway enzymes were determined. Furthermore, animal's neurologic outcome and hippocampal CA1 neuronal death was assessed after six days of reperfusion. NMN (62.5mg/kg) dramatically ameliorated the hippocampal CA1 injury and significantly improved the neurological outcome. Additionally, the post-ischemic NMN treatment prevented the increase in PAR formation and NAD(+) catabolism. Since the NMN administration did not affect animal's temperature, blood gases or regional cerebral blood flow during recovery, the protective effect was not a result of altered reperfusion conditions. These data suggest that administration of NMN at a proper dosage has a strong protective effect against ischemic brain injury.
Collapse
|
16
|
Yang LC, Guo H, Zhou H, Suo DQ, Li WJ, Zhou Y, Zhao Y, Yang WS, Jin X. Chronic oleoylethanolamide treatment improves spatial cognitive deficits through enhancing hippocampal neurogenesis after transient focal cerebral ischemia. Biochem Pharmacol 2015; 94:270-81. [PMID: 25748831 DOI: 10.1016/j.bcp.2015.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/24/2015] [Indexed: 11/28/2022]
Abstract
Oleoylethanolamide (OEA) has been shown to have neuroprotective effects after acute cerebral ischemic injury. The aim of this study was to investigate the effects of chronic OEA treatment on ischemia-induced spatial cognitive impairments, electrophysiology behavior and hippocampal neurogenesis. Daily treatments of 30 mg/kg OEA significantly ameliorated spatial cognitive deficits and attenuated the inhibition of long-term potentiation (LTP) in the middle cerebral artery occlusion (MCAO) rat model. Moreover, OEA administration improved cognitive function in a manner associated with enhanced neurogenesis in the hippocampus. Further study demonstrated that treatment with OEA markedly increased the expressions of brain-derived neurotrophic factor (BDNF) and peroxisome proliferator-activated receptors α (PPARα). Our data suggest that chronic OEA treatment can exert functional recovery of cognitive impairments and neuroprotective effects against cerebral ischemic insult in rats via triggering of neurogenesis in the hippocampus, which supports the therapeutic use of OEA for cerebral ischemia.
Collapse
Affiliation(s)
- Li-Chao Yang
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Han Guo
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Hao Zhou
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Da-Qin Suo
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Wen-Jun Li
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Yu Zhou
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Yun Zhao
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China
| | - Wu-Shuang Yang
- Department of Neurosurgery, Xiamen Traditional Chinese Medicine Hospital, Xiamen 361005, China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiang'an District, Xiamen 361102, China.
| |
Collapse
|
17
|
Malairaman U, Dandapani K, Katyal A. Effect of Ca2EDTA on zinc mediated inflammation and neuronal apoptosis in hippocampus of an in vivo mouse model of hypobaric hypoxia. PLoS One 2014; 9:e110253. [PMID: 25340757 PMCID: PMC4207758 DOI: 10.1371/journal.pone.0110253] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/16/2014] [Indexed: 12/17/2022] Open
Abstract
Background Calcium overload has been implicated as a critical event in glutamate excitotoxicity associated neurodegeneration. Recently, zinc accumulation and its neurotoxic role similar to calcium has been proposed. Earlier, we reported that free chelatable zinc released during hypobaric hypoxia mediates neuronal damage and memory impairment. The molecular mechanism behind hypobaric hypoxia mediated neuronal damage is obscure. The role of free zinc in such neuropathological condition has not been elucidated. In the present study, we investigated the underlying role of free chelatable zinc in hypobaric hypoxia-induced neuronal inflammation and apoptosis resulting in hippocampal damage. Methods Adult male Balb/c mice were exposed to hypobaric hypoxia and treated with saline or Ca2EDTA (1.25 mM/kg i.p) daily for four days. The effects of Ca2EDTA on apoptosis (caspases activity and DNA fragmentation), pro-inflammatory markers (iNOS, TNF-α and COX-2), NADPH oxidase activity, poly(ADP ribose) polymerase (PARP) activity and expressions of Bax, Bcl-2, HIF-1α, metallothionein-3, ZnT-1 and ZIP-6 were examined in the hippocampal region of brain. Results Hypobaric hypoxia resulted in increased expression of metallothionein-3 and zinc transporters (ZnT-1 and ZIP-6). Hypobaric hypoxia elicited an oxidative stress and inflammatory response characterized by elevated NADPH oxidase activity and up-regulation of iNOS, COX-2 and TNF-α. Furthermore, hypobaric hypoxia induced HIF-1α protein expression, PARP activation and apoptosis in the hippocampus. Administration of Ca2EDTA significantly attenuated the hypobaric hypoxia induced oxidative stress, inflammation and apoptosis in the hippocampus. Conclusion We propose that hypobaric hypoxia/reperfusion instigates free chelatable zinc imbalance in brain associated with neuroinflammation and neuronal apoptosis. Therefore, zinc chelating strategies which block zinc mediated neuronal damage linked with cerebral hypoxia and other neurodegenerative conditions can be designed in future.
Collapse
Affiliation(s)
- Udayabanu Malairaman
- Dr.B.R.Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Kumaran Dandapani
- Dr.B.R.Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Anju Katyal
- Dr.B.R.Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
- * E-mail:
| |
Collapse
|
18
|
Sriram CS, Jangra A, Kasala ER, Bodduluru LN, Bezbaruah BK. Targeting poly(ADP-ribose)polymerase1 in neurological diseases: A promising trove for new pharmacological interventions to enter clinical translation. Neurochem Int 2014; 76:70-81. [PMID: 25049175 DOI: 10.1016/j.neuint.2014.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022]
Abstract
The highly conserved abundant nuclear protein poly(ADP-ribose)polymerase1 (PARP1) functions at the center of cellular stress response and is mainly implied in DNA damage repair mechanism. Apart from its involvement in DNA damage repair, it does sway multiple vital cellular processes such as cell death pathways, cell aging, insulator function, chromatin modification, transcription and mitotic apparatus function. Since brain is the principal organ vulnerable to oxidative stress and inflammatory responses, upon stress encounters robust DNA damage can occur and intense PARP1 activation may result that will lead to various CNS diseases. In the context of soaring interest towards PARP1 as a therapeutic target for newer pharmacological interventions, here in the present review, we are attempting to give a silhouette of the role of PARP1 in the neurological diseases and the potential of its inhibitors to enter clinical translation, along with its structural and functional aspects.
Collapse
Affiliation(s)
- Chandra Shekhar Sriram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India.
| | - Ashok Jangra
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Eshvendar Reddy Kasala
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Lakshmi Narendra Bodduluru
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Babul Kumar Bezbaruah
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India; Department of Pharmacology, III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| |
Collapse
|
19
|
Gerace E, Masi A, Resta F, Felici R, Landucci E, Mello T, Pellegrini-Giampietro DE, Mannaioni G, Moroni F. PARP-1 activation causes neuronal death in the hippocampal CA1 region by increasing the expression of Ca(2+)-permeable AMPA receptors. Neurobiol Dis 2014; 70:43-52. [PMID: 24954469 DOI: 10.1016/j.nbd.2014.05.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/07/2014] [Accepted: 05/17/2014] [Indexed: 01/22/2023] Open
Abstract
An excessive activation of poly(ADP-ribose) polymerases (PARPs) may trigger a form of neuronal death similar to that occurring in neurodegenerative disorders. To investigate this process, we exposed organotypic hippocampal slices to N-methyl-N'-nitro-N'-nitrosoguanidine (MNNG, 100μM for 5min), an alkylating agent widely used to activate PARP-1. MNNG induced a pattern of degeneration of the CA1 pyramidal cells morphologically similar to that observed after a brief period of oxygen and glucose deprivation (OGD). MNNG exposure was also associated with a dramatic increase in PARP-activity and a robust decrease in NAD(+) and ATP content. These effects were prevented by PARP-1 but not PARP-2 inhibitors. In our experimental conditions, cell death was not mediated by AIF translocation (parthanatos) or caspase-dependent apoptotic processes. Furthermore, we found that PARP activation was followed by a significant deterioration of neuronal membrane properties. Using electrophysiological recordings we firstly investigated the suggested ability of ADP-ribose to open TRPM2 channels in MNNG-induced cells death, but the results we obtained showed that TRPM2 channels are not involved. We then studied the involvement of glutamate receptor-ion channel complex and we found that NBQX, a selective AMPA receptor antagonist, was able to effectively prevent CA1 neuronal loss while MK801, a NMDA antagonist, was not active. Moreover, we observed that MNNG treatment increased the ratio of GluA1/GluA2 AMPAR subunit expression, which was associated with an inward rectification of the IV relationship of AMPA sEPSCs in the CA1 but not in the CA3 subfield. Accordingly, 1-naphthyl acetyl spermine (NASPM), a selective blocker of Ca(2+)-permeable GluA2-lacking AMPA receptors, reduced MNNG-induced CA1 pyramidal cell death. In conclusion, our results show that activation of the nuclear enzyme PARP-1 may change the expression of membrane proteins and Ca(2+) permeability of AMPA channels, thus affecting the function and survival of CA1 pyramidal cells.
Collapse
Affiliation(s)
- E Gerace
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - A Masi
- Department of Neuroscience, Section of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - F Resta
- Department of Neuroscience, Section of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - R Felici
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - E Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - T Mello
- Department of Experimental and Biomedical Sciences, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - D E Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - G Mannaioni
- Department of Neuroscience, Section of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - F Moroni
- Department of Neuroscience, Section of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
20
|
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: 410] [Impact Index Per Article: 37.3] [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
|
21
|
Czubowicz K, Strosznajder R. Ceramide in the molecular mechanisms of neuronal cell death. The role of sphingosine-1-phosphate. Mol Neurobiol 2014; 50:26-37. [PMID: 24420784 PMCID: PMC4181317 DOI: 10.1007/s12035-013-8606-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/08/2013] [Indexed: 01/27/2023]
Abstract
Ceramide and sphingosine-1-phosphate (S1P), two important bioactive sphingolipids, have been suggested as being key players in the pathology of Alzheimer’s disease in inflammation and cancer. However, their role in the molecular mechanisms of neuronal death has not been fully elucidated. Our study indicated that ceramide significantly enhanced the level of free radicals and decreased the viability of the human neuroblastoma cell line (SH-SY5Y) through inhibition of the prosurvival PI3-K/Akt pathway. Ceramide also decreased anti-apoptotic (Bcl-2) and increased pro-apoptotic (Bax, Hrk) mRNA/protein levels. Concomitantly, our study indicated that ceramide induced poly(ADP-ribose) polymerase-1 (PARP-1) activation and accumulation of poly(ADP-ribose) PAR, a signalling molecule involved in mitochondria-nucleus cross-talk and mitochondria integrity. Ceramide treatment significantly decreased the level of apoptosis-inducing factor (AIF) in the mitochondria. The PARP-1 inhibitor (PJ-34) prevented AIF release from the mitochondria. In addition, our data showed that exogenously added S1P increased the viability of SH-SY5Y through the S1P (1,3) receptor-dependent mechanism. It was also revealed that the S1P and PARP-1 inhibitor (PJ-34) decreased oxidative stress, gene expression of the pro-apoptotic Hrk protein and up-regulated the anti-apoptotic Bcl-2 protein. Our data demonstrate that neuronal cell death evoked by ceramide is regulated by PARP/PAR/AIF and by S1P receptor signalling. In summary, our results suggest that PARP-1 inhibitor(s) and modulators of sphingosine-1-phosphate receptor(s) should be considered in potential therapeutic strategies directed at neurodegenerative diseases.
Collapse
Affiliation(s)
- Kinga Czubowicz
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | | |
Collapse
|
22
|
Curtin N, Szabo C. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol Aspects Med 2013; 34:1217-56. [PMID: 23370117 PMCID: PMC3657315 DOI: 10.1016/j.mam.2013.01.006] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/12/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
The aim of this article is to describe the current and potential clinical translation of pharmacological inhibitors of poly(ADP-ribose) polymerase (PARP) for the therapy of various diseases. The first section of the present review summarizes the available preclinical and clinical data with PARP inhibitors in various forms of cancer. In this context, the role of PARP in single-strand DNA break repair is relevant, leading to replication-associated lesions that cannot be repaired if homologous recombination repair (HRR) is defective, and the synthetic lethality of PARP inhibitors in HRR-defective cancer. HRR defects are classically associated with BRCA1 and 2 mutations associated with familial breast and ovarian cancer, but there may be many other causes of HRR defects. Thus, PARP inhibitors may be the drugs of choice for BRCA mutant breast and ovarian cancers, and extend beyond these tumors if appropriate biomarkers can be developed to identify HRR defects. Multiple lines of preclinical data demonstrate that PARP inhibition increases cytotoxicity and tumor growth delay in combination with temozolomide, topoisomerase inhibitors and ionizing radiation. Both single agent and combination clinical trials are underway. The final part of the first section of the present review summarizes the current status of the various PARP inhibitors that are in various stages of clinical development. The second section of the present review summarizes the role of PARP in selected non-oncologic indications. In a number of severe, acute diseases (such as stroke, neurotrauma, circulatory shock and acute myocardial infarction) the clinical translatability of PARP inhibition is supported by multiple lines of preclinical data, as well as observational data demonstrating PARP activation in human tissue samples. In these disease indications, PARP overactivation due to oxidative and nitrative stress drives cell necrosis and pro-inflammatory gene expression, which contributes to disease pathology. Accordingly, multiple lines of preclinical data indicate the efficacy of PARP inhibitors to preserve viable tissue and to down-regulate inflammatory responses. As the clinical trials with PARP inhibitors in various forms of cancer progress, it is hoped that a second line of clinical investigations, aimed at testing of PARP inhibitors for various non-oncologic indications, will be initiated, as well.
Collapse
Affiliation(s)
- Nicola Curtin
- Department of Experimental Cancer Therapy, Northern Institute for Cancer Research, Newcastle University, University of Newcastle Upon Tyne, UK
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
23
|
Mitochondrial dysfunction induced by nuclear poly(ADP-ribose) polymerase-1: a treatable cause of cell death in stroke. Transl Stroke Res 2013; 5:136-44. [PMID: 24323707 DOI: 10.1007/s12975-013-0283-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022]
Abstract
Many drugs targeting excitotoxic cell death have demonstrated robust neuroprotective effects in animal models of cerebral ischemia. However, these neuroprotective effects have almost universally required drug administration at relatively short time intervals after ischemia onset. This finding has translated to clinical trial results; interventions targeting excitotoxicity have had no demonstrable efficacy when initiated hours after ischemia onset, but beneficial effects have been reported with more rapid initiation. Consequently, there continues to be a need for interventions with efficacy at later time points after ischemia. Here, we focus on mitochondrial dysfunction as both a relatively late event in ischemic neuronal death and a recognized cause of delayed neuronal death. Activation of poly(ADP-ribose) polymerase-1 (PARP-1) is a primary cause of mitochondrial depolarization and subsequent mitochondria-triggered cell death in ischemia reperfusion. PARP-1 consumes cytosolic NAD(+), thereby blocking both glycolytic ATP production and delivery of glucose carbon to mitochondria for oxidative metabolism. However, ketone bodies such as pyruvate, beta- and gamma-hydroxybutyrate, and 1,4-butanediol can fuel mitochondrial metabolism in cells with depleted cytosolic NAD(+) as long as the mitochondria remain functional. Ketone bodies have repeatedly been shown to be highly effective in preventing cell death in animal models of ischemia, but a rigorous study of the time window of opportunity for this approach remains to be performed.
Collapse
|
24
|
Evaluation of the antioxidative properties of lipoxygenase inhibitors. Pharmacol Rep 2013; 64:1179-88. [PMID: 23238474 DOI: 10.1016/s1734-1140(12)70914-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 05/30/2012] [Indexed: 11/22/2022]
Abstract
BACKGROUND Oxidative stress is a component of many pathological conditions including neurodegenerative diseases and inflammation. An important source of reactive oxygen species (ROS) are lipoxygenases (LOX) - enzymes responsible for the metabolism of arachidonic acid and other polyunsaturated fatty acids. LOX inhibitors have a protective effect in inflammatory diseases and in neurodegenerative disorders because of their anti-inflammatory activity. However, the molecular mechanism of the protective action of LOX inhibitors has not yet been fully elucidated. METHODS The aim of this study was to compare the antioxidative potential of widely used LOX inhibitors: BWB70C, AA-861, zileuton, baicalein and NDGA. The antioxidative properties were evaluated in cell-free systems. We measured the effect of the tested compounds on iron/ascorbate-induced lipid peroxidation and on carbonyl group formation in the rat brain homogenate. Direct free radical scavenging was analyzed by using DPPH assay. RESULTS Our data showed that the inhibitor of all LOXs, i.e., NDGA, 5-LOX inhibitor BWB70C and the inhibitor of 12/15-LOX, baicalein, significantly decreased the level of lipid and protein oxidation. The free radical scavenging activity of these inhibitors was comparable to known ROS scavengers, i.e., resveratrol and trolox. Zileuton (the inhibitor of 5-LOX) slightly prevented lipid and protein oxidation, it also scavenged the DPPH radical. AA-861 (the inhibitor of 5 and 12/15-LOX) slightly protected lipids against Fe/asc-evoked lipid peroxidation at high concentrations, but had no effect on carbonyl group formation and DPPH scavenging. CONCLUSIONS Our results indicate that some LOX inhibitors demonstrate potent anti-oxidative, free radical scavenging properties. AA-861, whose antioxidative potential is very weak, may be a specific tool to be used in experimental and perhaps even clinical applications.
Collapse
|
25
|
Czapski GA, Adamczyk A, Strosznajder RP, Strosznajder JB. Expression and activity of PARP family members in the hippocampus during systemic inflammation: Their role in the regulation of prooxidative genes. Neurochem Int 2013; 62:664-73. [DOI: 10.1016/j.neuint.2013.01.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 12/21/2012] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
|
26
|
Docosahexaenoic acid and tetracyclines as promising neuroprotective compounds with poly(ADP-ribose) polymerase inhibitory activities for oxidative/genotoxic stress treatment. Neurochem Int 2013; 62:626-36. [PMID: 23439385 DOI: 10.1016/j.neuint.2013.02.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 02/07/2013] [Accepted: 02/13/2013] [Indexed: 11/24/2022]
Abstract
The human genome is exposed to oxidative/genotoxic stress by several endogenous and exogenous compounds. These events evoke DNA damage and activate poly(ADP-ribose) polymerase-1 (PARP-1), the key enzyme involved in DNA repair. The massive stress and over-activation of this DNA-bound enzyme can be responsible for an energy crisis and neuronal death. The last data indicated that product of PARP-1, i.e. poly(ADP-ribose) (PAR), acts as a signalling molecule and plays a significant role in nucleus-mitochondria cross-talk. PAR translocated to the mitochondria can be involved in mitochondrial permeability, the release of an apoptosis-inducing factor (AIF). Its translocation into the nucleus leads to chromatin condensation, fragmentation and cell death. The exact mechanism of this novel death pathway has not yet fully been understood. In this study the relationship between AIF and PARP/PAR in death signalling in the neuronal cell line (HT22) subjected to oxidative/genotoxic stress evoked by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was explored. The neuroprotective influence of docosahexaenoic acid (DHA), major dietary ω-3 long-chain polyunsaturated fatty acids as well as the action of tetracyclines, the novel suppressors of PARP-1, were examined. The effect of these all compounds was compared with specific PARP-1 inhibitors. The oxidative/genotoxic stress evoked by MNNG enhanced the level of PAR in a time-dependent manner with a concomitant significant decrease in the mitochondrial AIF protein level. Moreover, the down-regulation of the anti-apoptotic proteins (Bcl-2 and Bcl-xL) and the up-regulation of the Bax pro-apoptotic protein were presented. In these conditions massive HT22 cell death was observed. Both PARP-1 inhibitors: 3-aminobenzamide (3-AB) and PJ 34, tetracycline: doxocycline and minocycline, as well as DHA protected the cells against PAR formation and AIF translocation. Moreover, all of these compounds enhanced Bcl-xL gene expression and protected the cells against MNNG-induced death. Our data show that both DHA and tetracyclines offer a novel neuroprotective strategy for oxidative/genotoxic stress treatment.
Collapse
|
27
|
The early activation of PI3K strongly enhances the resistance of cortical neurons to hypoxic injury via the activation of downstream targets of the PI3K pathway and the normalization of the levels of PARP activity, ATP, and NAD⁺. Mol Neurobiol 2012; 47:757-69. [PMID: 23254998 DOI: 10.1007/s12035-012-8382-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 11/29/2012] [Indexed: 01/29/2023]
Abstract
Phosphatidylinositol 3-kinase (PI3K) plays several important roles in neuronal survival. Activation of the pathway is essential for the neuroprotective mechanisms of materials that shield neuronal cells from many stressful conditions. However, there have been no reports to date about the effect of the direct activation of the pathway in hypoxic injury of neuronal cells. We investigated whether the direct activation of the PI3K pathway inhibits neuronal cell death induced by hypoxia. Primary cultured cortical neurons (PCCNs) were exposed to hypoxic conditions (less than 1 mol% O2) and/or treated with PI3K activator. Hypoxia reduced the viability of PCCNs in a time-dependent manner, but treatment with PI3K significantly restored viability in a concentration-dependent manner. Among the signaling proteins involved in the PI3K pathway, those associated with survival, including Akt and glycogen synthase kinase-3β, were decreased shortly after exposure to hypoxia and those associated with cell death, including BAX, apoptosis-induced factor, cytochrome c, caspase-9, caspase-3, and poly(ADP-ribose) polymerase (PARP), were increased. However, treatment with PI3K activator normalized the expression levels of those signaling proteins. PARP activity and levels of ATP and NAD(+) altered by hypoxia were also normalized with direct PI3K activation. All these findings suggest that direct and early activation is important for protecting neuronal cells from hypoxic injury.
Collapse
|
28
|
Singh N, Sharma G, Singh N, Hanif K. A Comparative Study of Neuroprotective Effect of Single and Combined Blockade of AT1 Receptor and PARP-1 in Focal Cerebral Ischaemia in Rat. Int J Stroke 2012; 9:560-8. [DOI: 10.1111/j.1747-4949.2012.00916.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/01/2012] [Indexed: 11/30/2022]
Abstract
Background Cerebral ischaemia results in enhanced expression of type 1 angiotensin receptor and oxidative stress. Free radicals due to oxidative stress lead to excessive DNA damage causing overactivation of poly (ADP-ribose) polymerase-1 resulting in neuronal death. Activation of both type 1 angiotensin receptors and poly (ADP-ribose) polymerase-1 following cerebral ischaemia takes place simultaneously, but until now, no study has explored the effect of combined blockade of both angiotensin type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 in cerebral ischaemia. Aim Our purpose was to compare the effect of single and combined treatment with angiotensin type 1 angiotensin receptor blocker, candesartan, and the poly (ADP-ribose) polymerase-1 inhibitor, 1, 5 isoquinolinediol, on brain damage and oxidative stress in transient focal cerebral ischaemia in rats. Method Transient focal cerebral ischaemia was induced in Sprague-Dawley rats by an intraluminal technique for two-hours following 48 h of reperfusion. Candesartan (0·05 mg/kg) was administered just after initiation of ischaemia followed by a repeat administration at 24 h while 1, 5 isoquinolinediol (0·1 mg/kg) was given one-hour after of ischaemia. After 24 h of reperfusion, neurological deficit was evaluated in the different treatment groups. After 48 h of reperfusion, the rats were sacrificed and the brain was isolated. Ischaemic brain damage by 2,3,5 triphenyl tetrazolium chloride staining, oxidative stress markers, and levels of reactive oxygen species were determined biochemically. Result Single treatment with candesartan and 1, 5 isoquinolinediol significantly reduced neurological deficit, infarct, and oedema volume as compared to ischaemic control and different vehicle groups for each of the drugs. However, treatment with candesartan + 1, 5 isoquinolinediol offered greater reduction in neurological deficit, cerebral infarct volume, and oedema as compared to single-drug treatments. Furthermore, treatment with candesartan + 1, 5 isoquinolinediol significantly decreased oxidative stress as compared to single treatments with each drug. Conclusion The study suggests that blockade of either type 1 angiotensin receptor or poly (ADP-ribose) polymerase-1 alone provides neuroprotection, but the better result was achieved when both type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 were blocked together by the combined use of their pharmacological inhibitor in transient cerebral ischaemia in rat.
Collapse
|
29
|
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: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 03/01/2012] [Indexed: 01/08/2023]
|
30
|
Banasik M, Stedeford T, Strosznajder RP, Takehashi M, Tanaka S, Ueda K. Inhibition of poly(ADP-ribose) polymerase-1 attenuates the toxicity of carbon tetrachloride. J Enzyme Inhib Med Chem 2011; 26:883-9. [PMID: 21395487 PMCID: PMC3296519 DOI: 10.3109/14756366.2011.557315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Carbon tetrachloride (CCl4) is routinely used as a model compound for eliciting centrilobular hepatotoxicity. It can be bioactivated to the trichloromethyl radical, which causes extensive lipid peroxidation and ultimately cell death by necrosis. Overactivation of poly(ADP-ribose) polymerase-1 (PARP-1) can rapidly reduce the levels of (β-nicotinamide adenine dinucleotide and adenosine triphosphate and ultimately promote necrosis. The aim of this study was to determine whether inhibition of PARP-1 could decrease CCl4-induced hepatotoxicity, as measured by degree of poly(ADP-ribosyl)ation, serum levels of lactate dehydrogenase (LDH), lipid peroxidation,and oxidative DNA damage. For this purpose, male ICR mice were administered intraperitoneally a hepatotoxic dose of CCl4 with or without 6(5H)-phenanthridinone, a potent inhibitor of PARP-1. Animals treated with CCl4 exhibited extensive poly(ADP-ribosyl)ation in centrilobular hepatocytes, elevated serum levels of LDH, and increased lipid peroxidation. In contrast, animals treated concomitantly with CCl4 and 6(5H)-phenanthridinone showed significantly lower levels of poly(ADP-ribosyl) ation, serum LDH, and lipid peroxidation. No changes were observed in the levels of oxidative DNA damage regardless of treatment. These results demonstrated that the hepatotoxicity of CCl4is dependent on the overactivation of PARP-1 and that inhibition of this enzyme attenuates the hepatotoxicity of CCl4.
Collapse
Affiliation(s)
- Marek Banasik
- Institute of Public Health and Environmental Protection, Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The 12/15-lipoxygenase(s) (LOX), poly(ADP-ribose) polymerase (PARP-1) activity and mitochondrial apoptosis inducing factor (AIF) protein in the amyloid β (Aβ) toxicity were investigated in PC12 cells that express either wild-type (APPwt) or double Swedish mutation (APPsw) forms of human Aβ precursor protein. Different levels of Aβ secretion and free radicals formation characterize these cells. The results demonstrated a relationship between the Aβ levels and LOX protein expression and activity. High Aβ concentration in APPsw cells correlated with a significant increase in free radicals and LOX activation, which leads to translocation of p65/NF-κB into the nucleus. An increase in AIF expression in mitochondria was observed concurrently with inhibition of PARP-1 activity in the nuclear fraction of APPsw cells. We suggested that AIF accumulation in mitochondria may be involved in adaptive/protective processes. However, inhibition of PARP-1 may be responsible for the disturbances in transcription and DNA repair as well as the degeneration of APP cells. Under conditions of increased nitrosative stress, evoked by the nitric oxide donor, sodium nitroprusside (SNP, 0.5 mM), 70-80% of all cells types died after 24 h, significantly more in APPsw cells. There was no further significant change in mitochondrial AIF level and PARP-1 activity compared to corresponding non-treated cells. Only one exception was observed in PC12 control, where SNP significantly inhibits PARP-1 activity. Moreover, SNP significantly activated gene expression for 12/15-LOX in all types of investigated cells. Inhibitors of all LOX isoforms and specific inhibitor of 12-LOX enhanced the survival of cells that were subjected to SNP. We conclude that the LOX pathways may play a role in Aβ toxicity and in nitrosative-stress-induced cell death and that inhibition of these pathways offers novel protective strategies.
Collapse
|
32
|
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: 3.1] [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
|
33
|
Kauppinen TM, Suh SW, Berman AE, Hamby AM, Swanson RA. Inhibition of poly(ADP-ribose) polymerase suppresses inflammation and promotes recovery after ischemic injury. J Cereb Blood Flow Metab 2009; 29:820-9. [PMID: 19190653 DOI: 10.1038/jcbfm.2009.9] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The brain inflammatory response induced by stroke contributes to cell death and impairs neurogenesis. Poly(ADP-ribose) polymerase-1 (PARP-1) is a coactivator of the transcription factor NF-kappaB and required for NF-kappaB-mediated inflammatory responses. Here we evaluated PARP inhibition as a means of suppressing post-stroke inflammation and improving outcome after stroke. Rats were subjected to bilateral carotid occlusion-reperfusion, and treatment with the PARP inhibitor N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34) was begun 48 h later. PJ34 was found to rapidly suppress the ischemia-induced microglial activation and astrogliosis. Behavioral tests performed 6 to 8 weeks after ischemia showed deficits in spatial memory and learning that were lessened by the PJ34 treatment. Immunohistochemical evaluation of hippocampus at 8 weeks after ischemia showed increased neuronal density in CA1 layer of PJ34-treated animals relative to vehicle-treated animals. Bromodeoxyuridine labeling showed formation of new neurons in hippocampal CA1 area in PJ34-treated animals, but not in vehicle-treated animals. Together, these results suggest that treatment with a PARP inhibitor for several days after ischemia enhances long-term neuronal survival and neurogenesis by reducing inflammation.
Collapse
Affiliation(s)
- Tiina M Kauppinen
- Department of Neurology, University of California San Francisco, San Francisco, California 94121, USA.
| | | | | | | | | |
Collapse
|
34
|
Benzamide protects delayed neuronal death and behavioural impairment in a mouse model of global cerebral ischemia. Behav Brain Res 2008; 192:178-84. [PMID: 18501976 DOI: 10.1016/j.bbr.2008.03.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 03/28/2008] [Accepted: 03/31/2008] [Indexed: 12/21/2022]
Abstract
The present study is aimed at evaluating the functional and neuroprotective effect of benzamide, a poly-(ADP-ribose) polymerase (PARP) inhibitor on delayed neuronal death (DND) in hippocampus CA1 region and memory impairment following global cerebral ischemia (GCI) in a mouse model. GCI was induced by bilateral common carotid artery occlusion (BCAo) for 20 min followed by reperfusion for 9 days. Postischemic continuous treatment with benzamide (160 mg/kg b w i.p. for 9 days) significantly reversed the GCI-induced anterograde memory impairment in passive avoidance step through and elevated plus maze tasks. The observed memory impairment in vehicle treated ischemia group was found to be well correlated with DND and downregulation of cholinergic muscarinic receptor-1 expression, which was possibly mediated by inflammation and apoptosis, as revealed from inducible nitric oxide synthase (iNOS) expression and number of TUNEL positive neurons in hippocampus CA1 region. It is clear from the present experiment that benzamide treatment significantly decreases the iNOS expression and number of apoptotic neurons and thereby improves the neuronal survival and memory during GCI. Our present findings provide compelling evidence that multiple doses of benzamide treatment is a promising therapeutic approach for cerebrovascular and neurodegenerative diseases, which deserves further clinical evaluation.
Collapse
|
35
|
Czapski GA, Cakala M, Chalimoniuk M, Gajkowska B, Strosznajder JB. Role of nitric oxide in the brain during lipopolysaccharide-evoked systemic inflammation. J Neurosci Res 2007; 85:1694-703. [PMID: 17465018 DOI: 10.1002/jnr.21294] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Although the inducible isoform of nitric oxide synthase (iNOS) is a well-established source of nitric oxide (NO*) during inflammation of the central nervous system (CNS), little is known about the involvement of constitutive isoforms of NOS (cNOS) in the inflammatory process. The aim of this study was to compare the responses of the expression and activity of iNOS and the two cNOS isoforms, neuronal and endothelial (nNOS and eNOS, respectively), in the brain to systemic inflammation and their roles in the cascade of events leading to degeneration and apoptosis. A systemic inflammatory response in C57BL/6 mice was induced by intraperitoneal injection of lipopolysaccharide [LPS; 1 mg/kg body weight (b.w.)]. The relative roles of the NOS isoforms were evaluated after injection of NG-nitro-L-arginine (NNLA; 30 mg/kg b.w.), which preferentially inhibits cNOS, or 1400W (5 mg/kg b.w.), an inhibitor of iNOS. Biochemical and morphological alterations were analyzed up to 48 hr after administration of LPS. Systemic LPS administration evoked significant ultrastructural alterations in brain capillary vessels, neuropils, and intracellular organelles of neurons, astrocytes, and microglia. Apoptotic/autophagic processes occurred in many neurons of the substantia nigra (SN), which coincided with exclusive enhancement of iNOS expression and activity in this brain region. Moreover, inhibitors of both iNOS and cNOS prevented LPS-evoked release of apoptosis-inducing factor (AIF) from SN mitochondria. Collectively, the results indicate that synthesis of NO* by both the inducible and constitutive NOS isoforms contribute to the activation of apoptotic pathways in the brain during systemic inflammation.
Collapse
Affiliation(s)
- Grzegorz A Czapski
- Department of Cellular Signalling, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
36
|
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
|
37
|
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: 3.8] [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
|
38
|
Czapski GA, Cakala M, Kopczuk D, Strosznajder JB. Effect of poly(ADP-ribose) polymerase inhibitors on oxidative stress evoked hydroxyl radical level and macromolecules oxidation in cell free system of rat brain cortex. Neurosci Lett 2004; 356:45-8. [PMID: 14746898 DOI: 10.1016/j.neulet.2003.11.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme involved in DNA repair, replication and cell cycle. However, its overactivation leads to nicotinamide adenine dinucleotide and ATP depletion and cell death. The inhibitors of PARP-1 were successfully used in the basic studies and in animal models of different diseases. For this reason, it is important to discriminate between specific and non-specific antioxidant properties of PARP-1 inhibitors. The aim of this study was to investigate the effect of PARP-1 inhibitors on the free radical level and oxidation of macromolecules and to compare their properties with the efficacy of antioxidants. Oxidative stress was induced in the brain cortex homogenate by FeCl(2) or CuSO(4) at 25 microM during 15 min incubation at 37 degrees C. PARP-1 inhibitors 3-aminobenzamide (3-AB), 1,5-dihydroxyisoquinoline (DHIQ) and 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ), and the antioxidants alpha-tocopherol, resveratrol and Tempol were used at 0-5 mM. Free radical contents were estimated by spin-trapping using HPLC. Lipid and protein oxidation were determined by measuring thiobarbituric acid reactive substances and carbonyl groups or using fluorescent probe TyrFluo, respectively. Our data indicate that 3-AB and DHIQ are potent hydroxyl radical scavengers and inhibitors of protein oxidation. DHIQ additionally decreases lipid peroxidation. DPQ has no antioxidant properties and seems to be a specific PARP-1 inhibitor, however, it is a water insoluble compound. Among the investigated antioxidants, the most potent was resveratrol and then alpha-tocopherol and Tempol. These results indicate that 3-A beta, benzamide and DHIQ are potent hydroxyl radical scavengers and antioxidants. These data ought to be taken into consideration when properties of these compounds as PARP inhibitors are evaluated.
Collapse
Affiliation(s)
- Grzegorz A Czapski
- Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St, PL-02106 Warsaw, Poland.
| | | | | | | |
Collapse
|