1
|
El-Helaly A, Abou-El-Naga AM, Alshehri KM, El-Dein MA. Miracle Tree ( Moringa oleifera) Attuned GFAP and Synaptophysin Levels, Oxidative Stress and Biomarkers in Cerebellar Fluorosis of Pregnant Rats. Pak J Biol Sci 2023; 26:628-650. [PMID: 38334155 DOI: 10.3923/pjbs.2023.628.650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
<b>Background and Objective:</b> Cerebellar fluorosis is a health issue associated with excessive exposure to fluoride (F) either in direct or indirect ways as pesticides, drinking water and caries preventing prescriptions. It is characterized by elevation in oxidative stress, inflammation, demyelination and Purkinje cell loss. <i>Moringa oleifera</i> (M), is a widely cultivated plant used as a health-booster agent in modulating various disorders because of its high content of vitamins and minerals. The beneficial effect of moringa against fluoride-induced cerebellar toxicity in pregnant rats was investigated in this study. <b>Materials and Methods:</b> Twenty pregnant rats were administered daily 300 mg kg<sup></sup><sup>1</sup> <i>M. oleifera</i> aqueous extract incorporated with 10 mg kg<sup></sup><sup>1</sup> of F intoxication from the 1st day of gestation until the 20th day. Following the termination of the trial, sera were collected and cerebellar tissue was removed for further examinations, along with the assessment of maternity. <b>Results:</b> The <i>M. oleifera</i> significantly normalized serum FSH, LH, progesterone, dopamine and serotonin levels of F-intoxicated mothers. Additionally, <i>M. oleifera</i> markedly prevented the lipid peroxidation and DNA fragmentation indicated by the tail length and moment in comet assay (-34.4 and -75.3%, respectively, when compared to the fluoride intoxicated group), while sustaining the levels of SOD and CAT revealing its antioxidant activity. The <i>M. oleifera</i> regressed the cerebellar α-amylase (-25.4%) and acetylcholinesterase activity (-40.6%), also attenuated GFAP (-73.4%, p<0.0001), synaptophysin level (216.6%, p<0.0001) and IL-6 expression (-91.2%) comparing to fluoride only treated mothers. <b>Conclusion:</b> Histological and ultrastructural examinations confirmed the recuperating effects of <i>M. oleifera</i> on mothers' cerebellar tissue intoxicated with fluoride indicated by intact folia and restored Purkinje cells number and architecture. The maternal study emphasized the anti-abortifacient activity of moringa against fluoride induced-fetotoxicity.
Collapse
|
2
|
Lan XB, Ni YS, Liu N, Wei W, Liu Y, Yang JM, Ma L, Bai R, Zhang J, Yu JQ. Neuroprotective effects of oxymatrine on hypoxic-ischemic brain damage in neonatal rats by activating the Wnt/β-catenin pathway. Biomed Pharmacother 2023; 159:114266. [PMID: 36652736 DOI: 10.1016/j.biopha.2023.114266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Neuronal apoptosis is a major pathological process associated with neurological dysfunction in neonates after hypoxic-ischemic brain damage (HIBD). Our previous study demonstrated that oxymatrine (OMT) exerts potential neuroprotective effects on neonatal rats subjected to hypoxic-ischemic insult. However, the underlying molecular mechanism remains unclear. In this study, we investigated the effects of OMT-mediated neuroprotection on neonatal HIBD by attempting to determine its effect on the Wnt/β-catenin signaling pathway and explored the underlying mechanism. Both 7-day-old rat pups and primary hippocampus neurons were used to establish the HIBD and oxygen-glucose deprivation (OGD) injury models, respectively. Our results demonstrated that OMT treatment significantly increased cerebral blood flow and reduced S100B concentration, infarct volume, and neuronal apoptosis in neonatal rats. In vitro, OMT markedly increased cell viability and MMP level and decreased DNA damage. Moreover, OMT improved the mRNA and protein levels of Wnt1 and β-catenin, inhibited the expression of DKK1 and GSK-3β, enhanced the nuclear transfer of β-catenin, and promoted the binding activity of β-catenin with Tcf-4; however, it downregulated the expression of cleaved caspase-3 and cleaved caspase-9. Notably, the introduction of XAV-939 (a Wnt/β-catenin signaling inhibitor) reversed the positive effects of OMT both in vivo and in vitro. Collectively, our findings demonstrated that OMT exerted a neuroprotective effect on neonatal HIBD by inhibiting neuronal apoptosis, which was partly via the activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiao-Bing Lan
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yuan-Shu Ni
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ning Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Wei Wei
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yue Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jia-Mei Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Lin Ma
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ru Bai
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| | - Jian Zhang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China; State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jian-Qiang Yu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China; Ningxia Characteristic Traditional Chinese Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
3
|
Koehler RC, Dawson VL, Dawson TM. Targeting Parthanatos in Ischemic Stroke. Front Neurol 2021; 12:662034. [PMID: 34025565 PMCID: PMC8131834 DOI: 10.3389/fneur.2021.662034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parthanatos is a cell death signaling pathway in which excessive oxidative damage to DNA leads to over-activation of poly(ADP-ribose) polymerase (PARP). PARP then generates the formation of large poly(ADP-ribose) polymers that induce the release of apoptosis-inducing factor from the outer mitochondrial membrane. In the cytosol, apoptosis-inducing factor forms a complex with macrophage migration inhibitory factor that translocates into the nucleus where it degrades DNA and produces cell death. In a review of the literature, we identified 24 publications from 13 laboratories that support a role for parthanatos in young male mice and rats subjected to transient and permanent middle cerebral artery occlusion (MCAO). Investigators base their conclusions on the use of nine different PARP inhibitors (19 studies) or PARP1-null mice (7 studies). Several studies indicate a therapeutic window of 4-6 h after MCAO. In young female rats, two studies using two different PARP inhibitors from two labs support a role for parthanatos, whereas two studies from one lab do not support a role in young female PARP1-null mice. In addition to parthanatos, a body of literature indicates that PARP inhibitors can reduce neuroinflammation by interfering with NF-κB transcription, suppressing matrix metaloproteinase-9 release, and limiting blood-brain barrier damage and hemorrhagic transformation. Overall, most of the literature strongly supports the scientific premise that a PARP inhibitor is neuroprotective, even when most did not report behavior outcomes or address the issue of randomization and treatment concealment. Several third-generation PARP inhibitors entered clinical oncology trials without major adverse effects and could be repurposed for stroke. Evaluation in aged animals or animals with comorbidities will be important before moving into clinical stroke trials.
Collapse
Affiliation(s)
- Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, United States
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Physiology, The Johns Hopkins University, Baltimore, MD, United States
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
4
|
Zhao Y, Zhang LX, Jiang T, Long J, Ma ZY, Lu AP, Cheng Y, Cao DS. The ups and downs of Poly(ADP-ribose) Polymerase-1 inhibitors in cancer therapy–Current progress and future direction. Eur J Med Chem 2020; 203:112570. [DOI: 10.1016/j.ejmech.2020.112570] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
|
5
|
Fusco R, Cordaro M, Siracusa R, Peritore AF, Gugliandolo E, Genovese T, D’Amico R, Crupi R, Smeriglio A, Mandalari G, Impellizzeri D, Cuzzocrea S, Di Paola R. Consumption of Anacardium Occidentale L. (Cashew Nuts) Inhibits Oxidative Stress through Modulation of the Nrf2/HO-1 and NF-kB Pathways. Molecules 2020; 25:molecules25194426. [PMID: 32993187 PMCID: PMC7582295 DOI: 10.3390/molecules25194426] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
Ischemia/reperfusion injury is a severe disorder associated with a high mortality. Several antioxidant and pharmacological properties of cashew nuts (Anacardium occidentale L.) and its metabolites from different countries have recently been described. It is a medicinal plant with important therapeutic effects. This study aimed to verify the effect of an oral administration of cashew nuts in a rat model of ischemia/reperfusion (I/R). Adult male rats were subjected to intestinal I/R injury by clamping the superior mesenteric artery for 30 min and then allowing animals to 1 h of reperfusion. Rats subjected to I/R of the gut showed a significant increase in different biochemical markers. In particular, we evaluated lipid peroxidation, tissue myeloperoxidase activity, protein carbonyl content, reactive oxygen species generation and decreased antioxidant enzyme activities. Western blot analysis showed the activation of the NRF2 and NF-kB pathways. Increased immunoreactivity to nitrotyrosine, PARP, P-selectin, and ICAM-1 was observed in the ileum of rats subjected to I/R. Administration of cashew nuts (100 mg/kg) significantly reduced the mortality rate, the fall in arterial blood pressure, and oxidative stress and restored the antioxidant enzyme activities by a mechanism involving both NRF2 and NF-kB pathways. Cashew nuts treatments reduced cytokines plasma levels, nitrotyrosine, and PARP expression as well as adhesion molecules expressions. Additionally, cashew nuts decreased the intestinal barrier dysfunction and mucosal damage, the translocation of toxins and bacteria, which leads to systemic inflammation and associated organs injuries in particular of liver and kidney. Our study demonstrates that cashew nuts administration exerts antioxidant and pharmacological protective effects in superior mesenteric artery occlusion–reperfusion shock.
Collapse
Affiliation(s)
- Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Enrico Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
- Correspondence: (D.I.); (S.C.); Tel.: +39-090-676-5208 (D.I. & S.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
- Correspondence: (D.I.); (S.C.); Tel.: +39-090-676-5208 (D.I. & S.C.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (A.F.P.); (E.G.); (T.G.); (R.D.); (A.S.); (G.M.); (R.D.P.)
| |
Collapse
|
6
|
Dong Y, Zhu H, Shen Y, Zhang W, Zhang L. Antibacterial activity of silver nanoparticles of different particle size against Vibrio Natriegens. PLoS One 2019; 14:e0222322. [PMID: 31518380 PMCID: PMC6743781 DOI: 10.1371/journal.pone.0222322] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/28/2019] [Indexed: 11/19/2022] Open
Abstract
In this study, we describe the synthesis and characterization of silver nanoparticles (Ag-NPs) of different sizes and evaluated their antibacterial activity. Particles size and morphology were characterized by transmission electron microscopy. Evaluation of the bacteriostatic effects was performed by ultraviolet-visible spectrophotometry and comet assays. The smaller the particle size of Ag-NPs, the smaller the value of the minimum inhibitory concentration (MIC) and minimum bactericidal concentrations (MBC), indicating the greater the antibacterial activity. The antibacterial activity was determined by the generation of reactive oxygen species (ROS) by bacteria and by bacterial membrane damage. In this study, we determined ROS-induced damage of bacteria caused by Ag-NPs. In conclusion, our findings indicated that Ag-NPs were effective at different particle sizes and concentrations and that the smaller the particle size of Ag-NPs, the greater the antibacterial activity.
Collapse
Affiliation(s)
- Yaohua Dong
- College of Ocean Science and Engineering, Shanghai Maritime University, Shanghai, China
- School of Mechanical Engineering, Shanghai JiaoTong University, Shanghai, China
| | - Hongling Zhu
- College of Ocean Science and Engineering, Shanghai Maritime University, Shanghai, China
| | - Yuanyuan Shen
- College of Ocean Science and Engineering, Shanghai Maritime University, Shanghai, China
| | - Wenting Zhang
- College of Ocean Science and Engineering, Shanghai Maritime University, Shanghai, China
| | - Li Zhang
- College of Ocean Science and Engineering, Shanghai Maritime University, Shanghai, China
| |
Collapse
|
7
|
Şenel B, Demir N, Büyükköroğlu G, Yıldız M. Graphene quantum dots: Synthesis, characterization, cell viability, genotoxicity for biomedical applications. Saudi Pharm J 2019; 27:846-858. [PMID: 31516327 PMCID: PMC6733895 DOI: 10.1016/j.jsps.2019.05.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/20/2019] [Indexed: 12/21/2022] Open
Abstract
We report the synthesis and applications of a novel N-doped graphene quantum dots (GQDs) using hydrothermal reaction between citric acid and p-aminophenol. The synthesized N-doped GQDs have been characterized physico-chemically and evaluated its antioxidant, antimicrobial, DNA binding and cleavage activities. siRNA loading studies were performed and their effects on cells were evaluated. Obtained results indicate that monodisperse solution of N-doped GQDs has been obtained with particles size ca. ∼10.9 ± 1.3 nm. UV–Vis spectroscopy studies of the interactions between the N-doped GQDs and calf thymus DNA (CT-DNA) showed that the compound interact with CT-DNA via both intercalative and electrostatic binding. The DNA cleavage study showed that the N-doped GQDs cleaved DNA without any external agents. The antioxidant activity of N-doped GQDS was very active when compared to BHT. As the concentration of the compound increased, the antioxidant activity also increased. Cell viability assay demonstrated that the Ndoped GQDs showed cell viability (70%) when the concentration reached 200 μg/mL for A549 and also MDA-MB-231, 150 μg/mL for NIH-3T3 cell lines at 24 h incubation. N-doped GQDs were coated with Eudragit RS 100 and EphA2-siRNA was loaded. As a result of the studies on these formulations, it was concluded that there may be significant effects on A549 cells. The microscopy results revealed that N-doped GQDs was quickly internalized into the cell. Our novel N-doped-GQDs with siRNA are candidate for in situ tumor suppression via DNA and mRNA breakage.
Collapse
Affiliation(s)
- Behiye Şenel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Anadolu University, TR-26470 Tepebaşı-Eskişehir, Turkey
| | - Neslihan Demir
- Department of Biology, Faculty of Arts and Sciences, Çanakkale Onsekiz Mart University, 17100 Çanakkale, Turkey
| | - Gülay Büyükköroğlu
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Anadolu University, TR-26470 Tepebaşı-Eskişehir, Turkey
| | - Mustafa Yıldız
- Department of Chemistry, Faculty of Arts and Sciences, Çanakkale Onsekiz Mart University, 17100 Çanakkale, Turkey.,Nanoscience and Technology Research and Application Center (NANORAC), Faculty of Arts and Sciences, Çanakkale Onsekiz Mart University, 17100 Çanakkale, Turkey
| |
Collapse
|
8
|
Effects of Poly(ADP-Ribose) Polymerase-1 Inhibition in a Neonatal Rodent Model of Hypoxic-Ischemic Injury. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2924848. [PMID: 28698869 PMCID: PMC5494065 DOI: 10.1155/2017/2924848] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/11/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022]
Abstract
Background Hypoxia ischemia (HI) to the developing brain occurs in 1–6 in 1000 live births. Large numbers of survivors have neurological long-term sequelae. However, mechanisms of recovery after HI are not understood and preventive measures or clinical treatments are not effective. Poly(ADP-ribose) polymerase-1 is overactivated in response to ischemia. In neonatal mice HI activates PARP-1 but its role in perinatal brain injury remains uncertain. Objective Aim of this study was to explore the effect of TES448 (PARP-1-inhibitor) and hypothermia after an ischemic insult. Design and Methods 10-day-old Wistar rats underwent HI. TES448 was given 10 min, 3 hrs, and 6 hrs after hypoxia. Hypothermia was started 30 min after HI and brains were dissected at P12. Western blotting and histological staining were used to evaluate for degree of injury. Results Protein expression of PARP-1 levels was diminished after TES448 treatment. Cresyl violet and TUNEL staining revealed decreased injury in male rat pups following TES448 and combined treatment. Female rats showed increased numbers of TUNEL-positive cells after combined therapy. TES448 inhibited microglia activation after hypoxic-ischemic injury. A cellular response including NeuN, Olig2, and MBP was not affected by PARP-1-inhibition. Conclusions Inhibition of PARP-1 and hypothermia lead to an alteration of injury but this effect is sexually dimorphic.
Collapse
|
9
|
The Citrus Flavanone Naringenin Protects Myocardial Cells against Age-Associated Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9536148. [PMID: 28386313 PMCID: PMC5366223 DOI: 10.1155/2017/9536148] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
In recent years, the health-promoting effects of the citrus flavanone naringenin have been examined. The results have provided evidence for the modulation of some key mechanisms involved in cellular damage by this compound. In particular, naringenin has been revealed to have protective properties such as an antioxidant effect in cardiometabolic disorders. Very recently, beneficial effects of naringenin have been demonstrated in old rats. Because aging has been demonstrated to be directly related to the occurrence of cardiac disorders, in the present study, the ability of naringenin to prevent cardiac cell senescence was investigated. For this purpose, a cellular model of senescent myocardial cells was set up and evaluated using colorimetric, fluorimetric, and immunometric techniques. Relevant cellular senescence markers, such as X-gal staining, cell cycle regulator levels, and the percentage of cell cycle-arrested cells, were found to be reduced in the presence of naringenin. In addition, cardiac markers of aging-induced damage, including radical oxidative species levels, mitochondrial metabolic activity, mitochondrial calcium buffer capacity, and estrogenic signaling functions, were also modulated by the compound. These results suggested that naringenin has antiaging effects on myocardial cells.
Collapse
|
10
|
Lechaftois M, Dreano E, Palmier B, Margaill I, Marchand-Leroux C, Bachelot-Loza C, Lerouet D. Another "string to the bow" of PJ34, a potent poly(ADP-Ribose)polymerase inhibitor: an antiplatelet effect through P2Y12 antagonism? PLoS One 2014; 9:e110776. [PMID: 25329809 PMCID: PMC4203827 DOI: 10.1371/journal.pone.0110776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background Neuro- and vasoprotective effects of poly(ADP-ribose)polymerase (PARP) inhibition have been largely documented in models of cerebral ischemia, particularly with the potent PARP inhibitor PJ34. Furthermore, after ischemic stroke, physicians are faced with incomplete tissue reperfusion and reocclusion, in which platelet activation/aggregation plays a key role. Data suggest that certain PARP inhibitors could act as antiplatelet agents. In that context, the present in vitro study investigated on human blood the potential antiplatelet effect of PJ34 and two structurally different PARP inhibitors, DPQ and INO-1001. Methods and results ADP concentrations were chosen to induce a biphasic aggregation curve resulting from the successive activation of both its receptors P2Y1 and P2Y12. In these experimental conditions, PJ34 inhibited the second phase of aggregation; this effect was reduced by incremental ADP concentrations. In addition, in line with a P2Y12 pathway inhibitory effect, PJ34 inhibited the dephosphorylation of the vasodilator stimulated phosphoprotein (VASP) in a concentration-dependent manner. Besides, PJ34 had no effect on platelet aggregation induced by collagen or PAR1 activating peptide, used at concentrations inducing a strong activation independent on secreted ADP. By contrast, DPQ and INO-1001 were devoid of any effect whatever the platelet agonist used. Conclusions We showed that, in addition to its already demonstrated beneficial effects in in vivo models of cerebral ischemia, the potent PARP inhibitor PJ34 exerts in vitro an antiplatelet effect. Moreover, this is the first study to report that PJ34 could act via a competitive P2Y12 antagonism. Thus, this antiplatelet effect could improve post-stroke reperfusion and/or prevent reocclusion, which reinforces the interest of this drug for stroke treatment.
Collapse
Affiliation(s)
- Marie Lechaftois
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Elise Dreano
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Bruno Palmier
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Isabelle Margaill
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Catherine Marchand-Leroux
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Christilla Bachelot-Loza
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Dominique Lerouet
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
11
|
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
|
12
|
Martínez-Zamudio RI, Ha HC. PARP1 enhances inflammatory cytokine expression by alteration of promoter chromatin structure in microglia. Brain Behav 2014; 4:552-65. [PMID: 25161822 PMCID: PMC4128037 DOI: 10.1002/brb3.239] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 03/22/2014] [Accepted: 04/17/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase 1 (PARP1) is a chromatin-associated enzyme that participates in processes such as transcription and DNA repair through the regulation of chromatin structure. Accumulating evidence suggests an important role for PARP1 enzymatic activity in promoting CNS inflammation by facilitating the expression of inflammatory cytokines in glial cells. However, the molecular mechanisms by which PARP1 enzymatic activity mediates this process are not well understood. In this report we sought to determine the molecular mechanisms by which PARP1 enzymatic activity facilitates the expression of Il1β and TNF in LPS-stimulated BV2 cells. METHODS PARP1 enzymatic activity and histone ADP-ribosylation were measured in LPS-stimulated BV2 cells by radioactive labelling with (32)P-NAD(+). To assess the effect of histone ADP-ribosylation on nucleosome structure, in vitro nucleosome remodeling, nuclease accessibility and binding assays were performed. These studies were complemented by chromatin immunoprecipitation assays in resting and LPS-stimulated BV2 cells in order to determine the occupancy of PARP1, nucleosomes and the RelA subunit of NF-κB, as well as ADP-ribosylation, at the Il1β and Tnf promoters. Finally, we determined the effect of pharmacological inhibition of PARP1 enzymatic activity on the LPS stimulation-dependent induction of Il1β and Tnf mRNA. RESULTS Our results indicate that LPS stimulation induces PARP1 enzymatic activity and histone ADP-ribosylation in the chromatin compartment of BV2 cells. In vitro studies show that nucleosome-bound PARP1 disrupts nucleosome structure histone ADP-ribosylation, increasing the accessibility of nucleosomal DNA. Consistent with this PARP1 is constitutively associated with at the Il1β and Tnf promoters in resting BV2 cells. Upon stimulation with LPS, ADP-ribosylation is observed at these promoters, and this is correlated with increased recruitment of the transcription factor NF-κB, resulting in robust transcription of these inflammatory cytokines. Accordingly, pharmacological inhibition of PARP1 enzymatic activity reduces NF-κB recruitment, and Il1β and Tnf expression in LPS-stimulated microglia. CONCLUSIONS Collectively, our data suggest that PARP1 facilitates inflammatory cytokine expression in microglia by increasing the accessibility of promoter DNA via histone ADP-riboyslation.
Collapse
Affiliation(s)
- Ricardo Iván Martínez-Zamudio
- Department of Biochemistry and Molecular & Cellular Biology 337 Basic Science Building, 3900 Reservoir Road, Washington, District of Columbia, 20057
| | - Hyo Chol Ha
- Department of Biochemistry and Molecular & Cellular Biology 337 Basic Science Building, 3900 Reservoir Road, Washington, District of Columbia, 20057
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Gane BD, Bhat V, Rao R, Nandhakumar S, Harichandrakumar KT, Adhisivam B. Effect of therapeutic hypothermia on DNA damage and neurodevelopmental outcome among term neonates with perinatal asphyxia: a randomized controlled trial. J Trop Pediatr 2014; 60:134-40. [PMID: 24343823 DOI: 10.1093/tropej/fmt098] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To study the effect of therapeutic hypothermia (TH) on deoxyribonucleic acid (DNA) damage and the neurodevelopmental outcome in term babies with perinatal asphyxia. METHODS Babies in the hypothermia group were cooled for the first 72 h, using gel packs. Rectal temperature of 33-34°C was maintained. Blood sample was collected before, at 36 h and after completion of TH for assessment of comet assay and 8-hydroxy2-deoxyguanosine (8-OHdG). Infants were followed up till 12 months. RESULTS Baseline parameters were similar. After 72 h, the hypothermia group showed lower olive tail moment (12.88 ± 2.14) than the control group (22.16 ± 5.26) (p < 0.001). 8-HDG levels increased significantly in the control group (1252.87 ± 357.07) as compared to the hypothermia group (757.03 ± 198.49) (p < 0.001). Neurodevelopmental assessment at 12 months showed significantly low motor and mental developmental quotient in the control than hypothermia group. CONCLUSIONS TH reduces oxidative stress-induced DNA damage and improves neurodevelopmental outcome. <Trial registration No: CTRI/2011/10/002094>
Collapse
Affiliation(s)
- Bahubali D Gane
- Division of Neonatology, Department of Pediatrics, Jawaharlal Institute of Post-graduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | | | | | | | | | | |
Collapse
|
16
|
Moroni F, Cozzi A, Chiarugi A, Formentini L, Camaioni E, Pellegrini-Giampietro DE, Chen Y, Liang S, Zaleska MM, Gonzales C, Wood A, Pellicciari R. Long-lasting neuroprotection and neurological improvement in stroke models with new, potent and brain permeable inhibitors of poly(ADP-ribose) polymerase. Br J Pharmacol 2012; 165:1487-500. [PMID: 21913897 DOI: 10.1111/j.1476-5381.2011.01666.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSES Thienyl-isoquinolone (TIQ-A) is a relatively potent PARP inhibitor able to reduce post-ischaemic neuronal death in vitro. Here we have studied, in different stroke models in vivo, the neuroprotective properties of DAMTIQ and HYDAMTIQ, two TIQ-A derivatives able to reach the brain and to inhibit PARP-1 and PARP-2. EXPERIMENTAL APPROACH Studies were carried out in (i) transient (2 h) middle cerebral artery occlusion (tMCAO), (ii) permanent MCAO (pMCAO) and (iii) electrocoagulation of the distal portion of MCA in conjunction with transient (90 min) bilateral carotid occlusion (focal cortical ischaemia). KEY RESULTS In male rats with tMCAO, HYDAMTIQ (0.1-10 mg·kg(-1)) injected i.p. three times, starting 4 h after MCAO, reduced infarct volumes by up to 70%, reduced the loss of body weight by up to 60% and attenuated the neurological impairment by up to 40%. In age-matched female rats, HYDAMTIQ also reduced brain damage. Protection, however, was less pronounced than in the male rats. In animals with pMCAO, HYDAMTIQ administered 30 min after MCAO reduced infarct volumes by approximately 40%. In animals with focal cortical ischaemia, HYDAMTIQ treatment decreased post-ischaemic accumulation of PAR (the product of PARP activity) and the presence of OX42-positive inflammatory cells in the ischaemic cortex. It also reduced sensorimotor deficits for up to 90 days after MCAO. CONCLUSION AND IMPLICATIONS Our results show that HYDAMTIQ is a potent PARP inhibitor that conferred robust neuroprotection and long-lasting improvement of post-stroke neurological deficits.
Collapse
Affiliation(s)
- F Moroni
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Martínez-Romero R, Cañuelo A, Siles E, Oliver FJ, Martínez-Lara E. Nitric oxide modulates hypoxia-inducible factor-1 and poly(ADP-ribose) polymerase-1 cross talk in response to hypobaric hypoxia. J Appl Physiol (1985) 2012; 112:816-23. [DOI: 10.1152/japplphysiol.00898.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The physiological response to hypobaric hypoxia represents a complex network of biochemical pathways in which the nitrergic system plays an important role. Previous studies have provided evidence for an interplay between the hypoxia-inducible factor-1 (HIF-1) and poly(ADP-ribose) polymerase-1 (PARP-1) under hypoxia. Here, we evaluate the potential involvement of nitric oxide (NO) in the cross talk between these two proteins. With this aim, we studied comparatively the effect of pharmacological inhibitors of NO production or PARP activity in the response of the mouse cerebral cortex to 4 h of exposure to a simulated altitude of 31,000 ft. Particularly, we analyzed the NO and reactive oxygen species production, the expression of NO synthase (NOS) isoforms, PARP-1 activity, HIF-1α expression and HIF-1 transcriptional activity, the protein level of the factor inhibiting HIF, and, finally, beclin-1 and fractin expression, as markers of cellular damage. Our results demonstrate that the reduction of NO level did not affect reactive oxygen species production but significantly 1) dampened the posthypoxic increase in neuronal NOS and inducible NOS expression without altering endothelial NOS protein level; 2) prevented PARP activation; 3) decreased HIF-1α response to hypoxia; 4) achieved a higher long-term HIF-1 transcriptional activity by reducing factor inhibiting HIF expression; and 5) reduced hypoxic damage. The pharmacological inhibition of PARP reproduced the NOS expression pattern and the HIF-1α response observed in NOS-inhibited mice, supporting its involvement in the NO-dependent regulation of hypoxia. As a whole, these results provide new data about the molecular mechanism underlying the beneficial effects of controlling NO production under hypobaric hypoxic conditions.
Collapse
Affiliation(s)
| | - Ana Cañuelo
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - F. Javier Oliver
- Institute of Parasitology and Biomedicine, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | |
Collapse
|
18
|
Davies NJ, Batehup L, Thomas R. The role of diet and physical activity in breast, colorectal, and prostate cancer survivorship: a review of the literature. Br J Cancer 2011; 105 Suppl 1:S52-73. [PMID: 22048034 PMCID: PMC3251953 DOI: 10.1038/bjc.2011.423] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Evidence for the role of diet and physical activity in cancer incidence is well documented, but owing to increased cancer survivorship, an understanding of these lifestyle factors after a cancer diagnosis is of crucial importance. The purpose of this review was to update the literature in a review undertaken for the National Cancer Survivorship Initiative and to include observational studies that were not included in the WCRF survivorship systematic review. METHODS Evidence was initially gathered from pre-defined searches of the Cochrane Library Database and PubMed from March 2006 to February 2010. After a comprehensive review regarding lifestyle and cancer, for the purpose of this article, any studies not related to diet and physical activity, prognostic outcomes, and breast, colorectal or prostate cancers were excluded. Another search of 2011 literature was conducted to update the evidence. RESULTS A total of 43 records were included in this review. Evidence from observational studies suggests that a low-fat, high-fibre diet might be protective against cancer recurrence and progression. However, there is a paucity of RCTs substantiating this. There is more support for physical activity, with a dose response for better outcomes. When synthesized with findings from the World Cancer Research Fund review of RCTs investigating the effect of diet and physical activity interventions on cancer survival, evidence suggests that the mechanism of benefit from diet and physical activity pertains to body weight, with excess body weight being a risk factor, which is modifiable through lifestyle. IMPLICATIONS Cancer survivors would like to have a more active role in their health care and to know how to look after themselves after diagnosis, including what diet and lifestyle changes they should make. The challenge is in integrating lifestyle support into standardised models of aftercare.
Collapse
Affiliation(s)
- N J Davies
- Self-Management Support Programme, National Cancer Survivorship Initiative, Macmillan Cancer Support, 89 Albert Embankment, London SE1 7UQ, UK
| | - L Batehup
- Self-Management Support Programme, National Cancer Survivorship Initiative, Macmillan Cancer Support, 89 Albert Embankment, London SE1 7UQ, UK
| | - R Thomas
- Bedford Hospital and Addenbrooke's Hospital Cambridge University NHS Trusts, c/o The Primrose Unit, Bedford Hospital, Bedford MK42 9DJ, UK
| |
Collapse
|
19
|
Poly(ADP-ribose) polymerase-1 (PARP-1) and its therapeutic implications. Vascul Pharmacol 2010; 53:77-87. [DOI: 10.1016/j.vph.2010.06.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 05/03/2010] [Accepted: 06/16/2010] [Indexed: 01/24/2023]
|
20
|
Mondello S, Galuppo M, Mazzon E, Domenico I, Mondello P, Carmela A, Cuzzocrea S. Glutamine treatment attenuates the development of ischaemia/reperfusion injury of the gut. Eur J Pharmacol 2010; 643:304-15. [PMID: 20599905 DOI: 10.1016/j.ejphar.2010.06.044] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/04/2010] [Accepted: 06/10/2010] [Indexed: 01/17/2023]
Abstract
Intestinal ischemia/reperfusion causes tissue hypoxia and damage, leading to the pathophysiology of inflammation. The aim of this study was to investigate the effects of glutamine on the tissue injury caused by ischemia/reperfusion of the gut. Ischemia/reperfusion injury of the intestine was caused by clamping both the superior mesenteric artery and the celiac trunk for 30 min followed by the release of the clamp allowing reperfusion for 1h. This procedure results in splanchnic artery occlusion-injury. Based on our findings we propose that the amino acid glutamine, administered 15 min before reperfusion at the dose of 1.5mg/kg, i.v. may be useful in the treatment of various ischemia and reperfusion diseases. The present study was performed in order to determine the pharmacological effects of glutamine ischemia/reperfusion-induced intestinal injury in rats. In particular, to gain a better insight into the mechanism(s) of action of glutamine, we evaluated the following endpoints of the inflammatory response: (1) histological damage; (2) neutrophil infiltration of the reperfused intestine (MPO activity); (3) NF-kappaB activation and cytokines production; (4) expression of ICAM-1 and P-selectin during reperfusion; (5) nitrotyrosine and poly-ADP-ribose formation; (6) pro-inflammatory cytokine production; (7) inducible nitric oxide synthase expression; (8) apoptosis as shown by TUNEL staining and (9) Bax/Bcl-2 expression.
Collapse
|
21
|
Di Paola R, Esposito E, Mazzon E, Paterniti I, Galuppo M, Cuzzocrea S. GW0742, a selective PPAR-β/δ agonist, contributes to the resolution of inflammation after gut ischemia/reperfusion injury. J Leukoc Biol 2010; 88:291-301. [DOI: 10.1189/jlb.0110053] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
GW-0742 impacts the inflammatory process associated with intestinal ischemia reperfusion.
PPARs belong to a subfamily of transcription nuclear factors. Three isoforms of PPARs have been identified: α, β/δ, and γ, encoded by different genes and distributed in various tissues. They play important roles in metabolic processes, such as regulation of glucose and lipid redistribution. They also have antiatherogenic, anti-inflammatory, as well as antihypertensive functions. There is good evidence that ligands of PPARs reduce tissue injury associated with I/R. This study investigated the effects of GW0742, a potent and selective PPAR-β/δ agonist, on tissue injury, caused in a mouse model of SAO shock. IRI of the intestine was caused by clamping the superior mesenteric artery and the celiac trunk for 45 min, followed by release of the clamp, allowing reperfusion for 1 or 6 h. Only 10% of the SAO animals survived the entire 6-h reperfusion period. In a separate set of experiments after 60 min of reperfusion, animals were killed for histological examination and biochemical studies. Administration of GW0742 (0.1 mg/kg, i.p.), 5 min prior to reperfusion, significantly reduced the (1) mortality rate, (2) histological evidence of gut injury, (3) MPO activity, (4) proinflammatory cytokines (TNF-α, IL-1β), (5) adhesion molecules (ICAM-1, P-selectin), (6) nitrotyrosine formation, (7) NF-κB expression, (8) PAR formation, and (9) apoptosis (Bax, Bcl-2, Fas-L, and TUNEL). Based on these findings, we propose that GW0742 would be useful in the treatment of various I/R diseases.
Collapse
Affiliation(s)
| | - Emanuela Esposito
- IRCCS Centro Neurolesi “Bonino-Pulejo,” Messina , Italy
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina , Italy
| | | | - Irene Paterniti
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina , Italy
| | - Maria Galuppo
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina , Italy
| | - Salvatore Cuzzocrea
- IRCCS Centro Neurolesi “Bonino-Pulejo,” Messina , Italy
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina , Italy
| |
Collapse
|
22
|
Crisafulli C, Mazzon E, Galuppo M, Paterniti I, Caminiti R, Cuzzocrea S. Olprinone attenuates the development of ischemia/reperfusion injury of the gut. Intensive Care Med 2010; 36:1235-47. [PMID: 20349038 DOI: 10.1007/s00134-010-1798-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 11/03/2009] [Indexed: 11/29/2022]
|
23
|
Crisafulli C, Di Paola R, Mazzon E, Paterniti I, Galuppo M, Genovese T, Bramanti P, Cappellani A, Cuzzocrea S. Liver X receptor agonist treatment reduced splanchnic ischemia and reperfusion injury. J Leukoc Biol 2009; 87:309-21. [PMID: 20028773 DOI: 10.1189/jlb.0609438] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
LXR is another member of the superfamily of nuclear hormone receptors that heterodimerizes with RXR and regulates the intracellular levels of cholesterol through gene induction of enzymes and proteins involved in the cholesterol metabolism and transport. LXR ligands inhibit the gene expression of proinflammatory mediators in immunostimulated macrophages; in vivo studies have shown that activation of LXR reduces the inflammatory response in a murine model of contact dermatitis and atherosclerosis. No reports have addressed a role for LXRs in pathophysiology of intestinal ischemia. The aim of this study was to investigate the effects of T0901317, a potent LXR ligand, in a mouse model of SAO shock, which was induced by clamping the superior mesenteric artery and the celiac trunk, resulting in a total occlusion of these arteries for 30 min. After this period of occlusion, the clamps were removed. Mice were killed at 60 min after reperfusion. This study provides the evidence that T0901317, LXR agonist, modulates: the development of SAO shock; the infiltration of the tissue with PMNs; the expression of TNF-alpha and IL-1beta; the nitration of tyrosine residues; NF-kappaB expression; the MAPK phosphorylation (ERK, JNK, and p38); FasL; apoptosis; Bax and Bcl-2 expression; and the degree of tissue injury caused by SAO shock. Our results imply that LXR agonists may be useful in the therapy of inflammation.
Collapse
Affiliation(s)
- Concetta Crisafulli
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, 98100 Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Comet assay in evaluating DNA damage associated with ischaemia-reperfusion injury in patients undergoing coronary surgery. Arh Hig Rada Toksikol 2009; 60:307-15. [PMID: 19789160 DOI: 10.2478/10004-1254-60-2009-1934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ischaemia-reperfusion (I/R) injury is responsible for a number of conditions such as coronary bypass and myocardial infarction, and deaths. Oxygen-free radicals formed during I/R have been proposed as the leading causes of tissue injury, and they play an important role in I/R injury. I/R induces oxidative DNA damage (such as purinic and pyrimidinic base lesions). Comet assay is a suitable and sensitive method for early detection of low-level DNA damage. We used modified alkaline comet assay in peripheral blood lymphocytes and evaluated I/R-induced DNA damage in patients undergoing coronary artery bypass graft (CABG) operation (in vivo model for I/R). No statistically significant difference in DNA damage levels was found before surgery, after anaesthesia, ischemia, reperfusion, and surgery. However, blood lactate levels (assessed in parallel with the comet assay) increased after I/R and did not return to the baseline level. Our findings showed that I/R injury did not induce DNA damage, but increased the lactate levels. This finding suggests that there might be reversible and uncommon necrosis that did not reflect on overall DNA base damage. Further studies are needed using this approach.
Collapse
|
25
|
Martínez-Romero R, Cañuelo A, Martínez-Lara E, Javier Oliver F, Cárdenas S, Siles E. Poly(ADP-ribose) polymerase-1 modulation of in vivo response of brain hypoxia-inducible factor-1 to hypoxia/reoxygenation is mediated by nitric oxide and factor inhibiting HIF. J Neurochem 2009; 111:150-9. [PMID: 19656264 DOI: 10.1111/j.1471-4159.2009.06307.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear protein that once activated by genotoxic agents, modulates its own activity and that of several other nuclear proteins. The absence or pharmacological inhibition of this protein has been proven to be beneficial in the treatment of different diseases involving a hypoxic situation. We previously reported that PARP-1 modulates the hypoxia-inducible factor-1 (HIF-1) response in vitro, but this effect has not yet been demonstrated in vivo. The brain is especially susceptible to hypoxic injury, and the present study demonstrates that PARP-1 plays a major role in the post-hypoxic response of HIF-1alpha in the cerebral cortex. Immediate post-hypoxic HIF-1alpha accumulation was higher in the presence of PARP-1, and this differential response was mediated by nitric oxide and to a lesser extent, reactive oxygen species. PARP-1 was also found to induce a more rapid but less sustained HIF-1 transcriptional activity by up-regulating the factor inhibiting HIF. The implication of PARP-1 in these results was further demonstrated by pharmacologically inhibiting PARP in wild-type mice. In conclusion, our data suggest that PARP-1 has an important regulatory role in the in vivo response of brain HIF-1 to hypoxia/reoxygenation.
Collapse
Affiliation(s)
- Rubén Martínez-Romero
- Department of Experimental Biology. University of Jaén Paraje Las Lagunillas s/n, Jaén, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Peralta-Leal A, Rodríguez-Vargas JM, Aguilar-Quesada R, Rodríguez MI, Linares JL, de Almodóvar MR, Oliver FJ. PARP inhibitors: new partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med 2009; 47:13-26. [PMID: 19362586 DOI: 10.1016/j.freeradbiomed.2009.04.008] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 04/07/2009] [Accepted: 04/08/2009] [Indexed: 12/21/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are defined as cell signaling enzymes that catalyze the transfer of ADP-ribose units from NAD(+) to a number of acceptor proteins. PARP-1, the best characterized member of the PARP family, which currently comprises 18 members, is an abundant nuclear enzyme implicated in cellular responses to DNA injury provoked by genotoxic stress. PARP is involved in DNA repair and transcriptional regulation and is now recognized as a key regulator of cell survival and cell death as well as a master component of a number of transcription factors involved in tumor development and inflammation. PARP-1 is essential to the repair of DNA single-strand breaks via the base excision repair pathway. Inhibitors of PARP-1 have been shown to enhance the cytotoxic effects of ionizing radiation and DNA-damaging chemotherapy agents, such as the methylating agents and topoisomerase I inhibitors. There are currently at least five PARP inhibitors in clinical trial development. Recent in vitro and in vivo evidence suggests that PARP inhibitors could be used not only as chemo/radiotherapy sensitizers, but also as single agents to selectively kill cancers defective in DNA repair, specifically cancers with mutations in the breast cancer-associated genes (BRCA1 and BRCA2). PARP becomes activated in response to oxidative DNA damage and depletes cellular energy pools, thus leading to cellular dysfunction in various tissues. The activation of PARP may also induce various cell death processes and promotes an inflammatory response associated with multiple organ failure. Inhibition of PARP activity is protective in a wide range of inflammatory and ischemia-reperfusion-associated diseases, including cardiovascular diseases, diabetes, rheumatoid arthritis, endotoxic shock, and stroke. The aim of this review is to overview the emerging data in the literature showing the role of PARP in the pathogenesis of cancer and inflammatory diseases and unravel the solid body of literature that supports the view that PARP is an important target for therapeutic intervention in critical illness.
Collapse
Affiliation(s)
- Andreína Peralta-Leal
- Instituto de Parasitología y Biomedicina López Neyra, Consejo Superior de Investigaciones Cientificas (CSIC), Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Moroni F, Chiarugi A. Post-ischemic brain damage: targeting PARP-1 within the ischemic neurovascular units as a realistic avenue to stroke treatment. FEBS J 2008; 276:36-45. [DOI: 10.1111/j.1742-4658.2008.06768.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Teupolioside, a phenylpropanoid glycosides of Ajuga reptans, biotechnologically produced by IRBN22 plant cell line, exerts beneficial effects on a rodent model of colitis. Biochem Pharmacol 2008; 77:845-57. [PMID: 19070602 DOI: 10.1016/j.bcp.2008.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 10/30/2008] [Accepted: 11/05/2008] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to examine the effects of phenylpropanoid glycoside, teupolioside, biotechnologically produced by IRBN22 Ajuga reptans cell line, in rats subjected to experimental colitis. Colitis was induced in rats by intracolonic instillation of dinitrobenzene sulfonic acid (DNBS). Teupolioside was administered daily orally (0.2 or 2mgkg(-1)). On Day 4, animals were sacrificed and tissues were taken for histological and biochemical analysis. Four days after DNBS administration, colon TNF-alpha and IL-1beta productions were increased, associated with colon damage. Neutrophil infiltration, by myeloperoxidase activity, in the mucosa was associated with up-regulation of ICAM-1 and P-selectin and high levels of malondialdehyde. Immunohistochemistry for nitrotyrosine and poly (ADP-ribose) polymerase (PARP) showed an intense staining in the inflamed colon. Biochemical methods and zymography were used to analyze MMP-9 and -2 activities in colon tissues from DNBS-injured rats. Treatment with teupolioside significantly reduced the appearance of diarrhoea and the loss of body weight. This was associated with a remarkable amelioration in the disruption of the colonic architecture and a significant reduction in colonic myeloperoxidase activity and malondialdehyde levels. Teupolioside also reduced the pro-inflammatory cytokines release, the appearance of nitrotyrosine and PARP immunoreactivity in the colon and reduced the up-regulation of ICAM-1 and the expression of P-selectin. Therefore, teupolioside also reduced proMMP-9 and -2 activity induced in the colon by DNBS administration. The results of this study suggested that administration of teupolioside may be beneficial for treatment of inflammatory bowel disease.
Collapse
|
29
|
Protective effects of glycyrrhizin in a gut hypoxia (ischemia)-reoxygenation (reperfusion) model. Intensive Care Med 2008; 35:687-97. [PMID: 18953525 DOI: 10.1007/s00134-008-1334-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 10/04/2008] [Indexed: 11/27/2022]
|
30
|
Keretetse GS, Laubscher PJ, Du Plessis JL, Pretorius PJ, Van Der Westhuizen FH, Van Deventer E, Van Dyk E, Eloff FC, Van Aarde MN, Du Plessis LH. DNA damage and repair detected by the comet assay in lymphocytes of african petrol attendants: a pilot study. ACTA ACUST UNITED AC 2008; 52:653-62. [PMID: 18664513 DOI: 10.1093/annhyg/men047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Petrol attendants are exposed to petrol volatile organic compounds (VOCs) which may have genotoxic and carcinogenic effects. The single-cell gel electrophoresis assay (comet assay) is a method highly sensitive to DNA damage induced by environmental and occupational exposure to carcinogenic and mutagenic agents. The aim of this study was to evaluate the level of exposure of petrol attendants to petrol VOCs and also to determine their effect on DNA damage and repair in lymphocytes of African petrol attendants. The exposed group consisted of 20 subjects, randomly selected from three petrol stations. A control group of 20 unexposed subjects was also chosen and matched for age and smoking habits with the exposed group. Sorbent tubes were used to assess personal exposure of petrol attendants. The comet assay was used to investigate the basal DNA damage and repair capacity in isolated lymphocytes of petrol attendants and unexposed subjects. Blood samples were taken from the petrol attendants at the end of their 8-h working shift and also from the unexposed subjects. The petrol attendants were found to be exposed to levels of petrol VOCs lower than the South African occupational exposure limit for constituent chemicals. A significant relationship was found between the volume of petrol sold during the shift and the average concentrations of benzene, toluene and the total VOCs measured. However, relative humidity had a negative correlation with the average concentrations of benzene, toluene, xylene and the total VOCs. Significantly higher basal DNA damage was observed with the exposed group compared to the unexposed group. The period of exposure influenced the level of DNA damage and the calculated repair capacity. Smoking and age had a significant influence on the level of DNA damage. DNA repair capacity was delayed in smokers of both exposed and unexposed group.
Collapse
Affiliation(s)
- G S Keretetse
- Subject-Group Physiology, Faculty of Health Sciences, North-West University, Potchefstroom Campus, Private Bag X6001, Potchefstroom 2520, South Africa
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhao Y, Li W, Chow PC, Lau DT, Lee NT, Pang Y, Zhang X, Wang X, Han Y. Bis(7)-tacrine, a promising anti-Alzheimer's dimer, affords dose- and time-dependent neuroprotection against transient focal cerebral ischemia. Neurosci Lett 2008; 439:160-4. [DOI: 10.1016/j.neulet.2008.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 03/05/2008] [Accepted: 05/05/2008] [Indexed: 11/27/2022]
|
32
|
Hertz L. Bioenergetics of cerebral ischemia: a cellular perspective. Neuropharmacology 2008; 55:289-309. [PMID: 18639906 DOI: 10.1016/j.neuropharm.2008.05.023] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 05/14/2008] [Accepted: 05/14/2008] [Indexed: 12/27/2022]
Abstract
In cerebral ischemia survival of neurons, astrocytes, oligodendrocytes and endothelial cells is threatened during energy deprivation and/or following re-supply of oxygen and glucose. After a brief summary of characteristics of different cells types, emphasizing the dependence of all on oxidative metabolism, the bioenergetics of focal and global ischemia is discussed, distinguishing between events during energy deprivation and subsequent recovery attempt after re-circulation. Gray and white matter ischemia are described separately, and distinctions are made between mature and immature brains. Next comes a description of bioenergetics in individual cell types in culture during oxygen/glucose deprivation or exposure to metabolic inhibitors and following re-establishment of normal aerated conditions. Due to their expression of NMDA and non-NMDA receptors neurons and oligodendrocytes are exquisitely sensitive to excitotoxicity by glutamate, which reaches high extracellular concentrations in ischemic brain for several reasons, including failing astrocytic uptake. Excitotoxicity kills brain cells by energetic exhaustion (due to Na(+) extrusion after channel-mediated entry) combined with mitochondrial Ca(2+)-mediated injury and formation of reactive oxygen species. Many (but not all) astrocytes survive energy deprivation for extended periods, but after return to aerated conditions they are vulnerable to mitochondrial damage by cytoplasmic/mitochondrial Ca(2+) overload and to NAD(+) deficiency. Ca(2+) overload is established by reversal of Na(+)/Ca(2+) exchangers following Na(+) accumulation during Na(+)-K(+)-Cl(-) cotransporter stimulation or pH regulation, compensating for excessive acid production. NAD(+) deficiency inhibits glycolysis and eventually oxidative metabolism, secondary to poly(ADP-ribose)polymerase (PARP) activity following DNA damage. Hyperglycemia can be beneficial for neurons but increases astrocytic death due to enhanced acidosis.
Collapse
Affiliation(s)
- Leif Hertz
- College of Basic Medical Sciences, China Medical University, Shenyang, PR China.
| |
Collapse
|
33
|
Sivonová M, Kaplán P, Duracková Z, Dobrota D, Drgová A, Tatarková Z, Pavlíková M, Halasová E, Lehotský J. Time course of peripheral oxidative stress as consequence of global ischaemic brain injury in rats. Cell Mol Neurobiol 2008; 28:431-41. [PMID: 18058017 PMCID: PMC11515441 DOI: 10.1007/s10571-007-9246-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 11/17/2007] [Indexed: 10/22/2022]
Abstract
Free radicals play an important role in the pathogenesis of brain injury. This study evaluates the potential relationship between ischaemia/reperfusion (I/R)-induced brain injury, peripheral oxidative stress (lymphocyte DNA damage), plasma antioxidant potential and uric acid levels. We observed that 15 min of ischaemia were sufficient to significantly increase lymphocyte DNA damage that remained elevated at the end of early (3 h) reperfusion and at later (72 h) reperfusion time; this parameter was not significantly increased, when compared to preoperated levels. In parallel, antioxidant potential was elevated after 15 min of ischaemia, remained high at early (3 h) reperfusion and decreased again with longer (72 h) reperfusion. A close association between the plasma antioxidant status and the uric acid content has been confirmed by findings that changes in TRAP values positively correlate with uric acid concentration in rat plasma after ischaemic injury. Moreover, results of in vitro experiments with extra uric acid addition to control plasma have shown that uric acid contributes to a greater part of TRAP values. These results indicate a similar time course of brain I/R-associated oxidative stress and peripheral antioxidant defence status and/or oxidative stress in animal experiments.
Collapse
Affiliation(s)
- Monika Sivonová
- Jessenius Faculty of Medicine, Department of Medical Biochemistry, Comenius University, Malá hora 4, 036 01, Martin, Slovakia.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cuong DV, Warda M, Kim N, Park WS, Ko JH, Kim E, Han J. Dynamic changes in nitric oxide and mitochondrial oxidative stress with site-dependent differential tissue response during anoxic preconditioning in rat heart. Am J Physiol Heart Circ Physiol 2007; 293:H1457-65. [PMID: 17545474 DOI: 10.1152/ajpheart.01282.2006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this study, dynamic changes in nitric oxide (NO) and mitochondrial superoxide (O2•−) were examined during anoxic preconditioning (AP) in rat heart model. AP and anoxia-reoxygenation (A/R) were performed on isolated hearts and single cardiomyocytes. The cellular insult in the form of infarct size and DNA damage were localized and correlated with NO synthases (endothelial and inducible) expression levels. The results showed that endocardium was the most affected region in AP groups, whereas the larger area of infarct was confined to mid- and epicardium in the A/R group. Interestingly, a high-level expression of immunofluorescent NO synthases was restricted to viable areas in the AP. In contrast to the gradual increase in O2•− level that occurred in the AP group, a sudden massive increase in its level was demonstrated at the onset of reoxygenation in the A/R group. The observed increase in NO production during reoxygenation in the AP group was attenuated by inducible NO synthase inhibitor. The study revealed, on a real-time basis, the role played by preconditioning for modulating NO and O2•− levels on behalf of cell survival. The results afford a better understanding of cardiac-adapting mechanism during AP and the role of inducible NO synthase in this important phenomenon.
Collapse
Affiliation(s)
- Dang Van Cuong
- Mitochondrial Signaling Laboratory, Department of Physiology and Biophysics, College of Medicine, Mitochondrial Research Group-Frontier Inje Research Science and Technology Project, Inje University, Busanjin-Ku, Busan 614-735, Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Ranjan P, Heintz NH. S-phase arrest by reactive nitrogen species is bypassed by okadaic acid, an inhibitor of protein phosphatases PP1/PP2A. Free Radic Biol Med 2006; 40:247-59. [PMID: 16413407 DOI: 10.1016/j.freeradbiomed.2005.08.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 06/03/2005] [Accepted: 08/08/2005] [Indexed: 12/26/2022]
Abstract
In mammalian cells DNA damage activates a checkpoint that halts progression through S phase. To determine the ability of nitrating agents to induce S-phase arrest, mouse C10 cells synchronized in S phase were treated with nitrogen dioxide (NO(2)) or SIN-1, a generator of reactive nitrogen species (RNS). SIN-1 or NO(2) induced S-phase arrest in a dose- and time-dependent manner. As for the positive controls adozelesin and cisplatin, arrest was accompanied by phosphorylation of ATM kinase; dephosphorylation of pRB; decreases in RF-C, cyclin D1, Cdc25A, and Cdc6; and increases in p21. Comet assays indicated that RNS induce minimal DNA damage. Moreover, in a cell-free replication system, nuclei from cells treated with RNS were able to support control levels of DNA synthesis when incubated in cytosolic extracts from untreated cells, whereas nuclei from cells treated with cisplatin were not. Induction of phosphatase activity may represent one mechanism of RNS-induced arrest, for the PP1/PP2A phosphatase inhibitor okadaic acid inhibited dephosphorylation of pRB; prevented decreases in the levels of RF-C, cyclin D1, Cdc6, and Cdc25A; and bypassed arrest by SIN-1 or NO(2), but not cisplatin or adozelesin. Our studies suggest that RNS may induce S-phase arrest through mechanisms that differ from those elicited by classical DNA-damaging agents.
Collapse
Affiliation(s)
- Priya Ranjan
- Department of Pathology and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | |
Collapse
|
36
|
Cuong DV, Kim N, Youm JB, Joo H, Warda M, Lee JW, Park WS, Kim T, Kang S, Kim H, Han J. Nitric oxide-cGMP-protein kinase G signaling pathway induces anoxic preconditioning through activation of ATP-sensitive K+ channels in rat hearts. Am J Physiol Heart Circ Physiol 2005; 290:H1808-17. [PMID: 16339835 DOI: 10.1152/ajpheart.00772.2005] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Nitric oxide (NO) plays an important role in anoxic preconditioning to protect the heart against ischemia-reperfusion injuries. The present work was performed to study better the NO-cGMP-protein kinase G (PKG) signaling pathway in the activation of both sarcolemmal and mitochondrial ATP-sensitive K+ (KATP) channels during anoxic preconditioning (APC) and final influence on reducing anoxia-reperfusion (A/R)-induced cardiac damage in rat hearts. The upstream regulating elements controlling NO-cGMP-PKG signal-induced KATP channel opening that leads to cardioprotection were investigated. The involvement of both inducible and endothelial NO synthases (iNOS and eNOS) in the progression of this signaling pathway was followed. Final cellular outcomes of ischemia-induced injury after different preconditioning in the form of lactate dehydrogenase release, DNA strand breaks, and malondialdehyde formation as indexes of cell injury and lipid peroxidation, respectively, were investigated. The lactate dehydrogenase and malondialdehyde values decreased in the groups that underwent preconditioning periods with specific mitochondrial KATP channels opener diazoxide (100 microM), nonspecific mitochondrial KATP channels opener pinacidil (50 microM), S-nitroso-N-acetylpenicillamine (SNAP, 300 microM), or beta-phenyl-1,N2-etheno-8-bromoguanosine-3',5'-cyclicmonophosphorothioate, Sp-isomer (10 microM) before the A/R period. Preconditioning with SNAP significantly reduced the DNA damage. The effect was blocked by glibenclamide (50 microM), 5-hydroxydecanoate (100 microM), NG-nitro-L-arginine methyl ester (200 microM), and beta-phenyl-1,N2-etheno-8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rp-isomer (1 microM). The results suggest iNOS, rather than eNOS, as the major contributing NO synthase during APC treatment. Moreover, the PKG shows priority over NO as the upstream regulator of NO-cGMP-PKG signal-induced KATP channel opening that leads to cardioprotection during APC treatment.
Collapse
Affiliation(s)
- Dang Van Cuong
- Mitochondrial Signaling Laboratory, Department of Physiology and Biophysics, College of Medicine, Cardiovascular and Metabolic Disease Center, Biohealth Products Research Center, Inje University, Busan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
van Dyk E, Pretorius PJ. DNA damage and repair in mammalian cells exposed to p-hydroxyphenylpyruvic acid. Biochem Biophys Res Commun 2005; 338:815-9. [PMID: 16263080 DOI: 10.1016/j.bbrc.2005.10.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 10/19/2005] [Indexed: 11/26/2022]
Abstract
Tyrosinemia type 1 (HT1) is an autosomal recessive disorder of the tyrosine metabolism in which the fumarylacetoacetate hydrolase enzyme is defective. This disease is clinically heterogeneous and a chronic and acute form is discerned. Characteristic of the chronic form is the development of cellular hepatocarcinoma. Although p-hydroxyphenylpyruvic acid (pHPPA) is used as one of the diagnostic markers of this disease, it was suggested that it is unlikely to be involved in the pathophysiology of HT1 as it is present in other disorders that does not have hepatorenal symptoms. It was the aim of this study to investigate the possible effect of pHPPA on DNA damage and repair in mammalian cells. The comet assay was used to establish the genotoxicity of pHPPA in human peripheral blood lymphocytes and isolated rat hepatocytes after their exposure to pHPPA. At first glance the damage to DNA caused by pHPPA seemed reparable in both cell types, however, after challenging the DNA repair capacity of metabolite-treated cells with treatment with H(2)O(2), a marked impairment in the DNA repair capability of these cells was observed. We suggest that the main effect of pHPPA is the long-term impairment of the DNA repair machinery rather than the direct damage to DNA and that this effect of pHPPA, together with the other characteristic metabolites, e.g., FAA and MAA, causes cellular hepatocarcinoma to develop in the chronic form of HT1.
Collapse
Affiliation(s)
- Etresia van Dyk
- Division of Biochemistry, North-West University, Potchefstroom Campus, Potchefstroom 2520, South Africa
| | | |
Collapse
|
38
|
Pellicciari R, Camaioni E, Costantino G. 3. Life or death decisions: the cast of poly(ADP-ribose)polymerase (PARP) as a therapeutic target for brain ischaemia. PROGRESS IN MEDICINAL CHEMISTRY 2005; 42:125-69. [PMID: 15003720 DOI: 10.1016/s0079-6468(04)42003-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Affiliation(s)
- Roberto Pellicciari
- Dipartimento di Chimica e Tecnologia del Farmaco, Via del Liceo 1, 06123 Perugia, Italy
| | | | | |
Collapse
|
39
|
Abstract
Over the past decade, poly(ADP-ribosyl)ation has emerged as a crucial event in the pathogenesis of ischemic stroke. A large body of evidence unambiguously demonstrates that activity of poly(ADP-ribose) polymerase-1 (PARP-1) significantly increases during brain ischemia, and that inhibition of this enzymatic activity affords substantial neuroprotection from ischemic brain injury. This review strictly focuses on literature on poly(ADP-ribosyl)ation and ischemic stroke, highlighting the pathogenetic role of poly(ADP-ribose) in ischemic neuronal death, and the therapeutic relevance of drugs modulating its metabolism to pharmacological treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Alberto Chiarugi
- Department of Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
40
|
Mocali A, Giovannelli L, Dolara P, Paoletti F. The Comet Assay Approach to Senescent Human Diploid Fibroblasts Identifies Different Phenotypes and Clarifies Relationships Among Nuclear Size, DNA Content, and DNA Damage. J Gerontol A Biol Sci Med Sci 2005; 60:695-701. [PMID: 15983170 DOI: 10.1093/gerona/60.6.695] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The comet assay methodology was used to monitor nuclear changes occurring in MRC5 human fibroblasts during transition from young to senescent cultures and to study heterogeneity of senescent populations. Nuclear morphology and size, DNA content per nucleus, and DNA damage (basal strand break, total damage, and oxidized base levels) were evaluated; moreover, visually identified large and small nuclei were analyzed separately and arranged in classes of increasing DNA damage. Oxidized base levels were definitely lower in young versus senescent fibroblasts of which, however, a significant proportion showed negligible DNA damage. Nuclear size enlargement accompanying senescence was almost equally influenced by cell ploidy increase and also by a chromatin decondensation process involving diploid cells. It is noteworthy that DNA damage in senescent fibroblasts correlated significantly to nuclear size, but not to DNA content. The comet assay allowed us to identify different senescent phenotypes and to investigate changes in nuclear features and/or DNA damage irrespective of time elapsed in culture.
Collapse
Affiliation(s)
- Alessandra Mocali
- Department of Experimental Pathology and Oncology, Viale G.B. Morgagni 50, 50134 Firenze, Italy.
| | | | | | | |
Collapse
|
41
|
Warner DS, Sheng H, Batinić-Haberle I. Oxidants, antioxidants and the ischemic brain. ACTA ACUST UNITED AC 2004; 207:3221-31. [PMID: 15299043 DOI: 10.1242/jeb.01022] [Citation(s) in RCA: 415] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite numerous defenses, the brain is vulnerable to oxidative stress resulting from ischemia/reperfusion. Excitotoxic stimulation of superoxide and nitric oxide production leads to formation of highly reactive products, including peroxynitrite and hydroxyl radical, which are capable of damaging lipids, proteins and DNA. Use of transgenic mutants and selective pharmacological antioxidants has greatly increased understanding of the complex interplay between substrate deprivation and ischemic outcome. Recent evidence that reactive oxygen/nitrogen species play a critical role in initiation of apoptosis, mitochondrial permeability transition and poly(ADP-ribose) polymerase activation provides additional mechanisms for oxidative damage and new targets for post-ischemic therapeutic intervention. Because oxidative stress involves multiple post-ischemic cascades leading to cell death, effective prevention/treatment of ischemic brain injury is likely to require intervention at multiple effect sites.
Collapse
Affiliation(s)
- David S Warner
- Department of Anesthesiology, The Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
42
|
Kamanaka Y, Kondo K, Ikeda Y, Kamoshima W, Kitajima T, Suzuki Y, Nakamura Y, Umemura K. Neuroprotective effects of ONO-1924H, an inhibitor of poly ADP-ribose polymerase (PARP), on cytotoxicity of PC12 cells and ischemic cerebral damage. Life Sci 2004; 76:151-62. [PMID: 15519361 DOI: 10.1016/j.lfs.2004.04.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Accepted: 04/09/2004] [Indexed: 10/26/2022]
Abstract
N-[3-(4-Oxo-3,4-dihydro-phthalazin-1-yl)phenyl]-4-(morpholin-4-yl) butanamide methanesulfonate monohydrate (ONO-1924H) is a novel inhibitor of poly ADP-ribose polymerase (PARP). In this study, we examined the effects of ONO-1924H on cytotoxicity induced by hydrogen peroxide in PC12 cells in vitro and cerebral damage and neurological deficits induced by middle cerebral artery (MCA) thrombus occlusion in vivo in rat. In the in vitro cytotoxicity assay, exposure to 0.5 mmol/L hydrogen peroxide induced cell death in differentiated PC12 cells. ONO-1924H, a PARP inhibitor (Ki=0.21 micromol/L), reduced cell death in a concentration-dependent manner that was correlated with inhibition of PARP activation. A 50% reduction in cell death (EC50) was achieved with 2.4 micromol/L ONO-1924H. In the MCA occlusion model, ONO-1924H was injected intravenously at doses of 3, 10 and 30 mg/kg/h for 3 h, and cerebral damage and neurological deficits were estimated 24 h after MCA occlusion. ONO-1924H treatment led to a significant decrease in cerebral damage in the 10 mg/kg/h-treated group (P < 0.05) and the 30 mg/kg/h-treated group (P < 0.01). Further, ONO-1924H at doses of 30 mg/kg/h significantly (P < 0.05) improved neurological deficits. These findings suggest that the novel PARP inhibitor, ONO-1924H, affords effective neuroprotection and is a useful therapeutic candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yoshihisa Kamanaka
- Minase Research Institute, ONO Pharmaceutical Co. Ltd., 3-1-1, Sakurai,Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cemeli E, Smith IF, Peers C, Urenjak J, Godukhin OV, Obrenovitch TP, Anderson D. Oxygen-induced DNA damage in freshly isolated brain cells compared with cultured astrocytes in the Comet assay. ACTA ACUST UNITED AC 2003; Suppl 2:43-52. [PMID: 14691979 DOI: 10.1002/tcm.10079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Brain cells are continuously exposed to reactive oxygen species generated by oxidative metabolism, and in certain pathological conditions defence mechanisms against oxygen radicals may be weakened and/or overwhelmed. DNA is a potential target for oxidative damage, and genomic damage can contribute to neuropathogenesis. It is important, therefore, to identify tools for the quantitative analysis of DNA damage in models of neurological disorders. The aim of this study was to compare the susceptibility of DNA to oxidative stress in cells freshly dissociated from the mouse brain, to that in cultured brain cells. Both primary cultures and a continuous cell line of astrocytes were considered. All cells were treated by xanthine/xanthine oxidase, a superoxide generator or hydrogen peroxide, applied alone or in the presence of the oxygen radical scavengers, superoxide dismutase, catalase, or ascorbic acid. DNA damage, quantified with the Comet assay, was consistent in all the different cell preparations exposed to oxidative stress, and was attenuated in similar ways by superoxide dismutase and catalase, scavengers of superoxide anion and hydrogen peroxide, respectively. The results with ascorbic acid were more variable, presumably because this compound may switch from anti- to pro-oxidant status depending on its concentration and other experimental conditions. Overall, similar responses were found in freshly dissociated and cultured brain cells. These results suggest that the Comet assay can be directly applied to cells freshly dissociated from the brain of rodents, including models of neurological disorders, such as stroke models and animals with targeted mutations that mimic human diseases.
Collapse
Affiliation(s)
- Eduardo Cemeli
- Department of Biomedical Sciences, University of Bradford, Bradford, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
44
|
Yoshinaga H, Watanabe M, Manome Y. Possible role of nicaraven in neuroprotective effect on hippocampal slice culture. Can J Physiol Pharmacol 2003; 81:683-9. [PMID: 12897815 DOI: 10.1139/y03-060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicaraven is an agent that is especially beneficial in vasospasm or brain damage caused by subarachnoid hemorrhage. It ameliorates neurological deficits of patients and protects the central nervous system from ischemia. We investigated the neuroprotective effect of nicaraven against oxygen-glucose deprivation (OGD) induced or N-methyl-D-aspartic acid (NMDA) induced hippocampal neuronal cell death in organotypic brain slice cultures. The effect of nicaraven on hippocampal neuronal injury was evaluated by inhibition of uptake of propidium iodide (PI) into dead cells. The results demonstrated that nicaraven protected neuronal cells from both OGD- and NMDA-induced cell death. While nicaraven has a strong hydroxyl radical scavenging effect, another radical scavenger, N-acetyl-L-cysteine (NAC), inhibited cell death only caused by OGD. In contrast, the poly(ADP-ribose) synthetase (PARS) inhibitors 3-aminobenzamide (3-AB) and theophylline protected cells from both OGD- and NMDA-induced cell death. Since nicaraven has an inhibitory effect in PARS, as well as a radical scavenging effect, these results suggest that inhibition of hippocampal cell death caused by NMDA may be attributable to PARS inhibition by nicaraven.
Collapse
Affiliation(s)
- Hisao Yoshinaga
- Department of Microbiology, Jikei University School of Medicine, 3-25-8 Nishishinbashi, Minato-ku, Tokyo, Japan 105-8461
| | | | | |
Collapse
|
45
|
Giovannelli L, Decorosi F, Dolara P, Pulvirenti L. Vulnerability to DNA damage in the aging rat substantia nigra: a study with the comet assay. Brain Res 2003; 969:244-7. [PMID: 12676385 DOI: 10.1016/s0006-8993(03)02275-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Oxidative DNA damage was measured in the substantia nigra (SN), cortex, hippocampus, striatum and hypothalamus of 3- and 24-month-old rats, using single-cell gel electrophoresis (SCGE, 'comet' assay) which allows the detection of DNA breaks and oxidized bases. A significant increase in basal DNA damage was selectively found in the SN of aged rats. FPG-sensitive oxidative DNA damage was also significantly increased in the SN of aged rats and, to a lesser extent, in the cortex and hypothalamus. These data show a higher vulnerability of SN to oxidative damage with aging and indicate that the detection of DNA damage within discrete brain nuclei can provide a reliable tool for investigating oxidative damage in neurodegenerative processes.
Collapse
Affiliation(s)
- Lisa Giovannelli
- Department of Pharmacology, University of Florence, Florence, Italy
| | | | | | | |
Collapse
|