1
|
Uztimür M, Dörtbudak MB. Evaluation of brain injury in goats naturally infected with Coenurus cerebralis; brain specific biomarkers, acute inflammation, and DNA oxidation. Res Vet Sci 2023; 165:105043. [PMID: 37856943 DOI: 10.1016/j.rvsc.2023.105043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/14/2023] [Accepted: 10/01/2023] [Indexed: 10/21/2023]
Abstract
This investigate goals are to establish the utility of brain-specific biomarkers (GFAP and S100B) in vivo and to assess the brain damage in C. cerebralis-infected goats using histopathological and immunopathological methods. The animal material of the study consisted of 10 healthy and 20 Coenurus cerebralis infected female hair goats. Serum GFAP and S100B concentrations were measured to determine brain damage. Serum S100B (p < 0.037), GFAP (p < 0.012), urea (p < 0.045), GGT (p < 0.001) and ALT (p < 0.001) concentrations in the C.cerebralis group were significantly higher than the control group. There was no significant difference between the C.cerebralis group and the control group for hsTnI (p > 0.078), creatinine (p > 0.099) and CK-MB (p > 0.725). In the histopathological examination, pressure atrophy and related inflammatory changes were observed due to mechanical damage of the parasite. Immunohistochemical examinations revealed that the parasite stimulated inflammation with the expression of TNF-α and caused DNA damage with the expression of 8-OHdG. As a result, when the data collected for this study are assessed as a whole, it is thought that the use of brainspecific GFAP and S100B biomarkers may be beneficial in determining brain damage in naturally infected hair goats with C.cerebralis. Changes in the levels of brain-specific biomarkers contribute significantly to determining the prognosis of the disease in vivo. Measurement of GFAP and S100B concentrations from serum offers an important alternative to the CSF method.
Collapse
Affiliation(s)
- Murat Uztimür
- Bingöl University, Faculty of Veterinary Medicine, Department of Internal Medicine, Bingöl, Türkiye.
| | | |
Collapse
|
2
|
Lee LY, Chou W, Chen WP, Wang MF, Chen YJ, Chen CC, Tung KC. Erinacine A-Enriched Hericium erinaceus Mycelium Delays Progression of Age-Related Cognitive Decline in Senescence Accelerated Mouse Prone 8 (SAMP8) Mice. Nutrients 2021; 13:nu13103659. [PMID: 34684662 PMCID: PMC8537498 DOI: 10.3390/nu13103659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/20/2023] Open
Abstract
There have been many reports on the neuroprotective effects of Hericium erinaceus mycelium, in which the most well-known active compounds found are diterpenoids, such as erinacine A. Previously, erinacine A-enriched Hericeum erinaceus mycelium (EAHEM) was shown to decrease amyloid plaque aggregation and improve cognitive disability in Alzheimer’s disease model APP/PS1 mice. However, its effects on brain aging have not yet been touched upon. Here, we used senescence accelerated mouse prone 8 (SAMP8) mice as a model to elucidate the mechanism by which EAHEM delays the aging of the brain. Three-month-old SAMP8 mice were divided into three EAHEM dosage groups, administered at 108, 215 and 431 mg/kg/BW/day, respectively. During the 12th week of EAHEM feeding, learning and memory of the mice were evaluated by single-trial passive avoidance and active avoidance test. After sacrifice, the amyloid plaques, induced nitric oxidase synthase (iNOS) activity, thiobarbituric acid-reactive substances (TBARS) and 8-OHdG levels were analyzed. We found that the lowest dose of 108 mg/kg/BW EAHEM was sufficient to significantly improve learning and memory in the passive and active avoidance tests. In all three EAHEM dose groups, iNOS, TBARS and 8-OHdG levels all decreased significantly and showed a dose-dependent response. The results indicate that EAHEM improved learning and memory and delayed degenerative aging in mice brains.
Collapse
Affiliation(s)
- Li-Ya Lee
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402204, Taiwan;
| | - Wayne Chou
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (W.C.); (W.-P.C.)
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (W.C.); (W.-P.C.)
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung 433303, Taiwan;
| | - Ying-Ju Chen
- College of Humanities & Social Sciences, Providence University, Taichung 433303, Taiwan;
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 106319, Taiwan;
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei 104336, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Kwong-Chung Tung
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402204, Taiwan;
- Correspondence:
| |
Collapse
|
3
|
Jung HY, Kim W, Hahn KR, Kang MS, Kim TH, Kwon HJ, Nam SM, Chung JY, Choi JH, Yoon YS, Kim DW, Yoo DY, Hwang IK. Pyridoxine Deficiency Exacerbates Neuronal Damage after Ischemia by Increasing Oxidative Stress and Reduces Proliferating Cells and Neuroblasts in the Gerbil Hippocampus. Int J Mol Sci 2020; 21:ijms21155551. [PMID: 32759679 PMCID: PMC7432354 DOI: 10.3390/ijms21155551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 01/26/2023] Open
Abstract
We investigated the effects of pyridoxine deficiency on ischemic neuronal death in the hippocampus of gerbil (n = 5 per group). Serum pyridoxal 5′-phosphate levels were significantly decreased in Pyridoxine-deficient diet (PDD)-fed gerbils, while homocysteine levels were significantly increased in sham- and ischemia-operated gerbils. PDD-fed gerbil showed a reduction in neuronal nuclei (NeuN)-immunoreactive neurons in the medial part of the hippocampal CA1 region three days after. Reactive astrocytosis and microgliosis were found in PDD-fed gerbils, and transient ischemia caused the aggregation of activated microglia in the stratum pyramidale three days after ischemia. Lipid peroxidation was prominently increased in the hippocampus and was significantly higher in PDD-fed gerbils than in Control diet (CD)-fed gerbils after ischemia. In contrast, pyridoxine deficiency decreased the proliferating cells and neuroblasts in the dentate gyrus in sham- and ischemia-operated gerbils. Nuclear factor erythroid-2-related factor 2 (Nrf2) and brain-derived neurotrophic factor (BDNF) levels also significantly decreased in PDD-fed gerbils sham 24 h after ischemia. These results suggest that pyridoxine deficiency accelerates neuronal death by increasing serum homocysteine levels and lipid peroxidation, and by decreasing Nrf2 levels in the hippocampus. Additionally, it reduces the regenerated potentials in hippocampus by decreasing BDNF levels. Collectively, pyridoxine is an essential element in modulating cell death and hippocampal neurogenesis after ischemia.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Min Soo Kang
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea; (M.S.K.); (T.H.K.); (J.H.C.)
| | - Tae Hyeong Kim
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea; (M.S.K.); (T.H.K.); (J.H.C.)
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul 05030, Korea;
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea; (M.S.K.); (T.H.K.); (J.H.C.)
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
- Correspondence: (D.Y.Y.); (I.K.H.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (K.R.H.); (Y.S.Y.)
- Correspondence: (D.Y.Y.); (I.K.H.)
| |
Collapse
|
4
|
Cuprizone Affects Hypothermia-Induced Neuroprotection and Enhanced Neuroblast Differentiation in the Gerbil Hippocampus after Ischemia. Cells 2020; 9:cells9061438. [PMID: 32531881 PMCID: PMC7349804 DOI: 10.3390/cells9061438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
In the present study, we investigated the effects of cuprizone on cell death, glial activation, and neuronal plasticity induced by hypothermia after ischemia in gerbils. Food was supplemented with cuprizone at 0.2% ad libitum for eight weeks. At six weeks after diet feeing, gerbils received transient forebrain ischemia with or without hypothermic preconditioning. Cuprizone treatment for 8 weeks increased the number of astrocytes, microglia, and pro-inflammatory cytokine levels in the hippocampus. In addition, cuprizone treatment significantly decreased the number of proliferating cells and neuroblasts in the dentate gyrus. Brain ischemia caused cell death, disruption of myelin basic proteins, and reactive gliosis in CA1. In addition, ischemia significantly increased pro-inflammatory cytokines and the number of proliferating cells and differentiating neuroblasts in the dentate gyrus. In contrast, hypothermic conditioning attenuated these changes in CA1 and the dentate gyrus. However, cuprizone treatment decreased cell survival induced by hypothermic preconditioning after ischemia and increased the number of reactive microglia and astrocytes in CA1 as well as that of macrophages in the subcallosal zone. These changes occurred because the protective effect of hypothermia in ischemic damage was disrupted by cuprizone administration. Furthermore, cuprizone decreased ischemia-induced proliferating cells and neuroblasts in the dentate gyrus.
Collapse
|
5
|
Cho SB, Eum WS, Shin MJ, Kwon HJ, Park JH, Choi YJ, Park J, Han KH, Kang JH, Kim DS, Cho SW, Kim DW, Choi SY. Transduced Tat-aldose Reductase Protects Hippocampal Neuronal Cells against Oxidative Stress-induced Damage. Exp Neurobiol 2019; 28:612-627. [PMID: 31698553 PMCID: PMC6844837 DOI: 10.5607/en.2019.28.5.612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/06/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Aldose reductase (AR) protein, a member of the NADPH-dependent aldo-keto reductase family, reduces a wide range of aldehydes and enhances cell survival by inhibition of oxidative stress. Oxidative stress is known as one of the major pathological factor in ischemia. Since the precise function of AR protein in ischemic injury is fully unclear, we examined the function of AR protein in hippocampal neuronal (HT-22) cells and in an animal model of ischemia in this study. Cell permeable Tat-AR protein was produced by fusion of protein transduction domain in Tat for delivery into the cells. Tat-AR protein transduced into HT-22 cells and significantly inhibited cell death and regulated the mitogen-activate protein kinases (MAPKs), Bcl-2, Bax, and Caspase-3 under oxidative stress condition. In an ischemic animal model, Tat-AR protein transduced into the brain tissues through the blood-brain barrier (BBB) and drastically decreased neuronal cell death in hippocampal CA1 region. These results indicate that transduced Tat-AR protein has protective effects against oxidative stress-induced neuronal cell death in vitro and in vivo, suggesting that Tat-AR protein could be used as potential therapeutic agent in ischemic injury.
Collapse
Affiliation(s)
- Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Ju Hyeon Kang
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31538, Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31538, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
6
|
Risperidone Treatment after Transient Ischemia Induces Hypothermia and Provides Neuroprotection in the Gerbil Hippocampus by Decreasing Oxidative Stress. Int J Mol Sci 2019; 20:ijms20184621. [PMID: 31540405 PMCID: PMC6770640 DOI: 10.3390/ijms20184621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/06/2019] [Accepted: 09/15/2019] [Indexed: 12/11/2022] Open
Abstract
Compelling evidence from preclinical and clinical studies has shown that mild hypothermia is neuroprotective against ischemic stroke. We investigated the neuroprotective effect of post-risperidone (RIS) treatment against transient ischemic injury and its mechanisms in the gerbil brain. Transient ischemia (TI) was induced in the telencephalon by bilateral common carotid artery occlusion (BCCAO) for 5 min under normothermic condition (37 ± 0.2 °C). Treatment of RIS induced hypothermia until 12 h after TI in the TI-induced animals under uncontrolled body temperature (UBT) compared to that under controlled body temperature (CBT) (about 37 °C). Neuroprotective effect was statistically significant when we used 5 and 10 mg/kg doses (p < 0.05, respectively). In the RIS-treated TI group, many CA1 pyramidal neurons of the hippocampus survived under UBT compared to those under CBT. In this group under UBT, post-treatment with RIS to TI-induced animals markedly attenuated the activation of glial cells, an increase of oxidative stress markers [dihydroethidium, 8-hydroxy-2' -deoxyguanosine (8-OHdG), and 4-Hydroxynonenal (4-HNE)], and a decrease of superoxide dismutase 2 (SOD2) in their CA1 pyramidal neurons. Furthermore, RIS-induced hypothermia was significantly interrupted by NBOH-2C-CN hydrochloride (a selective 5-HT2A receptor agonist), but not bromocriptine mesylate (a D2 receptor agonist). Our findings indicate that RIS-induced hypothermia can effectively protect neuronal cell death from TI injury through attenuation of glial activation and maintenance of antioxidants, showing that 5-HT2A receptor is involved in RIS-induced hypothermia. Therefore, RIS could be introduced to reduce body temperature rapidly and might be applied to patients for hypothermic therapy following ischemic stroke.
Collapse
|
7
|
Jia L, Chen Y, Tian YH, Zhang G. MAPK pathway mediates the anti-oxidative effect of chicoric acid against cerebral ischemia-reperfusion injury in vivo. Exp Ther Med 2018; 15:1640-1646. [PMID: 29434748 PMCID: PMC5776621 DOI: 10.3892/etm.2017.5598] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 09/14/2017] [Indexed: 01/18/2023] Open
Abstract
The aim of the present study was to investigate the protective effect of chicoric acid on oxidative stress and inflammation in rats with cerebral ischemia-reperfusion injury. A cerebral ischemia-reperfusion injury rat model was created via transient middle cerebral artery occlusion (MCAO) and rats were treated with various doses of chicoric acid (0, 1, 10 and 100 mg/kg). Neurological deficits and infarct volume were used to estimate the protective effects of chicoric acid treatment. Levels of reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, nitric oxide (NO) and prostaglandin E2 (PGE2) were assessed. Western blot analysis was also used to measure the expression of cyclooxygenase (COX)-2, p38-mitogen activated protein kinase (MAPK), c-Jun, phosphorylated protein kinase B (p-AKT) and AKT. Chicoric acid exposure was observed to reduce neurological deficits and infarct volume in rats with cerebral ischemia-reperfusion injury. In addition, ROS production and inflammation were significantly suppressed following treatment with chicoric acid. Chicoric acid was demonstrated to significantly inhibit the upregulation of NO and PGE2 levels in rats following MCAO. Furthermore, chicoric acid significantly suppressed the MCAO-induced promotion of COX-2, p38-MAPK and c-Jun protein expression and enhanced the inhibition of p-AKT/AKT. These results suggest that chicoric acid has a protective effect, preventing oxidative stress and inflammation in rats with cerebral ischemia-reperfusion injury via the p38-MAPK, c-Jun and AKT signaling pathways.
Collapse
Affiliation(s)
- Linwei Jia
- Second Department of Neurosurgery, Hebei Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yonghan Chen
- Second Department of Neurosurgery, Hebei Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yao-Hui Tian
- Second Department of Neurosurgery, Hebei Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Gang Zhang
- Second Department of Neurosurgery, Hebei Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
8
|
Kim KH, Lee D, Lee HL, Kim CE, Jung K, Kang KS. Beneficial effects of Panax ginseng for the treatment and prevention of neurodegenerative diseases: past findings and future directions. J Ginseng Res 2017; 42:239-247. [PMID: 29989012 PMCID: PMC6035378 DOI: 10.1016/j.jgr.2017.03.011] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/15/2017] [Indexed: 01/20/2023] Open
Abstract
In recent years, several therapeutic drugs have been rationally designed and synthesized based on the novel knowledge gained from investigating the actions of biologically active chemicals derived from foods, plants, and medicinal herbs. One of the major advantages of these naturalistic chemicals is their ability to interact with multiple targets in the body resulting in a combined beneficial effect. Ginseng is a perennial herb (Araliaceae family), a species within the genus Panax, and a highly valued and popular medicinal plant. Evidence for the medicinal and health benefits of Panax ginseng and its components in preventing neurodegeneration has increased significantly in the past decade. The beneficial effects of P. ginseng on neurodegenerative diseases have been attributed primarily to the antioxidative and immunomodulatory activities of its ginsenoside components. Mechanistic studies on the neuroprotective effects of ginsenosides revealed that they act not only as antioxidants but also as modulators of intracellular neuronal signaling and metabolism, cell survival/death genes, and mitochondrial function. The goal of the present paper is to provide a brief review of recent knowledge and developments concerning the beneficial effects as well as the mechanism of action of P. ginseng and its components in the treatment and prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dahae Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hye Lim Lee
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Chang-Eop Kim
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
9
|
Yan J, Liu Y, Zhao Q, Li J, Mao A, Li H, Di C, Zhang H. 56Fe irradiation-induced cognitive deficits through oxidative stress in mice. Toxicol Res (Camb) 2016; 5:1672-1679. [PMID: 30090466 DOI: 10.1039/c6tx00282j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/12/2016] [Indexed: 12/23/2022] Open
Abstract
The rapid growth of manned space flight results in more concerns about health risks and an urgent need for health assessment for space travel. The cosmic environment is complicated and full of radiation. Because of their strong biological effects, heavy ions such as 56Fe ions are considered to be an important component of these lethal galactic rays. Due to the importance of brain function to astronauts, we explored the long-term effects and potential mechanisms of 56Fe ion radiation on mice brains containing the hippocampus. In our study, radiation doses were carried out with 0.5 Gy, 1 Gy or 2 Gy. One month after whole-body 56Fe ion exposure, the Morris water maze test was performed to assess the ability of spatial learning and memory. A histological study was used for pathology analysis of the hippocampus. Alteration of oxidative stress was reflected by MDA and GSH and oxidative DNA damage marked by 8-OHdG was detected by biochemical and immunofluorescence methods. In our results, irradiated groups exhibited significant changes in behavioral performance and also showed loose and edematous arrangement in the pathological characteristics. Furthermore, whole brain levels of MDA, GSH and 8-OHdG increased in the irradiated groups. In addition, increased expression of 8-OHdG can also be detected by immunofluorescence in the hippocampus. Our findings revealed a linkage between radiation-induced oxidative stress and behavioral deficits. This may suggest an underlying mechanism of brain tissue protection and risk assessment in manned space flight.
Collapse
Affiliation(s)
- Jiawei Yan
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China.,University of Chinese Academy of Sciences , Beijing 100039 , PR China
| | - Yang Liu
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China
| | - Qiuyue Zhao
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China.,University of Chinese Academy of Sciences , Beijing 100039 , PR China
| | - Jie Li
- School of Stomatology , Lanzhou University , Lanzhou 730000 , PR China
| | - Aihong Mao
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China.,University of Chinese Academy of Sciences , Beijing 100039 , PR China.,Institute of Gansu Medical Science Research , Lanzhou 730050 , PR China
| | - Hongyan Li
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China
| | - Cuixia Di
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China
| | - Hong Zhang
- Institute of Modern Physics , Chinese Academy of Sciences , Lanzhou 730000 , PR China . ; ; Tel: +86(931)496-9344.,Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences , Lanzhou 730000 , PR China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province , Lanzhou 730000 , PR China
| |
Collapse
|
10
|
Hu T, Wei G, Xi M, Yan J, Wu X, Wang Y, Zhu Y, Wang C, Wen A. Synergistic cardioprotective effects of Danshensu and hydroxysafflor yellow A against myocardial ischemia-reperfusion injury are mediated through the Akt/Nrf2/HO-1 pathway. Int J Mol Med 2016; 38:83-94. [PMID: 27176815 PMCID: PMC4899007 DOI: 10.3892/ijmm.2016.2584] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 04/27/2016] [Indexed: 01/27/2023] Open
Abstract
In clinical practice, the traditional Chinese medicinal herbs, Radix Salvia Miltiorrhiza and Carthamus tinctorius L., are usually prescribed in combination due to their significant cardioprotective effects. However, the mechanisms responsible for these combined effects remain unknown. Thus, in this study, we investigated the mechanisms responsible for the combined effects of Danshensu (DSS) and hydroxysafflor yellow A (HSYA) by establishing a rat model of myocardial ischemia/reperfusion (MI/R), as well as a model of hypoxia/reoxygenation (H/R) using H9c2 cells. The combination index (CI) was calculated using the median-effect method. DSS and HSYA in combination led to a CI value of <1 as regards infarct size in vivo and cell viability in vitro. The rats with MI/R injury that were treated with DSS and/or HSYA were found to have significantly lower levels of creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) and malondialdehyde (MDA), and a lower expressoin of 8-hydroxydeoxyguanosine (8-OHdG), and markedly enhanced superoxide dismutase (SOD) activity. Our in vitro experiments revealed that the cells treated with DSS and/or HSYA had a reduced lactate dehydrogenase (LDH) activity and a decreased percentage of cell apoptosis (increased Bcl-2/Bax ratio, decreased expression of cleaved caspase-3). DSS and HSYA increased the expression of heme oxygenase-1 (HO-1), the phosphorylation of Akt and the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Furthermore, the Akt inhibitor, LY294002, partially hampered the expression of Nrf2 and HO-1. The HO-1 inhibitor, zinc protoporphyrin IX (ZnPP‑IX), did not decrease the expression of p-Akt and Nrf2, although it abolished the anti-apoptotic and antioxidant effects of DSS and HSYA. The findings of our study thus demonstrate that DSS and HSYA confer synergistic cardioprotective effects through the Akt/Nrf2/HO-1 signaling pathway, to certain extent, by enhancing the antioxidant defense system and exerting anti-apoptotic effects.
Collapse
Affiliation(s)
- Tianxin Hu
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Guo Wei
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Miaomiao Xi
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jiajia Yan
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaoxiao Wu
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanhua Wang
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanrong Zhu
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Chao Wang
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
11
|
Hong S, Ahn JY, Cho GS, Kim IH, Cho JH, Ahn JH, Park JH, Won MH, Chen BH, Shin BN, Tae HJ, Park SM, Cho JH, Choi SY, Lee JC. Monocarboxylate transporter 4 plays a significant role in the neuroprotective mechanism of ischemic preconditioning in transient cerebral ischemia. Neural Regen Res 2015; 10:1604-11. [PMID: 26692857 PMCID: PMC4660753 DOI: 10.4103/1673-5374.167757] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Monocarboxylate transporters (MCTs), which carry monocarboxylates such as lactate across biological membranes, have been associated with cerebral ischemia/reperfusion process. In this study, we studied the effect of ischemic preconditioning (IPC) on MCT4 immunoreactivity after 5 minutes of transient cerebral ischemia in the gerbil. Animals were randomly designated to four groups (sham-operated group, ischemia only group, IPC + sham-operated group and IPC + ischemia group). A serious loss of neuron was found in the stratum pyramidale of the hippocampal CA1 region (CA1), not CA2/3, of the ischemia-only group at 5 days post-ischemia; however, in the IPC + ischemia groups, neurons in the stratum pyramidale of the CA1 were well protected. Weak MCT4 immunoreactivity was found in the stratum pyramidale of the CA1 in the sham-operated group. MCT4 immunoreactivity in the stratum pyramidale began to decrease at 2 days post-ischemia and was hardly detected at 5 days post-ischemia; at this time point, MCT4 immunoreactivity was newly expressed in astrocytes. In the IPC + sham-operated group, MCT4 immunoreactivity in the stratum pyramidale of the CA1 was increased compared with the sham-operated group, and, in the IPC + ischemia group, MCT4 immunoreactivity was also increased in the stratum pyramidale compared with the ischemia only group. Briefly, present findings show that IPC apparently protected CA1 pyramidal neurons and increased or maintained MCT4 expression in the stratum pyramidale of the CA1 after transient cerebral ischemia. Our findings suggest that MCT4 appears to play a significant role in the neuroprotective mechanism of IPC in the gerbil with transient cerebral ischemia.
Collapse
Affiliation(s)
- Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ji Yun Ahn
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea ; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Geum-Sil Cho
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Bai Hui Chen
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Seung Min Park
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea ; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
12
|
Kim UJ, Won R, Lee KH. Neuroprotective effects of okadaic acid following oxidative injury in organotypic hippocampal slice culture. Brain Res 2015; 1618:241-8. [DOI: 10.1016/j.brainres.2015.05.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/22/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
|
13
|
Cho YS, Cho JH, Shin BN, Cho GS, Kim IH, Park JH, Ahn JH, Ohk TG, Cho BR, Kim YM, Hong S, Won MH, Lee JC. Ischemic preconditioning maintains the immunoreactivities of glucokinase and glucokinase regulatory protein in neurons of the gerbil hippocampal CA1 region following transient cerebral ischemia. Mol Med Rep 2015; 12:4939-46. [PMID: 26134272 PMCID: PMC4581829 DOI: 10.3892/mmr.2015.4021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 06/15/2015] [Indexed: 01/06/2023] Open
Abstract
Glucokinase (GK) is involved in the control of blood glucose homeostasis. In the present study, the effect of ischemic preconditioning (IPC) on the immunoreactivities of GK and its regulatory protein (GKRP) following 5 min of transient cerebral ischemia was investigated in gerbils. The gerbils were randomly assigned to four groups (sham-operated group, ischemia-operated group, IPC + sham-operated group and IPC + ischemia-operated group). IPC was induced by subjecting the gerbils to 2 min of ischemia, followed by 1 day of recovery. In the ischemia-operated group, a significant loss of neurons was observed in the stratum pyramidale (SP) of the hippocampal CA1 region (CA1) at 5 days post-ischemia; however, in the IPC+ischemia-operated group, the neurons in the SP were well protected. Following immunohistochemical investigation, the immunoreactivities of GK and GKRP in the neurons of the SP were markedly decreased in the CA1, but not the CA2/3, from 2 days post-ischemia, and were almost undetectable in the SP 5 days post-ischemia. In the IPC + ischemia-operated group, the immunoreactivities of GK and GKRP in the SP of the CA1 were similar to those in the sham-group. In brief, the findings of the present study demonstrated that IPC notably maintained the immunoreactivities of GK and GKRP in the neurons of the SP of CA1 following ischemia-reperfusion. This indicated that GK and GKRP may be necessary for neuron survival against transient cerebral ischemia.
Collapse
Affiliation(s)
- Young Shin Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon, Gangwon 200‑702, Republic of Korea
| | - Geum-Sil Cho
- Department of Neuroscience, College of Medicine, Korea University, Seoul 136‑705, Republic of Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Byung-Ryul Cho
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| |
Collapse
|
14
|
Lee JC, Cho JH, Kim IH, Ahn JH, Park JH, Cho GS, Chen BH, Shin BN, Tae HJ, Park SM, Ahn JY, Kim DW, Cho JH, Bae EJ, Yong JH, Kim YM, Won MH, Lee YL. Ischemic preconditioning inhibits expression of Na + /H + exchanger 1 (NHE1) in the gerbil hippocampal CA1 region after transient forebrain ischemia. J Neurol Sci 2015; 351:146-153. [DOI: 10.1016/j.jns.2015.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/09/2015] [Accepted: 03/03/2015] [Indexed: 12/26/2022]
|
15
|
p63 Expression in the Gerbil Hippocampus Following Transient Ischemia and Effect of Ischemic Preconditioning on p63 Expression in the Ischemic Hippocampus. Neurochem Res 2015; 40:1013-22. [PMID: 25777256 DOI: 10.1007/s11064-015-1556-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/15/2015] [Accepted: 03/10/2015] [Indexed: 01/17/2023]
Abstract
p63 is a transcription factor of p53 gene family, which are involved in development, differentiation and cell response to stress; however, its roles in ischemic preconditioning (IPC) in the brain are not clear. In the present study, we investigated the effect of IPC on p63 immunoreactivity caused by 5 min of transient cerebral ischemia in gerbils. IPC was induced by subjecting the gerbils to 2 min of transie ischemia 1 day prior to 5 min of transient ischemia. The animals were randomly assigned to four groups (sham-operated-group, ischemia-operated-group, IPC plus (+)-sham-operated-group and IPC + ischemia-operated-group). The number of viable neurons in the stratum pyramidale of the hippocampal CA1 region (CA1) was significantly increased by IPC + ischemia-operated-group compared with that in the ischemia-operated-group 5 days after ischemic insult. We found that strong p63 immunoreactivity was detected in the CA1 pyramidal neurons in the sham-operated-group, and the immunoreactivity was decreased with time after ischemia-reperfusion. In addition, strong p63 immunoreactivity was newly expressed in microglial cells of the CA1 region from 2 days after ischemia-reperfusion. In all the IPC + sham-operated-groups, p63 immunoreactivity in the CA1 pyramidal neurons was similar to that in the sham-operated-group, and the immunoreactivity was well maintained in the IPC + ischemia-operated-groups after cerebral ischemia. In brief, our present findings show that IPC dramatically protected the reduction of p63 immunoreactivity in the pyramidal neurons of the CA1 region after ischemia-reperfusion, and this result suggests that the expression of p63 may be necessary for neurons to survive after transient cerebral ischemia.
Collapse
|
16
|
Kim MK, Choi WY, Lee HY. Enhancement of the neuroprotective activity of Hericium erinaceus mycelium co-cultivated with Allium sativum extract. Arch Physiol Biochem 2015; 121:19-25. [PMID: 25354984 DOI: 10.3109/13813455.2014.974618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study investigated the neuroprotective effects of Hericium erinaceus mycelium enriched with garlic extract (HGE) on rat pheochromocytoma nerve cells (PC12). The survival rates of the PC12 nerve cells and the neurite-bearing cells after the addition of HGE were estimated as 3.5 × 10(3) viable cells/ml and 2.3 × 10(3) viable cells/ml, respectively, which were 50% and 30% higher, respectively, compared with the untreated group. For the in vivo ischemia experiments, after treatment with the HGE extract, the hippocampal CA1 region was more strongly stained (>20%) than the control group, and the HGE extract also promoted higher staining levels than HFB, HM and HGEF, and even the garlic extract. This result indicates that HGE must have neuroprotective effects. Furthermore, HGE greatly decreased p21 gene expression to approximately 70% of the control and decreased p21 gene expression to even lower levels compared with HM, HGEF and the garlic extract. This work suggests that a synergistic effect of the H. erinaceus mycelium and the garlic extract (mainly allicin) exist because the amount of allicin in HGE (5.81 µg/ml) was lower than the garlic extract itself (6.89 µg/ml).
Collapse
Affiliation(s)
- Myong Ki Kim
- Department of Natural Medicine Resources, Semyung University , Chungbuk , South Korea
| | | | | |
Collapse
|
17
|
Lee JC, Kim IH, Park JH, Ahn JH, Cho JH, Cho GS, Tae HJ, Chen BH, Yan BC, Yoo KY, Choi JH, Lee CH, Hwang IK, Cho JH, Kwon YG, Kim YM, Won MH. Ischemic preconditioning protects hippocampal pyramidal neurons from transient ischemic injury via the attenuation of oxidative damage through upregulating heme oxygenase-1. Free Radic Biol Med 2015; 79:78-90. [PMID: 25483558 DOI: 10.1016/j.freeradbiomed.2014.11.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 11/05/2014] [Accepted: 11/25/2014] [Indexed: 01/17/2023]
Abstract
Ischemic preconditioning (IPC) provides neuroprotection against subsequent severe ischemic injury by activating specific mechanisms. In this study, we tested the hypothesis that IPC attenuates postischemic neuronal death via heme oxygenase-1 (HO-1). Animals used in this study were randomly assigned to 4 groups; sham-operated group, ischemia-operated group, IPC plus (+) sham-operated group and IPC+ischemia-operated group. IPC was induced by subjecting gerbils to 2min of ischemia followed by 1 day of recovery. A significant loss of neurons was observed in pyramidal neurons of the hippocampal CA1 region (CA1) in the ischemia-operated groups at 5 days postischemia. In the IPC+ischemia-operated groups, CA1 pyramidal neurons were well protected. The level of HO-1 protein and its activity increased significantly in the CA1 of the IPC+sham-operated group, and the level and activity was maintained in all the time after ischemia-reperfusion compared with the ischemia-operated groups. HO-1 immunoreactivity was induced in the CA1 pyramidal neurons in both IPC+sham-operated- and IPC+ischemia-operated groups. We also found that levels or immunoreactivities of superoxide anion, 8-hydroxy-2'-deoxyguanosine and 4-hydroxy-2-nonenal were significantly decreased in the CA1 of both IPC+sham-operated- and IPC+ischemia-operated groups. Whereas, treatment with zinc protoporphyrin IX (a HO-1 inhibitor) into the IPC+ischemia-operated groups did not preserve the IPC-mediated increase of HO-1 and lost beneficial effects of IPC by inhibiting ischemia-induced DNA damage and lipid peroxidation. In brief, IPC protects CA1 pyramidal neurons from ischemic injury by upregulating HO-1, and we suggest that the enhancement of HO-1 expression by IPC may be a legitimate strategy for a therapeutic intervention of cerebral ischemic damage.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Geum-Sil Cho
- Department of Neuroscience, College of Medicine, Korea University, Seoul 136-705, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, South Korea
| | - Bai Hui Chen
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 200-702, South Korea
| | - Bing Chun Yan
- Institute of Integrative Traditional & Western Medicine & Medical College, Yangzhou University, Yangzhou 225-001, China
| | - Ki-Yeon Yoo
- Department of Oral Anatomy, College of Dentistry, Gangneung-Wonju National University, Gangneung 210-702, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Choong Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 330-714, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea.
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea.
| |
Collapse
|
18
|
Di Foggia V, Zhang X, Licastro D, Gerli MFM, Phadke R, Muntoni F, Mourikis P, Tajbakhsh S, Ellis M, Greaves LC, Taylor RW, Cossu G, Robson LG, Marino S. Bmi1 enhances skeletal muscle regeneration through MT1-mediated oxidative stress protection in a mouse model of dystrophinopathy. ACTA ACUST UNITED AC 2014; 211:2617-33. [PMID: 25452464 PMCID: PMC4267246 DOI: 10.1084/jem.20140317] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Enhanced polycomb complex protein Bmi1 expression in adult stem cells of the skeletal muscle leads to improved muscle function in a model of Duchenne Muscular Dystrophy via metallothionein1-mediated protection from oxidative stress. The Polycomb group (PcG) protein Bmi1 is an essential epigenetic regulator of stem cell function during normal development and in adult organ systems. We show that mild up-regulation of Bmi1 expression in the adult stem cells of the skeletal muscle leads to a remarkable improvement of muscle function in a mouse model of Duchenne muscular dystrophy. The molecular mechanism underlying enhanced physiological function of Bmi1 depends on the injury context and it is mediated by metallothionein 1 (MT1)–driven modulation of resistance to oxidative stress in the satellite cell population. These results lay the basis for developing Bmi1 pharmacological activators, which either alone or in combination with MT1 agonists could be a powerful novel therapeutic approach to improve regeneration in muscle wasting conditions.
Collapse
Affiliation(s)
- Valentina Di Foggia
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| | | | - Mattia F M Gerli
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, England, UK
| | - Rahul Phadke
- The Dubowitz Neuromuscular Centre, Institute of Child Health and Great Ormond Street Hospital for Children, London WC1N 3JH, England, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Institute of Child Health and Great Ormond Street Hospital for Children, London WC1N 3JH, England, UK
| | - Philippos Mourikis
- Stem Cells and Development, Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS, URA 2578 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS, URA 2578 Paris, France
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, England, UK
| | - Laura C Greaves
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE4 2HH, England, UK
| | - Robert W Taylor
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE4 2HH, England, UK
| | - Giulio Cossu
- Institute for Inflammation and Repair, University of Manchester, Manchester M13 9PL, England, UK
| | - Lesley G Robson
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK
| |
Collapse
|
19
|
Park YS, Cho JH, Kim IH, Cho GS, Cho JH, Park JH, Ahn JH, Chen BH, Shin BN, Shin MC, Tae HJ, Cho YS, Lee YL, Kim YM, Won MH, Lee JC. Effects of ischemic preconditioning on VEGF and pFlk-1 immunoreactivities in the gerbil ischemic hippocampus after transient cerebral ischemia. J Neurol Sci 2014; 347:179-87. [PMID: 25300771 DOI: 10.1016/j.jns.2014.09.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/01/2014] [Accepted: 09/23/2014] [Indexed: 01/19/2023]
Abstract
Ischemia preconditioning (IPC) displays an important adaptation of the CNS to sub-lethal ischemia. In the present study, we examined the effect of IPC on immunoreactivities of VEGF-, and phospho-Flk-1 (pFlk-1) following transient cerebral ischemia in gerbils. The animals were randomly assigned to four groups (sham-operated-group, ischemia-operated-group, IPC plus (+) sham-operated-group, and IPC+ischemia-operated-group). IPC was induced by subjecting gerbils to 2 min of ischemia followed by 1 day of recovery. In the ischemia-operated-group, a significant loss of neurons was observed in the stratum pyramidale (SP) of the hippocampal CA1 region (CA1) alone 5 days after ischemia-reperfusion, however, in all the IPC+ischemia-operated-groups, pyramidal neurons in the SP were well protected. In immunohistochemical study, VEGF immunoreactivity in the ischemia-operated-group was increased in the SP at 1 day post-ischemia and decreased with time. Five days after ischemia-reperfusion, strong VEGF immunoreactivity was found in non-pyramidal cells, which were identified as pericytes, in the stratum oriens (SO) and radiatum (SR). In the IPC+sham-operated- and IPC+ischemia-operated-groups, VEGF immunoreactivity was significantly increased in the SP. pFlk-1 immunoreactivity in the sham-operated- and ischemia-operated-groups was hardly found in the SP, and, from 2 days post-ischemia, pFlk-1 immunoreactivity was strongly increased in non-pyramidal cells, which were identified as pericytes. In the IPC+sham-operated-group, pFlk-1 immunoreactivity was significantly increased in both pyramidal and non-pyramidal cells; in the IPC+ischemia-operated-groups, the similar pattern of VEGF immunoreactivity was found in the ischemic CA1, although the VEGF immunoreactivity was strong in non-pyramidal cells at 5 days post-ischemia. In brief, our findings show that IPC dramatically augmented the induction of VEGF and pFlk-1 immunoreactivity in the pyramidal cells of the CA1 after ischemia-reperfusion, and these findings suggest that the increases of VEGF and Flk-1 expressions may be necessary for neurons to survive from transient ischemic damage.
Collapse
Affiliation(s)
- Yoo Seok Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea; Department of Emergency Medicine, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Geum-Sil Cho
- Department of Neuroscience, College of Medicine, Korea University, Seoul 136-705, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Bai Hui Chen
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon 200-702, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon 200-702, South Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, South Korea
| | - Young Shin Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea; Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, Seoul 140-743, South Korea
| | - Yun Lyul Lee
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon 200-702, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea.
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea.
| |
Collapse
|
20
|
Ahmed MAE, El Morsy EM, Ahmed AAE. Pomegranate extract protects against cerebral ischemia/reperfusion injury and preserves brain DNA integrity in rats. Life Sci 2014; 110:61-9. [PMID: 25010842 DOI: 10.1016/j.lfs.2014.06.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/14/2014] [Accepted: 06/28/2014] [Indexed: 12/20/2022]
Abstract
AIM Interruption to blood flow causes ischemia and infarction of brain tissues with consequent neuronal damage and brain dysfunction. Pomegranate extract is well tolerated, and safely consumed all over the world. Interestingly, pomegranate extract has shown remarkable antioxidant and anti-inflammatory effects in experimental models. Many investigators consider natural extracts as novel therapies for neurodegenerative disorders. Therefore, this study was carried out to investigate the protective effects of standardized pomegranate extract against cerebral ischemia/reperfusion-induced brain injury in rats. MAIN METHODS Adult male albino rats were randomly divided into sham-operated control group, ischemia/reperfusion (I/R) group, and two other groups that received standardized pomegranate extract at two dose levels (250, 500 mg/kg) for 15 days prior to ischemia/reperfusion (PMG250+I/R, and PMG500+I/R groups). After I/R or sham operation, all rats were sacrificed and brains were harvested for subsequent biochemical analysis. KEY FINDINGS Results showed reduction in brain contents of MDA (malondialdehyde), and NO (nitric oxide), in addition to enhancement of SOD (superoxide dismutase), GPX (glutathione peroxidase), and GRD (glutathione reductase) activities in rats treated with pomegranate extract prior to cerebral I/R. Moreover, pomegranate extract decreased brain levels of NF-κB p65 (nuclear factor kappa B p65), TNF-α (tumor necrosis factor-alpha), caspase-3 and increased brain levels of IL-10 (interleukin-10), and cerebral ATP (adenosine triphosphate) production. Comet assay showed less brain DNA (deoxyribonucleic acid) damage in rats protected with pomegranate extract. SIGNIFICANCE The present study showed, for the first time, that pre-administration of pomegranate extract to rats, can offer a significant dose-dependent neuroprotective activity against cerebral I/R brain injury and DNA damage via antioxidant, anti-inflammatory, anti-apoptotic and ATP-replenishing effects.
Collapse
Affiliation(s)
- Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt.
| | - Engy M El Morsy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Helwan, Egypt
| | - Amany A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Helwan, Egypt
| |
Collapse
|
21
|
Changes and expressions of Redd1 in neurons and glial cells in the gerbil hippocampus proper following transient global cerebral ischemia. J Neurol Sci 2014; 344:43-50. [PMID: 24980938 DOI: 10.1016/j.jns.2014.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 06/07/2014] [Accepted: 06/11/2014] [Indexed: 12/22/2022]
Abstract
Redd1 (known as RTP801/Dig2/DDIT4) is a stress-induced protein, and it is known to be regulated in response to some stresses including hypoxia and oxidative stress. In the present study, we investigated the time-dependent changes in Redd1 immunoreactivity and its protein levels in the gerbil hippocampus proper (CA1-3 regions) after 5 min of transient global cerebral ischemia using immunohistochemistry and Western blot analysis. Redd1 immunoreactivity was apparently changed in the pyramidal neurons of the ischemic CA1 region, not in the pyramidal neurons of the ischemic CA2/3 region. Redd1 immunoreactivity in the CA1 pyramidal neurons was significantly increased at 6 h post-ischemia, decreased until 1 day post-ischemia, increased again at 2 days post-ischemia and weakly observed at 5 days post-ischemia. Especially, at 5 days after ischemic damage, Redd1 immunoreactivity was newly expressed in astrocytes and GABAergic interneurons in the CA1 region. Redd1 protein levels in the ischemic CA1 region were changed like the pattern of the Redd1 immunoreactivity. These results indicate that Redd1 immunoreactivity and protein levels are increased in the ischemic CA1 region at an early time after ischemic damage and that the increased Redd1 expression may be closely related to the delayed neuronal death of the CA1 pyramidal neurons following 5 min of transient global cerebral ischemia.
Collapse
|
22
|
Acquati S, Greco A, Licastro D, Bhagat H, Ceric D, Rossini Z, Grieve J, Shaked-Rabi M, Henriquez NV, Brandner S, Stupka E, Marino S. Epigenetic regulation of survivin by Bmi1 is cell type specific during corticogenesis and in gliomas. Stem Cells 2013; 31:190-202. [PMID: 23132836 DOI: 10.1002/stem.1274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 09/17/2012] [Indexed: 12/12/2022]
Abstract
Polycomb group proteins are essential regulators of stem cell function during embryonic development and in adult tissue homeostasis. Bmi1, a key component of the Polycomb Repressive Complex 1, is highly expressed in undifferentiated neural stem cells (NSC) as well as in several human cancers including high-grade gliomas--highly aggressive brain tumors. Using a conditional gene activation approach in mice, we show that overexpression of Bmi1 induces repressive epigenetic regulation of the promoter of Survivin, a well-characterized antiapoptotic protein. This phenomenon is cell type-specific and it leads to apoptotic death of progenitor cells exclusively upon commitment toward a neuronal fate. Moreover, we show that this is triggered by increased oxidative stress-induced DNA damage. In contrast, undifferentiated NSC as well as glioma-initiating cells display an open chromatin configuration at the Survivin promoter and do not undergo apoptotic death. These findings raise the possibility that normal and neoplastic stem cells depend on the same mechanism for surviving the hyperproliferative state induced by increased Bmi1 expression.
Collapse
Affiliation(s)
- Serena Acquati
- Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hamakawa M, Ishida A, Tamakoshi K, Shimada H, Nakashima H, Noguchi T, Toyokuni S, Ishida K. Repeated short-term daily exercise ameliorates oxidative cerebral damage and the resultant motor dysfunction after transient ischemia in rats. J Clin Biochem Nutr 2013; 53:8-14. [PMID: 23874064 PMCID: PMC3705152 DOI: 10.3164/jcbn.12-72] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 12/13/2012] [Indexed: 12/18/2022] Open
Abstract
Long-term exercise prior to brain ischemia enhances the activities of antioxidant enzymes and leads to a significant reduction in brain damage and neurological deficits in rats subjected to transient middle cerebral artery occlusion. However, it has not been established whether relatively short-term exercise generates similar results following middle cerebral artery occlusion. We aimed to determine whether short-term exercise could reduce oxidative damage and prevent sensori-motor dysfunction. Male Wistar rats were subjected to perform daily exercise on a treadmill for 30 min at a speed of 15 m/min for 3 weeks, followed by a 90-min middle cerebral artery occlusion. Animals were assessed after middle cerebral artery occlusion for neurological deficits and sensori-motor function. Brain tissues were processed to evaluate infarct volume and oxidative damage. Oxidative stress was assessed using immunohistochemistry for 4-hydroxy-2-nonenal-modified proteins and 8-hydroxy-2'-deoxyguanosine. Antioxidant enzymes were evaluated using immunohistochemistry for thioredoxin and activity assay for superoxide dismutase. Exercise for 3 weeks decreased the severity of paralysis and impairment in forelimb motor coordination. Furthermore, exercise had effect on superoxide dismutase and reduced the infarct volume and the number of cells immunopositive for 4-hydroxy-2-nonenal-modified proteins and 8-hydroxy-2'-deoxyguanosine. Our results suggest that pre-conditioning treadmill exercise for 3 weeks is useful for ameliorating ischemia-induced brain injury.
Collapse
Affiliation(s)
- Michiru Hamakawa
- Department of Physical Therapy, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-minami, Higashi-ku, Nagoya, Aichi 461-8673, Japan ; Okinawa Rehabilitation Center Hospital, 2-15-1 Hiyagon, Okinawa, Okinawa 904-2173, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Li H, Park JH, Lee JC, Yoo KY, Hwang IK, Lee CH, Choi JH, Kim JD, Kang IJ, Won MH. Neuroprotective effects of Alpinia katsumadai against experimental ischemic damage via control of oxidative stress. PHARMACEUTICAL BIOLOGY 2013; 51:197-205. [PMID: 23095174 DOI: 10.3109/13880209.2012.716853] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
CONTEXT Alpinia katsumadai (Zingiberaceae) has been identified by the National Plant Quarantine Service in Korea. The extract of Alpinia katsumadai seed (EAKS) has antioxidant activities. OBJECTIVE We investigated the neuroprotective effects of EAKS on ischemic damage in the gerbil hippocampal CA1 region after transient cerebral ischemia. MATERIALS AND METHODS The ethanol extract of EAKS was obtained by organic solvent, collected in Kangwon province (South Korea) and orally administered using a feeding needle once a day for one week before transient cerebral ischemia in gerbils. RESULT We adapted oral administration of 25 and 50 mg/kg EAKS because there are no data about the absorption and metabolism of EKAS. We found a significant neuroprotection in the 50 mg/kg EAKS-treated ischemia group, not in the 25 mg/kg EAKS-treated ischemia group, at 4 days ischemia-reperfusion (I-R). In the 50 mg/kg EAKS-treated ischemia group, about 68% of pyramidal neurons in the CA1 region were immunostained with neuronal nuclei (NeuN) 4 days after I-R, compared to the vehicle-treated ischemia group. 8-Hydroxy-2'-deoxyguanosine (a marker for DNA damage) and 4-hydroxy-2-nonenal (a marker for lipid peroxidation) immunoreactivity in the CA1 region of the EAKS-treated ischemia group were not markedly changed compared to the vehicle-treated ischemia group. In addition, Cu,Zn- and Mn-SOD immunoreactivity in the CA1 region of the EAKS-treated ischemia group were increased compared to the vehicle-treated ischemia group. DISCUSSION Repeated supplements of EAKS could protect neurons against ischemic damage, showing that DNA damage and lipid peroxidation are attenuated and SODs are increased in the ischemic CA1 region.
Collapse
Affiliation(s)
- Hua Li
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Effects of transient cerebral ischemia on the expression of DNA methyltransferase 1 in the gerbil hippocampal CA1 region. Neurochem Res 2012; 38:74-81. [PMID: 22987057 DOI: 10.1007/s11064-012-0890-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 08/20/2012] [Accepted: 09/10/2012] [Indexed: 12/25/2022]
Abstract
DNA methylation is a key epigenetic modification of DNA that is catalyzed by DNA methyltransferases (Dnmt). Increasing evidences suggest that DNA methylation in neurons regulates synaptic plasticity as well as neuronal network activity. In the present study, we investigated the changes in DNA methyltransferases 1 (Dnmt1) immunoreactivity and its protein levels in the gerbil hippocampal CA1 region after 5 min of transient global cerebral ischemia. CA1 pyramidal neurons were well stained with NeuN (a neuron-specific soluble nuclear antigen) antibody in the sham-group, Four days after ischemia-reperfusion (I-R), NeuN-positive ((+)) cells were significantly decreased in the stratum pyramidale (SP) of the CA1 region, and many Fluro-Jade B (a marker for neuronal degeneration)(+) cells were observed in the SP. Dnmt1 immunoreactivity was well detected in all the layers of the sham-group. Dnmt1 immunoreactivity was hardly detected only in the stratum pyramidale of the CA1 region from 4 days post-ischemia; however, at these times, Dnmt1 immunoreactivity was newly expressed in GABAergic interneurons or astrocytes in the ischemic CA1 region. In addition, the level of Dnmt1 was lowest at 4 days post-ischemia. In brief, both the Dnmt1 immunoreactivity and protein levels were distinctively decreased in the ischemic CA1 region 4 days after transient cerebral ischemia. These results indicate that the decrease of Dnmt1 expression at 4 days post-ischemia may be related to ischemia-induced delayed neuronal death.
Collapse
|
26
|
Kim GS, Jung JE, Narasimhan P, Sakata H, Yoshioka H, Song YS, Okami N, Chan PH. Release of mitochondrial apoptogenic factors and cell death are mediated by CK2 and NADPH oxidase. J Cereb Blood Flow Metab 2012; 32:720-30. [PMID: 22146192 PMCID: PMC3318149 DOI: 10.1038/jcbfm.2011.176] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 10/28/2011] [Accepted: 11/09/2011] [Indexed: 11/09/2022]
Abstract
Activation of the NADPH oxidase subunit, NOX2, and increased oxidative stress are associated with neuronal death after cerebral ischemia and reperfusion. Inhibition of NOX2 by casein kinase 2 (CK2) leads to neuronal survival, but the mechanism is unknown. In this study, we show that in copper/zinc-superoxide dismutase transgenic (SOD1 Tg) mice, degradation of CK2α and CK2α' and dephosphorylation of CK2β against oxidative stress were markedly reduced compared with wild-type (WT) mice that underwent middle cerebral artery occlusion. Inhibition of CK2 pharmacologically or by ischemic reperfusion facilitated accumulation of poly(ADP-ribose) polymers, the translocation of apoptosis-inducing factor (AIF), and cytochrome c release from mitochondria after ischemic injury. The eventual enhancement of CK2 inhibition under ischemic injury strongly increased 8-hydroxy-2'-deoxyguanosine and phosphorylation of H2A.X. Furthermore, CK2 inhibition by tetrabromocinnamic acid (TBCA) in SOD1 Tg and gp91 knockout (KO) mice after ischemia reperfusion induced less release of AIF and cytochrome c than in TBCA-treated WT mice. Inhibition of CK2 in gp91 KO mice subjected to ischemia reperfusion did not increase brain infarction compared with TBCA-treated WT mice. These results strongly suggest that NOX2 activation releases reactive oxygen species after CK2 inhibition, triggering release of apoptogenic factors from mitochondria and inducing DNA damage after ischemic brain injury.
Collapse
Affiliation(s)
- Gab Seok Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Joo Eun Jung
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Purnima Narasimhan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Hiroyuki Sakata
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Yun Seon Song
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Nobuya Okami
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Pak H Chan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
27
|
Ock CY, Kim EH, Choi DJ, Lee HJ, Hahm KB, Chung MH. 8-Hydroxydeoxyguanosine: Not mere biomarker for oxidative stress, but remedy for oxidative stress-implicated gastrointestinal diseases. World J Gastroenterol 2012; 18:302-8. [PMID: 22294836 PMCID: PMC3261525 DOI: 10.3748/wjg.v18.i4.302] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 08/26/2011] [Accepted: 09/02/2011] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) attack guanine bases in DNA easily and form 8-hydroxydeoxyguanosine (8-OHdG), which can bind to thymidine rather than cytosine, based on which, the level of 8-OHdG is generally regarded as a biomarker of mutagenesis consequent to oxidative stress. For example, higher levels of 8-OHdG are noted in Helicobacter pylori-associated chronic atrophic gastritis as well as gastric cancer. However, we have found that exogenous 8-OHdG can paradoxically reduce ROS production, attenuate the nuclear factor-κB signaling pathway, and ameliorate the expression of proinflammatory mediators such as interleukin (IL)-1, IL-6, cyclo-oxygenase-2, and inducible nitric oxide synthase in addition to expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX)-1, NOX organizer-1 and NOX activator-1 in various conditions of inflammation-based gastrointestinal (GI) diseases including gastritis, inflammatory bowel disease, pancreatitis, and even colitis-associated carcinogenesis. Our recent finding that exogenous 8-OHdG was very effective in either inflammation-based or oxidative-stress-associated diseases of stress-related mucosal damage has inspired the hope that synthetic 8-OHdG can be a potential candidate for the treatment of inflammation-based GI diseases, as well as the prevention of inflammation-associated GI cancer. In this editorial review, the novel fact that exogenous 8-OHdG can be a functional molecule regulating oxidative-stress-induced gastritis through either antagonizing Rac-guanosine triphosphate binding or blocking the signals responsible for gastric inflammatory cascade is introduced.
Collapse
|
28
|
Yan Y, Yang JY, Mou YH, Wang LH, Zhou YN, Wu CF. Differences in the activities of resveratrol and ascorbic acid in protection of ethanol-induced oxidative DNA damage in human peripheral lymphocytes. Food Chem Toxicol 2011; 50:168-74. [PMID: 22056336 DOI: 10.1016/j.fct.2011.10.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 08/16/2011] [Accepted: 10/07/2011] [Indexed: 01/24/2023]
Abstract
Previous studies have shown that ethanol induces oxidative DNA damage in human peripheral lymphocytes. In the present study, protective effect of resveratrol and ascorbic acid on ethanol-induced oxidative DNA damage in human peripheral lymphocytes in vitro were comparatively investigated. Pretreatments with resveratrol at 5, 25, and 50μM, which were in the concentration range of in vitro research, significantly inhibited ethanol-induced oxidative DNA damage in 24h, whereas ascorbic acid showed such DNA protective activity only in 1h. Further study showed that both compounds could directly scavenge hydroxyl radical produced during ethanol metabolism. Resveratrol significantly inhibited ethanol metabolism by regulating alcohol dehydrogenase 1B (ADH1B) and acetaldehyde dehydrogenase 2 (ALDH2) mRNA expressions. Moreover, resveratrol also activated the base excision repair (BER) system in mRNA and protein levels in DNA auto-repair process. However, ascorbic acid showed no effect on ethanol metabolic pathway and BER system. Thus, the present study provided the first evidence that even though both resveratrol and ascorbic acid are anti-oxidants, they possessed differential mechanisms of action in protection against ethanol-induced oxidative DNA damage in human peripheral lymphocytes.
Collapse
Affiliation(s)
- Yu Yan
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016 Shenyang, People's Republic of China
| | | | | | | | | | | |
Collapse
|
29
|
Zitka O, Krizkova S, Krejcova L, Hynek D, Gumulec J, Masarik M, Sochor J, Adam V, Hubalek J, Trnkova L, Kizek R. Microfluidic tool based on the antibody-modified paramagnetic particles for detection of 8-hydroxy-2'-deoxyguanosine in urine of prostate cancer patients. Electrophoresis 2011; 32:3207-20. [PMID: 22012838 DOI: 10.1002/elps.201100430] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 08/15/2011] [Accepted: 08/15/2011] [Indexed: 12/31/2022]
Abstract
Guanosine derivatives are important for diagnosis of oxidative DNA damage including 8-hydroxy-2'-deoxyguanosine (8-OHdG) as one of the most abundant products of DNA oxidation. This compound is commonly determined in urine, which makes 8-OHdG a good non-invasive marker of oxidation stress. In this study, we optimized and tested the isolation of 8-OHdG from biological matrix by using paramagnetic particles with an antibody-modified surface. 8-OHdG was determined using 1-naphthol generated by alkaline phosphatase conjugated with the secondary antibody. 1-Naphthol was determined by stopped flow injection analysis (SFIA) with electrochemical detector using a glassy carbon working electrode and by stationary electrochemical detection using linear sweep voltammetry. A special modular electrochemical SFIA system which needs only 10 μL of sample including working buffer for one analysis was completely designed and successfully verified. The recoveries in different matrices and analyte concentration were estimated. Detection limit (3 S/N) was estimated as 5 pg/mL of 8-OHdG. This method promises to be very easily modified to microfluidic systems as "lab on valve". The optimized method had sufficient selectivity and thus could be used for determination of 8-OHDG in human urine and therefore for estimation of oxidative DNA damage as a result of oxidation stress in prostate cancer patients.
Collapse
Affiliation(s)
- Ondrej Zitka
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Brno, Czech Republic
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kam KY, Yu SJ, Jeong N, Hong JH, Anthony Jalin AMA, Lee S, Choi YW, Lee CK, Kang SG. p-Hydroxybenzyl alcohol prevents brain injury and behavioral impairment by activating Nrf2, PDI, and neurotrophic factor genes in a rat model of brain ischemia. Mol Cells 2011; 31:209-15. [PMID: 21347705 PMCID: PMC3932695 DOI: 10.1007/s10059-011-0028-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 11/03/2010] [Accepted: 12/08/2010] [Indexed: 01/21/2023] Open
Abstract
The therapeutic goal in treating cerebral ischemia is to reduce the extent of brain injury and thus minimize neurological impairment. We examined the effects of p-hydroxybenzyl alcohol (HBA), an active component of Gastrodia elata Blume, on transient focal cerebral ischemia-induced brain injury with respect to the involvement of protein disulphide isomerase (PDI), nuclear factor-E2-related factor 2 (Nrf2), and neurotrophic factors. All animals were ovariectomized 14 days before ischemic injury. Ischemic injury was induced for 1 h by middle cerebral artery occlusion (MCAO) followed by 24-h reperfusion. Three days before MCAO, the vehicle-treated and the HBA-treated groups received intramuscular sesame oil and HBA (25 mg/kg BW), respectively. 2,3,5-Triphenyltetrazolium chloride (TTC) staining showed decreased infarct volume in the ischemic lesion of HBA-treated animals. HBA pretreatment also promoted functional recovery, as measured by the modified neurological severity score (mNSS; p < 0.05). Moreover, expression of PDI, Nrf2, BDNF, GDNF, and MBP genes increased by HBA treatment. In vitro, H(2)O(2)-induced PC12 cell death was prevented by 24 h HBA treatment, but bacitracin, a PDI inhibitor, attenuated this cytoprotective effect in a dose-dependent manner. HBA treatment for 2 h also induced nuclear translocation of Nrf2, possibly activating the intracellular antioxidative system. These results suggest that HBA protects against brain damage by modulating cytoprotective genes, such as Nrf2 and PDI, and neurotrophic factors.
Collapse
Affiliation(s)
- Kyung-Yoon Kam
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
- FIRST Research Group, Inje University, Gimhae 621-749, Korea
- Institute of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Seong Jin Yu
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Nahee Jeong
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Jeong Hwa Hong
- School of Food and Life Science , Inje University, Gimhae 621-749, Koreaof Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Angela M. A. Anthony Jalin
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Sungja Lee
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
| | - Yong Won Choi
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
| | - Chae Kwan Lee
- Institute of Environmental and Occupational Medicine, Department of Occu-pational and Environmental Medicine, Busan Paik Hospital, Inje University, Busan 614-735, Korea
| | - Sung Goo Kang
- FIRST Research Group, Inje University, Gimhae 621-749, Korea
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| |
Collapse
|
31
|
Yan Y, Yang JY, Mou YH, Wang LH, Zhang H, Wu CF. Possible Metabolic Pathways of Ethanol Responsible for Oxidative DNA Damage in Human Peripheral Lymphocytes. Alcohol Clin Exp Res 2010; 35:1-9. [DOI: 10.1111/j.1530-0277.2010.01316.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
32
|
Choi JH, Yoo KY, Lee CH, Park OK, Yan BC, Li H, Moon YS, Hwang IK, Lee YL, Shin HC, Won MH. Transient cerebral ischemia induces active astrocytosis without distinct neuronal death in the gerbil main olfactory bulb: a long-term analysis. Neurochem Res 2010; 35:1588-98. [PMID: 20593235 DOI: 10.1007/s11064-010-0219-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2010] [Indexed: 02/02/2023]
Abstract
In the present study, we examined ischemia-induced neuronal and glial changes in the gerbil MOB at various time points during 60 days after 5 min of transient cerebral ischemia. The number of neuronal neuclei-immunoreactive neurons was not changed after ischemia/reperfusion (I/R). Myelin basic protein immunoreaction was well preserved after I/R. Five days after I/R, reactive form of GFAP-immunoreactive astrocytes began to increase in the external plexiform layer and granule cell layer: These reactive astrocytes peaked 10 days after I/R, thereafter, they decreased with time after I/R. Iba-1-immunoreactive microglia were ubiquitously distributed in all layers of the MOB. After I/R, significant changes in their morphology and immunoreactivity were not detected. The results of western blot analyses for GFAP, Iba-1 and MBP were similar to the immunohistochemical data. In addition, 8-hydroxy-2'-deoxyguanosine (a marker for DNA damage) immunoreactivity and SOD1, an antioxidant, protein levels were not changed in the ischemic MOB. These results indicate that neurons in the MOB are resistant to ischemic insult, showing that astrocytes are activated late in the ischemic MOB.
Collapse
Affiliation(s)
- Jung Hoon Choi
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lee CH, Yoo KY, Choi JH, Park OK, Hwang IK, Choi SY, Kim DH, Won MH. Cyclin D1 immunoreactivity changes in CA1 pyramidal neurons and dentate granule cells in the gerbil hippocampus after transient forebrain ischemia. Neurol Res 2010; 33:93-100. [PMID: 20546683 DOI: 10.1179/016164110x12714125204399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Cyclin D1, a member of the G1 cyclin family, plays a critical role in the progression of the cell cycle. In the present study, we investigated chronological alterations in cyclin D1 immunoreactivity and its protein levels in the gerbil hippocampus after ischemia/reperfusion. METHODS Chronological alterations in cyclin D1 immunoreactivity and its levels were examined in the gerbil hippocampus after ischemia/reperfusion using immunohistochemistry and western blot analysis. RESULTS Changes in cyclin D1 immunoreactivity in the ischemic hippocampus were distinct in pyramidal neurons of the CA1 region and granule cells of the dentate gyrus. Cyclin D1 immunoreactivity in pyramidal neurons of the CA1 region was increased up to 1 day after ischemia/reperfusion, although a transient decrease of cyclin D1 immunoreactivity was detected at 12 hour after ischemia/reperfusion. Thereafter, cyclin D1 immunoreactivity in the CA1 pyramidal neurons was very weak 2 days and disappeared nearly 4 and 7 days after ischemia/reperfusion. However, 4 days after ischemia/reperfusion, the cyclin D1 immunoreactivity in non-pyramidal neurons of the CA1 region was very strong. In the CA2/3 region, cyclin D1 immunoreactivity was higher than that in the CA1 region and not changed after ischemia/reperfusion. In the dentate gyrus, chronological change in cyclin D1 immunoreactivity was observed. Cells in the granule cell layer showed distinct change in cyclin D1 immunoreactivity after ischemia/reperfusion: the cyclin D1 immunoreactivity was lowest at 12 hours and strong 1 and 4 days after ischemia/reperfusion. In addition, change in cyclin D1 protein level was found in the ischemic hippocampus. CONCLUSION Our results indicate that cyclin D1 may play an important role in cellular events related with neuronal damage following ischemia/reperfusion.
Collapse
Affiliation(s)
- Choong Hyun Lee
- Department of Anatomy and Neurobiology, and Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon, Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim DW, Jeong HJ, Kang HW, Shin MJ, Sohn EJ, Kim MJ, Ahn EH, An JJ, Jang SH, Yoo KY, Won MH, Kang TC, Hwang IK, Kwon OS, Cho SW, Park J, Eum WS, Choi SY. Transduced human PEP-1-catalase fusion protein attenuates ischemic neuronal damage. Free Radic Biol Med 2009; 47:941-52. [PMID: 19577641 DOI: 10.1016/j.freeradbiomed.2009.06.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 05/11/2009] [Accepted: 06/24/2009] [Indexed: 10/20/2022]
Abstract
Antioxidant enzymes are considered to have beneficial effects against various diseases mediated by reactive oxygen species (ROS). Ischemia is characterized by both oxidative stress and changes in the antioxidant defense system. Catalase (CAT) and superoxide dismutase (SOD) are major antioxidant enzymes by which cells counteract the deleterious effects of ROS. To investigate the protective effects of CAT, we constructed PEP-1-CAT cell-permeative expression vectors. When PEP-1-CAT fusion proteins were added to the culture medium of neuronal cells, they rapidly entered the cells and protected them against oxidative stress-induced neuronal cell death. Immunohistochemical analysis revealed that PEP-1-CAT prevented neuronal cell death in the hippocampus induced by transient forebrain ischemia. Moreover, we showed that the protective effect of PEP-1-CAT was observed in neuronal cells treated with PEP-1-SOD. Therefore, we suggest that transduced PEP-1-CAT and PEP-1-SOD fusion proteins could be useful as therapeutic agents for various human diseases related to oxidative stress, including stroke.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, College of Medicine, Hallym University, Chunchon 200-702, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hwang IK, Yoo KY, Li H, Park OK, Lee CH, Choi JH, Jeong YG, Lee YL, Kim YM, Kwon YG, Won MH. Indole-3-propionic acid attenuates neuronal damage and oxidative stress in the ischemic hippocampus. J Neurosci Res 2009; 87:2126-37. [DOI: 10.1002/jnr.22030] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
36
|
Platycodin D and 2''-O-acetyl-polygalacin D2 isolated from Platycodon grandiflorum protect ischemia/reperfusion injury in the gerbil hippocampus. Brain Res 2009; 1279:197-208. [PMID: 19433075 DOI: 10.1016/j.brainres.2009.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 04/29/2009] [Accepted: 05/02/2009] [Indexed: 12/31/2022]
Abstract
Platycodi radix is used as a folk remedy for several conditions. In this study, we investigated the neuroprotective effects of five major extracts; deapioplatycoside E (DPE), platycoside E (PE), platyconic acid A (PA), platycodin D (PD) and 2''-o-acetyl-polygalacin D2 (PD2) isolated from the P.radix in the hippocampal CA1 region (CA1) 4 or 10 days after ischemia/reperfusion (I/R). Each extract was administered into gerbils with intraperitoneal injection (5 mg/kg/day) 10 days before ischemic surgery and the gerbils were sacrificed 4 or 10 days after I/R. Fluoro-Jade B (F-J B, a marker for neurodegeneration) positive ((+)) neurons increased significantly in the stratum pyramidale of the CA1 region in the vehicle-treated group after I/R. A similar pattern was observed in the DPE-, PE- and PA-treated groups; however, in the PD- and PD2-treated groups, F-J B(+) neurons were small in number. We also observed that activations of astrocytes and microglia in the CA1 region after I/R were blocked by the PD- and PD2 treatments. In addition, we found that Cu,Zn-superoxide dismutase (SOD1) immunoreactivity in the pyramidal layer of the PD- and PD2-treated groups was similar to that of the sham group and COX-2(+) and NF-kappaB(+) cells were significantly lower in the PD- and PD2-treated group than those in the vehicle-treated group after I/R. These results suggest that PD and PD2 rescue neurons in the CA1 region from an ischemic damage.
Collapse
|
37
|
Hwang IK, Yoo KY, Suh HW, Kim YS, Kwon DY, Kwon YG, Yoo JH, Won MH. Folic acid deficiency increases delayed neuronal death, DNA damage, platelet endothelial cell adhesion molecule-1 immunoreactivity, and gliosis in the hippocampus after transient cerebral ischemia. J Neurosci Res 2008; 86:2003-15. [PMID: 18335523 DOI: 10.1002/jnr.21647] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Folic acid deficiency increases stroke risk. In the present study, we examined whether folic acid deficiency enhances neuronal damage and gliosis via oxidative stress in the gerbil hippocampus after transient forebrain ischemia. Animals were exposed to a folic acid-deficient diet (FAD) for 3 months and then subjected to occlusion of both common carotid arteries for 5 min. Exposure to an FAD increased plasma homocysteine levels by five- to eightfold compared with those of animals fed with a control diet (CD). In CD-treated animals, most neurons were dead in the hippocampal CA1 region 4 days after ischemia/reperfusion, whereas, in FAD-treated animals, this occurred 3 days after ischemia/reperfusion. Immunostaining for 8-hydroxy-2'-deoxyguanosine (8-OHdG) was performed to examine DNA damage in CA1 neurons in both groups after ischemia, and it was found that 8-OHdG immunoreactivity in both FAD and CD groups peaked at 12 hr after reperfusion, although the immunoreactivity in the FAD group was much greater than that in the CD group. Platelet endothelial cell adhesion molecule-1 (PECAM-1; a final mediator of neutrophil transendothelial migration) immunoreactivity in both groups increased with time after ischemia/reperfusion: Its immunoreactivity in the FAD group was much higher than that in the CD group 3 days after ischemia/reperfusion. In addition, reactive gliosis in the ischemic CA1 region increased with time after ischemia in both groups, but astrocytosis and microgliosis in the FAD group were more severe than in the CD group at all times after ischemia. Our results suggest that folic acid deficiency enhances neuronal damage induced by ischemia.
Collapse
Affiliation(s)
- In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang Q, Sun AY, Simonyi A, Miller DK, Smith RE, Luchtefeld RG, Korthuis RJ, Sun GY. Oral administration of grape polyphenol extract ameliorates cerebral ischemia/reperfusion-induced neuronal damage and behavioral deficits in gerbils: comparison of pre- and post-ischemic administration. J Nutr Biochem 2008; 20:369-77. [PMID: 18602816 DOI: 10.1016/j.jnutbio.2008.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Revised: 04/17/2008] [Accepted: 04/18/2008] [Indexed: 10/21/2022]
Abstract
Oxidative stress has been regarded as an important underlying cause for the delayed neuronal death (DND) after cerebral ischemia. In this study, the effects of short-term oral administration of grape polyphenol extract (GPE) on ischemia/reperfusion (I/R) injury in a gerbil global ischemia model were determined. Ischemia was induced by occlusion of the common carotid arteries for 5 min. GPE (30 mg/ml)-containing formula or formula without GPE was administered daily via gavage for 4 days prior to and/or for 4 days after I/R. I/R resulted in hyperlocomotion, extensive DND, oxidative and fragmented DNA damage, and an increase in reactive astrocytes and microglial cells in the hippocampal CA1 region. GPE administration for 4 days prior to I/R and for 4 days after I/R attenuated DND, DNA damage and glial cell activation. However, neuroprotection was more pronounced when GPE was administered for 4 days after I/R than when administered for 4 days prior to I/R. GPE administration after I/R attenuated I/R-induced hyperlocomotion. These findings indicate that oral GPE intake may confer protection against I/R injury and emphasize that early intervention may be an effective therapeutic measure for ameliorating brain injury in stroke.
Collapse
Affiliation(s)
- Qun Wang
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Duong TTH, Antao S, Ellis NA, Myers SJ, Witting PK. Supplementation with a synthetic polyphenol limits oxidative stress and enhances neuronal cell viability in response to hypoxia-re-oxygenation injury. Brain Res 2008; 1219:8-18. [PMID: 18538307 DOI: 10.1016/j.brainres.2008.04.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 03/28/2008] [Accepted: 04/09/2008] [Indexed: 12/31/2022]
Abstract
Oxidative stress is associated with the pathology of acute and chronic neurodegenerative disease. Cultured human neuronal cells exposed to experimental hypoxia-re-oxygenation (H/R) injury responded with an increased production of reactive oxygen species (ROS) and a significant decrease in intracellular ATP. Expression of genes encoding for hypoxia-inducible factor 1-alpha (HIF1-alpha), inducible haemoxygenase-1 (HO-1), glucose transporter-1 (Glut-1), the oxygen-sensor neuroglobin (Nb) and Cu,Zn-superoxide dismutase (SOD1), catalase (CAT) and glutathione peroxidase-1 (Gpx-1) increased significantly in response to the insult. Enhanced expression of HO-1, SOD1 and CAT correlated with an increase in the corresponding protein activity. Despite the cellular response to bolster antioxidant capacity, apoptosis and necrosis increased following H/R injury. In contrast, ROS accumulation, the endogenous gene response and cell death was limited in neuronal cells pre-incubated with 50 or 100, but not 10 microM of the phenolic antioxidant 3,3',5,5'-tetra-t-butyl-biphenyl-4,4'-diol (BP) prior to H/R injury. These data indicate that the early endogenous gene response to H/R injury is unable to inhibit neuronal dysfunction and that increasing cellular antioxidant capacity with a synthetic polyphenol (>10 microM) is potentially neuro-protective.
Collapse
Affiliation(s)
- T T Hong Duong
- Vascular Biology Group, ANZAC Research Institute, Concord Hospital, Concord, NSW 2139, Australia
| | | | | | | | | |
Collapse
|
41
|
An JJ, Lee YP, Kim SY, Lee SH, Lee MJ, Jeong MS, Kim DW, Jang SH, Yoo KY, Won MH, Kang TC, Kwon OS, Cho SW, Lee KS, Park J, Eum WS, Choi SY. Transduced human PEP-1-heat shock protein 27 efficiently protects against brain ischemic insult. FEBS J 2008; 275:1296-308. [DOI: 10.1111/j.1742-4658.2008.06291.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
Wakade C, Khan MM, De Sevilla LM, Zhang QG, Mahesh VB, Brann DW. Tamoxifen neuroprotection in cerebral ischemia involves attenuation of kinase activation and superoxide production and potentiation of mitochondrial superoxide dismutase. Endocrinology 2008; 149:367-79. [PMID: 17901229 PMCID: PMC2194601 DOI: 10.1210/en.2007-0899] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to enhance our understanding of the mechanisms of neuronal death after focal cerebral ischemia and the neuroprotective effects of tamoxifen (TMX). The phosphorylation state of 31 protein kinases/signaling proteins and superoxide anion (O(2)(-)) production in the contralateral and ipsilateral cortex was measured after permanent middle cerebral artery occlusion (pMCAO) in ovariectomized rats treated with placebo or TMX. The study revealed that pMCAO modulated the phosphorylation of a number of kinases/proteins in the penumbra at 2 h after pMCAO. Of significant interest, phospho-ERK1/2 (pERK1/2) was elevated significantly after pMCAO. TMX attenuated the elevation of pERK1/2, an effect correlated with reduced infarct size. In situ detection of O(2)(-) production showed a significant elevation at 1-2 h after pMCAO in the ischemic cortex with enhanced oxidative damage detected at 24 h. ERK activation may be downstream of free radicals, a suggestion supported by the findings that cells positive for O(2)(-) had high pERK activation and that a superoxide dismutase (SOD) mimetic, tempol, significantly attenuated pERK activation after MCAO. TMX treatment significantly reduced the MCAO-induced elevation of O(2)(-) production, oxidative damage, and proapoptotic caspase-3 activation. Additionally, pMCAO induced a significant reduction in the levels of manganese SOD (MnSOD), which scavenge O(2)(-), an effect largely prevented by TMX treatment, thus providing a potential mechanistic basis for the antioxidant effects of TMX. As a whole, these studies suggest that TMX neuroprotection may be achieved via an antioxidant mechanism that involves enhancement of primarily MnSOD levels, with a corresponding reduction of O(2)(-) production, and downstream kinase and caspase-3 activation.
Collapse
Affiliation(s)
- Chandramohan Wakade
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, 1120 15th Street, Augusta, Georgia 30912, USA
| | | | | | | | | | | |
Collapse
|
43
|
Hwang IK, Yoo KY, Kim DW, Kim SY, Park JH, Ryoo ZY, Kim J, Choi SY, Won MH. Ischemia-induced ribosomal protein S3 expressional changes and the neuroprotective effect against experimental cerebral ischemic damage. J Neurosci Res 2008; 86:1823-35. [DOI: 10.1002/jnr.21621] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Wang Q, Sun AY, Simonyi A, Kalogeris TJ, Miller DK, Sun GY, Korthuis RJ. Ethanol preconditioning protects against ischemia/reperfusion-induced brain damage: role of NADPH oxidase-derived ROS. Free Radic Biol Med 2007; 43:1048-60. [PMID: 17761301 PMCID: PMC2173699 DOI: 10.1016/j.freeradbiomed.2007.06.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Revised: 05/04/2007] [Accepted: 06/20/2007] [Indexed: 12/21/2022]
Abstract
Ethanol preconditioning (EtOH-PC) refers to a phenomenon in which tissues are protected from the deleterious effects of ischemia/reperfusion (I/R) by prior ingestion of ethanol at low to moderate levels. In this study, we tested whether prior (24 h) administration of ethanol as a single bolus that produced a peak plasma concentration of 42-46 mg/dl in gerbils would offer protective effects against neuronal damage due to cerebral I/R. In addition, we also tested whether reactive oxygen species (ROS) derived from NADPH oxidase played a role as initiators of these putative protective effects. Groups of gerbils were administered either ethanol or the same volume of water by gavage 24 h before transient global cerebral ischemia induced by occlusion of both common carotid arteries for 5 min. In some experiments, apocynin, a specific inhibitor of NADPH oxidase, was administered (5 mg/kg body wt, i.p.) 10 min before ethanol administration. EtOH-PC ameliorated behavioral deficit induced by cerebral I/R and protected the brain against I/R-induced delayed neuronal death, neuronal and dendritic degeneration, oxidative DNA damage, and glial cell activation. These beneficial effects were attenuated by apocynin treatment coincident with ethanol administration. Ethanol ingestion was associated with translocation of the NADPH oxidase subunit p67(phox) from hippocampal cytosol fraction to membrane, increased NADPH oxidase activity in hippocampus within the first hour after gavage, and increased lipid peroxidation (4-hydroxy-2-nonenal) in plasma and hippocampus within the first 2 h after gavage. These effects were also inhibited by concomitant apocynin treatment. Our data are consistent with the hypothesis that antecedent ethanol ingestion at socially relevant levels induces neuroprotective effects in I/R by a mechanism that is triggered by ROS produced through NADPH oxidase. Our results further suggest the possibility that preconditioning with other pharmacological agents that induce a mild oxidative stress may have similar therapeutic value for suppressing stroke-mediated damage in brain.
Collapse
Affiliation(s)
- Qun Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Albert Y. Sun
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Agnes Simonyi
- Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Theodore J. Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Dennis K. Miller
- Department of Psychological Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Grace Y. Sun
- Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Corresponding author: Ronald J. Korthuis, Ph.D., Department of Medical Pharmacology and Physiology, School of Medicine, One Hospital Drive, MA415, University of Missouri-Columbia, Columbia, MO 65212, Phone: (573) 882-8059, Fax: (573) 884-4276, E-mail:
| |
Collapse
|
45
|
Kim HJ, Hwang IK, Won MH. Vanillin, 4-hydroxybenzyl aldehyde and 4-hydroxybenzyl alcohol prevent hippocampal CA1 cell death following global ischemia. Brain Res 2007; 1181:130-41. [PMID: 17945203 DOI: 10.1016/j.brainres.2007.08.066] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 08/20/2007] [Accepted: 08/25/2007] [Indexed: 02/04/2023]
Abstract
Mongolian gerbils subjected to transient global ischemia exhibit neuroprotection against ischemic neuronal cell death in the hippocampal CA1 region when treated with vanillin, 4-hydroxybenzyl aldehyde (4-HBAL) and 4-hydroxybenzyl alcohol (4-HBA), which are active components of Gastrodia elata Blume. Pre- and post-insult vanillin, 4-HBAL and 4-HBA treated-animals showed a significant increase in neuronal survival (66.32%, 43.21% and 64.58%, respectively) compared to vehicle-treated animals. Animals exhibited a gender difference in this neuroprotective effect. To study the neuroprotective mechanism of 4-HBA, we investigated N-methyl-d-aspartate (NMDA) receptor 1 (NR1), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and gamma-aminobutyric acid transaminase (GABA-T) immunoreactivity at various times after ischemic insults. Treatment with 4-HBA did not affect NR1 expression levels, down-regulated 8-OHdG immunoreactivity, and increased GABA-T expression levels after global ischemia, suggesting that 4-HBA inhibited NR1 stimulation. Moreover, GABA-T was rapidly increased in the early stage after ischemia, which might enhance the survival of cells by supplying energy to the CA1 region. These results suggest that 4-HBA inhibits oxidative stress and excitotoxicity for at least 12 h and suppresses neuronal death in CA1 region. Diethyl ether fractions of GE scavenged hydroxyl radical (OH.) and showed antioxidant activity on lipid peroxidation. Vanillin and 4-HBA treatment blocked oxidative damage in PC12 cells. The neuroprotective effect has therapeutic significance and these compounds need to be evaluated for potential use in protecting against neuronal cell damage during stroke.
Collapse
Affiliation(s)
- Hyeon Ju Kim
- Department of Pharmacology, College of Pharmacy, Seoul National University, Seoul 110-799, South Korea.
| | | | | |
Collapse
|
46
|
Dmitriev LF. Shortage of lipid-radical cycles in membranes as a possible prime cause of energetic failure in aging and Alzheimer disease. Neurochem Res 2007; 32:1278-91. [PMID: 17541743 DOI: 10.1007/s11064-007-9322-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2006] [Accepted: 02/27/2007] [Indexed: 01/02/2023]
Abstract
Polyunsaturated fatty acids (PUFA) and alpha-tocopherol (alpha-TOH) are the most oxygen-sensitive constituents of cells. alpha-TOH is a member of the vitamin E family that is considered the most important lipophilic antioxidant in cell membranes. Its importance is emphasized by the involvement of oxidative stress in injury to the central nervous system and neurodegenerative diseases. Currently, alpha-TOH transfer protein (TTP), is believed to play a significant role in maintaining the vitamin status but the presence of alpha-TOH in membranes is required but not sufficient to protect the membranes against lipid hydroperoxides (LOOH) formation. The lipid-radical theory presented in this review considers the role of two membrane factors--alpha-tocopherol and cytochrome b5; these factors secure the functioning of lipid-radical cycles and the participation of lipid-radical reactions in the key membrane processes. The prominent intermembrane reaction realized via a protein-lipid interaction, during which electron transport from cytochrome b5--located in the outer membrane--to peroxyl radical (LOO*)--located in inner membrane--causes reduction of the peroxyl radical: cyt.b5red + LOO* --> cyt.b5ox + LOO(-). This secures an interaction of alpha-TOH with other intermediate, LOO(- )excepting the LOOH formation. The discussion will be focused on the consequences of ineffective electron transfer to LOO* and excessive oxidative pathway of metabolism of the PUFA (LOO* --> LOOH). Assuming the operation of cytochrome b5/alpha-tocopherol-controlled lipid-radical cycles and considering the role of the cycles in membrane bioenergetics we arrive at a model for effective function of adenine nucleotide translocator and ATP synthesis in mitochondria. This paper summarizes our experimental evidence that the oxidative and non-oxidative pathways of metabolism of PUFA via their respective intermediates occur in the cells. While this fact is not widely appreciated it may be relevant to elucidation of new mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leonid F Dmitriev
- Inst Exp Cardiology, Cardiology Research Center, Cherepkovskaya, Moscow, Russian Federation.
| |
Collapse
|
47
|
Hwang ES, Bowen PE. DNA damage, a biomarker of carcinogenesis: its measurement and modulation by diet and environment. Crit Rev Food Sci Nutr 2007; 47:27-50. [PMID: 17364694 DOI: 10.1080/10408390600550299] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Free radicals and other reactive oxygen or nitrogen species are constantly generated in vivo and can cause oxidative damage to DNA. This damage has been implicated to be important in many diseases, including cancer. The assessment of damage in various biological matrices, such as tissues, cells, and urine, is vital to understanding this role and subsequently devising intervention strategies. During the last 20 years, many analytical techniques have been developed to monitor oxidative DNA base damage. High-performance liquid chromatography-electrochemical detection and gas chromatography-mass spectrometry are the two pioneering contributions to the field. Currently, the arsenal of methods available include the promising high-performance liquid chromatography-tandem mass spectrometry technique, capillary electrophoresis, 32P-postlabeling, antibody-base immunoassays, and assays involving the use of DNA repair glycosylases such as the comet assay. The objective of this review is to discuss the biological significance of oxidative DNA damage, evaluate the effectiveness of several techniques for measurement of oxidative DNA damage in various biological samples and review current research on factors (dietary and non-dietary) that influence DNA oxidative damage using these techniques.
Collapse
Affiliation(s)
- Eun-Sun Hwang
- Department of Human Nutrition, University of Illinois at Chicago. Chicago, IL, 60612, USA
| | | |
Collapse
|
48
|
Chen SD, Wu HY, Yang DI, Lee SY, Shaw FZ, Lin TK, Liou CW, Chuang YC. Effects of rosiglitazone on global ischemia-induced hippocampal injury and expression of mitochondrial uncoupling protein 2. Biochem Biophys Res Commun 2006; 351:198-203. [PMID: 17052689 DOI: 10.1016/j.bbrc.2006.10.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Accepted: 10/04/2006] [Indexed: 11/18/2022]
Abstract
We investigate the effect of rosiglitazone, a ligand for peroxisome proliferator-activated receptor-gamma (PPARgamma) with anti-inflammatory and anti-oxidative actions, on hippocampal injury and its roles in mitochondrial uncoupling protein 2 (UCP2) expression caused by transient global ischemia (TGI) in rats. Increased UCP2 expression was observed in mitochondria of hippocampal CA1 2-24h after TGI/reperfusion, with maximal expression levels at 6-18h. Administration of rosiglitazone to hippocampus 30min prior to the onset of TGI further enhanced mitochondrial UCP2 expression 2-6h following TGI/reperfusion. Rats subjected to TGI/reperfusion displayed a significant increase in lipid peroxidation, based on increased malondialdehyde (MDA) levels, in hippocampal CA1 mitochondria 2-6 h after reperfusion. Rosiglitazone significantly attenuated TGI/reperfusion-induced lipid peroxidation and suppressed hippocampal CA1 neuronal death based on the surviving neuronal counts. In conclusion, our results provide correlative evidence for the "PPARgamma-->UCP2-->neuroprotection" cascade in ischemic brain injury.
Collapse
Affiliation(s)
- Shang-Der Chen
- Department of Neurology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim DS, Kwak SE, Kim JE, Won MH, Kang TC. The co-treatments of vigabatrin and P2X receptor antagonists protect ischemic neuronal cell death in the gerbil hippocampus. Brain Res 2006; 1120:151-60. [PMID: 16979598 DOI: 10.1016/j.brainres.2006.08.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Revised: 08/12/2006] [Accepted: 08/15/2006] [Indexed: 10/24/2022]
Abstract
During transient global ischemia, the excessive accumulation of intracellular Ca2+ induced by several episodes triggers delayed neuronal death within the vulnerable CA1 region of the hippocampus after ischemia-reperfusion insults. Although P2X receptors provide an additional source of Ca2+ entry, little data are available that these receptors could modulate the performance of the ischemic neuronal death. Therefore, we investigated the roles of the P2X receptor in the ischemic neuronal damage associated with various sequelae of transient ischemia, and the effects of their antagonist on the ischemic insults. As the results, ischemic insults increased P2X receptor expression in the gerbil hippocampus. Neither vigabatrin (VGB) nor P2X receptor antagonists (suramin, pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid) protected against the delayed neuronal death in the CA1 region of the hippocampus after ischemia. However, the co-treatments of VGB and P2X receptor antagonists effectively prevent ischemia-induced neurodegeneration. Therefore, these findings suggest that blockade of the P2X receptor accompanied by activation of GABAergic inhibition may play an important role in the neuroprotection against ischemic insults.
Collapse
Affiliation(s)
- Duk-Soo Kim
- Department of Anatomy, College of Medicine, Hallym University, Chunchon, Kangwon-Do 200-702, South Korea
| | | | | | | | | |
Collapse
|
50
|
Choi SH, Kim SY, An JJ, Lee SH, Kim DW, Won MH, Kang TC, Park J, Eum WS, Kim J, Choi SY. Immunohistochemical Studies of Human Ribosomal Protein S3 (rpS3). BMB Rep 2006; 39:208-15. [PMID: 16584637 DOI: 10.5483/bmbrep.2006.39.2.208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human ribosomal protein S3 (rpS3) was expressed in E. coli using the pET-15b vector and the monoclonal antibodies (mAbs) were produced and characterized. A total of five hybridoma cell lines were established and the antibodies recognized a single band of molecular weight of 33 kDa on immunoblot with purified rpS3. When the purified rpS3 was incubated with the mAbs, the UV endonuclease activity of rpS3 was inhibited up to a maximum of 49%. The binding affinity of mAbs to rpS3 determined by using a biosensor technology showed that they have similar binding affinities. Using the anti-rpS3 antibodies as probes, we investigated the cross-reactivities of various other mammalian brain tissues and cell lines, including human. The immunoreactive bands on Western blots appeared to be the same molecular mass of 33 kDa in all animal species tested. They also appear to be extensively cross-reactive among different organs in rat. These results demonstrated that only one type of immunologically similar rpS3 protein is present in all of the mammalian brain tissues including human. Furthermore, these antibodies were successfully applied in immunohistochemistry in order to detect rpS3 in the gerbil brain tissues. Among the various regions in the brain tissues, the rpS3 positive neurons were predominantly observed in the ependymal cells, hippocampus and stantia nigra pars compacta. The different distributions of rpS3 in brain tissues reply that rpS3 protein may play an important second function in the neuronal cells.
Collapse
Affiliation(s)
- Soo Hyun Choi
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|