1
|
Fanoudi S, Alavi MS, Hosseini M, Sadeghnia HR. Nigella sativa and thymoquinone attenuate oxidative stress and cognitive impairment following cerebral hypoperfusion in rats. Metab Brain Dis 2019; 34:1001-1010. [PMID: 31016464 DOI: 10.1007/s11011-019-00394-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 02/04/2019] [Indexed: 11/28/2022]
Abstract
Nigella sativa, a plant widely used in traditional medicine, possesses anti-inflammatory, antioxidant and neuroprotective properties. In the present study, we investigated the effect of hydroalcoholic extract of N. sativa seeds (NSE) and its active constituent, thymoquinone (TQ), on learning and memory deficits, hippocampal acetylcholine esterase (AChE) activity, and markers of redox status, mainly lipid peroxidation and superoxide dismutase (SOD) activity following cerebral hypoperfusion in rats. Cerebral hypoperfusion was induced by permanent occlusion of bilateral common carotid arteries (2VO). Male Wistar rats were administered either a vehicle (sham group: 10 ml/kg/day, ip), NSE (100, 200, and 400 mg/kg/day, ip), TQ (10, 20, and 40 mg/kg/day, ip), or donepezil (5 mg/kg/day, ip) for 10 days (three days before and seven days after ligation). Spatial learning and memory deficits were investigated using the Morris water maze (MWM) task. 2VO produced significant learning and memory deficits as evidenced by increased latency time to reach the hidden platform, increased swimming time, and decreased time spent in the target quadrant in the probe trial in the MWM task. There was also a significant increase in the lipid peroxidation level and AChE activity, and a significant decrease in SOD activity in the hippocampal portion of hypoperfused rats, as compared with the sham group. Treatment with NSE (400 mg/kg/day; p < 0.001) and TQ (40 mg/kg/day; p < 0.001), as well as donepezil significantly prevented learning and memory impairments and alleviated changes in the hippocampal lipid peroxide level and SOD and AChE activities in this model. In conclusion, our data suggest that N. sativa and thymoquinone have a beneficial role in cerebrovascular insufficiency states and dementia.
Collapse
Affiliation(s)
- Sahar Fanoudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh S Alavi
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid R Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Park KW, Kim EJ, Han HJ, Shim YS, Kwon JC, Ku BD, Park KH, Yi HA, Kim KK, Yang DW, Lee HW, Kang H, Kwon OD, Kim S, Lee JH, Chung EJ, Park SW, Park MY, Yoon B, Kim BC, Seo SW, Choi SH. Efficacy and tolerability of rivastigmine patch therapy in patients with mild-to-moderate Alzheimer's dementia associated with minimal and moderate ischemic white matter hyperintensities: A multicenter prospective open-label clinical trial. PLoS One 2017; 12:e0182123. [PMID: 28786987 PMCID: PMC5546604 DOI: 10.1371/journal.pone.0182123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/12/2017] [Indexed: 12/04/2022] Open
Abstract
Background and objective Studies investigating the impact of white matter hyperintensities (WMHs) on the response of acetylcholinesterase inhibitors in patients with Alzheimer’s disease (AD) have presented inconsistent results. We aimed to compare the effects of the rivastigmine patch between patients with AD with minimal WMHs and those with moderate WMHs. Methods Three hundred patients with mild to moderate AD were enrolled in this multicenter prospective open-label study and divided into two groups. Group 1 comprised patients with AD with minimal WMHs and group 2 comprised those with moderate WMHs. The patients were treated with a rivastigmine patch for 24 weeks. Efficacy measures were obtained at baseline and after 24 weeks. The primary endpoint was the change in the AD Assessment Scale-Cognitive subscale (ADAS-Cog) from the baseline to the end of the study. Results Of the 300 patients, there were 206 patients in group 1 and 94 patients in group 2. The intention-to-treat group comprised 198 patients (group 1, n = 136; group 2, n = 46) during the 24-week study period. Demographic factors did not differ between group 1 and group 2. There were no significant differences in change in ADAS-cog between group 1 (-0.62±5.70) and group 2 (-0.23±5.98) after the 24-week rivastigmine patch therapy (p = 0.378). The patients in group 1 had a 0.63-point improvement from baseline on the Frontal Assessment Battery, while group 2 had a 0.16-point decline compared to baseline at the end of the study (p = 0.037). The rates of adverse events (AEs) (42.6 vs. 40.3%) and discontinuation due to AEs (10.3% vs. 4.3%) did not differ between the groups. Conclusions Although the efficacy and tolerability of rivastigmine patch therapy were not associated with WMH severity in patients with AD, some improvement in frontal function was observed in those with minimal WMHs. Trial registration ClinicalTrials.gov NCT01380288
Collapse
Affiliation(s)
- Kyung Won Park
- Department of Neurology, Cognitive Disorders and Dementia Center, Dong-A University College of Medicine and Institute of Convergence Bio-Health, Busan, South Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital and Biomedical Research Institute, Pusan National University School of Medicine, Busan, South Korea
| | - Hyun Jeong Han
- Department of Neurology, Seonam University College of Medicine, Myongji Hospital, Goyang, South Korea
| | - Yong S. Shim
- Department of Neurology, Holy Family Hospital, The Catholic University of Korea, School of Medicine, Bucheon, South Korea
| | - Jae C. Kwon
- Department of Neurology, Changwon Fatima Hospital, Changwon, South Korea
| | - Bon D. Ku
- Department of Neurology, Catholic Kwandong University College of Medicine, Gangneung, South Korea
| | - Kee Hyung Park
- Department of Neurology, Gachon University Gil Hospital, Incheon, South Korea
| | - Hyon-Ah Yi
- Department of Neurology, Keimyung University College of Medicine, Daegu, South Korea
| | - Kwang K. Kim
- Department of Neurology, Dongguk University College of Medicine, Seoul, South Korea
| | - Dong Won Yang
- Department of Neurology, The Catholic University of Korea, School of Medicine, Seoul, South Korea
| | - Ho-Won Lee
- Department of Neurology, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Heeyoung Kang
- Department of Neurology, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Oh Dae Kwon
- Department of Neurology, Catholic University of Daegu School of Medicine, Daegu, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jae-Hyeok Lee
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Eun Joo Chung
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Sang-Won Park
- Department of Neurology, Daegu Fatima Hospital, Daegu, South Korea
| | - Mee Young Park
- Department of Neurology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Bora Yoon
- Department of Neurology, Konyang University College of Medicine, Daejeon, South Korea
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University School of Medicine, Gwangju, South Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, South Korea
- * E-mail:
| |
Collapse
|
3
|
Xi Y, Wang M, Zhang W, Bai M, Du Y, Zhang Z, Li Z, Miao J. Neuronal damage, central cholinergic dysfunction and oxidative damage correlate with cognitive deficits in rats with chronic cerebral hypoperfusion. Neurobiol Learn Mem 2013; 109:7-19. [PMID: 24315928 DOI: 10.1016/j.nlm.2013.11.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 11/14/2013] [Accepted: 11/24/2013] [Indexed: 12/31/2022]
Abstract
Chronic cerebral hypoperfusion has been identified to be a risk factor for cognitive decline in aging, vascular dementia, and Alzheimer's disease. Substantial evidence has shown that chronic cerebral hypoperfusion may cause cognitive impairment, but the underlying neurobiological mechanism is poorly understood so far. In this study, we used a rat model of chronic cerebral hypoperfusion by permanent bilateral common carotid artery occlusion (BCCAO) to investigate the alterations of neuronal damage, glial activation oxidative stress and central cholinergic dysfunction, and their causal relationship with the cognitive deficits induced by chronic cerebral hypoperfusion. We found that BCCAO rats exhibited spatial learning and memory impairments and working memory dysfunction 12 weeks after BCCAO compared with sham-operated rats, simultaneously accompanied by significantly increased neuronal damage and glial cell activation in the cerebral cortex and hippocampus. Twelve weeks of BCCAO treatment in rats resulted in central cholinergic dysfunction and increased oxidative damage compared with sham-operated rats. Correlational analyses revealed that spatial learning and memory impairments and working memory dysfunction were significantly correlated with the measures of neuronal damage, central cholinergic dysfunction and oxidative damage in the cerebral cortex and hippocampus of rats with BCCAO. Moreover, the measures of neuronal damage and central cholinergic dysfunction were significantly correlated with the indexes of oxidative damage in rats with BCCAO. Collectively, this study provides novel evidence that neuronal damage and central cholinergic dysfunction is likely due to increased oxidative stress under the condition of chronic cerebral hypoperfusion. Furthermore, the results of the present study suggest that neuronal damage, central cholinergic dysfunction and oxidative damage in the brain following the reduction of cerebral blood flow could be involved in cognitive deficits induced by chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Ye Xi
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Man Wang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China; Institute of Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Wei Zhang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Miao Bai
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China; Institute of Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Ying Du
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China; Institute of Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Zhuo Zhang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | - Zhuyi Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China; Institute of Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China.
| | - Jianting Miao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China.
| |
Collapse
|
4
|
Shu XJ, Liu W, Zhang L, Yang R, Yi HL, Li CL, Ye YJ, Ai YX. Effect of bis(7)-tacrine on cognition in rats with chronic cerebral ischemia. Neurosci Lett 2012; 512:103-8. [DOI: 10.1016/j.neulet.2012.01.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 12/27/2011] [Accepted: 01/26/2012] [Indexed: 11/26/2022]
|
5
|
Zakharova EI, Dudchenko AM, Svinov MM, Fedorova MM, Germanova EL. Cholinergic systems of the rat brain and neuronal reorganization under conditions of acute hypoxia. NEUROCHEM J+ 2010. [DOI: 10.1134/s1819712410040082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
Cho H, Kwon JH, Seo HJ, Kim JS. The short-term effect of acetylcholinesterase inhibitor on the regional cerebral blood flow of Alzheimer's disease. Arch Gerontol Geriatr 2010; 50:222-6. [DOI: 10.1016/j.archger.2009.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 03/19/2009] [Accepted: 03/24/2009] [Indexed: 11/26/2022]
|
7
|
Xu RX, Wu Q, Luo Y, Gong QH, Yu LM, Huang XN, Sun AS, Shi JS. PROTECTIVE EFFECTS OF ICARIIN ON COGNITIVE DEFICITS INDUCED BY CHRONIC CEREBRAL HYPOPERFUSION IN RATS. Clin Exp Pharmacol Physiol 2009; 36:810-5. [DOI: 10.1111/j.1440-1681.2009.05149.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
Barone P, Burn DJ, van Laar T, Hsu C, Poewe W, Lane RM. Rivastigmine versus placebo in hyperhomocysteinemic Parkinson's disease dementia patients. Mov Disord 2009; 23:1532-40. [PMID: 18581467 DOI: 10.1002/mds.21997] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The effects of rivastigmine versus placebo in Parkinson's disease dementia (PDD) patients with elevated or normal/low plasma homocysteine were determined. In this prospective analysis of a 24-week, randomly assigned, placebo-controlled study of rivastigmine in PDD, subpopulations comprised patients with plasma homocysteine >or=14 micromol/L (elevated) or <14 micromol/L (normal/low). Coprimary outcomes were the Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-cog) and Alzheimer Disease Cooperative Society-Clinical Global Impression of Change (ADCS-CGIC). Secondary outcomes included additional measures of cognition, including attention and executive function, daily function, and neuropsychiatric symptoms. Adverse events (AEs) were monitored. In total, 342 of 541 patients provided samples for analysis, from which 72% had elevated plasma homocysteine. Hyperhomocysteinemic patients showed treatment differences (rivastigmine vs. placebo) of 4.0 on ADAS-cog and 0.7 on ADCS-CGIC (both P < 0.01), and significant treatment differences on secondary outcomes. Rivastigmine- and placebo-treated hyperhomocysteinemic patients (16.5% and 14.6%) discontinued the study because of AEs. Patients with normal/low homocysteine showed no treatment differences on primary or secondary outcomes (1.4 on the ADAS-cog and 0.1 on ADCS-CGIC, both P = ns); 16.7% and 10.3% rivastigmine- and placebo-treated patients discontinued because of AEs. Elevated homocysteine was associated with greater rivastigmine treatment differences than normal/low homocysteine.
Collapse
Affiliation(s)
- Paolo Barone
- Dipartimento di Scienze Neurologiche, Università Federico II di Napoli, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
9
|
Wang ZF, Wang J, Zhang HY, Tang XC. Huperzine A exhibits anti-inflammatory and neuroprotective effects in a rat model of transient focal cerebral ischemia. J Neurochem 2008; 106:1594-603. [PMID: 18513368 DOI: 10.1111/j.1471-4159.2008.05504.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Huperzine A, a reversible and selective acetylcholinesterase (AChE) inhibitor, has been reported to display neuroprotective properties. The present study investigated the protective effects of huperzine A in a rat model of transient focal cerebral ischemia created by middle cerebral artery occlusion (MCAO). Huperzine A (0.1 mg/kg), administrated intraperitoneally at the onset of occlusion and 6 h later, markedly restored regional cerebral blood flow, reduced infarct size, and decreased neurological deficit score at 24 h after reperfusion. Along with inhibiting AChE activity, huperzine A inhibited nuclear translocation of transcription factor nuclear factor-kappa B, decreased overexpression of proinflammatory factors in both ipsilateral cortex and striatum, and suppressed activation of glial cells in the ischemic penumbra. Neurological deficit and glial cells activation were also reduced by daily administration of huperzine A for 14 days. Mecamylamine, a nicotinic acetylcholine receptor (nAChR) antagonist, totally abolished the inhibitory effects of huperzine A on ischemia-induced glial cells activation. Meanwhile, mecamylamine partially reversed the infarct size-reducing effects of huperzine A. In conclusion, our results demonstrate that huperzine A exhibits neuroprotective effects against transient focal cerebral ischemia-induced brain injury and suggest that the protection mechanism may involve a cholinergic anti-inflammatory pathway, in which nAChR plays an essential role.
Collapse
Affiliation(s)
- Zhi-Fei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhangjiang Hi-Tech Park, Shanghai, P.R. China
| | | | | | | |
Collapse
|
10
|
Ji X, Li C, Lu Y, Chen Y, Guo L. Post-ischemic continuous administration of galantamine attenuates cognitive deficits and hippocampal neurons loss after transient global ischemia in gerbils. Neurosci Lett 2007; 416:92-5. [PMID: 17314008 DOI: 10.1016/j.neulet.2007.01.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Revised: 01/25/2007] [Accepted: 01/25/2007] [Indexed: 10/23/2022]
Abstract
The deficits due to global cerebral ischemia show a close correlation with hippocampal CA1 neurons damage. Here we presented that galantamine showed effective protection against ischemic insults. In a transient brain global ischemic model conducted in gerbils, we investigated the alterations of performance in passive avoidance test and the histological changes between sham-operated animals (as controls) and those received post-ischemic continuous administration of saline (vehicle) or galantamine. Compared to the controls, gerbils received ischemic insults with saline treatment showed significant impairment in performance in passive avoidance test and dramatic loss of cells in hippocampal CA1 region. However, gerbils that received the same insults with galantamine (2mg/kg/day) treatment performed similarly to the controls. Consistently, most of the cells in hippocampal CA1 region which were vulnerable to ischemic insults, survived after galantamine treatment. Our results suggested that continuous application of galantamine might be effective in treatment of ischemic injury.
Collapse
Affiliation(s)
- Xunhe Ji
- Laboratory of Genetic Engineering, Institute of Biochemistry and Cell Biology, Graduate School of the Chinese Academy of Sciences, Shanghai, PR China
| | | | | | | | | |
Collapse
|
11
|
Wang ZF, Tang XC. Huperzine A protects C6 rat glioma cells against oxygen-glucose deprivation-induced injury. FEBS Lett 2007; 581:596-602. [PMID: 17257593 DOI: 10.1016/j.febslet.2007.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 12/28/2006] [Accepted: 01/10/2007] [Indexed: 10/23/2022]
Abstract
The protective effects of huperzine A against oxygen-glucose deprivation (OGD)-induced injury in C6 cells were investigated. OGD for 6h and reoxygenation for 6h enhanced phosphorylation and degradation of IkappaBalpha and nuclear translocation of nuclear factor-kappa B (NF-kappaB), triggered overexpression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and nitric oxide (NO) in C6 cells. Along with inhibiting acetylcholinesterase activity, treatment with 1 microM huperzine A inhibited activation of NF-kappaB, attenuated iNOS, COX-2 and NO overexpression, and promoted survival in C6 cells subjected to OGD insult. The protective effects of huperzine A were partly mediated by "cholinergic anti-inflammatory pathway" through alpha7 nicotinic acetylcholine receptor.
Collapse
Affiliation(s)
- Zhi Fei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, People's Republic of China
| | | |
Collapse
|
12
|
Román GC, Kalaria RN. Vascular determinants of cholinergic deficits in Alzheimer disease and vascular dementia. Neurobiol Aging 2006; 27:1769-85. [PMID: 16300856 DOI: 10.1016/j.neurobiolaging.2005.10.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Revised: 09/02/2005] [Accepted: 10/03/2005] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are widely accepted as the most common forms of dementia. Cerebrovascular lesions frequently coexist with AD, creating an overlap in the clinical and pathological features of VaD and AD. This review assembles evidence for a role for cholinergic mechanisms in the pathogenesis of VaD, as has been established for AD. We first consider the anatomy and vascularization of the basal forebrain cholinergic neuronal system, emphasizing its susceptibility to the effects of arterial hypertension, sustained hypoperfusion, and ischemic cerebrovascular disease. The impact of aging and consequences of disruption of the cholinergic system in cognition and in control of cerebral blood flow are further discussed. We also summarize preclinical and clinical evidence supporting cholinergic deficits and the use of cholinesterase inhibitors in patients with VaD. We postulate that vascular pathology likely plays a common role in initiating cholinergic neuronal abnormalities in VaD and AD.
Collapse
Affiliation(s)
- Gustavo C Román
- University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | |
Collapse
|
13
|
Claassen JAHR, Jansen RWMM. Cholinergically Mediated Augmentation of Cerebral Perfusion in Alzheimer's Disease and Related Cognitive Disorders: The Cholinergic-Vascular Hypothesis. J Gerontol A Biol Sci Med Sci 2006; 61:267-71. [PMID: 16567376 DOI: 10.1093/gerona/61.3.267] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The treatment of Alzheimer's disease (AD) with cholinesterase inhibitors (ChEIs) is based on the cholinergic hypothesis. This hypothesis fails to account for the global nature of the clinical effects of ChEIs, for the replication of these effects in other dementias, and for the strong and unpredictable intraindividual variation in response to treatment. These findings may be better explained by the premise that ChEIs primarily act by augmenting cerebral perfusion: the cholinergic-vascular hypothesis. This article will review the evidence from preclinical and clinical investigations on the vascular role of the cholinergic neural system. The clinical relevance of this hypothesis is discussed with respect to its interactions with the vascular and amyloid hypotheses of AD. Implications for treatment are indicated. Finally, we propose that the role of the cholinergic system in neurovascular regulation and functional hyperemia elucidates how the cholinergic deficit in AD contributes to the clinical and pathological features of this disease.
Collapse
Affiliation(s)
- Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | | |
Collapse
|
14
|
Zhao Q, Murakami Y, Tohda M, Watanabe H, Matsumoto K. Preventive effect of chotosan, a Kampo medicine, on transient ischemia-induced learning deficit is mediated by stimulation of muscarinic M1 but not nicotinic receptor. Biol Pharm Bull 2006; 28:1873-8. [PMID: 16204938 DOI: 10.1248/bpb.28.1873] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown using a water maze task that transient 2 vessel occlusion (T2VO) induced learning deficit in mice and that the deficit was prevented by pre-treatment of mice with chotosan, a Kampo prescription. In this study, we investigated the mechanism underlying the preventive effect of chotosan on T2VO-induced learning deficit. Chotosan administration 1 h before T2VO operation prevented learning impairment. The extract of Uncaria, a major constituent of chotosan, also had a protective effect on learning impairment in T2VO mice, whereas Uncaria-free chotosan had no beneficial effect on maze performance of T2VO mice. The ameliorative effect of chotosan was blocked by pirenzepine, a muscarinic M1 antagonist, but not by mecamylamine, a nicotinic receptor antagonist. Acetylcholine (ACh) content in the hippocampus of T2VO mice was significantly lower than that in the hippocampus of sham-operated control mice. Chotosan and Uncaria administration attenuated T2VO-induced reduction of ACh levels in the brain. These results suggest that the preventive effect of chotosan on transient ischemia-induced learning impairment is mainly attributable to the effect of Uncaria and that the ameliorative effect is mediated by stimulation of muscarinic M1 receptor.
Collapse
Affiliation(s)
- Qi Zhao
- Division of Medicinal Pharmacology, Institute of Natural Medicine, Toyama Medical and Pharmaceutical University; 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | | | |
Collapse
|
15
|
Borlongan CV, Sumaya IC, Moss DE. Methanesulfonyl fluoride, an acetylcholinesterase inhibitor, attenuates simple learning and memory deficits in ischemic rats. Brain Res 2005; 1038:50-8. [PMID: 15748872 DOI: 10.1016/j.brainres.2005.01.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2004] [Revised: 11/17/2004] [Accepted: 01/01/2005] [Indexed: 11/15/2022]
Abstract
Methanesulfonyl fluoride (MSF), a highly selective CNS inhibitor of acetylcholinesterase, has been recently demonstrated to promote improvement in cognitive performance in patients with senile dementia of Alzheimer type. Because a similar cognitive impairment may accompany stroke, we investigated in the present study whether treatment with MSF could produce beneficial effects in adult rats subjected to an experimental stroke model. Sprague-Dawley rats received transient 60 min intraluminal occlusion of the right middle cerebral artery (MCAo) and were given i.p. injections of either MSF (1 mg/kg at 24 and 48 h post-MCAo and 0.3 mg/kg thereafter every other day) or the vehicle, peanut oil, for 4 weeks. Behavioral tests and biochemical assays were performed at 28 days post-surgery. MSF treatment produced about 90% inhibition of acetylcholinesterase in the brain. Ischemic animals that received the vehicle displayed significant elevated body swing biased activity (84.8 +/- 10%) and significantly prolonged acquisition (398 +/- 62 s) and shortened retention (79 +/- 26 s) of the passive avoidance task. Interestingly, while the ischemic animals that received the MSF exhibited elevated body swing biased activity (87.7 +/- 8%), they performed significantly better in the passive avoidance task (255 +/- 36 s and 145 +/- 18 s in acquisition and retention) than the vehicle-treated animals. Moreover, whereas brains from both groups of animals revealed similar extent and degree of cerebral infarction, the MSF-treated ischemic animals showed more intense immunoreactivity, as well as a significantly higher number (10-15% increase) of septal choline acetyltransferase-positive cells than the vehicle-treated ischemic animals. These results show that MSF, possibly by preserving a functional cholinergic system, attenuated stroke-induced deficits in a simple learning and memory task.
Collapse
Affiliation(s)
- Cesario V Borlongan
- National Institutes of Health, National Institute on Drug Abuse, Intramural, Research Program, Cellular Neurophysiology, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
16
|
Farkas E, Institóris A, Domoki F, Mihály A, Luiten PGM, Bari F. Diazoxide and dimethyl sulphoxide prevent cerebral hypoperfusion-related learning dysfunction and brain damage after carotid artery occlusion. Brain Res 2004; 1008:252-60. [PMID: 15145763 DOI: 10.1016/j.brainres.2004.02.037] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2004] [Indexed: 11/21/2022]
Abstract
Chronic cerebral hypoperfusion, a mild ischemic condition is associated with advancing age and severity of dementia; however, no unanimous therapy has been established to alleviate related neurological symptoms. We imposed a permanent, bilateral occlusion of the common carotid arteries of rats (n=18) to create cerebral hypoperfusion. A mitochondrial ATP-sensitive K+ channel opener diazoxide (DZ, 5 mg/kg) or its solvent dimethyl sulphoxide (DMSO) were administered i.p. (0.25 ml) on five consecutive days after surgery. Sham-operated animals (n=18) served as control for the surgery, while nontreated rats were used as control for the treatments. Three months after the onset of cerebral hypoperfusion, the rats were tested in a hippocampus-related learning paradigm, the Morris water maze. Subsequently, the animals were sacrificed and neurons, astrocytes and microglia were labeled with immunocytochemistry in the dorsal hippocampus. DMSO and diazoxide dissolved in DMSO restored cerebral hypoperfusion-related learning dysfunction and prevented cyclooxygenase-2-positive neuron loss in the dentate gyrus. Cerebral hypoperfusion led to reduced astrocyte proliferation, which was not clearly affected by the treatment. Microglia activation was considerably enhanced by cerebral hypoperfusion, which was completely prevented by diazoxide dissolved in DMSO, but not by DMSO alone. We conclude that diazoxide can moderate ischemia-related neuroinflammation by suppressing microglial activation. Furthermore, we suggest that DMSO is a neuroprotective chemical in ischemic conditions, and it must be considerately used as a solvent for water-insoluble compounds in experimental animal models.
Collapse
Affiliation(s)
- Eszter Farkas
- Department of Anatomy, School of Medicine, University of Szeged, P.O. Box 427, Szeged 6701, Hungary.
| | | | | | | | | | | |
Collapse
|
17
|
Lojkowska W, Ryglewicz D, Jedrzejczak T, Minc S, Jakubowska T, Jarosz H, Bochynska A. The effect of cholinesterase inhibitors on the regional blood flow in patients with Alzheimer's disease and vascular dementia. J Neurol Sci 2003; 216:119-26. [PMID: 14607313 DOI: 10.1016/s0022-510x(03)00229-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effects of therapy with cholinesterase inhibitors (ChE-I) on regional cerebral blood flow (rCBF) disturbances were investigated by means of single photon emission computed tomography (SPECT). The changes in rCBF were compared with the results of the medical examination and neuropsychological tests. The sample consisted of 41 patients with the Alzheimer's dementia (AD) and vascular dementia (VaD). The effect of ChE-I (rivastigmine) treatment was studied on 33 patients, while the nontreated control group consisted of 8 patients. In the treated patients, an increase in the rCBF was observed, while the scores of the neuropsychological tests decreased slightly. In the VaD group, the increase in rCBF was more significant in the frontal regions, whereas in the group with AD in the temporal regions, respectively. In the nontreated patients, a decrease of both rCBF and scores of neuropsychological tests were observed. The scores of the neuropsychological tests correlated with the results of rCBF. Increased levels of acetylcholine in the brain after ChE-I treatment may support the cholinergic regulation of rCBF, and in result increase it. Such effects seem to be more pronounced in the more affected brain regions.
Collapse
Affiliation(s)
- Wanda Lojkowska
- Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957, Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
18
|
Iliev AI, Traykov VB, Mantchev GT, Stoykov I, Prodanov D, Yakimova KS, Krushkov IM. A post-ischaemic single administration of galanthamine, a cholinesterase inhibitor, improves learning ability in rats. J Pharm Pharmacol 2000; 52:1151-6. [PMID: 11045897 DOI: 10.1211/0022357001774921] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Transient forebrain ischaemia is widely observed in clinical practice. We have examined the effect of a single administration of the cholinesterase inhibitor galanthamine (2mg kg(-1) i.p.) 25 min after reperfusion in male Sprague-Dawley rats (180 +/- 20 g) after a 20-min common carotid artery occlusion. Twenty-four-hours post-ischaemia there was no difference in motor co-ordination or muscle tonus of the rats treated with or without galanthamine as assessed by the rota-rod test. Learning ability was examined using the shuttle-box test, evaluating the latency time and the number of errors for six days in succession. The performance of the ischaemic saline-injected rats was significantly impaired on days 4, 5, 6 (latency time) compared with the non-ischaemic rats and with the ischaemic animals administered galanthamine (P < 0.05). Similar results were obtained when counting the number of errors (failure to cross the cage during conditioned or unconditioned stimulus). The monitoring of body temperature during the first 12-h post-ischaemia did not show any significant difference between the groups. The data showed a beneficial effect of galanthamine on the recovery of learning ability when administered once only post-ischaemia. This suggests a direct effect on the early pathologic mechanisms of CNS damage. Cholinesterase inhibitors may prove useful in the early clinical treatment of ischaemic conditions.
Collapse
Affiliation(s)
- A I Iliev
- Department of Pharmacology and Toxicology, Medical University, Sofia, Bulgaria
| | | | | | | | | | | | | |
Collapse
|
19
|
Kumar V, Anand R, Messina J, Hartman R, Veach J. An efficacy and safety analysis of Exelon in Alzheimer's disease patients with concurrent vascular risk factors. Eur J Neurol 2000; 7:159-69. [PMID: 10809936 DOI: 10.1046/j.1468-1331.2000.00046.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We evaluated the efficacy and safety of the centrally acting cholinesterase inhibitor, rivastigmine tartrate, for patients with mild to moderately severe Alzheimer's disease (AD) with or without concurrent vascular risk factors (VRF). Patients (45-90 years of age) were randomized to placebo (n = 235), low-dose rivastigmine (1-4 mg/day, n = 233), or high-dose rivastigmine (6-12 mg/day, n = 231) for 26 weeks. Efficacy measures included the Alzheimer's Disease Assessment Scale-Cognitive subscale (ADAS-Cog), the Clinician's Interview Based Impression of Change (CIBIC-Plus), the Progressive Deterioration Scale (PDS), the Global Deterioration Scale (GDS), and the Mini-Mental State Examination (MMSE). For efficacy and safety analysis, patients were categorized by baseline Modified Hachinski Ischemic Score (MHIS) for the determination of VRF (MHIS > 0: presence of VRF; MHIS = 0: absence of VRF). As early as 12 weeks, the mean change from the baseline ADAS-Cog score was significantly different for those patients treated with high-dose rivastigmine compared with placebo controls in both MHIS categories. However, the treatment difference between high-dose rivastigmine and placebo at each time-point was larger for patients with MHIS > 0. The proportion of responders was significantly greater in the high-dose rivastigmine group for each level of improvement. No differences were noted between treatment groups regarding safety evaluations. Rivastigmine is effective in both categories of patients, and those with VRF experience greater clinical benefit (cognition, activities of daily living, and disease severity).
Collapse
Affiliation(s)
- V Kumar
- Florida Institute of Neurosciences and Clinical Research, 530 S. Nokomis Avenue, Suite 14, Venice, FL 34284, USA
| | | | | | | | | |
Collapse
|
20
|
Jiang ZW, Gong QZ, Di X, Zhu J, Lyeth BG. Dicyclomine, an M1 muscarinic antagonist, reduces infarct volume in a rat subdural hematoma model. Brain Res 2000; 852:37-44. [PMID: 10661493 DOI: 10.1016/s0006-8993(99)02230-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The rat subdural hematoma (SDH) model produces a zone of ischemic brain damage within the hemisphere beneath the SDH. Previous studies have measured large increases in extracellular acetylcholine during cerebral ischemia in the rat. We examined infarct volume after selectively blocking muscarinic M1 receptors with dicyclomine during SDH. Rats were anesthetized with isoflurane (2%), intubated, and femoral artery and vein cannulated. Autologous blood (0.375 ml) was injected (0.05 ml/min) under the dura of the right parietal cortex. Dicyclomine (5 mg/kg, i.v.) was injected at 5 min after and again at 2 h after completion of the subdural blood infusion. Blood pressure and intracranial pressure (ICP) were continuously measured. At 4 h after SDH rats were euthanized, brains sectioned, and immunoreacted with glia fibrillary acidic protein. Cortical infarct volume was quantified in coronal brain sections at 0.7-mm intervals from +1.0 mm to -3.9 mm relative to bregma. Infarct volume in drug-treated rats (n = 10) 22.1 +/- 6.99 mm3 was significantly smaller (p < 0.02) than vehicle treated rats (n = 10) 56.7 +/- 9.59 mm3. ICP, blood pressure and cerebral perfusion pressure were not significantly different between groups. These data suggest that activation of M1 muscarinic receptors during an ischemic event may contribute to the development of subsequent pathology.
Collapse
Affiliation(s)
- Z W Jiang
- Division of Neurosurgery, Medical College of Virginia Campus, Virginia Commonwealth University Richmond, USA
| | | | | | | | | |
Collapse
|
21
|
Park CH, Lee YJ, Lee SH, Choi SH, Kim HS, Jeong SJ, Kim SS, Suh YH. Dehydroevodiamine.HCl prevents impairment of learning and memory and neuronal loss in rat models of cognitive disturbance. J Neurochem 2000; 74:244-53. [PMID: 10617126 DOI: 10.1046/j.1471-4159.2000.0740244.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We previously reported that dehydroevodiamine.HCl (DHED) has anticholinesterase and antiamnesic activities. To verify the effects of DHED on cognitive deficits further, we tested it on the scopolamine-induced amnesia model of the rat using the passive avoidance and eight-arm radial maze tests. A single (20 mg/kg p.o.) and repeated (10 mg/kg p.o.) administrations of DHED could significantly reverse the latency time shortened by scopolamine (1 mg/kg i.p.) to control level. The impaired spatial working memory induced by scopolamine (1 mg/kg i.p.) was also improved significantly by a single injection (6.25 mg/kg i.p.) and repeated administrations of DHED (10 mg/kg p.o.) in the eight-arm radial maze test. In addition, we examined the effects of DHED on the memory impairment and the histological changes of the brain after unilateral electrolytic lesion of the entorhinal cortex (EC) and middle cerebral artery occlusion in rats. The cognitive deficits caused by EC lesion and middle cerebral artery occlusion were improved significantly by repeated administrations of DHED (6.25 mg/kg i.p.) after EC lesion or ischemic insult once a day for 7 days in the passive avoidance test. Histological analysis showed that the neuronal loss in the DHED-treated group was notably reduced in the hippocampal area (CA1) of ischemic rats and in the dentate gyrus and hippocampal area (CA1 and CA3) of EC-lesioned rats compared with the nontreated group. The infarction area was decreased significantly by a single administration of DHED (6.25 mg/kg i.p.) 30 min before ischemic insult for 6 h. These results suggest that DHED might be an effective drug for not only the Alzheimer's disease type, but also the vascular type of dementia.
Collapse
Affiliation(s)
- C H Park
- Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Seoul National University, Chongno-gu, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Low PA, Novak V, Spies JM, Novak P, Petty GW. Cerebrovascular Regulation in the Postural Orthostatic Tachycardia Syndrome (POTS). Am J Med Sci 1999. [DOI: 10.1016/s0002-9629(15)40486-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Low PA, Novak V, Spies JM, Novak P, Petty GW. Cerebrovascular regulation in the postural orthostatic tachycardia syndrome (POTS). Am J Med Sci 1999; 317:124-33. [PMID: 10037116 DOI: 10.1097/00000441-199902000-00007] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Patients with the postural orthostatic tachycardia syndrome (POTS) have symptoms of orthostatic intolerance despite having a normal orthostatic blood pressure (BP), which suggests some impairment of cerebrovascular regulation. Cerebrovascular autoregulation refers to the maintenance of normal cerebral blood flow in spite of changing BP. Mechanisms of autoregulation include myogenic, metabolic and neurogenic vasoregulation. Beat-to-beat recording of blood-flow velocity (BFV) is possible using transcranial Doppler imaging. It is possible to evaluate autoregulation by regressing deltaBFV to deltaBP during head-up tilt. A number of dynamic methods, relating deltaBFV to deltaBP during sudden induced changes in BP by occluding then releasing peripheral arterial flow or by the Valsalva maneuver. The deltaBFV to deltaBP provides an index of autoregulation. In orthostatic hypotension, the autoregulated range is typically expanded. In contrast, paradoxical vasoconstriction occurs in POTS because of an increased depth of respiration, resulting in hypocapnic cerebrovascular constriction, and impaired autoregulation.
Collapse
Affiliation(s)
- P A Low
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
UNLABELLED Rivastigmine (SDZ ENA 713) is a carbamylating, long-acting reversible and noncompetitive carbamate acetylcholinesterase inhibitor that is indicated as an oral treatment for patients with mild to moderately severe Alzheimer's disease. The drug has been evaluated for this use in 3 well designed, adequately powered, phase II/III, 26-week clinical trials that included a total of 1479 rivastigmine and 647 placebo recipients. Most of these patients had concomitant disorders that were being treated with numerous other drugs. Individual and pooled results of these trials indicate that rivastigmine 6 to 12 mg/day usually produces cognitive, global and functional changes that indicate significantly less deterioration than was observed with placebo in patients with mild to moderately severe Alzheimer's disease. Individual results of the 2 pivotal trials and pooled analysis also show that, compared with placebo recipients, significantly more rivastigmine 6 to 12 mg/day recipients respond to therapy. Indeed, after 26 weeks of therapy in the 2 pivotal trials, significantly more rivastigmine 6 to 12 mg/day than placebo recipients achieved clinically meaningful improvements as defined by 3 separate response criteria. The lower dosage range of 1 to 4 mg/day was not as effective as 6 to 12 mg/day, as measured using these criteria and other efficacy parameters. Rivastigmine causes adverse events that are generally those expected from an acetylcholinesterase inhibitor. They are usually mild to moderate, of short duration and responsive to dosage reduction. Unpublished data from 3989 patients indicate that rivastigmine and placebo were associated with similar incidences of serious adverse events and changes in laboratory parameters, ECG and cardiorespiratory vital signs. The most common events were gastrointestinal, central and peripheral nervous system and whole body adverse events. However, compared with placebo, rivastigmine more commonly caused adverse events resulting in treatment withdrawal. These events were most frequently gastrointestinal and were more common in women. CONCLUSION Rivastigmine is a useful option for the treatment of patients with mild to moderately severe Alzheimer's disease. Although only short term (6- month) comparisons with placebo are available, given the lack of established treatment options it should be considered for first-line use in this population.
Collapse
Affiliation(s)
- C M Spencer
- Adis International Limited, Auckland, New Zealand.
| | | |
Collapse
|
25
|
Chen Y, Shohami E, Constantini S, Weinstock M. Rivastigmine, a brain-selective acetylcholinesterase inhibitor, ameliorates cognitive and motor deficits induced by closed-head injury in the mouse. J Neurotrauma 1998; 15:231-7. [PMID: 9555969 DOI: 10.1089/neu.1998.15.231] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The effects of Rivastigmine, a novel centrally-acting anticholinesterase agent, were evaluated on cerebral edema, neurological and motor deficits, and impairment of spatial memory induced in mice by closed-head injury (CHI). Severe injury was induced in the left hemisphere of mice under ether anesthesia. Rivastigmine (1 or 2 mg/kg) or saline (10 ml/kg) was injected SC 5 min later. Rivastigmine (2 mg/kg) reduced cerebral edema by at least 50% (p < 0.01), 24 h after CHI and accelerated the recovery of motor function 7 and 14 days after CHI. Control mice (n = 24), previously trained to find the goal platform in a Morris water maze failed to recall or relearn its position for at least 11 days post-injury. Those given a single injection of Rivastigmine (2 mg/kg) regained their pre-test latencies by the third day after CHI. The neuroprotective effects of Rivastigmine on brain edema, neurological and motor function, and performance in the Morris water maze were completely antagonized by simultaneous SC injection of either scopolamine (0.5 mg/kg) or mecamylamine (2.5 mg/kg). The antagonists alone had no significant effect on any of these parameters. These data show that the reduction by Rivastigmine of the immediate and long-term sequelae of brain injury are mediated by increased cholinergic activity at both muscarinic and nicotinic receptors.
Collapse
Affiliation(s)
- Y Chen
- Department of Pharmacology, School of Pharmacy, The Hebrew University of Jerusalem, Israel
| | | | | | | |
Collapse
|
26
|
Chen Y, Shohami E, Bass R, Weinstock M. Cerebro-protective effects of ENA713, a novel acetylcholinesterase inhibitor, in closed head injury in the rat. Brain Res 1998; 784:18-24. [PMID: 9518537 DOI: 10.1016/s0006-8993(97)00982-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Focal ischemic brain damage and diffuse brain swelling occur in severe cases of traumatic head injury. Ischemia decreases brain acetylcholine (ACh) levels and head trauma upregulates acetylcholinesterase (AChE) in experimental animal models. The present study determined whether a brain-selective AChE inhibitor, ENA713, given once, up to 2 h after closed head injury (CHI) could reduce the vasogenic edema and accelerate recovery from neurological deficits induced by the injury in rats. ENA713 1-5 mg/kg produced a dose-related inhibition of AChE ranging from 40-85% in the cortex and hippocampus. Doses of 1, 2 and 5 mg/kg, significantly reduced the motor and neurological deficits and speeded recovery, as indicated by measurements made 7 and 14 days after injury. The two larger doses were still effective when injected 1 or 2 h after CHI. The acceleration by ENA713 of recovery of motor function was independent of its reduction in body temperature and was prevented by the simultaneous injection of mecamylamine (2.5 mg/kg), but not by scopolamine (0.2 or 1 mg/kg). Edema in the contused hemisphere (24 h after injury) and disruption of the blood brain barrier (4 h after injury) were significantly reduced (about 50%) by doses of 2 and 5 mg/kg, but not by 1 mg/kg. The data support the hypothesis that ENA713 exerts a neuroprotective effect in brain injury by preventing the decrease in cholinergic activity in cerebral vessels and in neurones.
Collapse
Affiliation(s)
- Y Chen
- Department of Pharmacology, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | | | | |
Collapse
|
27
|
Sadoshima S, Nagao T, Okada Y, Fujii K, Ibayashi S, Fujishima M. L-arginine ameliorates recirculation and metabolic derangement in brain ischemia in hypertensive rats. Brain Res 1997; 744:246-52. [PMID: 9027384 DOI: 10.1016/s0006-8993(96)01063-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The effects of L-arginine (a precursor of nitric oxide, NO) on cerebral blood flow (CBF), cerebrovascular resistance (CVR) and metabolites in the ischemic brain were examined in spontaneously hypertensive rats with bilateral carotid artery occlusion for 30 min followed by 60 min-recirculation. The administration of L-arginine (300 mg/kg, i.v.) increased the CBF by an average of 11 ml x 100 g-1 x min-1 (P < 0.05 vs. at rest), and N(omega)-nitro-L-arginine (L-NNA, an inhibitor of NO synthase, 5 mg/kg, i.v.) reduced the CBF by 5-6 ml x 100 g-1.min-1 with increase in the mean arterial pressure by 26 mmHg. During ischemia the CBF significantly decreased to below 8% of the resting values in all rats. The largest blood flow in postischemic hyperemia was 171 +/- 9% of the resting CBF in the rats with L-arginine (P < 0.05 vs. L-NNA and saline), followed by 126 +/- 5 with saline and 109 +/- 3 with L-NNA. The CVR at 60 min of recirculation was 3.291 +/- 0.144 mmHg . ml-1. 100 g-1 .min-1 in the rats with saline, remained low level of 2.711 +/- 0.124 with L-arginine (P < 0.01 vs. L-NNA and P < 0.05 vs. saline) and in contrast, significantly increased to 5.732 +/- 0.184 with L-NNA (P < 0.01 vs. L-arginine and saline, respectively). Tissue lactate with saline increased 2.3-fold at 60 min of recirculation, whereas the increase was inhibited to 1.4-fold after L-arginine treatment (P < 0.01 vs. L-NNA) and in contrast, significantly increased 5.7-fold with L-NNA. The ATP and glucose levels were better preserved in the rats with L-arginine than in those with L-NNA or saline. These findings support that the enhanced postischemic hyperemia is beneficial to the ischemic brain and the administration of L-arginine may be potentially useful for the treatment of acute stroke.
Collapse
Affiliation(s)
- S Sadoshima
- Second Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|