1
|
Poore CP, Yang J, Wei S, Fhu CK, Bichler Z, Wang J, Soong TW, Liao P. Enhanced isradipine sensitivity in vascular smooth muscle cells due to hypoxia-induced Ca v1.2 splicing and RbFox1/Fox2 downregulation. FEBS J 2024; 291:4265-4285. [PMID: 38794806 DOI: 10.1111/febs.17159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/15/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Calcium influx via the L-type voltage-gated Cav1.2 calcium channel in smooth muscle cells regulates vascular contraction. Calcium channel blockers (CCBs) are widely used to treat hypertension by inhibiting Cav1.2 channels. Using the vascular smooth muscle cell line, A7r5 and primary culture of cerebral vascular smooth muscle cells, we found that the expression and function of Cav1.2 channels are downregulated during hypoxia. Furthermore, hypoxia induces structural changes in Cav1.2 channels via alternative splicing. The expression of exon 9* is upregulated, whereas exon 33 is downregulated. Such structural alterations of Cav1.2 channels are caused by the decreased expression of RNA-binding proteins RNA-binding protein fox-1 homolog 1 and 2 (RbFox1 and RbFox2). Overexpression of RbFox1 and RbFox2 prevents hypoxia-induced exon 9* inclusion and exon 33 exclusion. Importantly, such structural alterations of the Cav1.2 channel partly contribute to the enhanced sensitivity of Cav1.2 to isradipine (a CCB) under hypoxia. Overexpression of RbFox1 and RbFox2 successfully reduces isradipine sensitivity in hypoxic smooth muscle cells. Our results suggest a new strategy to manage ischemic diseases such as stroke and myocardial infarction.
Collapse
MESH Headings
- Calcium Channels, L-Type/metabolism
- Calcium Channels, L-Type/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- RNA Splicing Factors/genetics
- RNA Splicing Factors/metabolism
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Alternative Splicing
- Down-Regulation
- Rats
- Cell Hypoxia/genetics
- Exons/genetics
- Mice
- Calcium Channel Blockers/pharmacology
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
Collapse
Affiliation(s)
| | - Jialei Yang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Shunhui Wei
- Calcium Signaling Laboratory, National Neuroscience Institute, Singapore City, Singapore
| | - Chee Kong Fhu
- Calcium Signaling Laboratory, National Neuroscience Institute, Singapore City, Singapore
| | - Zoë Bichler
- Neurobehavioural Phenotyping Core, Center for Biometric Analysis, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Tuck Wah Soong
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Ping Liao
- Calcium Signaling Laboratory, National Neuroscience Institute, Singapore City, Singapore
| |
Collapse
|
2
|
Serag I, Abouzid M, Elmoghazy A, Sarhan K, Alsaad SA, Mohamed RG. An updated systematic review of neuroprotective agents in the treatment of spinal cord injury. Neurosurg Rev 2024; 47:132. [PMID: 38546884 DOI: 10.1007/s10143-024-02372-6] [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: 09/16/2023] [Revised: 01/03/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
This systematic review aims to summarize the findings from all clinical randomized trials assessing the efficacy of potential neuroprotective agents in influencing the outcomes of acute spinal cord injuries (SCI). Following the PRISMA guidelines, we conducted comprehensive searches in four electronic databases (PubMed, Scopus, Cochrane Library, and Web of Science) up to September 5th, 2023. Our analysis included a total of 30 studies. We examined the effects of 15 substances/drugs: methylprednisolone, tirilazad mesylate, erythropoietin, nimodipine, naloxone, Sygen, Rho protein antagonist, granulocyte colony-stimulating factor, autologous macrophages, autologous bone marrow cells, vitamin D, progesterone, riluzole, minocycline, and blood alcohol concentration. Notable improvements in neurological outcomes were observed with progesterone plus vitamin D and granulocyte colony-stimulating factor. In contrast, results for methylprednisolone, erythropoietin, Sygen, Rho Protein, and Riluzole were inconclusive, primarily due to insufficient sample size or outdated evidence. No significant differences were found in the remaining evaluated drugs. Progesterone plus vitamin D, granulocyte colony-stimulating factor, methylprednisolone, Sygen, Rho Protein, and Riluzole may enhance neurological outcomes in acute SCI cases. It is worth noting that different endpoints or additional subgroup analyses may potentially alter the conclusions of individual trials. Therefore, certain SCI grades may benefit more from these treatments than others, while the overall results may remain inconclusive.
Collapse
Affiliation(s)
- Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St, 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | | | - Khalid Sarhan
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Rashad G Mohamed
- Mansoura Manchester Program for Medical Education, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
3
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
Li J, Xiang H, Huang C, Lu J. Pharmacological Actions of Myricetin in the Nervous System: A Comprehensive Review of Preclinical Studies in Animals and Cell Models. Front Pharmacol 2021; 12:797298. [PMID: 34975495 PMCID: PMC8716845 DOI: 10.3389/fphar.2021.797298] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/01/2022] Open
Abstract
Myricetin is a natural flavonoid extracted from a variety of plants, such as medicinal herbs, vegetables, berries, and tea leaves. A growing body of evidence has reported that myricetin supplementation display therapeutic activities in a lot of nervous system disorders, such as cerebral ischemia, Alzheimer’s disease, Parkinson’s disease, epilepsy, and glioblastoma. Myricetin supplementation can also protect against pathological changes and behavioral impairment induced by multiple sclerosis and chronic stress. On the basis of these pharmacological actions, myricetin could be developed as a potential drug for the prevention and/or treatment of nervous system disorders. Mechanistic studies have shown that inhibition of oxidative stress, cellular apoptosis, and neuroinflammatory response are common mechanisms for the neuroprotective actions of myricetin. Other mechanisms, including the activation of the nuclear factor E2-related factor 2 (Nrf2), extracellular signal-regulated kinase 1/2 (ERK1/2), protein kinase B (Akt), cyclic adenosine monophosphate-response element binding protein (CREB), and brain-derived neurotrophic factor (BDNF) signaling, inhibition of intracellular Ca2+ increase, inhibition of c-Jun N-terminal kinase (JNK)-p38 activation, and suppression of mutant protein aggregation, may also mediate the neuroprotective effects of myricetin. Furthermore, myricetin treatment has been shown to promote the activation of the inhibitory neurons in the hypothalamic paraventricular nucleus, which subsequently produces anti-epilepsy effects. In this review, we make a comprehensive understanding about the pharmacological effects of myricetin in the nervous system, aiming to push the development of myricetin as a novel drug for the treatment of nervous system disorders.
Collapse
Affiliation(s)
- Jie Li
- Department of Gastroenterology, The People’s Hospital of Taizhou, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| | - Haitao Xiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Jiashu Lu
- Department of Pharmacy, The People’s Hospital of Taizhou, The Fifth Affiliated Hospital of Nantong University, Taizho, China
- *Correspondence: Jiashu Lu,
| |
Collapse
|
5
|
Chen Y, Guo Z, Peng X, Xie W, Chen L, Tan Z. Nimodipine represses AMPK phosphorylation and excessive autophagy after chronic cerebral hypoperfusion in rats. Brain Res Bull 2018; 140:88-96. [PMID: 29625150 DOI: 10.1016/j.brainresbull.2018.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/29/2018] [Accepted: 03/30/2018] [Indexed: 12/29/2022]
Abstract
Chronic cerebral hypofusion (CCH) after bilateral carotid artery occlusion (2VO) causes cognitive damage and neuronal degeneration in the cortex and hippocampal CA1 area, and influences the oxygen and glucose supply in the brain which often results in metabolic alterations and oxidative stress. AMP-activated protein kinase (AMPK) phosphorylation, a sensor of cellular energy status, directs metabolic adaptation to support cellular growth and survival after CCH. Autophagy is also likely to be involved in metabolic adaptation and plays an important role in neuronal deterioration and cognitive decline after CCH. Nimodipine, an L-type calcium channel antagonist, has been reported to exert neuroprotective effects. However, the potential role of nimodipine in autophagy and the energy sensing AMPK signal is not well understood. In addition, little is known about the relationship between autophagy and AMPK signal. Here, we designed a way to evaluate these issues. Adult male Wistar rats were subjected to 2VO and randomly divided into three groups: the Vehicle (2VO), Nimodipine (2VO + nimodipine 10 mg/kg) groups. A third group served as sham controls. Each group was investigated at 2 and 4 weeks post gavage and tested using the Morris water maze. The activities of LC3B and AMPK signal were examined using immunohistochemistry and western blotting. Nimodipine significantly alleviated spatial learning and memory impairments and the number of lesion neurons. At 2 weeks of durg administration, these drug effects, suppressing AMPK activation and excessive autophagy, were more pronounced at the cortex than at hippocampal CA1 area. The effects of nimodipine were significant in the hippocampal CA1 area after 4 weeks of administration. Furthermore, nimodipine inhibited expression of eIF2α/ATF4 signaling related to energy deficit stress in 2VO rats. These results suggest that excessive autophagy has promoted neuronal and tissue injury after 2VO in rats. Nimodipine protected the brain from CCH by inhibiting the autophagy activity. The p-AMPK and eIF2α/ATF4 pathway is likely part of an integrated pro-autophagy signaling network after CCH.
Collapse
Affiliation(s)
- Yan Chen
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China; Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Zhenli Guo
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Xingming Peng
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Wenting Xie
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Lizhu Chen
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Zihu Tan
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Province Academy of Traditional Chinese Medicine, Wuhan, Hubei 430074, China.
| |
Collapse
|
6
|
Tan Z, Chen Y, Xie W, Liu X, Zhu Y, Zhu Y. Nimodipine attenuates tau phosphorylation at Ser396 via miR-132/GSK-3β pathway in chronic cerebral hypoperfusion rats. Eur J Pharmacol 2017; 819:1-8. [PMID: 29042207 DOI: 10.1016/j.ejphar.2017.10.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/20/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022]
Abstract
Chronic cerebral hypofusion (CCH) promotes hyperphosphorylation of tau proteins, a key neuropathological trait that reflects neurodegenerative disorders. Nimodipine, an L-type calcium channel antagonist, has been reported to show neuroprotective effects. In this study, we investigated the potential mechanism of nimodipine in tauopathies induced by CCH. MiR-132 is downregulated in tauopathies such as AD and directly targets tau mRNA to regulate its expression. Here, we report that CCH induced miR-132 deficiency and increased tau phosphorylation at Ser396 while tau expression was not influenced. Nimodipine treatment attenuated CCH induced tau phosphorylation by up-regulating expression of miR-132. Furthermore, nimodipine inhibited activation of GSK-3β and neuronal apoptosis induced by CCH. Interestingly, GSK-3βmRNA level negatively correlated with the expression of miR-132. These findings support a role for nimodipine inhibiting tau phosphorylation at Ser396 via miR-132/GSK-3β. Therefore, nimodipine may be a candidate for the treatment of tauopathy present in CCH.
Collapse
Affiliation(s)
- Zihu Tan
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Province Academy of Traditional Chinese Medicine, Wuhan, Hubei 430074, China
| | - Yan Chen
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China.
| | - Wenting Xie
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Xi Liu
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Yuanyue Zhu
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Yan Zhu
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
7
|
Hu M, Liu Z, Lv P, Wang H, Zhu Y, Qi Q, Xu J, Gao L. Nimodipine activates neuroprotective signaling events and inactivates autophages in the VCID rat hippocampus. Neurol Res 2017; 39:904-909. [PMID: 28782464 DOI: 10.1080/01616412.2017.1356157] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Autophagy and phosphatidylinositol 3-kinase (PI3K)/Akt kinase pathways are implicated in cognitive decline associated with cerebrovascular lesions. This decline is reflected in the concept of vascular cognitive impairment and dementia (VCID). However, the underlying molecular mechanism and specific details regarding these types of cognitive deficits induced by chronic brain hypoperfusion have not been elucidated. METHODS We designed a method to evaluate these mechanisms. Adult male Sprague-Dawley rats were subjected to permanent bilateral occlusion of the common carotid artery (2VO) and randomly divided into three groups: Sham, Vehicle (2VO), and Nimodipine10 (2VO + nimodipine 10 mg/kg). Each group was studied for 4 weeks postoperatively and assessed by the Morris water maze. RESULTS The results of this study show that chronic brain hypoperfusion significantly increased the number of autophagic vacuoles with high LC3 II levels, but it decreased p-Akt and p-CREB levels, which were involved in the PI3K/Akt kinase pathway in the hippocampi of rats. Additionally, significant cognitive losses were observed following 2VO. Further analysis showed that, in VCID rats subjected to 2VO, nimodipine administration decreased autophagy, increased the Akt/CREB signaling pathway and significantly reduced brain damage. CONCLUSIONS We concluded that neuronal pathology and activation of the autophagic and Akt/CREB signaling pathway caused by chronic brain hypoperfusion could suppress cognitive behavior, which may provide a novel way for the prevention of VCID. The results of this study indicate that nimodipine protected the brain from chronic brain hypoperfusion damage by suppressing autophagy and activating the Akt/CREB signaling pathway.
Collapse
Affiliation(s)
- Ming Hu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Zhijuan Liu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Peiyuan Lv
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Hebo Wang
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Yifei Zhu
- b Department of Neurology , the Second Hospital of Hebei Medical University , Shijiazhuang , People's Republic of China
| | - Qianqian Qi
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Jing Xu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Lei Gao
- c Department of Ultrasonography , the First Central Hospital of Baoding , Baoding , People's Republic of China
| |
Collapse
|
8
|
Hu M, Liu Z, Lv P, Wang H, Zhu Y, Qi Q, Xu J. Autophagy and Akt/CREB signalling play an important role in the neuroprotective effect of nimodipine in a rat model of vascular dementia. Behav Brain Res 2017; 325:79-86. [PMID: 27923588 DOI: 10.1016/j.bbr.2016.11.053] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/26/2022]
Abstract
The Akt/CREB signalling pathway is involved in neuronal survival and protection. Autophagy is also likely to be involved in survival mechanisms. Nimodipine is an L-type calcium channel antagonist that reduces excessive calcium influx during pathological conditions (contributing to its neuroprotective properties). However, the potential role of nimodipine in autophagic and Akt/CREB signalling is not well understood. In addition, little is known about the relationship between autophagic and Akt/CREB signalling. Here, we designed a way to evaluate these issues. Adult male Sprague-Dawley rats were subjected to permanent bilateral occlusion of the common carotid artery (2VO) and randomly divided into three groups: the Vehicle (2VO), Nimodipine10 (2VO+nimodipine 10mg/kg), and Nimodipine20 (2VO+nimodipine 20mg/kg) groups. A fourth group of animals served as Sham controls. Each group was investigated at 4 and 8 weeks post-operatively and assessed using the Morris water maze. Nimodipine significantly alleviated spatial learning and memory impairments and inhibited the loss of neurons in the CA1 region of the hippocampus. These drug effects were more pronounced at 8 weeks than at 4 weeks. The activities of LC3 II p-Akt and p-CREB were examined using immunohistochemistry and western blotting. Suppressing autophagy induced pyramidal cell death without affecting increased pro-survival signalling induced by nimodipine. Nimodipine protected the brain from chronic cerebral hypoperfusion by activating the Akt/CREB signalling pathway. Autophagy has a neuroprotective effect on rats after 2VO. Autophagy is likely part of an integrated survival signalling network involving the Akt/CREB pathway.
Collapse
Affiliation(s)
- Ming Hu
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China
| | - Zhijuan Liu
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China
| | - Peiyuan Lv
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China.
| | - Hebo Wang
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China
| | - Yifei Zhu
- Department of Neurology, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Xinhua District, Shijiazhuang 050000, Hebei Province, People's Republic of China
| | - Qianqian Qi
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei Province, People's Republic of China
| |
Collapse
|
9
|
Villa RF, Ferrari F, Moretti A. Effects of Neuroprotectants Before and After Stroke: Statins and Anti-hypertensives. SPRINGER SERIES IN TRANSLATIONAL STROKE RESEARCH 2017. [DOI: 10.1007/978-3-319-45345-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
10
|
Young LM, Geldenhuys WJ, Domingo OC, Malan SF, Van der Schyf CJ. Synthesis and Biological Evaluation of Pentacycloundecylamines and Triquinylamines as Voltage-Gated Calcium Channel Blockers. Arch Pharm (Weinheim) 2016; 349:252-67. [PMID: 26892182 PMCID: PMC5482218 DOI: 10.1002/ardp.201500293] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 01/21/2023]
Abstract
Preclinical studies for neurodegenerative diseases have shown a multi-targeted approach to be successful in the treatment of these complex disorders with several pathoetiological pathways. Polycyclic compounds, such as NGP1-01 (7a), have demonstrated the ability to target multiple mechanisms of the complex etiology and are referred to as multifunctional compounds. These compounds have served as scaffolds with the ability to attenuate Ca(2+) overload and excitotoxicity through several pathways. In this study, our focus was on mitigating Ca(2+) overload through the L-type calcium channels (LTCC). Here, we report the synthesis and biological evaluation of several novel polycyclic compounds. We determined the IC50 values for both the pentacycloundecylamines and the triquinylamines by means of a high-throughput fluorescence calcium flux assay utilizing Fura-2/AM. The potential of these compounds to offer protection against hydrogen peroxide-induced cell death was also evaluated. Overall, 8-benzylamino-8,11-oxapentacyclo[5.4.0.0(2,6) .0(3,10) .0(5,9) ]undecane (NGP1-01, 7a) had the most favorable pharmacological profile with an IC50 value of 86 µM for LTCC inhibition and significant reduction of hydrogen peroxide-induced cell death. In general, the triquinylamines were more active as LTCC blockers than the oxa-pentacycloundecylamines. The aza-pentacycloundecylamines were potent LTCC inhibitors, with 8-hydroxy-N-phenylethyl-8,11-azapentacyclo[5.4.0.0(2,6) .0(3,10) .0(5,9) ]undecane (8b) also able to offer significant protection in the cell viability assays.
Collapse
Affiliation(s)
- Lois-May Young
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Werner J. Geldenhuys
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Olwen C. Domingo
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Sarel F. Malan
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Potchefstroom, South Africa
- School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Cornelis J. Van der Schyf
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
11
|
Wang W, Ma X, Han J, Zhou M, Ren H, Pan Q, Zheng C, Zheng Q. Neuroprotective Effect of Scutellarin on Ischemic Cerebral Injury by Down-Regulating the Expression of Angiotensin-Converting Enzyme and AT1 Receptor. PLoS One 2016; 11:e0146197. [PMID: 26730961 PMCID: PMC4711585 DOI: 10.1371/journal.pone.0146197] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 12/14/2015] [Indexed: 01/08/2023] Open
Abstract
Background and Purpose Previous studies have demonstrated that angiotensin-converting enzyme (ACE) is involved in brain ischemic injury. In the present study, we investigated whether Scutellarin (Scu) exerts neuroprotective effects by down-regulating the Expression of Angiotensin-Converting Enzyme and AT1 receptor in a rat model of permanent focal cerebral ischemia. Methods Adult Sprague–Dawley rats were administrated with different dosages of Scu by oral gavage for 7 days and underwent permanent middle cerebral artery occlusion (pMCAO). Blood pressure was measured 7 days after Scu administration and 24 h after pMCAO surgery by using a noninvasive tail cuff method. Cerebral blood flow (CBF) was determined by Laser Doppler perfusion monitor and the neuronal dysfunction was evaluated by analysis of neurological deficits before being sacrificed at 24 h after pMCAO. Histopathological change, cell apoptosis and infarct area were respectively determined by hematoxylin–eosin staining, terminal deoxynucleotidyl transfer-mediated dUTP nick end labeling (TUNEL) analysis and 2,3,5-triphenyltetrazolium chloride staining. Tissue angiotensin II (Ang II) and ACE activity were detected by enzyme-linked immunosorbent assays. The expression levels of ACE, Ang II type 1 receptor (AT1R), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) were measured by Western blot and real-time PCR. ACE inhibitory activity of Scu in vitro was detected by the photometric determination. Results Scu treatment dose-dependently decreased neurological deficit score, infarct area, cell apoptosis and morphological changes induced by pMCAO, which were associated with reductions of ACE and AT1R expression and the levels of Ang II, TNF-α, IL-6, and IL-1β in ischemic brains. Scu has a potent ACE inhibiting activity. Conclusion Scu protects brain from acute ischemic injury probably through its inhibitory effect on the ACE/Ang II/AT1 axis, CBF preservation and proinflammation inhibition.
Collapse
Affiliation(s)
- Wenjuan Wang
- Pharmacy School, Shihezi University, Shihezi, China
- Department of Pharmacy, the First Division Hospital of Xinjiang Production and Construction Corps, Aksu, Xinjiang, China
| | - Xiaotang Ma
- Institute of Neurological Disease, Zhanjiang Medical College, Zhanjiang, Guangdong, China
| | - Jichun Han
- Pharmacy School, Shihezi University, Shihezi, China
| | | | - Huanhuan Ren
- Pharmacy School, Shihezi University, Shihezi, China
| | - Qunwen Pan
- Institute of Neurological Disease, Zhanjiang Medical College, Zhanjiang, Guangdong, China
| | - Chunli Zheng
- College of Life Sciences, Northwest A&F University, Yangling, Shanxi, China
| | - Qiusheng Zheng
- Pharmacy School, Shihezi University, Shihezi, China
- Binzhou Medical University, Yantai, China
- * E-mail:
| |
Collapse
|
12
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
13
|
Cha JB, Kwon MY, Chung SW, Woo JM. Neuroprotective Effects of Betaxolol Mediated by Heme Oxygenase-1 Induction in RGC-5. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2016. [DOI: 10.3341/jkos.2016.57.1.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Jae Bong Cha
- Department of Ophthalmology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Min Young Kwon
- Biological Sciences, University of Ulsan College of Natural Science, Ulsan, Korea
| | - Su Wol Chung
- Biological Sciences, University of Ulsan College of Natural Science, Ulsan, Korea
| | - Je Moon Woo
- Department of Ophthalmology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| |
Collapse
|
14
|
Nimodipine Dose Reductions in the Treatment of Patients with Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care 2015; 25:29-39. [DOI: 10.1007/s12028-015-0230-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Estes S, Zhong L, Artinian L, Rehder V. Regulation of electrical activity and neuronal excitability in Helisoma trivolvis by carbon monoxide. Neuroscience 2015; 311:453-63. [DOI: 10.1016/j.neuroscience.2015.10.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 10/23/2015] [Accepted: 10/28/2015] [Indexed: 11/30/2022]
|
16
|
Koskimäki J, Matsui N, Umemori J, Rantamäki T, Castrén E. Nimodipine activates TrkB neurotrophin receptors and induces neuroplastic and neuroprotective signaling events in the mouse hippocampus and prefrontal cortex. Cell Mol Neurobiol 2015; 35:189-96. [PMID: 25204460 DOI: 10.1007/s10571-014-0110-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/01/2014] [Indexed: 12/14/2022]
Abstract
The L-type calcium channel blocker nimodipine improves clinical outcome produced by delayed cortical ischemia or vasospasm associated with subarachnoid hemorrhage. While vasoactive mechanisms are strongly implicated in these therapeutic actions of nimodipine, we sought to test whether nimodipine might also regulate neurotrophic and neuroplastic signaling events associated with TrkB neurotrophin receptor activation. Adult male mice were acutely treated with vehicle or nimodipine (10 mg/kg, s.c., 1.5 h) after which the phosphorylation states of TrkB, cyclic-AMP response element binding protein (CREB), protein kinase B (Akt), extracellular regulated kinase (ERK), mammalian target of rapamycin (mTor) and p70S6 kinase (p70S6k) from prefrontal cortex and hippocampus were assessed. Nimodipine increased the phosphorylation of the TrkB catalytic domain and the phosphoslipase-Cγ1 (PLCγ1) domain, whereas phosphorylation of the TrkB Shc binding site remained unaltered. Nimodipine-induced TrkB phosphorylation was associated with increased phosphorylation levels of Akt and CREB in the prefrontal cortex and the hippocampus whereas phosphorylation of ERK, mTor and p70S6k remained unaltered. Nimodipine-induced TrkB signaling was not associated with changes in BDNF mRNA or protein levels. These nimodipine-induced changes on TrkB signaling mimic those produced by antidepressant drugs and thus propose common mechanisms and long-term functional consequences for the effects of these medications. This work provides a strong basis for investigating the role of TrkB-associated signaling underlying the neuroprotective and neuroplastic effects of nimodipine in translationally relevant animal models of brain trauma or compromised synaptic plasticity.
Collapse
Affiliation(s)
- Janne Koskimäki
- Neuroscience Center, University of Helsinki, P.O. Box 56 (Viikinkaari 4), 00014, Helsinki, Finland
| | | | | | | | | |
Collapse
|
17
|
Nimodipine enhances neurite outgrowth in dopaminergic brain slice co-cultures. Int J Dev Neurosci 2014; 40:1-11. [PMID: 25447789 DOI: 10.1016/j.ijdevneu.2014.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 11/24/2022] Open
Abstract
Calcium ions (Ca(2+)) play important roles in neuroplasticity and the regeneration of nerves. Intracellular Ca(2+) concentrations are regulated by Ca(2+) channels, among them L-type voltage-gated Ca(2+) channels, which are inhibited by dihydropyridines like nimodipine. The purpose of this study was to investigate the effect of nimodipine on neurite growth during development and regeneration. As an appropriate model to study neurite growth, we chose organotypic brain slice co-cultures of the mesocortical dopaminergic projection system, consisting of the ventral tegmental area/substantia nigra and the prefrontal cortex from neonatal rat brains. Quantification of the density of the newly built neurites in the border region (region between the two cultivated slices) of the co-cultures revealed a growth promoting effect of nimodipine at concentrations of 0.1μM and 1μM that was even more pronounced than the effect of the growth factor NGF. This beneficial effect was absent when 10μM nimodipine were applied. Toxicological tests revealed that the application of nimodipine at this higher concentration slightly induced caspase 3 activation in the cortical part of the co-cultures, but did neither affect the amount of lactate dehydrogenase release or propidium iodide uptake nor the ratio of bax/bcl-2. Furthermore, the expression levels of different genes were quantified after nimodipine treatment. The expression of Ca(2+) binding proteins, immediate early genes, glial fibrillary acidic protein, and myelin components did not change significantly after treatment, indicating that the regulation of their expression is not primarily involved in the observed nimodipine mediated neurite growth. In summary, this study revealed for the first time a neurite growth promoting effect of nimodipine in the mesocortical dopaminergic projection system that is highly dependent on the applied concentrations.
Collapse
|
18
|
The β-adrenergic system as a possible new target for pharmacologic treatment of neovascular retinal diseases. Prog Retin Eye Res 2014; 42:103-29. [DOI: 10.1016/j.preteyeres.2014.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 12/31/2022]
|
19
|
Behravan E, Razavi BM, Hosseinzadeh H. Review of plants and their constituents in the therapy of cerebral ischemia. Phytother Res 2014; 28:1265-74. [PMID: 24919707 DOI: 10.1002/ptr.5187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 05/08/2014] [Accepted: 05/18/2014] [Indexed: 11/08/2022]
Abstract
Cerebral ischemia is a condition in which there is insufficient blood flow to the brain to meet metabolic demand. This leads to cerebral hypoxia and thus to the death of neuronal cells or stroke. The limited number of medicines currently available for patients following ischemic stroke and insufficient data on efficiency of these chemicals in the treatment of stroke led us to the search for novel therapeutic approaches. Recent studies have focused on the possible capacity of natural compounds extracted from vegetables and fruits, to prevent human disabilities caused by cerebral ischemia. In this review, we will discuss some plants and their constituents that may protect brain ischemia or delay the neurological disorders following a stroke. We have reviewed different studies in scientific databases that investigate herbal compounds and their effects on cerebral ischemia.
Collapse
Affiliation(s)
- Effat Behravan
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | |
Collapse
|
20
|
Neuroprotective agents in ischemic stroke: past failures and future opportunities. ACTA ACUST UNITED AC 2013. [DOI: 10.4155/cli.13.91] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Holmes S, Abbassi B, Su C, Singh M, Cunningham RL. Oxidative stress defines the neuroprotective or neurotoxic properties of androgens in immortalized female rat dopaminergic neuronal cells. Endocrinology 2013; 154:4281-92. [PMID: 23959938 PMCID: PMC3800758 DOI: 10.1210/en.2013-1242] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Males have a higher risk for developing Parkinson's disease and parkinsonism after ischemic stroke than females. Although estrogens have been shown to play a neuroprotective role in Parkinson's disease, there is little information on androgens' actions on dopamine neurons. In this study, we examined the effects of androgens under conditions of oxidative stress to determine whether androgens play a neuroprotective or neurotoxic role in dopamine neuronal function. Mitochondrial function, cell viability, intracellular calcium levels, and mitochondrial calcium influx were examined in response to androgens under both nonoxidative and oxidative stress conditions. Briefly, N27 dopaminergic cells were exposed to the oxidative stressor, hydrogen peroxide, and physiologically relevant levels of testosterone or dihydrotestosterone, applied either before or after oxidative stress exposure. Androgens, alone, increased mitochondrial function via a calcium-dependent mechanism. Androgen pretreatment protected cells from oxidative stress-induced cell death. However, treatment with androgens after the oxidative insult increased cell death, and these effects were, in part, mediated by calcium influx into the mitochondria. Interestingly, the negative effects of androgens were not blocked by either androgen or estrogen receptor antagonists. Instead, a putative membrane-associated androgen receptor was implicated. Overall, our results indicate that androgens are neuroprotective when oxidative stress levels are minimal, but when oxidative stress levels are elevated, androgens exacerbate oxidative stress damage.
Collapse
Affiliation(s)
- Shaletha Holmes
- PhD, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, Texas 76107-2699.
| | | | | | | | | |
Collapse
|
22
|
Young LM, Zeller M, Geldenhuys WJ, Malan SF, Van der Schyf CJ. Triquinane scaffolds: Shape and geometry as a function of saturation and bridgehead groups. J Mol Struct 2013. [DOI: 10.1016/j.molstruc.2013.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Sendrowski K, Rusak M, Sobaniec P, Iłendo E, Dąbrowska M, Boćkowski L, Koput A, Sobaniec W. Study of the protective effect of calcium channel blockers against neuronal damage induced by glutamate in cultured hippocampal neurons. Pharmacol Rep 2013; 65:730-6. [DOI: 10.1016/s1734-1140(13)71052-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 12/13/2012] [Indexed: 11/28/2022]
|
24
|
Li S, Wu C, Zhu L, Gao J, Fang J, Li D, Fu M, Liang R, Wang L, Cheng M, Yang H. By improving regional cortical blood flow, attenuating mitochondrial dysfunction and sequential apoptosis galangin acts as a potential neuroprotective agent after acute ischemic stroke. Molecules 2012; 17:13403-23. [PMID: 23143152 PMCID: PMC6268745 DOI: 10.3390/molecules171113403] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 10/31/2012] [Accepted: 11/06/2012] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a devastating disease with a complex pathophysiology. Galangin is a natural flavonoid isolated from the rhizome of Alpina officinarum Hance, which has been widely used as an antioxidant agent. However, its effects against ischemic stroke have not been reported and its related neuroprotective mechanism has not really been explored. In this study, neurological behavior, cerebral infarct volumes and the improvement of the regional cortical blood flow (rCBF) were used to evaluate the therapeutic effect of galangin in rats impaired by middle cerebral artery occlusion (MCAO)-induced focal cerebral ischemia. Furthermore, the determination of mitochondrial function and Western blot of apoptosis-related proteins were performed to interpret the neuroprotective mechanism of galangin. The results showed that galangin alleviated the neurologic impairments, reduced cerebral infarct at 24 h after MCAO and exerted a protective effect on the mitochondria with decreased production of mitochondrial reactive oxygen species (ROS). These effects were consistent with improvements in the membrane potential level (Dym), membrane fluidity, and degree of mitochondrial swelling in a dose-dependent manner. Moreover, galangin significantly improved the reduced rCBF after MCAO. Western blot analysis revealed that galangin also inhibited apoptosis in a dose-dependent manner concomitant with the up-regulation of Bcl-2 expression, down-regulation of Bax expression and the Bax/Bcl-2 ratio, a reduction in cytochrome c release from the mitochondria to the cytosol, the reduced expression of activated caspase-3 and the cleavage of poly(ADP-ribose) polymerase (PARP). All these data in this study demonstrated that galangin might have therapeutic potential for ischemic stroke and play its protective role through the improvement in rCBF, mitochondrial protection and inhibiting caspase-dependent mitochondrial cell death pathway for the first time.
Collapse
Affiliation(s)
- Shaojing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chuanhong Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Li Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Jian Gao
- College of Pharmaceutical Science, Hebei University, Baoding, Hebei 071002, China
| | - Jing Fang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Defeng Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Meihong Fu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Rixin Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lan Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ming Cheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
25
|
Lecht S, Rotfeld E, Arien‐Zakay H, Tabakman R, Matzner H, Yaka R, Lelkes PI, Lazarovici P. Neuroprotective effects of nimodipine and nifedipine in the NGF‐differentiated PC12 cells exposed to oxygen‐glucose deprivation or trophic withdrawal. Int J Dev Neurosci 2012; 30:465-9. [DOI: 10.1016/j.ijdevneu.2012.05.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 05/26/2012] [Accepted: 05/26/2012] [Indexed: 02/08/2023] Open
Affiliation(s)
- Shimon Lecht
- Integrated Cellular Tissue Engineering & Regenerative Medicine LaboratorySchool of Biomedical Engineering, Science and Health SystemsDrexel UniversityPhiladelphiaPA19102USA
| | - Elena Rotfeld
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Hadar Arien‐Zakay
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Rinat Tabakman
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Henry Matzner
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Rami Yaka
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Peter I. Lelkes
- Integrated Cellular Tissue Engineering & Regenerative Medicine LaboratorySchool of Biomedical Engineering, Science and Health SystemsDrexel UniversityPhiladelphiaPA19102USA
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| |
Collapse
|
26
|
Haile M, Galoyan S, Li YS, Cohen BH, Quartermain D, Blanck T, Bekker A. Nimodipine-induced hypotension but not nitroglycerin-induced hypotension preserves long- and short-term memory in adult mice. Anesth Analg 2012; 114:1034-41. [PMID: 22366846 DOI: 10.1213/ane.0b013e31824b2b05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Acute hypotension may be implicated in cognitive dysfunction. L-type calcium channel blockers in the setting of hypoxia are protective of learning and memory. We tested the hypothesis that hypotension induced by nimodipine (NIMO) and nicardipine (NICA) would be protective of long- and short-term memory compared to hypotension induced by nitroglycerin (NTG). METHODS Forty Swiss-Webster mice (30 to 35 g, 6 to 8 weeks) were randomized into 4 groups for i.p. injection immediately after passive avoidance (PA) learning on day 0: (1) NTG (30 mg/kg); (2) NICA (40 mg/kg); (3) NIMO (40 mg/kg); and (4) saline. PA training latencies (seconds) were recorded for entry from a suspended platform into a Plexiglas tube where a shock (0.3 mA; 2-second duration) was automatically delivered. On day 2 latencies were recorded during a testing trial during which no shock was delivered. Latencies >900 seconds were assigned this value. Lower testing latency is indicative of an impairment of long-term associative memory. Forty-nine additional mice were randomized into similar groups for object recognition testing (ORT) and given i.p. injections on day 0. ORT measures short-term memory by exploiting the tendency of mice to prefer novel objects where a familiar object is present. On day 5 during training, 2 identical objects were placed in a circular arena and mice explored both for 15 minutes. A testing trial was conducted 1 hour later for 3 minutes after a novel object replaced a familiar one. Mice with intact memory spend about 65% of the time exploring the novel object. Mice with impaired memory devote equal time to each object. Recognition index (RI) is defined as the ratio of time spent exploring the novel object to time spent exploring both objects was the measure of memory. Mean arterial blood pressure (MAP), cerebral bloodflow, and body and brain oxygenation (PO(2)) studies were done in separate groups of mice to determine the dosages for matched degrees of hypotension and the physiological profile of each treatment. RESULTS The median PA latencies for the different conditions were as follows: NTG (219.5 ± 93.5 second semi-interquartile range [SIQR]), NICA (372.5 ± 75.5 second SIQR), NIMO (540 ± 200 second SIQR) and saline (804 ± 257.5 second SIQR). Rank methods were used to analyze the PA latencies for significant differences. NTG latency was significantly shorter than NIMO latency (P = 0.012) and saline latency (P = 0.006), but not NICA latency (P = 0.126). ORT RI values showed a similar pattern. We found that NTG RI (47.2 ± 5.9% SEM) was different from NIMO RI (60.2 ± 4.6% SEM, P = 0.031) and different from saline RI (66.9 + 3.7% SEM, P = 0.006). Physiological experiments showed that MAP decreased to 45 to 50 mm Hg in all animals who became minimally responsive to external stimuli within 10 to 15 minutes of injection. Intergroup differences for MAP, body and brain oxygenation, and cerebral bloodflow were not statistically significant. CONCLUSION Acute hypotension induced by NIMO was protective of 2 categories of memory formation relevant to the clinical posttreatment period. Both immediate long-term associative memory consolidation as measured by the PA learning paradigm and delayed short-term working memory function as measured by the ORT paradigm were significantly improved compared to matched levels of hypotension induced by NTG. These results indicate the utility of further investigation of l-type calcium channel blockers as a potential means of preserving cognition in the setting of hypotensive and low flow states.
Collapse
Affiliation(s)
- Michael Haile
- Department of Anesthesiology, New York University Langone Medical Center, 550 First Avenue, Rusk 605, New York, NY 10016, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Wu WN, Wu PF, Chen XL, Zhang Z, Gu J, Yang YJ, Xiong QJ, Ni L, Wang F, Chen JG. Sinomenine protects against ischaemic brain injury: involvement of co-inhibition of acid-sensing ion channel 1a and L-type calcium channels. Br J Pharmacol 2012; 164:1445-59. [PMID: 21585344 DOI: 10.1111/j.1476-5381.2011.01487.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Sinomenine (SN), a bioactive alkaloid, has been utilized clinically to treat rheumatoid arthritis in China. Our preliminary experiments indicated that it could protect PC12 cells from oxygen-glucose deprivation-reperfusion (OGD-R), we thus investigated the possible effects of SN on cerebral ischaemia and the related mechanism. EXPERIMENTAL APPROACH Middle cerebral artery occlusion in rats was used as an animal model of ischaemic stroke in vivo. The mechanisms of the effects of SN were investigated in vitro using whole-cell patch-clamp recording, calcium imaging in PC12 cells and rat cortical neurons subjected to OGD-R. KEY RESULTS Pretreatment with SN (10 and 30 mg·kg(-1) , i.p.) significantly decreased brain infarction and the overactivation of calcium-mediated events in rats subjected to 2 h ischaemia followed by 24 h reperfusion. Extracellular application of SN inhibited the currents mediated by acid-sensing ion channel 1a and L-type voltage-gated calcium channels, in the rat cultured neurons, in a concentration-dependent manner. These inhibitory effects contribute to the neuroprotection of SN against OGD-R and extracellular acidosis-induced cytotoxicity. More importantly, administration of SN (30 mg·kg(-1) , i.p.) at 1 and 2 h after cerebral ischaemia also decreased brain infarction and improved functional recovery. CONCLUSION AND IMPLICATIONS SN exerts potent protective effects against ischaemic brain injury when administered before ischaemia or even after the injury. The inhibitory effects of SN on acid-sensing ion channel 1a and L-type calcium channels are involved in this neuroprotection.
Collapse
Affiliation(s)
- Wen-Ning Wu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
He P, Liu Q, Wu J, Shen Y. Genetic deletion of TNF receptor suppresses excitatory synaptic transmission via reducing AMPA receptor synaptic localization in cortical neurons. FASEB J 2011; 26:334-45. [PMID: 21982949 DOI: 10.1096/fj.11-192716] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The distribution of postsynaptic glutamate receptors has been shown to be regulated by proimmunocytokine tumor necrosis factor α (TNF-α) signaling. The role of TNF-α receptor subtypes in mediating glutamate receptor expression, trafficking, and function still remains unclear. Here, we report that TNF receptor subtypes (TNFR1 and TNFR2) differentially modulate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) clustering and function in cultured cortical neurons. We find that genetic deletion of TNFR1 decreases surface expression and synaptic localization of the AMPAR GluA1 subunit, reduces the frequency of miniature excitatory postsynaptic current (mEPSC), and reduces AMPA-induced maximal whole-cell current. In addition, these results are not observed in TNFR2-deleted neurons. The decreased AMPAR expression and function in TNFR1-deleted cells are not significantly restored by short (2 h) or long (24 h) term exposure to TNF-α. In TNFR2-deleted cells, TNF-α promotes AMPAR trafficking to the synapse and increases mEPSC frequency. In the present study, we find no significant change in the GluN1 subunit of NMDAR clusters, location, and mEPSC. This includes applying or withholding the TNF-α treatment in both TNFR1- and TNFR2-deleted neurons. Our results indicate that TNF receptor subtype 1 but not 2 plays a critical role in modulating AMPAR clustering, suggesting that targeting TNFR1 gene might be a novel approach to preventing neuronal AMPAR-mediated excitotoxicity.
Collapse
Affiliation(s)
- Ping He
- Center for Advanced Therapeutic Strategies for Brain Disorders, Roskamp Institute, Sarasota, FL, USA
| | | | | | | |
Collapse
|
29
|
Yamaguchi M, Nakayama T, Fu Z, Sato N, Soma M, Morita A, Hinohara S, Doba N, Mizutani T. The haplotype of the CACNA1B gene associated with cerebral infarction in a Japanese population. Hereditas 2011; 147:313-9. [PMID: 21166801 DOI: 10.1111/j.1601-5223.2009.02115.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cerebral infarction (CI) is thought to be a multifactorial disease that is affected by several environmental factors and genetic variants. N-type voltage-gated calcium channels (VGCCs), which are expressed primarily in the neurons, have various roles in neuronal functions and are especially involved with neurotransmitter release at the sympathetic nerve terminals. We considered the α1B subunit of the N-type voltage-gated calcium channel (CACNA1B) to be representative of the general characteristics of this channel type. The aim of the present study was to assess the association of the human CACNA1B gene with the occurrence of CI via a haplotype-based case-control study that used single nucleotide polymorphisms (SNPs) from the Japanese population. A total of 165 CI patients and 314 controls were enrolled in the case-controlled studies that examined three SNPs of the human CACNA1B gene (rs7042521, rs11137351, rs10780199). There were significant differences between the CI and control groups for the overall distribution of the genotypes and the presence of the recessive rs10780199. Multiple logistic regression analyses revealed that even after adjusting for confounding factors (odds ratio: 1.716), the frequencies of the A/G and G/G genotypes of rs10780199 in the CI group were significantly higher than those observed in the control group (p = 0.021). Furthermore, the C-C-G and G-G-G haplotypes of rs7042521-rs11137351-rs10780199 were significantly more frequent in the CI group than in the control group (p = 0.024 and p < 0.000). In conclusion, significant differences were noted between the CI and control patients for the specific SNPs and haplotypes in the CACNA1B gene. The results indicate that these polymorphisms and haplotypes might be genetic markers for CI.
Collapse
Affiliation(s)
- Mai Yamaguchi
- Division of Neurology, Dept of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Calcium overload is associated with lipofuscin formation in human retinal pigment epithelial cells fed with photoreceptor outer segments. Eye (Lond) 2011; 25:519-27. [PMID: 21311572 DOI: 10.1038/eye.2011.7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To investigate the role of Ca²(+) in lipofuscin formation in human retinal pigment epithelial (RPE) cells that phagocytize bovine photoreceptor outer segments (POSs). METHODS Cultured human RPE cells fed with 2 × 10⁷per l bovine POS were treated with flunarizine, an antagonist of Ca²(+) channel, or/and centrophenoxine, a lipofuscin scavenger. The Ca²(+) changes and lipofuscin formation were measured with fluoresence dye Fluo-3/AM ester, laser scanning confocal microscopy (LSCM) and flow cytometry (FCM). The activity of RPE cells was measured by methyl thiazolyl tetrazolium (MTT) assay and argyrophilic nucleolar organizer regions (AgNORs) assay. RESULTS The Ca²(+) fluorescence intensity (CFI) of RPE cells fed with POS was significantly increased compared with the controls (165.36 ± 29.92 U). It reached a peak with 777.33 ± 63.86 U (P<0.01) at 12 h, and then decreased but still maintained a high level of 316.90 ± 36.07 U (P<0.01) for 4 days. Flunarizine and centrophenoxine significantly decreased the Ca²(+) overload to 227.18 ± 14.00 U at 12 h and 211.06 ± 20.45 U at 4 days. FCM confirmed these changes. The drugs also showed an inhibitory effect on the lipofuscin formation. The proliferation rate of the cells fed with POS increased significantly. Both drugs had inhibitory effects on the activity of the cultured cells. This tendency was confirmed by AgNORs assay. CONCLUSIONS The Ca²(+) inflow initiated lipofuscin accumulation in RPE cells fed with POS. Flunarizine and centrophenoxine can decrease Ca²(+) overload and lipofuscin formation in RPE cells, accompanied by maintaining cellular vitality.
Collapse
|
31
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10053-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Liu Y, Lo YC, Qian L, Crews FT, Wilson B, Chen HL, Wu HM, Chen SH, Wei K, Lu RB, Ali S, Hong JS. Verapamil protects dopaminergic neuron damage through a novel anti-inflammatory mechanism by inhibition of microglial activation. Neuropharmacology 2010; 60:373-80. [PMID: 20950631 DOI: 10.1016/j.neuropharm.2010.10.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 09/23/2010] [Accepted: 10/05/2010] [Indexed: 01/09/2023]
Abstract
Verapamil has been shown to be neuroprotective in several acute neurotoxicity models due to blockade of calcium entry into neurons. However, the potential use of verapamil to treat chronic neurodegenerative diseases has not been reported. Using rat primary mesencephalic neuron/glia cultures, we report that verapamil significantly inhibited LPS-induced dopaminergic neurotoxicity in both pre- and post-treatment experiments. Reconstituted culture studies revealed that the presence of microglia was essential in verapamil-elicited neuroprotection. Mechanistic studies showed that decreased production of inflammatory mediators from LPS-stimulated microglia underlay neuroprotective property of verapamil. Further studies demonstrated that microglial NADPH oxidase (PHOX), the key superoxide-producing enzyme, but not calcium channel in neurons, is the site of action for the neuroprotective effect of verapamil. This conclusion was supported by the following two observations: 1) Verapamil failed to show protective effect on LPS-induced dopaminergic neurotoxicity in PHOX-deficient (deficient in the catalytic subunit of gp91(phox)) neuron/glia cultures; 2) Ligand binding studies showed that the binding of [(3)H]Verapamil onto gp91(phox) transfected COS7 cell membranes was higher than the non-transfected control. The calcium channel-independent neuroprotective property of verapamil was further supported by the finding that R(+)-verapamil, a less active form in blocking calcium channel, showed the same potency in neuroprotection, inhibition of pro-inflammatory factors production and binding capacity to gp91(phox) membranes as R(-)-verapamil, the active isomer of calcium channel blocker. In conclusion, our results demonstrate a new indication of verapamil-mediated neuroprotection through a calcium channel-independent pathway and provide a valuable avenue for the development of therapy for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuxin Liu
- Neuropharmacology Section, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
de Los Ríos C, Egea J, Marco-Contelles J, León R, Samadi A, Iriepa I, Moraleda I, Gálvez E, García AG, López MG, Villarroya M, Romero A. Synthesis, inhibitory activity of cholinesterases, and neuroprotective profile of novel 1,8-naphthyridine derivatives. J Med Chem 2010; 53:5129-43. [PMID: 20575555 DOI: 10.1021/jm901902w] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
1,8-Naphthyridine derivatives related to 17 (ITH4012), a neuroprotective compound reported by our research group, have been synthesized. In general, they have shown better inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) than most tacrine derivatives previously synthesized in our laboratory. The compounds presented an interesting neuroprotective profile in SH-SY5Y neuroblastoma cells stressed with rotenone/oligomycin A. Moreover, compound 14 (ethyl 5-amino-2-methyl-6,7,8,9-tetrahydrobenzo[b][1,8]naphthyridine-3-carboxylate) also caused protection in cells stressed with okadaic acid (OA) or amyloid beta 1-42 peptide (Abeta(1-42)). Interestingly, compound 14 prevented the OA-induced PP2A inhibition, one of the enzymes implicated in tau dephosphorylation. This compound also exhibited neuroprotection against neurotoxicity elicited by oxygen and glucose deprivation in hippocampal slices. Because these stressors caused neuronal damage related to physiopathological hallmarks found in the brain of Alzheimer's disease (AD) patients, we conclude that compound 14 deserves further in vivo studies in AD models to test its therapeutic potential in this disease.
Collapse
Affiliation(s)
- Cristóbal de Los Ríos
- Departamento de Farmacologia y Terapeutica, Facultad de Medicina, Instituto Teofilo Hernando, Universidad Autonoma de Madrid, C/Arzobispo Morcillo 4, 28029 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
McNeish AJ, Jimenez Altayo F, Garland CJ. Evidence both L-type and non-L-type voltage-dependent calcium channels contribute to cerebral artery vasospasm following loss of NO in the rat. Vascul Pharmacol 2010; 53:151-9. [PMID: 20601125 PMCID: PMC3191278 DOI: 10.1016/j.vph.2010.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 06/01/2010] [Accepted: 06/12/2010] [Indexed: 11/25/2022]
Abstract
We recently found block of NO synthase in rat middle cerebral artery caused spasm, associated with depolarizing oscillations in membrane potential (Em) similar in form but faster in frequency (circa 1 Hz) to vasomotion. T-type voltage-gated Ca2+ channels contribute to cerebral myogenic tone and vasomotion, so we investigated the significance of T-type and other ion channels for membrane potential oscillations underlying arterial spasm. Smooth muscle cell membrane potential (Em) and tension were measured simultaneously in rat middle cerebral artery. NO synthase blockade caused temporally coupled depolarizing oscillations in cerebrovascular Em with associated vasoconstriction. Both events were accentuated by block of smooth muscle BKCa. Block of T-type channels or inhibition of Na+/K+-ATPase abolished the oscillations in Em and reduced vasoconstriction. Oscillations in Em were either attenuated or accentuated by reducing [Ca2+]o or block of KV, respectively. TRAM-34 attenuated oscillations in both Em and tone, apparently independent of effects against KCa3.1. Thus, rapid depolarizing oscillations in Em and tone observed after endothelial function has been disrupted reflect input from T-type calcium channels in addition to L-type channels, while other depolarizing currents appear to be unimportant. These data suggest that combined block of T and L-type channels may represent an effective approach to reverse cerebral vasospasm.
Collapse
Affiliation(s)
- A J McNeish
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | |
Collapse
|
35
|
Li XY, Liang J, Tang YB, Zhou JG, Guan YY. Ginsenoside Rd prevents glutamate-induced apoptosis in rat cortical neurons. Clin Exp Pharmacol Physiol 2009; 37:199-204. [PMID: 19719747 DOI: 10.1111/j.1440-1681.2009.05286.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
1. The role of voltage-independent Ca(2+) entry in cell apoptosis has recently received considerable attention. It has been found that ginsenoside Rd significantly inhibits voltage-independent Ca(2+) entry. The aim of the present study was to investigate the protective effects of ginsenoside Rd against glutamate-induced apoptosis of rat cortical neurons. 2. Ginsenoside Rd significantly reduced glutamate-induced apoptotic morphological changes and DNA laddering. In comparison, nimodipine only had a weak effect. 3. Ginsenoside Rd (1, 3 and 10 micromol/L) concentration-dependently inhibited caspase 3 activation and expression of the p20 subunit of active caspase 3 (by 30 +/- 10%, 41 +/- 9% and 62 +/- 19%, respectively, compared with glutamate alone; P < 0.05), whereas 1 micromol/L nimodipine had no effect. 4. Glutamate decreased cell viability to 37.4 +/- 4.7 (n = 8) and evoked cell apoptosis. Ginsenoside Rd (1, 3, 10 and 30 micromol/L) concentration-dependently inhibited glutamate-induced cell death, increased cell viability and reduced apoptotic percentage (from 47.5 +/- 4.9% to 37.4 +/- 6.9%, 28.3 +/- 5.2% and 22.5 +/- 5.6%, respectively; P < 0.05). At 1 micromol/L, nimodipine had no effect on cell viability. Furthermore, although 1, 3, 10, 30 and 60 micromol/L ginsenoside Rd concentration-dependently inhibited glutamate-induced Ca(2+) entry by 8 +/- 2%, 24 +/- 4%, 40 +/- 7%, 49 +/- 8% and 50 +/- 8% (P < 0.05), respectively, nimodipine had no effect. 5. In conclusion, the results indicate that ginsenoside Rd prevents glutamate-induced apoptosis in rat cortical neurons and provide further evidence of the potential of voltage-independent Ca(2+) channel blockers as new neuroprotective drugs for the prevention of neuronal apoptosis and death induced by cerebral ischaemia.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Department of Pharmacy, Third Affiliated Hospital, Guangzhou, China
| | | | | | | | | |
Collapse
|
36
|
Hou ST, Jiang SX, Smith RA. Permissive and repulsive cues and signalling pathways of axonal outgrowth and regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 267:125-81. [PMID: 18544498 DOI: 10.1016/s1937-6448(08)00603-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Successful axonal outgrowth in the adult central nervous system (CNS) is central to the process of nerve regeneration and brain repair. To date, much of the knowledge on axonal guidance and outgrowth comes from studies on neuritogenesis and patterning during development where distal growth cones constantly sample the local environment and respond to specific physical and trophic influences. Opposing permissive (e.g., growth factors) and hostile signals (e.g., repulsive cues) are processed, leading to growth cone remodelling, and a concomitant restructuring of the cytoskeleton, thereby permitting pioneering extension and a potential for establishing synaptic connections. Repulsive cues, such as semaphorins, ephrins and myelin-secreted inhibitory glycoproteins, act through their respective receptors to affect the collapsing or turning of growth cones via several pathways, such as the Rho GTPases signalling which precipitates the cytoskeletal changes. One of the direct modulators of microtubules is the family of brain-specific proteins, collapsin response mediator protein (CRMP). Exciting evidence emerged recently that cleavage of CRMPs in response to injury-activated proteases, such as calpain, signals axonal retraction and neuronal death in adult post-mitotic neurons, while blocking this signal transduction prevents axonal retraction and death following excitotoxic insult and cerebral ischemia. Regeneration is minimal in injured postnatal CNS, albeit the occurrence of some limited remodelling in areas where synaptic plasticity is prevalent. Frequently in the absence of axonal regeneration, there is not only an inevitable loss of functional connections, but also a loss of neurons, such as through the actions of dependence receptors. Deciphering the cues and signalling pathways of axonal guidance and outgrowth may hold the key to fully understanding nerve regeneration and brain repair, thereby opening the way for developing potential therapeutics.
Collapse
Affiliation(s)
- Sheng T Hou
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, K1A 0R6, Canada
| | | | | |
Collapse
|
37
|
Mielcarek J, Khmelinskii I, Sikorski M, Stefaniak H. Photolability of potential calcium channel antagonists: Hexahydroquinoline derivatives. J Photochem Photobiol A Chem 2007. [DOI: 10.1016/j.jphotochem.2007.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
38
|
Shen J, Yip S, Wang Z, Wang W, Xing D, Du L. Brazilein-induced contraction of rat arterial smooth muscle involves activation of Ca2+ entry and ROK, ERK pathways. Eur J Pharmacol 2007; 580:366-71. [PMID: 18177858 DOI: 10.1016/j.ejphar.2007.11.012] [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: 07/05/2007] [Revised: 10/01/2007] [Accepted: 11/07/2007] [Indexed: 11/18/2022]
Abstract
Brazilein (6a,7-dihydro-3,6a,10-trihydroxy-benz[b]indeno[1,2-d]pyran-9(6H)-one) is a compound isolated from Caesalpinia sappan. The vasoactivities of brazilein were evaluated in isolated rat thoracic aorta. The results showed that brazilein can dose-dependently induce contraction of rat thoracic aorta in the resting and phenylephrine pre-evoked state. The average response to 100 microM of brazilein was 30% of the 50 mM KCl contraction, 26% of the 10 muM phenylephrine and 116% of the 20 mM caffeine contraction in comparison. The effects of vasocontraction were proved not to be endothelial dependent and could not be inhibited by alpha-adrenergic receptor blocker phentolamine, beta-adrenergic receptor blocker propranolol, M-adrenaline receptor blocker atropine, angiotensin II receptor blocker losartan or the non-selective nitric oxide synthase (NOS) inhibitor NG-Nitro-L-Arginine Methyl Ester (L-NAME). However the influx of extracellular calcium seemed to be required for this action, because depletion of extracellular calcium and the addition of L-type calcium ion channel antagonist (nimodipine and diltiazem), calcium ion channel activator (BAY-K8644) and potassium ion channel opener (pinacidil) could significantly affect the contraction induced by brazilein. We also investigated the possible signal mechanisms underlying brazilein-induced contraction using selective inhibitors. The inhibitors of myosin light chain kinase (MLCK), Rho-kinase (ROK) and extracellular signal regulated kinase (ERK) can suppress the effect of brazilein respectively, whereas inhibitors of other signaling or receptor molecules such as protein kinase C (PKC) and inositol 1,4,5-triphosphate (IP3) receptor had no effect. All these results demonstrated that brazilein can induce contraction of rat aorta, that the Ca2+ influx, ROK and ERK signal pathways and MLCK activation must be involved in the contractile processes.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiology
- Benzopyrans/chemistry
- Benzopyrans/pharmacology
- Caesalpinia/chemistry
- Caffeine/pharmacology
- Calcium/metabolism
- Diltiazem/pharmacology
- Dose-Response Relationship, Drug
- Extracellular Signal-Regulated MAP Kinases/metabolism
- In Vitro Techniques
- Indenes/chemistry
- Indenes/pharmacology
- Male
- Molecular Structure
- Muscle Contraction/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- NG-Nitroarginine Methyl Ester/pharmacology
- Nimodipine/pharmacology
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/metabolism
- Phenylephrine/pharmacology
- Pinacidil/pharmacology
- Potassium Chloride/pharmacology
- Rats
- Rats, Wistar
- Signal Transduction/drug effects
- Vasoconstriction/drug effects
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Jia Shen
- Protein Science Laboratory of the Ministry of Education, Laboratory of Pharmaceutical Sciences, Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | |
Collapse
|
39
|
A modelling approach to explore some hypotheses of the failure of neuroprotective trials in ischemic stroke patients. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2007; 97:60-78. [PMID: 18076975 DOI: 10.1016/j.pbiomolbio.2007.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ischemic stroke is the third cause of death in industrialised countries, but no satisfactory treatment is currently available. The hundreds of neuroprotective drugs developed to block the ischemic cascade gave very promising results in animal models but the clinical trials performed with these drugs showed no beneficial effects in stroke patients. Many hypotheses were advanced to explain this discrepancy, among which the morphological and functional differences between human and rodent brains. This discrepancy could be partly due to the differences in white matter and glial cell proportions between human and rodent brains. In order to test this hypothesis, we built a mathematical model of the main early pathophysiological mechanisms of stroke in rodent and in human brains. This model is a two-scale model and relies on a set of ordinary differential equations. We built two versions of this model (for human and rodent brains) differing in their white matter and glial cell proportions. Then, we carried out in silico experiments with various neuroprotective drugs. The simulation results obtained with a sodium channel blocker show that the proportion of penumbra recovery is much higher in rodent than in human brain and the results are similar with some other neuroprotective drugs tested during phase III trials. This in silico investigation suggests that the proportions of glial cells and white matter have an influence on neuroprotective drug efficacy. It reinforces the hypothesis that histological and morphological differences between rodent and human brains can partly explain the failure of these agents in clinical trials.
Collapse
|
40
|
Fan Y, Shi L, Gu Y, Zhao Y, Xie J, Qiao J, Yang GY, Wang Y, Lu CZ. Pretreatment with PTD-calbindin D 28k alleviates rat brain injury induced by ischemia and reperfusion. J Cereb Blood Flow Metab 2007; 27:719-28. [PMID: 16868556 DOI: 10.1038/sj.jcbfm.9600373] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium toxicity remains the central focus of ischemic brain injury. Calcium channel antagonists have been reported to be neuroprotective in ischemic animal models but have failed in clinical trials. Rather than block the calcium channels, calbindin proteins can buffer excessive intracellular Ca2+, and as a result, maintain the calcium homeostasis. In the present study, we investigated the effect of calbindin D 28k (CaBD) in ischemic brain using the novel technique protein transduction domain (PTD)-mediated protein transduction. We generated PTD-CaBD in Escherichia coli, tested its biologic activity in N-methyl-D-aspartate (NMDA)- and oxygen-glucose deprivation (OGD)-induced hippocampal injury models, and examined the protection of the fusion protein using a rat brain focal ischemia model. Infarct volume was determined using 2,3,5-triphenyl-tetrazolium chloride staining; neuronal injury was examined using terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining and cleaved caspase-3 assay. The results showed that the PTD-CaBD was efficiently delivered into Cos7 cells, hippocampal slice cells, and brain tissue. Pretreatment with PTD-CaBD decreased intracellular free calcium concentration and reduced cell death in NMDA- or OGD-exposed hippocampal slices (P<0.05). Intraperitoneal administration of PTD-CaBD before transient middle cerebral artery occlusion decreased brain infarct volume (280+/-47 versus 166+/-70 mm3, P<0.05), and improved neurologic outcomes compared with the control. Further studies showed that, compared with the control animals, PTD-CaBD decreased TUNEL (58%+/-7% versus 29%+/-3%, P<0.05)- and cleaved caspase-3 (62+/-4/field versus 31+/-6/field, P<0.05)-positive cells in the ischemic boundary zone. These results indicate that systemic administration of PTD-CaBD could attenuate ischemic brain injury, suggesting that PTD-mediated protein transduction might provide a promising and effective approach for the therapies of brain diseases, including cerebral ischemia.
Collapse
Affiliation(s)
- Yongfeng Fan
- Department of Neurology, Institute of Neurology, Hua-Shan Hospital, Fudan University, Shanghai, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nisticò R, Piccirilli S, Sebastianelli L, Nisticò G, Bernardi G, Mercuri NB. The Blockade of K+‐ATP Channels has Neuroprotective Effects in an In Vitro Model of Brain Ischemia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:383-95. [PMID: 17678973 DOI: 10.1016/s0074-7742(07)82021-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is a common belief that the opening of K(+)-ATP channels during an ischemic episode has protective effects on neuronal functions by inducing a reduction in energy consumption. However, recent studies have also proposed that activation of these channels might have deleterious effects on cell's survival possibly after a stroke or during long-lasting neurodegenerative processes. Considering these contrasting results, we have used a hippocampal in vitro slice preparation in order to investigate the possible effects of K(+)-ATP channel blockers on the electrophysiological and morphological changes induced by a transient episode of ischemia (oxygen and glucose deprivation) on CA1 pyramidal neurons. Therefore, we found that tolbutamide and glibenclamide, both nonselective K(+)-ATP channel blockers, produce neuroprotective effects against in vitro ischemia. Interestingly, the mitochondrial K(+)-ATP channel blocker 5-hydroxydecanoate and various K(+) channel blockers did not exert neuroprotection. Our results are consistent with the concept that a decreased activity of the plasmalemmal K(+)-ATP conductances may have a protective effect during episodes of transient cerebral ischemia.
Collapse
Affiliation(s)
- Robert Nisticò
- Department of Experimental Neurology, S. Lucia Foundation IRCCS, Rome, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Yamamoto T, Niwa S, Iwayama S, Koganei H, Fujita SI, Takeda T, Kito M, Ono Y, Saitou Y, Takahara A, Iwata S, Yamamoto H, Shoji M. Discovery, structure–activity relationship study, and oral analgesic efficacy of cyproheptadine derivatives possessing N-type calcium channel inhibitory activity. Bioorg Med Chem 2006; 14:5333-9. [PMID: 16616501 DOI: 10.1016/j.bmc.2006.03.040] [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: 02/14/2006] [Revised: 03/21/2006] [Accepted: 03/22/2006] [Indexed: 11/29/2022]
Abstract
Antiallergic drug cyproheptadine (Cyp) is known to have inhibitory activities for L-type calcium channels in addition to histamine and serotonin receptors. Since we found that Cyp had an inhibitory activity against N-type calcium channel, Cyp was optimized to obtain more selective N-type calcium channel blocker with analgesic action. As a consequence of the optimization, we found 13 with potent N-type calcium channel inhibitory activity which had lower inhibitory activities against L-type calcium channel, histamine (H1), and serotonin (5-HT2A) receptors than those of Cyp. 13 showed an oral analgesic activity in rat formalin-induced pain model.
Collapse
Affiliation(s)
- Takashi Yamamoto
- Pharmaceutical Research Laboratory, Ajinomoto company Inc., 1-1, Suzuki-cho, Kawasaki-ku, Kawasaki-shi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hainsworth AH, Stefani A, Calabresi P, Smith TW, Leach MJ. Sipatrigine (BW 619C89) is a Neuroprotective Agent and a Sodium Channel and Calcium Channel Inhibitor. CNS DRUG REVIEWS 2006. [DOI: 10.1111/j.1527-3458.2000.tb00141.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Ohana E, Sekler I, Kaisman T, Kahn N, Cove J, Silverman WF, Amsterdam A, Hershfinkel M. Silencing of ZnT-1 expression enhances heavy metal influx and toxicity. J Mol Med (Berl) 2006; 84:753-63. [PMID: 16741752 DOI: 10.1007/s00109-006-0062-4] [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] [Received: 05/27/2005] [Accepted: 02/14/2006] [Indexed: 10/24/2022]
Abstract
ZnT-1 reduces intracellular zinc accumulation and confers resistance against cadmium toxicity by a mechanism which is still unresolved. A functional link between the L-type calcium channels (LTCC) and ZnT-1 has been suggested, indicating that ZnT-1 may regulate ion permeation through this pathway. In the present study, immunohistochemical analysis revealed a striking overlap of the expression pattern of LTCC and ZnT-1 in cardiac tissue and brain. Using siRNA to silence ZnT-1 expression, we then assessed the role of ZnT-1 in regulating cation permeation through the L-type Ca(2+) channels in cells that are vulnerable to heavy metal permeation. Transfection of cortical neurons with ZnT-1 siRNA resulted in about 70% reduction of ZnT-1 expression and increased Ca(2+) influx via LTCC by approximately fourfold. Moreover, ZnT-1 siRNA transfected neurons showed approximately 30% increase in synaptic release, monitored using the FM1-43 dye. An increased cation influx rate, through the LTCC, was also recorded for Zn(2+) and Cd(2+) in cells treated with the ZnT-1 siRNA. Furthermore, Cd(2+)-induced neuronal death increased by approximately twofold after transfection with ZnT-1 siRNA. In addition, ZnT-1 siRNA transfection of the ovarian granulosa cell line, POGRS1, resulted in a twofold increase in Cd(2+) influx rate via the LTCC. Finally, a robust nimodipine-sensitive Cd(2+) influx was observed using a low extracellular Cd(2+) concentration (5 muM) in neurons and testicular slice cultures, attesting to the relevance of the LTCC pathway to heavy metal toxicity. Taken together, our results indicate that endogenously-expressed ZnT-1, by modulating LTCC, has a dual role: regulating calcium influx, and attenuating Cd(2+) and Zn(2+) permeation and toxicity in neurons and other cell types.
Collapse
Affiliation(s)
- Ehud Ohana
- Department of Physiology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Broadley KJ, Penson PE. Effects of hypoxia on the vasodilator activity of nifedipine and evidence of secondary pharmacological properties. Eur J Pharmacol 2006; 536:279-86. [PMID: 16584727 DOI: 10.1016/j.ejphar.2006.02.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 02/15/2006] [Accepted: 02/21/2006] [Indexed: 10/25/2022]
Abstract
The effects of hypoxia on the vasodilator response of endothelium-denuded rat aortic rings to the calcium channel blocker, nifedipine, were examined. Under normoxic conditions, nifedipine (10(-8)-3x10(-6) M) attenuated the contractility of noradrenaline precontracted rings in a concentration-dependent manner, although the sensitivity was less than what occurs with K+ precontracted tissues. Under hypoxic conditions there was no relaxation by nifedipine. When a concentration-response curve to noradrenaline was constructed before and in the presence of a high concentration of nifedipine (10(-5) M), the response to noradrenaline was unaffected in both normoxic and hypoxic conditions. When noradrenaline was replaced by phenylephrine (10(-8)-10(-5) M), the maximum tension was reduced in the presence of nifedipine to 59+/-6% of the pre-nifedipine value. Repetition of the experiment in the presence of cocaine (10(-5) M) revealed the inhibitory effect of nifedipine on noradrenaline-induced contraction, the maximum contraction in the presence of nifedipine falling significantly (P<0.005) to 67+/-6% of the pre-nifedipine response. When propranolol (10(-7) M) was present in the bath, the maximum contraction to noradrenaline was significantly (P<0.05) reduced by nifedipine to 55+/-4% of its previous value. The fact that nifedipine was able to inhibit phenylephrine-induced contractions and relax noradrenaline-precontracted aortic rings confirms its calcium channel blocking activity. The failure to inhibit noradrenaline when added prior to the noradrenaline-induced contractions suggests an opposing effect in addition to calcium channel blockade, which cancels out the attenuation of noradrenaline--but not phenylephrine-induced contractions. When neuronal uptake of noradrenaline was blocked with cocaine or beta-adrenoceptors were blocked with propranolol, the inhibitory effect of nifedipine against noradrenaline-induced contractions was revealed. This suggests that the additional property was due to blockade of neuronal reuptake or antagonism at beta-adrenoceptors. This study also showed that nifedipine is ineffective as a vasodilator in the rat aorta under hypoxic conditions.
Collapse
Affiliation(s)
- Kenneth J Broadley
- Division of Pharmacology, Welsh School of Pharmacy, Cardiff University, King Edward VII Avenue, Cathays Park, Cardiff, CF10 3XF, UK.
| | | |
Collapse
|
46
|
Franco R, Bortner CD, Cidlowski JA. Potential Roles of Electrogenic Ion Transport and Plasma Membrane Depolarization in Apoptosis. J Membr Biol 2006; 209:43-58. [PMID: 16685600 DOI: 10.1007/s00232-005-0837-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Indexed: 12/15/2022]
Abstract
Apoptosis is characterized by the programmed activation of specific biochemical pathways leading to the organized demise of cells. To date, aspects of the intracellular signaling machinery involved in this phenomenon have been extensively dissected and characterized. However, recent studies have elucidated a novel role for changes in the intracellular milieu of the cells as important modulators of the cell death program. Specially, intracellular ionic homeostasis has been reported to be a determinant in both the activation and progression of the apoptotic cascade. Several apoptotic insults trigger specific changes in ionic gradients across the plasma membrane leading to depolarization of the plasma membrane potential (PMP). These changes lead to ionic imbalance early during apoptosis. Several studies have also suggested the activation and/or modulation of specific ionic transport mechanisms including ion channels, transporters and ATPases, as mediators of altered intracellular ionic homeostasis leading to PMP depolarization during apoptosis. However, the role of PMP depolarization and of the changes in ionic homeostasis during the progression of apoptosis are still unclear. This review summarizes the current knowledge regarding the causes and consequences of PMP depolarization during apoptosis. We also review the potential electrogenic ion transport mechanisms associated with this event, including the net influx/efflux of cations and anions. An understanding of these mechamisms could lead to the generation of new therapeutic approaches for a variety of diseases involving apoptosis.
Collapse
Affiliation(s)
- R Franco
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | |
Collapse
|
47
|
Costa C, Martella G, Picconi B, Prosperetti C, Pisani A, Di Filippo M, Pisani F, Bernardi G, Calabresi P. Multiple mechanisms underlying the neuroprotective effects of antiepileptic drugs against in vitro ischemia. Stroke 2006; 37:1319-26. [PMID: 16574927 DOI: 10.1161/01.str.0000217303.22856.38] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND PURPOSE The possible neuroprotective effects of classic and new antiepileptic drugs on the electrophysiological changes induced by in vitro ischemia on striatal neurons were investigated. In particular, the aim of the study was to correlate the putative neuroprotective effects with the action of these drugs on fast sodium (Na+) and high-voltage-activated (HVA) calcium (Ca2+) currents. METHODS Extracellular field potentials were recorded from rat corticostriatal brain-slice preparations. In vitro ischemia was delivered by switching to an artificial cerebrospinal fluid solution in which glucose and oxygen were omitted. Na+ and HVA Ca2+ currents were analyzed by whole-cell patch-clamp recordings from acutely isolated rat striatal neurons. Excitatory postsynaptic potential was measured following synaptic stimulation in corticostriatal slices by sharp intracellular microelectrodes. RESULTS Neuroprotection against in vitro ischemia was observed in slices treated with carbamazepine (CBZ), valproic acid (VPA), and topiramate (TPM), whereas it was not achieved by using levetiracetam (LEV). Fast Na+ conductances were inhibited by CBZ and TPM, whereas VPA and LEV showed no effect. HVA Ca2+ conductances were reduced by CBZ, TPM, and LEV. VPA had no effect on this current. All antiepileptic drugs induced a small reduction of excitatory postsynaptic potential amplitude at concentrations higher than 100 microm without changes of paired-pulse facilitation. CONCLUSIONS The concomitant inhibition of fast Na+ and HVA Ca2+ conductances is critically important for the neuroprotection, whereas the presynaptic inhibition on glutamate transmission does not seem to play a major role.
Collapse
Affiliation(s)
- Cinzia Costa
- Clinica Neurologica, Università di Perugia, Ospedale Silvestrini, S. Andrea delle Fratte, Perugia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cheon EW, Park CH, Kim YS, Cho CH, Chung YC, Kwon JG, Yoo JM, Choi WS, Cho GJ. Protective effects of betaxolol in eyes with kainic acid-induced neuronal death. Brain Res 2006; 1069:75-85. [PMID: 16387283 DOI: 10.1016/j.brainres.2005.10.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 10/26/2005] [Accepted: 10/31/2005] [Indexed: 12/31/2022]
Abstract
In the present study, we investigated whether betaxolol, a selective beta1-adrenoceptor antagonist, has neuroprotective effect on kainic acid (KA)-induced retinal damage. Neurotoxicities were induced in adult male rats by intravitreal injection of KA (total amount, 6 nmol). To examine the neuroprotective effects of betaxolol, rats were pretreated with betaxolol topically 60 min before KA injection to the rat eyes and twice daily for 1, 3, and 7 days after KA injection. The neuroprotective effects of betaxolol were estimated by measuring the thickness of the various retinal layers, and by counting the number of choline acetyltransferase (ChAT)- and tyrosine hydroxylase (TH)-positive cells in each retinal layer. The retina is highly vulnerable to KA-induced neuronal damage. Morphometric analysis of retinal damage in KA injected eyes, the thickness of the retinal layers decreased markedly after KA injection period of both 3 and 7 days. Furthermore, the numbers of ChAT- and TH-positive cells were significantly reduced by intravitreal injection of KA. However, when two drops of betaxolol, once before KA injection and twice daily for 7 days after KA injection, were continuously administered, the reductions in the retinal thickness and the retinal ChAT- and TH-positive cells were significantly attenuated. The present study suggests that topically applied betaxolol has neuroprotective effect on the retinal cell damage due to KA-induced neurotoxicity.
Collapse
Affiliation(s)
- Eun Woo Cheon
- Division of Food Science, Jinju International University, 270 San Sangmun-ri Munsan-eup, Jinju, Kyungnam 660-759, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schauer E, Wronski R, Patockova J, Moessler H, Doppler E, Hutter-Paier B, Windisch M. Neuroprotection of Cerebrolysin in tissue culture models of brain ischemia: post lesion application indicates a wide therapeutic window. J Neural Transm (Vienna) 2005; 113:855-68. [PMID: 16362636 DOI: 10.1007/s00702-005-0384-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Accepted: 09/10/2005] [Indexed: 01/24/2023]
Abstract
All attempts to reduce neuronal damage after acute brain ischemia by the use of neuroprotective compounds have failed to prove efficacy in clinical trials so far. One of the main reasons might be the relatively narrow time window for intervention. In this study 2 different tissue culture models of ischemia, excitotoxic lesion by the use of glutamate and oxygen-glucose deprivation (OGD), were used to investigate the effects of delayed application of Cerebrolysin (Cere) on neuronal survival. This drug consists of low molecular weight peptides with neuroprotective and neurotrophic properties similar to naturally occurring growth factors. After both types of lesion, acute as well as delayed treatment with Cere resulted in a dose dependent and significant rescue of neurons. In the model of excitotoxic cell death significant drug effects were found even when the treatment started with a delay of 96 hours after addition of glutamate. In the OGD model pronounced effects were found after 48 hours delay of treatment, and even after 72 hours a small but significant rescue of neurons was detected. The neuroprotective effects of a single addition of Cerebrolysin to the culture medium resulted in significant protection until end of the experiments which was up to 2 weeks after the initial lesion. A shift of the efficacious dosages from low to high concentrations indicates that most likely active compounds are used up, indicating that multiple dosing might even increase the effect size. In conclusion the results indicate that Cere displays a relatively wide therapeutic time window which might be explained by a combination of acute neuroprotective properties and neurotrophic efficacy.
Collapse
Affiliation(s)
- E Schauer
- JSW-Research, Institute of Experimental Pharmacology, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
50
|
Saito S, Ohashi M, Naito A, Fukaya Y, Suzuki Y, Araie M. Neuroprotective effect of the novel Na+/Ca2+ channel blocker NS-7 on rat retinal ganglion cells. Jpn J Ophthalmol 2005; 49:371-6. [PMID: 16187036 DOI: 10.1007/s10384-005-0210-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Accepted: 01/12/2005] [Indexed: 01/31/2023]
Abstract
PURPOSE To investigate whether NS-7, 4-(4-fluorophenyl)-2-methyl-6-(5-piperidinopentyloxy) pyrimidine hydrochloride, a novel Na(+)/Ca(2+) channel blocker, can protect the rat retina subjected to ischemia-reperfusion insult. METHODS To evaluate the protective effect of NS-7 against retinal damage, the drug was administered before and after ischemia-reperfusion. Damage to the retina was assessed by measuring the thickness of the inner plexiform layer (IPL) and the outer nuclear layer (ONL) of each eye. In a subsequent experiment, electroretinographic (ERG) evaluation was also used. RESULTS In histopathologic evaluation, ischemia-reperfusion injury caused a significant reduction of IPL thickness (measured as the IPL/ONL ratio). In the NS-7-treated group, retinal damage was partially prevented by a concentration of 0.25 mg/kg per day. In the ERG evaluation, ischemia-reperfusion injury caused a reduction of A- and B-wave amplitudes. NS-7 treatment significantly prevented the reduction of the B wave at a concentration of 0.1 or 0.3 mg/kg, while the reduction of the A wave was not significantly affected. CONCLUSIONS NS-7 has neuroprotective effects against retinal damage resulting from subjection to ischemia. In addition, NS-7 can be used as an agent for treating acute ischemic retinopathy, including diseases associated with very high intraocular pressure, such as acute angle-closure glaucoma.
Collapse
Affiliation(s)
- Shinichiro Saito
- Department of Ophthalmology, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|