1
|
Ravi R, Das S, Hakami T, B. M. P, Pushparajan L. Pharmacotherapy for Poststroke Cognitive Impairment and Poststroke Cognitive Impairment With Dementia: A Review. Stroke Res Treat 2025; 2025:6893801. [PMID: 40421258 PMCID: PMC12103972 DOI: 10.1155/srat/6893801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 10/27/2024] [Accepted: 04/08/2025] [Indexed: 05/28/2025] Open
Abstract
Poststroke cognitive impairment (PSCI) refers to any level of cognitive decline occurring after a stroke, ranging from mild to severe impairments, while PSCI with dementia describes a more severe form where the cognitive decline significantly affects daily functioning and meets the clinical criteria for dementia. PSCI occurs in more than half of individuals who have had a stroke. Despite its high prevalence, the pharmacotherapeutic options for PSCI are limited. Several pharmacotherapeutic options like cholinesterase inhibitors (e.g., donepezil, galantamine, and rivastigmine) and N-methyl-d-aspartate receptor antagonists (e.g., memantine) have shown potential in improving cognitive functions. However, their overall effectiveness remains inconsistent across different studies and patient populations. Newer drugs such as citicoline, cilostazol, and antidepressants have shown promise, but further research is needed to validate their efficacy and safety specifically for PSCI management.
Collapse
Affiliation(s)
- Renju Ravi
- Department of Clinical Pharmacology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Saibal Das
- Indian Council of Medical Research-Centre for Ageing and Mental Health, Kolkata, India
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Tahir Hakami
- Department of Clinical Pharmacology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Prakash B. M.
- Department of Clinical Pharmacology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Libby Pushparajan
- Department of Neurology, St. Gregorios Medical Mission Hospital, Parumala, Kerala, India
| |
Collapse
|
2
|
Shadman J, Panahpour H, Alipour MR, Salimi A, Shahabi P, Azar SS. Investigating the therapeutic effects of nimodipine on vasogenic cerebral edema and blood-brain barrier impairment in an ischemic stroke rat model. Neuropharmacology 2024; 257:110054. [PMID: 38950691 DOI: 10.1016/j.neuropharm.2024.110054] [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: 03/24/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Vasogenic brain edema, a potentially life-threatening consequence following an acute ischemic stroke, is a major clinical problem. This research aims to explore the therapeutic benefits of nimodipine, a calcium channel blocker, in mitigating vasogenic cerebral edema and preserving blood-brain barrier (BBB) function in an ischemic stroke rat model. In this research, animals underwent the induction of ischemic stroke via a 60-min blockage of the middle cerebral artery and treated with a nonhypotensive dose of nimodipine (1 mg/kg/day) for a duration of five days. The wet/dry method was employed to identify cerebral edema, and the Evans blue dye extravasation technique was used to assess the permeability of the BBB. Furthermore, immunofluorescence staining was utilized to assess the protein expression levels of matrix metalloproteinase-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1). The study also examined mitochondrial function by evaluating mitochondrial swelling, succinate dehydrogenase (SDH) activity, the collapse of mitochondrial membrane potential (MMP), and the generation of reactive oxygen species (ROS). Post-stroke administration of nimodipine led to a significant decrease in cerebral edema and maintained the integrity of the BBB. The protective effects observed were associated with a reduction in cell apoptosis as well as decreased expression of MMP-9 and ICAM-1. Furthermore, nimodipine was observed to reduce mitochondrial swelling and ROS levels while simultaneously restoring MMP and SDH activity. These results suggest that nimodipine may reduce cerebral edema and BBB breakdown caused by ischemia/reperfusion. This effect is potentially mediated through the reduction of MMP-9 and ICAM-1 levels and the enhancement of mitochondrial function.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | | | - Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saied Salimpour Azar
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
3
|
Chidambaram SB, Anand N, Varma SR, Ramamurthy S, Vichitra C, Sharma A, Mahalakshmi AM, Essa MM. Superoxide dismutase and neurological disorders. IBRO Neurosci Rep 2024; 16:373-394. [PMID: 39007083 PMCID: PMC11240301 DOI: 10.1016/j.ibneur.2023.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/21/2023] [Indexed: 07/16/2024] Open
Abstract
Superoxide dismutase (SOD) is a common antioxidant enzyme found majorly in living cells. The main physiological role of SOD is detoxification and maintain the redox balance, acts as a first line of defence against Reactive nitrogen species (RNS), Reactive oxygen species (ROS), and other such potentially hazardous molecules. SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide dismutases (SODs) are expressed in neurons and glial cells throughout the CNS both intracellularly and extracellularly. Endogenous oxidative stress (OS) linked with enlarged production of reactive oxygen metabolites (ROMs), inflammation, deregulation of redox balance, mitochondrial dysfunction and bioenergetic crisis are found to be prerequisite for neuronal loss in neurological diseases. Clinical and genetic studies indicate a direct correlation between mutations in SOD gene and neurodegenerative diseases, like Amyotrophic Lateral Sclerosis (ALS), Huntington's disease (HD), Parkinson's Disease (PD) and Alzheimer's Disease (AD). Therefore, inhibitors of OS are considered as an optimistic approach to prevent neuronal loss. SOD mimetics like Metalloporphyrin Mn (II)-cyclic polyamines, Nitroxides and Mn (III)- Salen complexes are designed and used as therapeutic extensively in the treatment of neurological disorders. SODs and SOD mimetics are promising future therapeutics in the field of various diseases with OS-mediated pathology.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department of Pharmacology, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Antigua, Antigua and Barbuda
| | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Ajman University, 346 Ajman, the United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, 346 Ajman, the United Arab Emirates
| | - Srinivasan Ramamurthy
- College of Pharmacy & Health Sciences, University of Science and Technology of Fujairah, 2202 Fujairah, the United Arab Emirates
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ambika Sharma
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
4
|
Akarasereenont P, Pattanapholkornsakul S, Limsuvan S, Mamaethong D, Booranasubkajorn S, Pakaprot N, Tripatara P, Pilakasiri K. Therapeutic potential of Thai herbal formula for cognitive impairment: A metabolomics approach for Comprehensive Insights. Heliyon 2024; 10:e28027. [PMID: 38560220 PMCID: PMC10981045 DOI: 10.1016/j.heliyon.2024.e28027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Chronic cerebral ischemia hypoperfusion plays a role in the initiation and progression of vascular dementia, which causes changes in metabolites. Currently, there is no standard treatment to treat, prevent and reduce the severity of this condition. Thai herbal Yahom no.20 (YHF20) is indicated for fatigue and dizziness. The components of YHF20 have been found to have pharmacological effects related to the pathology of chronic cerebral ischemia hypoperfusion. This study aimed to investigate metabolomic changes after YHF20 administration in a rat model of permanent bilateral common carotid artery occlusion (2-VO) induced chronic cerebral ischemia hypoperfusion, and to explore its impact on spatial learning and memory. Albino Wistar rats were randomly allocated to 5 groups; sham, 2-VO, 2-VO+ 100 mg/kg YHF20, 2-VO+300 mg/kg YHF20, and 2-VO+1000 mg/kg YHF20. The rats were administered YHF20 daily by oral gavage for 56 days after 2-VO induction. Plasma was collected weekly for metabolome change analysis using LC-MS/QTof and toxicity study. The rats were evaluated for spatial learning and memory using the Morris water maze. The results showed that 78 known metabolites and 10 tentative pathways altered after chronic cerebral hypoperfusion, although it was not able to determine the effect on memory and learning behaviors of rats. Glutathione and glutathione metabolism might be metabolite-pathway that were the affect after YHF20 administration in cerebral ischemic condition. The 4 known metabolites may be the metabolites from the constituents of YHF20 could be considered and confirmed for quality control purpose. In conclusion, YHF20 administration might contribute to metabolic changes related to cerebral ischemia condition without the effect on spatial learning and memory, including hepatotoxicity and nephrotoxicity after 56 days of treatment. Alterations in the potential metabolites may provide data support for elucidating dementia pathogenesis and selecting pathways for intervention.
Collapse
Affiliation(s)
- Pravit Akarasereenont
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Saracha Pattanapholkornsakul
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Suveerawan Limsuvan
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Dollaporn Mamaethong
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Suksalin Booranasubkajorn
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Pinpat Tripatara
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Kajee Pilakasiri
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| |
Collapse
|
5
|
Li Z, Zou X, Lu R, Wan X, Sun S, Wang S, Qu Y, Zhang Y, Li Z, Yang L, Fang S. Arsenic trioxide alleviates atherosclerosis by inhibiting CD36-induced endocytosis and TLR4/NF-κB-induced inflammation in macrophage and ApoE -/- mice. Int Immunopharmacol 2024; 128:111452. [PMID: 38237221 DOI: 10.1016/j.intimp.2023.111452] [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: 08/10/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Inflammation and lipid accumulation are key events in atherosclerosis progression. Despite arsenic trioxide's (ATO) toxicity, at appropriate doses, it is a useful treatment for various diseases treatment. ATO prevents vascular restenosis; however, its effects on atherosclerotic plaque development and instability remain unclear. METHODS ApoE-/- mice were fed high-fat diet for 4 months, and starting at the third month, ATO was intravenously administered every other day. Atherosclerotic lesion size, histological characteristics, and related protein and lipid profiles were assessed using samples from the aorta, carotid artery, and serum. The anti-inflammatory and anti-pyroptosis effects of ATO were investigated by stimulating RAW264.7 and THP-1 cell lines with oxidized low-density lipoprotein (ox-LDL) or lipopolysaccharide (LPS). RESULTS ATO reduced atherosclerotic lesion formation and plasma lipid levels in ApoE-/- mice. In the serum and aortic plaques, ATO reduced the levels of pro-inflammatory factors, including interleukin (IL) 6 and tumor necrosis factor α, but increased IL-10 levels. Mechanistically, ATO promoted the CD36-mediated internalization of ox-LDL in a peroxisome proliferator-activated receptor γ-dependent manner. Furthermore, ATO downregulated Toll-like receptor 4 (TLR4) expression in plaques and macrophages and inhibited p65 nuclear translocation and IκBα degradation. ATO reduced macrophage pyroptosis by downregulating NLR family pyrin domain-containing 3 (NLRP3) expression and caspase 1 activation. CONCLUSION ATO has potential atheroprotective effects, especially in macrophages. The mechanisms were inhibition of CD36-mediated foam cell formation and suppression of inflammatory responses and pyroptosis mediated by TLR4/nuclear factor κB and NLRP3 activation. Our findings provide evidence supporting the potential atheroprotective value of ATO.
Collapse
Affiliation(s)
- Zhaoying Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaoyi Zou
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Rongzhe Lu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xin Wan
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Song Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shanjie Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yinan Qu
- Department of Cardiac Function, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Yun Zhang
- Univ Texas MD Anderson Canc Ctr, Dept Clin Canc Prevent, Houston, TX 77030 USA
| | - Zhangyi Li
- Department of biochemistry and life sciences, Faculty of Arts and Sciences, Queen's University, Kingston, Ontario, Canada
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Shaohong Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
6
|
Krishnamoorthy N, Kalyan M, Hediyal TA, Anand N, Kendaganna PH, Pendyala G, Yelamanchili SV, Yang J, Chidambaram SB, Sakharkar MK, Mahalakshmi AM. Role of the Gut Bacteria-Derived Metabolite Phenylacetylglutamine in Health and Diseases. ACS OMEGA 2024; 9:3164-3172. [PMID: 38284070 PMCID: PMC10809373 DOI: 10.1021/acsomega.3c08184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024]
Abstract
Over the past few decades, it has been well established that gut microbiota-derived metabolites can disrupt gut function, thus resulting in an array of diseases. Notably, phenylacetylglutamine (PAGln), a bacterial derived metabolite, has recently gained attention due to its role in the initiation and progression of cardiovascular and cerebrovascular diseases. This meta-organismal metabolite PAGln is a byproduct of amino acid acetylation of its precursor phenylacetic acid (PAA) from a range of dietary sources like egg, meat, dairy products, etc. The microbiota-dependent metabolism of phenylalanine produces PAA, which is a crucial intermediate that is catalyzed by diverse microbial catalytic pathways. PAA conjugates with glutamine and glycine in the liver and kidney to predominantly form phenylacetylglutamine in humans and phenylacetylglycine in rodents. PAGln is associated with thrombosis as it enhances platelet activation mediated through the GPCRs receptors α2A, α2B, and β2 ADRs, thereby aggravating the pathological conditions. Clinical evidence suggests that elevated levels of PAGln are associated with pathology of cardiovascular, cerebrovascular, and neurological diseases. This Review further consolidates the microbial/biochemical synthesis of PAGln and discusses its role in the above pathophysiologies.
Collapse
Affiliation(s)
- Naveen
Kumar Krishnamoorthy
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Manjunath Kalyan
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Tousif Ahmed Hediyal
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Nikhilesh Anand
- Department
of Pharmacology, College of Medicine, American
University of Antigua, P. O. Box W-1451, Saint John’s, Antigua and Barbuda
| | - Pavan Heggadadevanakote Kendaganna
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Gurudutt Pendyala
- Department
of Anesthesiology, University of Nebraska
Medical Center (UNMC), Omaha, Nebraska 68198, United States
- Department
of Genetics, Cell Biology, and Anatomy, UNMC, Omaha, Nebraska 68198, United States
- Child Health
Research Institute, UNMC, Omaha, Nebraska 68198, United States
- National
Strategic Research Institute, UNMC, Omaha, Nebraska 68198, United States
| | - Sowmya V. Yelamanchili
- Department
of Anesthesiology, University of Nebraska
Medical Center (UNMC), Omaha, Nebraska 68198, United States
- Department
of Genetics, Cell Biology, and Anatomy, UNMC, Omaha, Nebraska 68198, United States
- National
Strategic Research Institute, UNMC, Omaha, Nebraska 68198, United States
| | - Jian Yang
- Drug
Discovery and Development Research Group, College of Pharmacy and
Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Meena Kishore Sakharkar
- Drug
Discovery and Development Research Group, College of Pharmacy and
Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Arehally M. Mahalakshmi
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| |
Collapse
|
7
|
Zhao S, Zhang P, Yan Y, Xu W, Li J, Wang L, Wang N, Huang Y. Network pharmacology-based prediction and validation of the active ingredients and potential mechanisms of the Huangxiong formula for treating ischemic stroke. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116507. [PMID: 37080367 DOI: 10.1016/j.jep.2023.116507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/30/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangxiong Formula (HXF) is composed of four herbs: Rheum palmatum L., Ligusticum striatum DC., Curcuma aromatica Salisb., and Acorus gramineus Aiton. HXF is clinically used for the treatment of ischemic stroke (IS). However, its molecular mechanism remains unclear. AIM OF THE STUDY A network pharmacology-based strategy combined with experimental study in vivo and in vitro to were used to investigate the bioactive components, potential targets, and molecular mechanisms of HXF in the treatment of IS. MATERIALS AND METHODS The components of HXF were detected by ultra-performance liquid chromatography (UPLC). The potential active ingredients of HXF were acquired from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and literature, and corresponding targets were discerned through the Swiss TargetPrediction database. IS-related targets were obtained from Genecards, Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), and DisGeNET. The intersection of ingredient and disease targets was screened, and a herbal-compound-target network was constructed. A protein-protein interaction (PPI) network was created, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. Based on these analyses, we established a compound-target-pathway (C-T-P) network. A cerebral ischemia-reperfusion (I/R) animal model was established, and the cerebral protective effect of HXF was assessed. The accuracy of the predicted targets was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Hippocampal neuronal injury cell model induced by oxygen-glucose deprivation and reperfusion (OGD/R) was used to evaluate the protective effect of α-Asarone. Furthermore, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to verify whether α-Asarone can bind to PI3K. RESULTS A total of 44 active ingredients and 795 gene targets were identified through network pharmacology. Network analysis showed that naringenin, eupatin, kaempferol, and α-Asarone were possible drug candidates. SRC, AKT1, TP53, MAPK3, STAT3, HRAS, CTNNB1, EGFR, VEGFA, PIK3R1 could serve as potential drug targets. KEGG analysis implied that the PI3K/AKT signaling pathway might play an important role in treating IS by HXF. Moreover, HXF significantly reduced neurological impairment, cerebral infarct volume, brain index, and brain histopathological damage in I/R rats. The mRNA expression of the top 10 potential targets was verified in the brain tissue. The C-T-P network and UPLC analysis suggested that α-Asarone might be an important component of HXF and can inhibit oxidative stress and apoptosis in HT22 cells by activating the PI3K/AKT signaling pathway. Molecular docking, DARTS, and CETSA assay analysis confirmed that there were direct interactions between α-Asarone and PI3K. CONCLUSION HXF had a therapeutic effect in IS with multi-component, multi-target, and multi-approach features. α-Asarone, identified as one of the major active components of HXF, could alleviate oxidative stress and apoptosis by targeting PI3K/AKT pathway.
Collapse
Affiliation(s)
- Saihong Zhao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Pingping Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yonghuan Yan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Weifang Xu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jiacheng Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ning Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China; Institute for the Evaluation of the Efficacy and Safety of Chinese Medicines, Anhui Academy of Chinese Medicine, Hefei, China.
| | - Yingying Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China; Institute for the Evaluation of the Efficacy and Safety of Chinese Medicines, Anhui Academy of Chinese Medicine, Hefei, China.
| |
Collapse
|
8
|
Fakharaldeen Z, Al-Mudhafar A, Radhi A, Hadi N. POTENTIAL PROTECTIVE EFFECTS OF NIMODIPINE FROM CEREBRAI ISCHEMIA REPERFUSION INJURY IN RATS. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 75:3094-3101. [PMID: 36723333 DOI: 10.36740/wlek202212134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim: To see whether nimodipine had neuroprotective effects in cerebral ischemia/reperfusion injury. PATIENTS AND METHODS Materials and methods: A total of 28 adult male Sprauge-dawley rats weighting 200-300 g were distributed randomly into 4 groups (7 animals in each group): sham (neck dissection without bilateral common carotid artery occlusion), control (bilateral common carotid artery occlusion for 30 minutes and reperfusion for 1 hour), vehicle (7 days of daily carboxymethylcellulose by oral gavage followed by bilateral carotid artery occlusion and reperfusion), and nimodipine-treated rats (7 days of 3 mg/kg/day of oral Azelnidipine pretreatment then bilateral common carotid artery occlusion and reperfusion). Besides assessment of histological changes and brain infarct volume, the brain tissues were sectioned to estimate NF-κB p65, IL-6, IL-10, TNF-α, ICAM-1 and total anti-oxidant capacity. RESULTS Results: Cerebral NF-κB p65, IL-6, IL-10, TNF-α, ICAM-1, in addition to cerebral infarct size were markedly increased in control and vehicle related to sham rats, while total anti-oxidant capacity was considerably decreased. Treatment with nimodipine resulted in remarkable increment of total anti-oxidant capacity, while NF-κB p65, IL-6, TNF-α, and ICAM-1 showed great reduction. Cerebral IL-10 levels didn't change by nimodipine treatment. Histologically, control and vehicle rats showed severe brain ischemic changes which is dramatically reduced by nimodipine treatment. CONCLUSION Conclusions: Our study results revealed that nimodipine can greatly decrease cerebral infarct size and reduce histological ischemic injury in male rats subjected to cerebral ischemia/ reperfusion. The neuroprotective actions of nimodipine possibly originated from its anti-inflammatory and antioxidative effects. Nimodipine protection was unrelated to IL-10.
Collapse
Affiliation(s)
- Zainab Fakharaldeen
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| | - Ahmed Al-Mudhafar
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| | - Ali Radhi
- AL-HAKEEM HOSPITAL, AL-NAJAF AL-ASHRAF, NAJAF, IRAQ
| | - Najah Hadi
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| |
Collapse
|
9
|
Syringic Acid Ameliorates Cardiac, Hepatic, Renal and Neuronal Damage Induced by Chronic Hyperglycaemia in Wistar Rats: A Behavioural, Biochemical and Histological Analysis. Molecules 2022; 27:molecules27196722. [PMID: 36235257 PMCID: PMC9573038 DOI: 10.3390/molecules27196722] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
This study investigated the effects of syringic acid (SA) on renal, cardiac, hepatic, and neuronal diabetic complications in streptozotocin-induced neonatal (nSTZ) diabetic rats. STZ (110 mg/kg i.p) was injected into Wistar rat neonates as a split dose (second and third postnatal day). Diabetes mellitus was diagnosed in adults by measuring fasting blood glucose levels, urine volume, and food and water intake. The treatment of SA (25 mg/kg, 50 mg/kg p.o) was given from the 8th to 18th postnatal week. To assess the development of diabetic complications and the effect of therapy, biochemical indicators in serum and behavioural parameters were recorded at specific intervals during the study period. SA (25 mg/kg, 50 mg/kg p.o) treatment reduced hyperglycaemia, polydipsia, polyphagia, polyuria, relative organ weight, cardiac hypertrophic indices, inflammatory markers, cell injury markers, glycated haemoglobin, histopathological score, and oxidative stress, and increased Na/K ATPase activity. These findings suggest that SA might significantly alleviate diabetic complications and/or renal, neuronal, cardiac, and hepatic damage in nSTZ diabetic rats.
Collapse
|
10
|
The Role of a Gut Microbial-Derived Metabolite, Trimethylamine N-Oxide (TMAO), in Neurological Disorders. Mol Neurobiol 2022; 59:6684-6700. [DOI: 10.1007/s12035-022-02990-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
|
11
|
Wang Y, He S, Liu X, Li Z, Zhu L, Xiao G, Du X, Du H, Zhang W, Zhang Y, Orgah J, Feng Y, Zhang B, Zhu Y. Galectin-3 Mediated Inflammatory Response Contributes to Neurological Recovery by QiShenYiQi in Subacute Stroke Model. Front Pharmacol 2021; 12:588587. [PMID: 33953667 PMCID: PMC8089377 DOI: 10.3389/fphar.2021.588587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/29/2021] [Indexed: 11/13/2022] Open
Abstract
Effective therapies for stroke are still limited due to its complex pathological manifestations. QiShenYiQi (QSYQ), a component-based Chinese medicine capable of reducing organ injury caused by ischemia/reperfusion, may offer an alternative option for stroke treatment and post-stroke recovery. Recently, we reported a beneficial effect of QSYQ for acute stroke via modulation of the neuroinflammatory response. However, if QSYQ plays a role in subacute stroke remains unknown. The pharmacological action of QSYQ was investigated in experimental stroke rats which underwent 90 min ischemia and 8 days reperfusion in this study. Neurological and locomotive deficits, cerebral infarction, brain edema, and BBB integrity were assessed. TMT-based quantitative proteomics were performed to identify differentially expressed proteins following QSYQ treatment. Immunohistochemistry, western blot analysis, RT-qPCR, and ELISA were used to validate the proteomics data and to reveal the action mechanisms. Therapeutically, treatment with QSYQ (600 mg/kg) for 7 days significantly improved neurological recovery, attenuated infarct volume and brain edema, and alleviated BBB breakdown in the stroke rats. Bioinformatics analysis indicated that protein galectin-3 and its mediated inflammatory response was closely related to the beneficial effect of QSYQ. Specially, QSYQ (600 mg/kg) markedly downregulated the mRNA and protein expression levels of galectin-3, TNF-α, and IL-6 in CI/RI brain as well as serum levels of TNF-α and IL-6. Overall, our findings showed that the effective action of QSYQ against the subacute phase of CI/RI occurs partly via regulating galectin-3 mediated inflammatory reaction.
Collapse
Affiliation(s)
- Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China.,Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Lin Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Xiaoli Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China.,Inner Mongolia Medical University, Jinshan Economic and Technological Development District, Inner Mongolia, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Wen Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China
| | - Yiqian Zhang
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin Tasly Holding Group Co., Ltd., Tianjin, China
| | - John Orgah
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Boli Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| |
Collapse
|
12
|
Hopp SC. Targeting microglia L-type voltage-dependent calcium channels for the treatment of central nervous system disorders. J Neurosci Res 2021; 99:141-162. [PMID: 31997405 PMCID: PMC9394523 DOI: 10.1002/jnr.24585] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+ ) is a ubiquitous mediator of a multitude of cellular functions in the central nervous system (CNS). Intracellular Ca2+ is tightly regulated by cells, including entry via plasma membrane Ca2+ permeable channels. Of specific interest for this review are L-type voltage-dependent Ca2+ channels (L-VDCCs), due to their pleiotropic role in several CNS disorders. Currently, there are numerous approved drugs that target L-VDCCs, including dihydropyridines. These drugs are safe and effective for the treatment of humans with cardiovascular disease and may also confer neuroprotection. Here, we review the potential of L-VDCCs as a target for the treatment of CNS disorders with a focus on microglia L-VDCCs. Microglia, the resident immune cells of the brain, have attracted recent attention for their emerging inflammatory role in several CNS diseases. Intracellular Ca2+ regulates microglia transition from a resting quiescent state to an "activated" immune-effector state and is thus a valuable target for manipulation of microglia phenotype. We will review the literature on L-VDCC expression and function in the CNS and on microglia in vitro and in vivo and explore the therapeutic landscape of L-VDCC-targeting agents at present and future challenges in the context of Alzheimer's disease, Parkinson's disease, Huntington's disease, neuropsychiatric diseases, and other CNS disorders.
Collapse
Affiliation(s)
- Sarah C. Hopp
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
13
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
14
|
Bhat A, Ray B, Mahalakshmi AM, Tuladhar S, Nandakumar DN, Srinivasan M, Essa MM, Chidambaram SB, Guillemin GJ, Sakharkar MK. Phosphodiesterase-4 enzyme as a therapeutic target in neurological disorders. Pharmacol Res 2020; 160:105078. [PMID: 32673703 DOI: 10.1016/j.phrs.2020.105078] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/08/2023]
Abstract
Phosphodiesterases (PDE) are a diverse family of enzymes (11 isoforms so far identified) responsible for the degradation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which are involved in several cellular and biochemical functions. Phosphodiesterase 4 (PDE4) is the major isoform within this group and is highly expressed in the mammalian brain. An inverse association between PDE4 and cAMP levels is the key mechanism in various pathophysiological conditions like airway inflammatory diseases-chronic obstruction pulmonary disease (COPD), asthma, psoriasis, rheumatoid arthritis, and neurological disorders etc. In 2011, roflumilast, a PDE4 inhibitor (PDE4I) was approved for the treatment of COPD. Subsequently, other PDE4 inhibitors (PDE4Is) like apremilast and crisaborole were approved by the Food and Drug Administration (FDA) for psoriasis, atopic dermatitis etc. Due to the adverse effects like unbearable nausea and vomiting, dose intolerance and diarrhoea, PDE4 inhibitors have very less clinical compliance. Efforts are being made to develop allosteric modulation with high specificity to PDE4 isoforms having better efficacy and lesser adverse effects. Interestingly, repositioning PDE4Is towards neurological disorders including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS) and sleep disorders, is gaining attention. This review is an attempt to summarize the data on the effects of PDE4 overexpression in neurological disorders and the use of PDE4Is and newer allosteric modulators as therapeutic options. We have also compiled a list of on-going clinical trials on PDE4 inhibitors in neurological disorders.
Collapse
Affiliation(s)
- Abid Bhat
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Bipul Ray
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Sunanda Tuladhar
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - D N Nandakumar
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Malathi Srinivasan
- Department of Lipid Science, CSIR - Central Food Technological Research Institute (CFTRI), CFTRI Campus, Mysuru, 570020, India
| | - Musthafa Mohamed Essa
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman; Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman.
| | - Saravana Babu Chidambaram
- Dept. of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India; Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India.
| | - Gilles J Guillemin
- Neuroinflammation group, Faculty of Medicine and Health Sciences, Macquarie University, NSW, 2109, Australia.
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK, S7N 5C9, Canada
| |
Collapse
|
15
|
Ilesanmi OB, Akinmoladun AC, Josiah SS, Olaleye MT, Akindahunsi AA. Modulation of key enzymes linked to Parkinsonism and neurologic disorders by Antiaris africana in rotenone-toxified rats. J Basic Clin Physiol Pharmacol 2019; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0014/jbcpp-2019-0014.xml. [PMID: 31800394 DOI: 10.1515/jbcpp-2019-0014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 10/18/2019] [Indexed: 01/17/2023]
Abstract
Background The physiopathologies of many neurologic diseases are characterized by related biochemical dysfunctions that could be explored as drug targets. This study evaluated the effect of a methanol leaf extract of Antiaris africana (MEA) on critical bioindices of Parkinsonism and related neurologic dysfunctions in rats with rotenone-induced neurotoxicity. Methods Animals were administered 50 or 100 mg/kg MEA for 14 consecutive days. Rotenone (1.5 mg/kg) was administered three times per day on days 13 and 14. Coenzyme Q10 (5 mg/kg) was the reference drug. Complex I activity, dopamine level, activities of acetylcholinesterase, myeloperoxidase, Na+/K+ ATPase and glutamine synthetase, as well as oxidative stress indices were evaluated at the end of the period of treatment. Results Rotenone-intoxicated group showed disruption of complex 1 activity, dopamine level, and glutamine synthetase activity with negative alterations to activities of acetylcholinesterase, myeloperoxidase, and Na+/K+ ATPase as well as heightened cerebral oxidative stress. MEA restored brain mitochondria functionality, mitigated altered neurochemical integrity, and ameliorated cerebral oxidative stress occasioned by rotenone neurotoxicity. The activity of A. Africana was comparable with that of 5 mg/kg coenzyme Q10. Conclusions These results indicated that A. africana displayed therapeutic potential against Parkinsonism and related neurologic dysfunctions and support its ethnobotanical use for the treatment of neurologic disorders.
Collapse
Affiliation(s)
- Omotayo B Ilesanmi
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, The Federal University of Technology Akure, Akure, Nigeria.,Department of Biochemistry, Faculty of Science, Federal University Otuoke, P.M.B. 126, Yenagoa, Bayelsa State, Nigeria
| | - Afolabi C Akinmoladun
- Department of Biochemistry, Faculty of Science, Federal University Otuoke, P.M.B. 126, Yenagoa, Bayelsa State, Nigeria, Phone: +2348034445893
| | - Sunday S Josiah
- Department of Biochemistry, Faculty of Science, Federal University Otuoke, P.M.B. 126, Yenagoa, Bayelsa State, Nigeria
| | - Mary Tolulope Olaleye
- Department of Biochemistry, Faculty of Science, Federal University Otuoke, P.M.B. 126, Yenagoa, Bayelsa State, Nigeria
| | - Afolabi A Akindahunsi
- Department of Biochemistry, Faculty of Science, Federal University Otuoke, P.M.B. 126, Yenagoa, Bayelsa State, Nigeria
| |
Collapse
|
16
|
Chidambaram SB, Rathipriya AG, Bolla SR, Bhat A, Ray B, Mahalakshmi AM, Manivasagam T, Thenmozhi AJ, Essa MM, Guillemin GJ, Chandra R, Sakharkar MK. Dendritic spines: Revisiting the physiological role. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:161-193. [PMID: 30654089 DOI: 10.1016/j.pnpbp.2019.01.005] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/04/2019] [Accepted: 01/12/2019] [Indexed: 12/11/2022]
Abstract
Dendritic spines are small, thin, specialized protrusions from neuronal dendrites, primarily localized in the excitatory synapses. Sophisticated imaging techniques revealed that dendritic spines are complex structures consisting of a dense network of cytoskeletal, transmembrane and scaffolding molecules, and numerous surface receptors. Molecular signaling pathways, mainly Rho and Ras family small GTPases pathways that converge on actin cytoskeleton, regulate the spine morphology and dynamics bi-directionally during synaptic activity. During synaptic plasticity the number and shapes of dendritic spines undergo radical reorganizations. Long-term potentiation (LTP) induction promote spine head enlargement and the formation and stabilization of new spines. Long-term depression (LTD) results in their shrinkage and retraction. Reports indicate increased spine density in the pyramidal neurons of autism and Fragile X syndrome patients and reduced density in the temporal gyrus loci of schizophrenic patients. Post-mortem reports of Alzheimer's brains showed reduced spine number in the hippocampus and cortex. This review highlights the spine morphogenesis process, the activity-dependent structural plasticity and mechanisms by which synaptic activity sculpts the dendritic spines, the structural and functional changes in spines during learning and memory using LTP and LTD processes. It also discusses on spine status in neurodegenerative diseases and the impact of nootropics and neuroprotective agents on the functional restoration of dendritic spines.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai, Tamil Nadu, India
| | - Srinivasa Rao Bolla
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Damam, Saudi Arabia
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Arehally Marappa Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Arokiasamy Justin Thenmozhi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | - Gilles J Guillemin
- Neuropharmacology Group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory, Macquarie University, Sydney, NSW 2109, Australia
| | - Ramesh Chandra
- Department of Chemistry, Ambedkar Centre for BioMedical Research, Delhi University, Delhi 110007, India
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK S7N 5C9, Canada.
| |
Collapse
|
17
|
Guo WL, Qu WR, Zeng LN, Qi ZP, Huang C, Zhu Z, Li R. l-Theanine and NEP1-40 promote nerve regeneration and functional recovery after brachial plexus root avulsion. Biochem Biophys Res Commun 2019; 508:1126-1132. [DOI: 10.1016/j.bbrc.2018.11.124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/20/2018] [Indexed: 01/19/2023]
|
18
|
Huebner EA, Budel S, Jiang Z, Omura T, Ho TSY, Barrett L, Merkel JS, Pereira LM, Andrews NA, Wang X, Singh B, Kapur K, Costigan M, Strittmatter SM, Woolf CJ. Diltiazem Promotes Regenerative Axon Growth. Mol Neurobiol 2018; 56:3948-3957. [PMID: 30232777 DOI: 10.1007/s12035-018-1349-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 12/31/2022]
Abstract
Axotomy results in permanent loss of function after brain and spinal cord injuries due to the minimal regenerative propensity of the adult central nervous system (CNS). To identify pharmacological enhancers of axon regeneration, 960 compounds were screened for cortical neuron axonal regrowth using an in vitro cortical scrape assay. Diltiazem, verapamil, and bromopride were discovered to facilitate axon regeneration in rat cortical cultures, in the presence of chondroitin sulfate proteoglycans (CSPGs). Diltiazem, an L-type calcium channel blocker (L-CCB), also promotes axon outgrowth in adult primary mouse dorsal root ganglion (DRG) and induced human sensory (iSensory) neurons.
Collapse
Affiliation(s)
- Eric A Huebner
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Stéphane Budel
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Zhaoxin Jiang
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Takao Omura
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tammy Szu-Yu Ho
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Lee Barrett
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Janie S Merkel
- Yale Center for Molecular Discovery, Yale University, West Haven, CT, 06516, USA
| | - Luis M Pereira
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Andrews
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kush Kapur
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Costigan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Gonçalves LV, Herlinger AL, Ferreira TAA, Coitinho JB, Pires RGW, Martins-Silva C. Environmental enrichment cognitive neuroprotection in an experimental model of cerebral ischemia: biochemical and molecular aspects. Behav Brain Res 2018; 348:171-183. [DOI: 10.1016/j.bbr.2018.04.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/07/2018] [Accepted: 04/16/2018] [Indexed: 01/25/2023]
|
20
|
Rosenberg EC, Lippman-Bell JJ, Handy M, Soldan SS, Rakhade S, Hilario-Gomez C, Folweiler K, Jacobs L, Jensen FE. Regulation of seizure-induced MeCP2 Ser421 phosphorylation in the developing brain. Neurobiol Dis 2018; 116:120-130. [PMID: 29738885 DOI: 10.1016/j.nbd.2018.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/23/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
Neonatal seizures disrupt normal synaptic maturation and often lead to later-life epilepsy and cognitive deficits. During early life, the brain exhibits heightened synaptic plasticity, in part due to a developmental overabundance of CaV1.2 L-type voltage gated calcium (Ca2+) channels (LT-VGCCs) and Ca2+-permeable AMPARs (CP-AMPARs) lacking GluA2 subunits. We hypothesized that early-life seizures overactivate these channels, in turn dysregulating Ca2+-dependent signaling pathways including that of methyl CPG binding protein 2 (MeCP2), a transcription factor implicated in the autism spectrum disorder (ASD) Rett Syndrome. Here, we show that in vivo hypoxia-induced seizures (HS) in postnatal day (P)10 rats acutely induced phosphorylation of the neuronal-specific target of activity-dependent MeCP2 phosphorylation, S421, as well as its upstream activator CaMKII T286. We next identified mechanisms by which activity-dependent Ca2+ influx induced MeCP2 phosphorylation using in vitro cortical and hippocampal neuronal cultures at embryonic day (E)18 + 10 days in vitro (DIV). In contrast to the prevalent role of NMDARs in the adult brain, we found that both CP-AMPARs and LT-VGCCs mediated MeCP2 S421 and CaMKII T286 phosphorylation induced by kainic acid (KA) or high potassium chloride (KCl) stimulation. Furthermore, in vivo post-seizure treatment with the broad-spectrum AMPAR antagonist NBQX, the CP-AMPAR blocker IEM-1460, or the LT-VGCC antagonist nimodipine blocked seizure-induced MeCP2 phosphorylation. Collectively, these results demonstrate that early-life seizures dysregulate critical activity-dependent developmental signaling pathways, in part via CP-AMPAR and LT-VGCC activation, providing novel age-specific therapeutic targets for convergent pathways underlying epilepsy and ASDs.
Collapse
Affiliation(s)
- Evan C Rosenberg
- Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States; New York University Langone Medical Center, New York, NY 10016, United States
| | - Jocelyn J Lippman-Bell
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States; Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States; Philadelphia College of Osteopathic Medicine, Department of Biomedical Sciences, Philadelphia, PA 19131, United States
| | - Marcus Handy
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Samantha S Soldan
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Sanjay Rakhade
- Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States
| | | | - Kaitlyn Folweiler
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Leah Jacobs
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States
| | - Frances E Jensen
- Perelman School of Medicine, University of Pennsylvania, Department of Neurology, Philadelphia, PA 19104, United States; Boston Children's Hospital, Department of Neurology, Boston, MA 02115, United States.
| |
Collapse
|
21
|
Wessell A, Kole MJ, Badjatia N, Parikh G, Albrecht JS, Schreibman DL, Simard JM. High Compliance with Scheduled Nimodipine Is Associated with Better Outcome in Aneurysmal Subarachnoid Hemorrhage Patients Cotreated with Heparin Infusion. Front Neurol 2017. [PMID: 28649230 PMCID: PMC5465287 DOI: 10.3389/fneur.2017.00268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Introduction We sought to determine whether compliance with scheduled nimodipine in subarachnoid hemorrhage patients impacted patient outcomes, with the intent of guiding future nimodipine management in patients who experience nimodipine-induced hypotension. Methods We performed a retrospective analysis of 118 consecutive aneurysmal subarachnoid hemorrhage patients treated with the Maryland Low-Dose IV Heparin Infusion Protocol. Patients were categorized into three independent nimodipine compliance groups: ≥1 dose held, ≥1 dose split, and no missed or split-doses. A split-dose was defined as 30 mg of nimodipine administered every 2 h. Our primary outcome was discharge to home. Bivariate and multivariable logistic regression analyses were used to assess predictors of discharge disposition as a function of nimodipine compliance. Results Of the 118 patients, 20 (17%) received all nimodipine doses, 6 (5%) received split-doses but never had a full dose held, and 92 (78%) had ≥1 dose held. Forty-five percent of patients were discharged to home, including 75% who received all doses, 67% who received ≥1 split-doses, and 37% with ≥1 missed doses (p = 0.003). Multivariable analysis showed that, along with age and World Federation of Neurosurgical Societies grade, nimodipine compliance was an independent predictor of clinical outcome; compared to missing one or more nimodipine doses, full dosing compliance was associated with increased odds of discharge to home (odds ratio 5.20; 95% confidence intervals 1.46–18.56). Conclusion In aneurysmal subarachnoid hemorrhage patients with modified Fisher scores 2 through 4 who are cotreated with a low-dose heparin infusion, full compliance with nimodipine dosing was associated with increased odds of discharge to home.
Collapse
Affiliation(s)
- Aaron Wessell
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Matthew J Kole
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj Badjatia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Gunjan Parikh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jennifer S Albrecht
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - David L Schreibman
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Buendia I, Tenti G, Michalska P, Méndez-López I, Luengo E, Satriani M, Padín-Nogueira F, López MG, Ramos MT, García AG, Menéndez JC, León R. ITH14001, a CGP37157-Nimodipine Hybrid Designed to Regulate Calcium Homeostasis and Oxidative Stress, Exerts Neuroprotection in Cerebral Ischemia. ACS Chem Neurosci 2017; 8:67-81. [PMID: 27731633 DOI: 10.1021/acschemneuro.6b00181] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During brain ischemia, oxygen and glucose deprivation induces calcium overload, extensive oxidative stress, neuroinflammation, and, finally, massive neuronal loss. In the search of a neuroprotective compound to mitigate this neuronal loss, we have designed and synthesized a new multitarget hybrid (ITH14001) directed at the reduction of calcium overload by acting on two regulators of calcium homeostasis; the mitochondrial Na+/Ca2+ exchanger (mNCX) and L-type voltage dependent calcium channels (VDCCs). This compound is a hybrid of CGP37157 (mNCX inhibitor) and nimodipine (L-type VDCCs blocker), and its pharmacological evaluation revealed a moderate ability to selectively inhibit both targets. These activities conferred concentration-dependent neuroprotection in two models of Ca2+ overload, such as toxicity induced by high K+ in the SH-SY5Y cell line (60% protection at 30 μM) and veratridine in hippocampal slices (26% protection at 10 μM). It also showed neuroprotective effect against oxidative stress, an activity related to its nitrogen radical scavenger effect and moderate induction of the Nrf2-ARE pathway. Its Nrf2 induction capability was confirmed by the increase of the expression of the antioxidant and anti-inflammatory enzyme heme-oxygenase I (3-fold increase). In addition, the multitarget profile of ITH14001 led to anti-inflammatory properties, shown by the reduction of nitrites production induced by lipopolysaccharide in glial cultures. Finally, it showed protective effect in two acute models of cerebral ischemia in hippocampal slices, excitotoxicity induced by glutamate (31% protection at 10 μM) and oxygen and glucose deprivation (76% protection at 10 μM), reducing oxidative stress and iNOS deleterious induction. In conclusion, our hybrid derivative showed improved neuroprotective properties when compared to its parent compounds CGP37157 and nimodipine.
Collapse
Affiliation(s)
- Izaskun Buendia
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Giammarco Tenti
- Departamento
de Química Orgánica y Farmacéutica, Facultad
de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Patrycja Michalska
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Iago Méndez-López
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Enrique Luengo
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Michele Satriani
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Departamento
de Química Orgánica y Farmacéutica, Facultad
de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Fernando Padín-Nogueira
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Manuela G. López
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - M. Teresa Ramos
- Departamento
de Química Orgánica y Farmacéutica, Facultad
de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Antonio G. García
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - J. Carlos Menéndez
- Departamento
de Química Orgánica y Farmacéutica, Facultad
de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Rafael León
- Instituto
Teófilo Hernando y Departamento de Farmacología y Terapéutica,
Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto
de Investigación Sanitaria, Servicio de Farmacología
Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| |
Collapse
|
23
|
Rcom-H'cheo-Gauthier AN, Osborne SL, Meedeniya ACB, Pountney DL. Calcium: Alpha-Synuclein Interactions in Alpha-Synucleinopathies. Front Neurosci 2016; 10:570. [PMID: 28066161 PMCID: PMC5167751 DOI: 10.3389/fnins.2016.00570] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/25/2016] [Indexed: 11/20/2022] Open
Abstract
Aggregation of the pre-synaptic protein, α-synuclein (α-syn), is the key etiological factor in Parkinson's disease (PD) and other alpha-synucleinopathies, such as multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB). Various triggers for pathological α-syn aggregation have been elucidated, including post-translational modifications, oxidative stress, and binding of metal ions, such as calcium. Raised neuronal calcium levels in PD may occur due to mitochondrial dysfunction and/or may relate to calcium channel dysregulation or the reduced expression of the neuronal calcium buffering protein, calbindin-D28k. Recent results on human tissue and a mouse oxidative stress model show that neuronal calbindin-D28k expression excludes α-syn inclusion bodies. Previously, cell culture model studies have shown that transient increases of intracellular free Ca(II), such as by opening of the voltage-gated plasma calcium channels, could induce cytoplasmic aggregates of α-syn. Raised intracellular free calcium and oxidative stress also act cooperatively to promote α-syn aggregation. The association between raised neuronal calcium, α-syn aggregation, oxidative stress, and neurotoxicity is reviewed in the context of neurodegenerative α-syn disease and potential mechanism-based therapies.
Collapse
Affiliation(s)
| | | | | | - Dean L. Pountney
- Menzies Health Institute Queensland, Griffith UniversityGold Coast, QLD, Australia
| |
Collapse
|
24
|
Liu B, Li F, Shi J, Yang D, Deng Y, Gong Q. Gastrodin ameliorates subacute phase cerebral ischemia‑reperfusion injury by inhibiting inflammation and apoptosis in rats. Mol Med Rep 2016; 14:4144-4152. [PMID: 27748849 PMCID: PMC5101922 DOI: 10.3892/mmr.2016.5785] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/26/2016] [Indexed: 11/08/2022] Open
Abstract
Gastrodin (GAS), which is extracted from the Chinese herbal medicine Gastrodia elata Blume, has long been used to improve stroke, epilepsy, dizziness and dementia. However, the effects and underlying mechanisms of GAS on subacute phase cerebral ischemia-reperfusion (I/R) injury remain unknown. The aim of the present study was to investigate the effects and mechanisms of GAS on cerebral I/R injury in rats. The rats were pretreated with GAS by gavage for 7 days followed by I/R surgery, and were then treated with GAS for 7 days after I/R surgery. Neurological deficits were assessed on days 1, 3 and 7 post-cerebral I/R injury. 2,3,5-Triphenyltetrazolium chloride staining was using to measure the infarct volume; morphological alterations were observed by hematoxylin and eosin staining under an optical microscope; apoptosis in the hippocampus and cortex was observed by terminal deoxynucleotidyl transferase dUTP nick end labeling staining; and the level of mRNA and protein expression was tested by reverse transcription-quantitative polymerase chain reation and western blot analysis, respectively. GAS markedly attenuated I/R-induced disability and histological damage, alleviated neuronal apoptosis, and reduced the mRNA and protein expression levels of inflammatory and proapoptotic factors, including interleukin-1β, cyclooxygenase-2, inducible nitric oxide synthase and cleaved caspase-3. These findings suggested that GAS may ameliorate subacute phase cerebral I/R injury by inhibiting inflammation and apoptosis in rats; therefore, GAS may be considered a potential candidate for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Bo Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Fei Li
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yuanyuan Deng
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education, Department of Pharmacology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
25
|
Ai J, Wan H, Shu M, Zhou H, Zhao T, Fu W, He Y. Guhong injection protects against focal cerebral ischemia-reperfusion injury via anti-inflammatory effects in rats. Arch Pharm Res 2016; 40:610-622. [PMID: 27624481 DOI: 10.1007/s12272-016-0835-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 09/07/2016] [Indexed: 11/30/2022]
Abstract
Guhong injection (GHI), composed of aceglutamide and safflower aqueous extract, has been used clinically for the treatment of cerebrovascular diseases such as cerebral embolism, hemorrhage and mental deterioration. In this paper, we reported the results of the first study on the anti-inflammatory effects of GHI in murine focal cerebral ischemia-reperfusion (I/R) injury. Adult male SD rats were randomly divided into six groups: Sham group, I/R group, GHI-L group (2.5 mL/kg), GHI-M group (5 mL/kg), GHI-H group (10 mL/kg) and Nimodipine group. I/R injury was induced by middle cerebral artery occlusion (MCAO) for 1.5 h followed by reperfusion for 24 h. Compared with I/R group, rats treated with GHI showed dose dependent reductions in neurological defect scores and cerebral infarct volume. GHI obviously down-regulated nitric oxide (NO), inducible NO synthase (iNOS), myeloperoxidase (MPO), interleukin-1β (IL-1β), TNF-α (tumor necrosis factor-α) and C reactive protein (CRP) levels in serum. Moreover, histological examination by H&E staining showed that clear cell outline, less vacuolated spaces and largely surviving neurons were observed in GHI-treated rats. The immunohistochemical staining revealed that GHI administration significantly diminished the positive expressions of intercellular cell adhesion molecule-1 (ICAM-1) and nuclear factor-κB p65 (NF-κB p65) in brain tissues. Western blot analysis for ICAM, NF-κB p65 and iNOS further solidified the above findings. All these results demonstrate that GHI exerts a strong and ameliorative effect on cerebral I/R injury in rats possibly through the inhibition of inflammation.
Collapse
Affiliation(s)
- Jinchao Ai
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Mingchun Shu
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Tao Zhao
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Wei Fu
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Yu He
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
26
|
Jia SW, Liu XY, Wang SC, Wang YF. Vasopressin Hypersecretion-Associated Brain Edema Formation in Ischemic Stroke: Underlying Mechanisms. J Stroke Cerebrovasc Dis 2016; 25:1289-300. [PMID: 27068863 DOI: 10.1016/j.jstrokecerebrovasdis.2016.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Brain edema formation is a major cause of brain damages and the high mortality of ischemic stroke. The aim of this review is to explore the relationship between ischemic brain edema formation and vasopressin (VP) hypersecretion in addition to the oxygen and glucose deprivation and the ensuing reperfusion injury. METHODS Pertinent studies involving ischemic stroke, brain edema formation, astrocytes, and VP were identified by a search of the PubMed and the Web of Science databases in January 2016. Based on clinical findings and reports of animal experiments using ischemic stroke models, this systematic review reanalyzes the implication of individual reports in the edema formation and then establishes the inherent links among them. RESULTS This systematic review reveals that cytotoxic edema and vasogenic brain edema in classical view are mainly under the influence of a continuous malfunction of astrocytic plasticity. Adaptive VP secretion can modulate membrane ion transport, water permeability, and blood-brain barrier integrity, which are largely via changing astrocytic plasticity. Maladaptive VP hypersecretion leads to disruptions of ion and water balance across cell membranes as well as the integrity of the blood-brain barrier. This review highlights our current understandings of the cellular mechanisms underlying ischemic brain edema formation and its association with VP hypersecretion. CONCLUSIONS VP hypersecretion promotes brain edema formation in ischemic stroke by disrupting hydromineral balance in the neurovascular unit; suppressing VP hypersecretion has the potential to alleviate ischemic brain edema.
Collapse
Affiliation(s)
- Shu-Wei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiao-Yu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Stephani C Wang
- Department of Surgery, Albany Medical Center, Albany, New York
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Aşcı S, Demirci S, Aşcı H, Doğuç DK, Onaran İ. Neuroprotective Effects of Pregabalin on Cerebral Ischemia and Reperfusion. Balkan Med J 2016; 33:221-7. [PMID: 27403394 DOI: 10.5152/balkanmedj.2015.15742] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/01/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Stroke is one of the most common causes of death and the leading cause of disability in adults. Cerebral ischemia/reperfusion injury causes cerebral edema, hemorrhage, and neuronal death. AIMS In post-ischemic reperfusion, free radical production causes brain tissue damage by oxidative stress. Pregabalin, an antiepileptic agent was shown to have antioxidant effects. The aim of this study was to evaluate the neuroprotective and antioxidant effects of pregabalin on ischemia and reperfusion in rat brain injury. STUDY DESIGN Animal experimentation. METHODS Male Wistar rats weighing (250-300 g) were randomly divided into six groups, each consisting of 6 rats: control (C), pregabalin (P), ischemia (I), pregabalin + ischemia (PI), ischemia + reperfusion (IR) and ischemia + reperfusion + pregabalin (PIR). Rats were initially pre-treated with 50 mg/kg/d pregabalin orally for two days. Then, animals that applied ischemia in I, PI, IR and PIR groups were exposed to carotid clamping for 30 minutes and 20 minutes reperfusion was performed in the relevant reperfusion groups. RESULTS NR2B receptor levels were significantly lower in the PIR group in comparison to the IR group. In the PIR group, Thiobarbituric acid reactive substance (TBARS) level had statistically significant decrease compared with IR group. Glutathione peroxidase (GSH-PX) levels were also significantly increased in the PIR group compared with I, IR and control groups. In the PI and PIR groups, catalase (CAT) levels were also significantly increased compared with I and IR groups (p=0.03 and p=0.07, respectively). CONCLUSION Pregabalin may protect the damage of oxidative stress after ischemia + reperfusion. This result would illuminate clinical studies in the future.
Collapse
Affiliation(s)
- Sanem Aşcı
- Neurology Service, Gülkent State Hospital, Isparta, Turkey
| | - Serpil Demirci
- Department of Neurology, Süleyman Demirel University School of Medicine, Isparta, Turkey
| | - Halil Aşcı
- Department of Pharmacology, Süleyman Demirel University School of Medicine, Isparta, Turkey
| | - Duygu Kumbul Doğuç
- Department of Biochemistry, Süleyman Demirel University School of Medicine, Isparta, Turkey
| | - İbrahim Onaran
- Department of Medical Biology and Genetic, Süleyman Demirel University School of Medicine, Isparta, Turkey
| |
Collapse
|
28
|
Kelestemur T, Yulug B, Caglayan AB, Beker MC, Kilic U, Caglayan B, Yalcin E, Gundogdu RZ, Kilic E. Targeting different pathophysiological events after traumatic brain injury in mice: Role of melatonin and memantine. Neurosci Lett 2015; 612:92-97. [PMID: 26639427 DOI: 10.1016/j.neulet.2015.11.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/12/2015] [Accepted: 11/25/2015] [Indexed: 01/08/2023]
Abstract
The tissue damage that emerges during traumatic brain injury (TBI) is a consequence of a variety of pathophysiological events, including free radical generation and over-activation of N-methyl-d-aspartate-type glutamate receptors (NMDAR). Considering the complex pathophysiology of TBI, we hypothesized that combination of neuroprotective compounds, targeting different events which appear during injury, may be a more promising approach for patients. In this context, both NMDAR antagonist memantine and free radical scavenger melatonin are safe in humans and promising agents for the treatment of TBI. Herein, we examined the effects of melatonin administered alone or in combination with memantine on the activation of signaling pathways, injury development and DNA fragmentation. Both compounds reduced brain injury moderately and the density of DNA fragmentation significantly. Notably, melatonin/memantine combination decreased brain injury and DNA fragmentation significantly, which was associated with reduced p38 and ERK-1/2 phosphorylation. As compared with melatonin and memantine groups, SAPK/JNK-1/2 phosphorylation was also reduced in melatonin/memantine combined animals. In addition, melatonin, memantine and their combination decreased iNOS activity significantly. Here, we provide evidence that melatonin/memantine combination protects brain from traumatic injury, which was associated with decreased DNA fragmentation, p38 phosphorylation and iNOS activity.
Collapse
Affiliation(s)
- Taha Kelestemur
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Burak Yulug
- Department of Neurology, University of Istanbul Medipol, Turkey
| | - Ahmet Burak Caglayan
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Mustafa Caglar Beker
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, University of Istanbul Medipol, Turkey
| | - Berrak Caglayan
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Esra Yalcin
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Reyhan Zeynep Gundogdu
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey
| | - Ertugrul Kilic
- Department of Physiology, Regenerative and Restorative Medical Research Center, University of Istanbul Medipol, Turkey.
| |
Collapse
|
29
|
Prathab Balaji S, Vijay Chand C, Justin A, Ramanathan M. Telmisartan mediates anti-inflammatory and not cognitive function through PPAR-γ agonism via SARM and MyD88 signaling. Pharmacol Biochem Behav 2015; 137:60-8. [DOI: 10.1016/j.pbb.2015.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 10/23/2022]
|
30
|
Jeitner TM, Battaile K, Cooper AJL. Critical Evaluation of the Changes in Glutamine Synthetase Activity in Models of Cerebral Stroke. Neurochem Res 2015; 40:2544-56. [DOI: 10.1007/s11064-015-1667-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/14/2015] [Accepted: 05/20/2015] [Indexed: 01/04/2023]
|
31
|
Li R. Hot spots and future directions of research on the neuroprotective effects of nimodipine. Neural Regen Res 2015; 9:1933-8. [PMID: 25558246 PMCID: PMC4281435 DOI: 10.4103/1673-5374.145365] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2014] [Indexed: 01/17/2023] Open
Abstract
Calcium antagonists are widely used in the clinical treatment of ischemic cerebrovascular disease because of their vascular and neuroprotective effects. Nimodipine, a typical calcium antagonist, can cross the blood-brain barrier and act selectively at neurons and blood vessels of target tissues, thus exerting neuroprotective effects. The aim of the present study was to explore the hot spots and future trends of research on the neuroprotective effects of nimodipine. We retrieved 425 articles on the neuroprotective effects of nimodipine that were indexed in the Web of the Science database between 2000 and 2014. The retrieved articles were analyzed using document analysis reporting and the derived information function in the Web of Science, and the information visualization software CiteSpace III. The reference co-citation network was plotted, and the high frequency key words in these publications were used to analyze the research fronts and development trends for nimodipine neuroprotection. According to these co-citation clusters, the research front of nimodipine neuroprotection is the use of randomized controlled trials to study nimodipine intervention of subarachnoid hemorrhage. Using time zone view analysis on hot spots labeled with a key word, the areas of interest in the field of nimodipine neuroprotection are nimodipine pharmacology and therapeutics, blood-brain barrier, trials, and anti-angiospasm.
Collapse
Affiliation(s)
- Runhui Li
- Department of Neurology, Affiliated Central Hospital of Shenyang Medical College, Shenyang, Liaoning Province, China
| |
Collapse
|
32
|
Wang F, Liang W, Lei C, Kinden R, Sang H, Xie Y, Huang Y, Qu Y, Xiong L. Combination of HBO and Memantine in Focal Cerebral Ischemia: Is There a Synergistic Effect? Mol Neurobiol 2014; 52:1458-1466. [DOI: 10.1007/s12035-014-8949-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/20/2014] [Indexed: 11/30/2022]
|
33
|
Nuñez-Figueredo Y, Ramírez-Sánchez J, Hansel G, Simões Pires EN, Merino N, Valdes O, Delgado-Hernández R, Parra AL, Ochoa-Rodríguez E, Verdecia-Reyes Y, Salbego C, Costa SL, Souza DO, Pardo-Andreu GL. A novel multi-target ligand (JM-20) protects mitochondrial integrity, inhibits brain excitatory amino acid release and reduces cerebral ischemia injury in vitro and in vivo. Neuropharmacology 2014; 85:517-27. [DOI: 10.1016/j.neuropharm.2014.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/26/2014] [Accepted: 06/10/2014] [Indexed: 12/22/2022]
|
34
|
Singhal K, Sandhir R. L-type calcium channel blocker ameliorates diabetic encephalopathy by modulating dysregulated calcium homeostasis. J Neurosci Res 2014; 93:296-308. [DOI: 10.1002/jnr.23478] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 07/20/2014] [Accepted: 08/07/2014] [Indexed: 01/02/2023]
Affiliation(s)
- Kirti Singhal
- Department of Biochemistry; Panjab University; Chandigarh India
| | - Rajat Sandhir
- Department of Biochemistry; Panjab University; Chandigarh India
| |
Collapse
|
35
|
Ogundele OM, Adeniyi PA, Ajonijebu DC, Abdulbasit A, Cobham AE, Ishola AO, Balogun GW. Motor and memory function in rat models of cyanide toxicity and vascular occlusion induced ischemic injury. PATHOPHYSIOLOGY 2014; 21:191-8. [DOI: 10.1016/j.pathophys.2014.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/07/2014] [Accepted: 07/20/2014] [Indexed: 02/04/2023] Open
|
36
|
|
37
|
Sun JH, Tan L, Yu JT. Post-stroke cognitive impairment: epidemiology, mechanisms and management. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:80. [PMID: 25333055 PMCID: PMC4200648 DOI: 10.3978/j.issn.2305-5839.2014.08.05] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 07/18/2014] [Indexed: 01/26/2023]
Abstract
Post-stroke cognitive impairment occurs frequently in the patients with stroke. The prevalence of post-stroke cognitive impairment ranges from 20% to 80%, which varies for the difference between the countries, the races, and the diagnostic criteria. The risk of post-stroke cognitive impairment is related to both the demographic factors like age, education and occupation and vascular factors. The underlying mechanisms of post-stroke cognitive impairment are not known in detail. However, the neuroanatomical lesions caused by the stroke on strategic areas such as the hippocampus and the white matter lesions (WMLs), the cerebral microbleeds (CMBs) due to the small cerebrovascular diseases and the mixed AD with stroke, alone or in combination, contribute to the pathogenesis of post-stroke cognitive impairment. The treatment of post-stroke cognitive impairment may benefit not only from the anti-dementia drugs, but also the manage measures on cerebrovascular diseases. In this review, we will describe the epidemiological features and the mechanisms of post-stroke cognitive impairment, and discuss the promising management strategies for these patients.
Collapse
|
38
|
Non-hypotensive dose of telmisartan and nimodipine produced synergistic neuroprotective effect in cerebral ischemic model by attenuating brain cytokine levels. Pharmacol Biochem Behav 2014; 122:61-73. [DOI: 10.1016/j.pbb.2014.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/30/2014] [Accepted: 03/12/2014] [Indexed: 02/04/2023]
|
39
|
Huang BR, Chang PC, Yeh WL, Lee CH, Tsai CF, Lin C, Lin HY, Liu YS, Wu CYJ, Ko PY, Huang SS, Hsu HC, Lu DY. Anti-neuroinflammatory effects of the calcium channel blocker nicardipine on microglial cells: implications for neuroprotection. PLoS One 2014; 9:e91167. [PMID: 24621589 PMCID: PMC3951295 DOI: 10.1371/journal.pone.0091167] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 02/11/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/OBJECTIVE Nicardipine is a calcium channel blocker that has been widely used to control blood pressure in severe hypertension following events such as ischemic stroke, traumatic brain injury, and intracerebral hemorrhage. However, accumulating evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play important roles in neurodegeneration, and the effect of nicardipine on microglial activation remains unresolved. METHODOLOGY/PRINCIPAL FINDINGS In the present study, using murine BV-2 microglia, we demonstrated that nicardipine significantly inhibits microglia-related neuroinflammatory responses. Treatment with nicardipine inhibited microglial cell migration. Nicardipine also significantly inhibited LPS plus IFN-γ-induced release of nitric oxide (NO), and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, nicardipine also inhibited microglial activation by peptidoglycan, the major component of the Gram-positive bacterium cell wall. Notably, nicardipine also showed significant anti-neuroinflammatory effects on microglial activation in mice in vivo. CONCLUSION/SIGNIFICANCE The present study is the first to report a novel inhibitory role of nicardipine on neuroinflammation and provides a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Bor-Ren Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Neurosurgery Department, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, United States of America
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Caren Yu-Ju Wu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Pei-Ying Ko
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Horng-Chaung Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
40
|
Figueredo YN, Rodríguez EO, Reyes YV, Domínguez CC, Parra AL, Sánchez JR, Hernández RD, Verdecia MP, Pardo Andreu GL. Characterization of the anxiolytic and sedative profile of JM-20: a novel benzodiazepine–dihydropyridine hybrid molecule. Neurol Res 2013; 35:804-12. [DOI: 10.1179/1743132813y.0000000216] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Estael Ochoa Rodríguez
- Laboratorio de Síntesis Orgánica de La Facultad de Química de La Universidad de La Habana Ciudad de la Habana, Cuba
| | - Yamila Verdecia Reyes
- Centro de Estudio para las Investigaciones y Evaluaciones Biológicas, Instituto de Farmacia y Alimentos, Universidad de La Habana, Ciudad Habana, Cuba
| | | | | | | | | | | | | |
Collapse
|
41
|
Linden J, Fassotte L, Tirelli E, Plumier JC, Ferrara A. Assessment of behavioral flexibility after middle cerebral artery occlusion in mice. Behav Brain Res 2013; 258:127-37. [PMID: 24157337 DOI: 10.1016/j.bbr.2013.10.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 02/04/2023]
Abstract
Middle cerebral artery occlusion (MCAO) is the most common animal model of cerebral ischemia and induces various functional impairments. Long-lasting deficits resulting from MCAO however, remain insufficiently characterized, especially regarding cognition. Yet, behavioral flexibility, a prominent cognitive process is found impaired after stroke in humans. We thus used an operant-based task to assess behavioral flexibility in mice after MCAO. Three weeks after 30 min MCAO surgery, mice were subjected to a battery of sensorimotor tests (rotarod, vertical pole test, spontaneous locomotion and grip-strength test). Behavioral flexibility was then assessed in an operant task, in which mice, rewarded according to a FR5 schedule of reinforcement, had to alternate their operant responses between two levers from trial to trial. Regarding sensory and motor functioning, only the pole test yielded a significant difference between MCAO and sham mice. In the operant flexibility task, results showed a behavioral flexibility deficit in MCAO mice; neither the operant response acquisition nor the appeal for food rewards was altered. In conclusion, our operant-based task revealed a long-lasting behavioral flexibility deficit after MCAO in mice.
Collapse
Affiliation(s)
- Jérôme Linden
- Département de Psychologie, Cognition et Comportement, Université de Liège, 4000 Liège, Belgium.
| | | | | | | | | |
Collapse
|
42
|
Gurkoff G, Shahlaie K, Lyeth B, Berman R. Voltage-gated calcium channel antagonists and traumatic brain injury. Pharmaceuticals (Basel) 2013; 6:788-812. [PMID: 24276315 PMCID: PMC3816709 DOI: 10.3390/ph6070788] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 01/17/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States. Despite more than 30 years of research, no pharmacological agents have been identified that improve neurological function following TBI. However, several lines of research described in this review provide support for further development of voltage gated calcium channel (VGCC) antagonists as potential therapeutic agents. Following TBI, neurons and astrocytes experience a rapid and sometimes enduring increase in intracellular calcium ([Ca2+]i). These fluxes in [Ca2+]i drive not only apoptotic and necrotic cell death, but also can lead to long-term cell dysfunction in surviving cells. In a limited number of in vitro experiments, both L-type and N-type VGCC antagonists successfully reduced calcium loads as well as neuronal and astrocytic cell death following mechanical injury. In rodent models of TBI, administration of VGCC antagonists reduced cell death and improved cognitive function. It is clear that there is a critical need to find effective therapeutics and rational drug delivery strategies for the management and treatment of TBI, and we believe that further investigation of VGCC antagonists should be pursued before ruling out the possibility of successful translation to the clinic.
Collapse
Affiliation(s)
- Gene Gurkoff
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
- NSF Center for Biophotonics Science and Technology, Suite 2700 Stockton Blvd, Suite 1400, Sacramento, CA, 95817, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-530-754-7501; Fax: +1-530-754-5125
| | - Kiarash Shahlaie
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Bruce Lyeth
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Robert Berman
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| |
Collapse
|
43
|
Kilic U, Yilmaz B, Reiter R, Yüksel A, Kilic E. Effects of memantine and melatonin on signal transduction pathways vascular leakage and brain injury after focal cerebral ischemia in mice. Neuroscience 2013; 237:268-76. [DOI: 10.1016/j.neuroscience.2013.01.059] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 12/09/2022]
|
44
|
Mestriner RG, Miguel PM, Bagatini PB, Saur L, Boisserand LSB, Baptista PPA, Xavier LL, Netto CA. Behavior outcome after ischemic and hemorrhagic stroke, with similar brain damage, in rats. Behav Brain Res 2013; 244:82-9. [DOI: 10.1016/j.bbr.2013.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/29/2013] [Accepted: 02/01/2013] [Indexed: 01/13/2023]
|
45
|
Zukhurova M, Prosvirnina M, Daineko A, Simanenkova A, Petrishchev N, Sonin D, Galagudza M, Shamtsyan M, Juneja LR, Vlasov T. L-theanine Administration Results in Neuroprotection and Prevents Glutamate Receptor Agonist-Mediated Injury in the Rat Model of Cerebral Ischemia-Reperfusion. Phytother Res 2012; 27:1282-7. [DOI: 10.1002/ptr.4868] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/10/2012] [Accepted: 10/02/2012] [Indexed: 01/21/2023]
Affiliation(s)
- Mavdzhuda Zukhurova
- Department of Pathophysiology; I. P. Pavlov Federal Medical University; St-Petersburg; Russian Federation
| | - Maria Prosvirnina
- Department of Pathophysiology; I. P. Pavlov Federal Medical University; St-Petersburg; Russian Federation
| | - Anastasia Daineko
- Department of Pathophysiology; I. P. Pavlov Federal Medical University; St-Petersburg; Russian Federation
| | - Anna Simanenkova
- Department of Pathophysiology; I. P. Pavlov Federal Medical University; St-Petersburg; Russian Federation
| | | | | | | | - Mark Shamtsyan
- Department of Technology of Microbiological Synthesis; St-Petersburg State Institute of Technology (Technical University); St-Petersburg; Russian Federation
| | - Lekh R. Juneja
- Division of Nutrition; Taiyo Kagaku Co. Ltd.; Yokkaichi; Japan
| | | |
Collapse
|
46
|
Abstract
Cryonics technology seeks to cryopreserve the anatomical basis of the mind so that future medicine can restore legally dead cryonics patients to life, youth, and health. Most cryonics patients experience varying degrees of ischemia and reperfusion injury. Neurons can survive ischemia and reperfusion injury more than is generally believed, but blood vessels are more vulnerable, and such injury can impair perfusion of vitrifying cryoprotectant solution intended to eliminate ice formation in the brain. Forms of vascular and neuronal damage are reviewed, along with means of mitigating that damage. Recommendations are also made for preventing such damage.
Collapse
Affiliation(s)
- Benjamin P Best
- Cryonics Institute, 24355 Sorrentino Court, Clinton Township, MI 48035, USA.
| |
Collapse
|
47
|
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: 2.2] [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
|
48
|
Inhibition of human astrocyte and microglia neurotoxicity by calcium channel blockers. Neuropharmacology 2012; 63:685-91. [PMID: 22659089 DOI: 10.1016/j.neuropharm.2012.05.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/12/2012] [Accepted: 05/24/2012] [Indexed: 11/24/2022]
Abstract
We examined the effects of L-type calcium channel blockers (CCBs) on toxicity exerted by activated human astrocytes and microglia towards SH-SY5Y human neuronal cells. The CCBs nimodipine (NDP) and verapamil (VPM) both significantly suppressed toxic secretions from human astrocytes and astrocytoma U-373 MG cells that were induced by interferon (IFN)-γ. NDP also inhibited neurotoxic secretions of human microglia and monocytic THP-1 cells that were induced by the combination of lipopolysaccharide and IFN-γ. In human astrocytes, both NDP and VPM reduced IFN-γ-induced phosphorylation of signal transducer and activator of transcription (STAT) 3. They also inhibited the astrocytic production of IFN-γ-inducible T cell α chemoattractant (I-TAC). These results suggest that CCBs attenuate IFN-γ-induced neurotoxicity of human astrocytes through inhibition of the STAT3 signaling pathway. L-type CCBs, especially NDP, might be a useful treatment option for a broad spectrum of neurodegenerative diseases, including Alzheimer disease, where the pathology is believed to be exacerbated by neurotoxic glial activation.
Collapse
|