1
|
Tabassum S, Shorter S, Ovsepian SV. Analysis of the action mechanisms and targets of herbal anticonvulsants highlights opportunities for therapeutic engagement with refractory epilepsy. J Mol Med (Berl) 2024; 102:761-771. [PMID: 38653825 PMCID: PMC11106186 DOI: 10.1007/s00109-024-02445-5] [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: 12/11/2023] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Epilepsy is a neurological disorder characterized by spontaneous and recurring seizures. It poses significant therapeutic challenges due to diverse etiology, pathobiology, and pharmacotherapy-resistant variants. The anticonvulsive effects of herbal leads with biocompatibility and toxicity considerations have attracted much interest, inspiring mechanistic analysis with the view of their use for engagement of new targets and combination with antiseizure pharmacotherapies. This article presents a comprehensive overview of the key molecular players and putative action mechanisms of the most common antiepileptic herbals demonstrated in tissue culture and preclinical models. From the review of the literature, it emerges that their effects are mediated via five distinct mechanisms: (1) reduction of membrane excitability through inhibition of cation channels, (2) improvement of mitochondrial functions with antioxidant effects, (3) enhancement in synaptic transmission mediated by GABAA receptors, (4) improvement of immune response with anti-inflammatory action, and (5) suppression of protein synthesis and metabolism. While some of the primary targets and action mechanisms of herbal anticonvulsants (1, 3) are shared with antiseizure pharmacotherapies, herbal leads also engage with distinct mechanisms (2, 4, and 5), suggesting new drug targets and opportunities for their integration with antiseizure medications. Addressing outstanding questions through research and in silico modeling should facilitate the future use of herbals as auxiliary therapy in epilepsy and guide the development of treatment of pharmacoresistant seizures through rigorous trials and regulatory approval.
Collapse
Affiliation(s)
- Sobia Tabassum
- Department of Biological Sciences, Faculty of Sciences, International Islamic University, Islamabad, Pakistan
| | - Susan Shorter
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK.
- Faculty of Medicine, Tbilisi State University, Tbilisi, 0177, Republic of Georgia.
| |
Collapse
|
2
|
He S, Shi J, Chai H, Ma L, Pei H, Zhang P, Shi D, Li H. Mechanisms with network pharmacology approach of Ginsenosides in Alzheimer's disease. Heliyon 2024; 10:e26642. [PMID: 38434355 PMCID: PMC10906400 DOI: 10.1016/j.heliyon.2024.e26642] [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: 08/08/2023] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss, cognitive disorder, language dysfunction, and mental disability. The main neuropathological changes in AD mainly include amyloid plaque deposition, neurofibrillary tangles, synapse loss, and neuron reduction. However, the current anti-AD drugs do not demonstrate a favorable effect in altering the pathological course of AD. Moreover, long-term use of these drugs is usually accompanied with various side effects. Ginsenosides are the major active constituents of ginseng and have protective effects on AD through various mechanisms in both in vivo and in vitro studies. In this review, we focused on discussing the therapeutic potential effects and the mechanisms of pharmacological activities of ginsenosides in AD, to provide new insight for further research and clinical application of ginsenosides in the future. Recent studies on the pharmacological effects and mechanisms of ginsenosides were retrieved from Chinese National Knowledge Infrastructure, National Science and Technology Library, Wanfang Data, Elsevier, ScienceDirect, PubMed, SpringerLink, and the Web of Science database up to April 2023 using relevant keywords. Network pharmacology and bioinformatics analysis were used to predict the therapeutic effects and mechanisms of ginsenosides against AD. Ginsenosides presented a wide range of therapeutic and biological activities, including alleviating Aβ deposition, decreasing tau hyperphosphorylation, regulating the cholinergic system, resisting oxidative stress, modulating Ca2+ homeostasis, as well as anti-inflammation and anti-apoptosis in neurons, respectively. For further developing the therapeutic potential as well as clinical applications, the network pharmacology approach was combined with a summary of published studies.
Collapse
Affiliation(s)
- Shan He
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junhe Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Chai
- Hepingli Hospital, Beijing, China
| | - Lina Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Cuong Tran NK, Jeong JH, Sharma N, Doan Nguyen YN, Phi Tran HY, Dang DK, Park JH, Byun JK, Jin D, Xiaoyan Z, Ko SK, Nah SY, Kim HC, Shin EJ. Ginsenoside Re blocks Bay k-8644-induced neurotoxicity via attenuating mitochondrial dysfunction and PKCδ activation in the hippocampus of mice: Involvement of antioxidant potential. Food Chem Toxicol 2023:113869. [PMID: 37308051 DOI: 10.1016/j.fct.2023.113869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
Although the anticonvulsant effects of ginsenosides are recognized, little is known about their effects on the convulsive behaviors induced by the activation of L-type Ca2+ channels. Here, we investigated whether ginsenoside Re (GRe) modulates excitotoxicity induced by the L-type Ca2+ channel activator Bay k-8644. GRe significantly attenuated Bay k-8644-induced convulsive behaviors and hippocampal oxidative stress in mice. GRe-mediated antioxidant potential was more pronounced in the mitochondrial fraction than cytosolic fraction. As L-type Ca2+ channels are thought to be targets of protein kinase C (PKC), we investigated the role of PKC under excitotoxic conditions. GRe attenuated Bay k-8644-induced mitochondrial dysfunction, PKCδ activation, and neuronal loss. The PKCδ inhibition and neuroprotection mediated by GRe were comparable to those by the ROS inhibitor N-acetylcysteine, the mitochondrial protectant cyclosporin A, the microglial inhibitor minocycline, or the PKCδ inhibitor rottlerin. Consistently, the GRe-mediated PKCδ inhibition and neuroprotection were counteracted by the mitochondrial toxin 3-nitropropionic acid or the PKC activator bryostatin-1. GRe treatment did not have additional effects on PKCδ gene knockout-mediated neuroprotection, suggesting that PKCδ is a molecular target of GRe. Collectively, our results suggest that GRe-mediated anticonvulsive/neuroprotective effects require the attenuation of mitochondrial dysfunction and altered redox status and inactivation of PKCδ.
Collapse
Affiliation(s)
- Ngoc Kim Cuong Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yen Nhi Doan Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Hoang-Yen Phi Tran
- Physical Chemistry Department, University of Medicine and Pharmacy, Ho Chi Minh City, 760000, Viet Nam
| | - Duy-Khanh Dang
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Jung Hoon Park
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Jae Kyung Byun
- Korea Society of Forest Environmental Research, Namyangju, 12106, Republic of Korea
| | - Dezhong Jin
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Zeng Xiaoyan
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
4
|
Wu J, Wang S, Zhao W, Li M, Li S. Ginsenoside Rh2 inhibits CBP/p300-mediated FOXO3a acetylation and epilepsy-induced oxidative damage via the FOXO3a-KEAP1-NRF2 pathway. Eur J Pharmacol 2023; 940:175391. [PMID: 36400161 DOI: 10.1016/j.ejphar.2022.175391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Epilepsy is a chronic disease that affects a wide range of people. Furthermore, a third of patients suffering from epileptic seizures do not respond to antiepileptic drugs. In recent years, increasing attention has focused on the role of oxidative stress in acquired epilepsy, and adjuvant antiepileptic drugs to reduce oxidative stress may be a new therapeutic strategy. In this study ginsenoside Rh2 was resistant to oxidative stress induced by epileptic activity in vivo and in vitro. Using online databases, we identified forkhead box O3a (FOXO3a) overexpression in epilepsy tissue and validated this in vitro, in vivo, and in clinical tissues of patients with epilepsy. An in vitro epilepsy model revealed that the overexpression of FOXO3a led to more severe oxidative stress, while the knockdown of FOXO3a had a protective effect on SH-SY5Y cells. Moreover, our results showed that the positive effect of FOXO3a on oxidative stress was caused by the transcriptional activation of Kelch-like ECH-associated protein 1 (KEAP1), a negative regulator of nuclear factor erythroid 2-related factor 2 (NRF2). We also found that ginsenoside Rh2 can directly inhibit the activation of FOXO3a by selectively blocking CREB-binding protein (CBP)/p300-mediated FOXO3a acetylation and play a role in regulating the KEAP1-NRF2 pathway to resist oxidative stress.
Collapse
Affiliation(s)
- Jingheng Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Shuai Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Wujun Zhao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Miaomiao Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Shaoyi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China.
| |
Collapse
|
5
|
He LY, Hu MB, Li RL, Zhao R, Fan LH, He L, Lu F, Ye X, Huang YL, Wu CJ. Natural Medicines for the Treatment of Epilepsy: Bioactive Components, Pharmacology and Mechanism. Front Pharmacol 2021; 12:604040. [PMID: 33746751 PMCID: PMC7969896 DOI: 10.3389/fphar.2021.604040] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
Epilepsy is a chronic disease that can cause temporary brain dysfunction as a result of sudden abnormal discharge of the brain neurons. The seizure mechanism of epilepsy is closely related to the neurotransmitter imbalance, synaptic recombination, and glial cell proliferation. In addition, epileptic seizures can lead to mitochondrial damage, oxidative stress, and the disorder of sugar degradation. Although the mechanism of epilepsy research has reached up to the genetic level, the presently available treatment and recovery records of epilepsy does not seem promising. Recently, natural medicines have attracted more researches owing to their low toxicity and side-effects as well as the excellent efficacy, especially in chronic diseases. In this study, the antiepileptic mechanism of the bioactive components of natural drugs was reviewed so as to provide a reference for the development of potential antiepileptic drugs. Based on the different treatment mechanisms of natural drugs considered in this review, it is possible to select drugs clinically. Improving the accuracy of medication and the cure rate is expected to compensate for the shortage of the conventional epilepsy treatment drugs.
Collapse
Affiliation(s)
- Li-Ying He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mei-Bian Hu
- Institute of Pharmaceutical and Food engineering, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Ruo-Lan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin-Hong Fan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Lu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xun Ye
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong-Liang Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chun-Jie Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Liu L, Xu FR, Wang YZ. Traditional uses, chemical diversity and biological activities of Panax L. (Araliaceae): A review. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:112792. [PMID: 32311488 DOI: 10.1016/j.jep.2020.112792] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/22/2020] [Accepted: 03/22/2020] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax L. (Araliaceae) is globally-recognized plant resource suitable for the globalization of traditional Chinese medicines. It has traditionally been used as tonic agents in various ethnomedicinal systems of East Asia, especially in China. It is often used to regulate bodily functions and considered as adjuvant therapy for tumor, resuscitation of traumatic hemorrhagic shock, etc. AIM OF THIS REVIEW: This review systematically summarized the information on distributions, botanical characteristics, traditional uses, chemical components and biological activities of the genus Panax, in order to explore and exploit the therapeutic potential of this plant. MATERIALS AND METHODS The available information about genus Panax was collected via the online search on Web of Science, Google Scholar, PubMed, Baidu Scholar, Science Direct, China National Knowledge Infrastructure and Springer search. The keywords used include Panax, saponin, secondary metabolites, chemical components, biological activity, pharmacology, traditional medicinal uses, safety and other related words. The Plant List (www.theplantlist.org) and Catalogue of Life: 2019 Annual Checklist (www.catalogueoflife.org/col/) databases were used to provide the scientific names, subspecies classification and distribution information of Panax. RESULTS Panax is widely assessed concerning its phytochemistry and biological activities. To date, at least 748 chemical compounds from genus Panax were isolated, including saponins, flavonoids, polysaccharides, steroids and phenols. Among them, triterpenoid saponins and polysaccharides were the representative active ingredients of Panax plants, which have been widely investigated. Modern pharmacological studies showed that these compounds exhibited a wide range of biological activities in vitro and in vivo including antineoplastic, anti-inflammatory, hepatorenal protective, neuroprotective, immunoregulatory, cardioprotective and antidiabetic activities. Many studies also confirmed that the mechanisms of organ-protective were closely related to molecular signaling pathways, the expression of related proteins and antioxidant reactions. To sum up, genus Panax has high medicinal and social value, deserving further investigation. CONCLUSIONS The genus Panax is very promising to be fully utilized in the development of nutraceutical and pharmaceutical products. However, there is a lack of in-depth studies on ethnomedicinal uses of Panax plants. In addition, further studies of single chemical component should be performed based on the diversity of chemical structure, significant biological activities and clinical application. If the bioactive molecules and multicomponent interactions are discovered, it will be of great significance to the clinical application of Panax plants. It is an urgent requirement to carry out detailed phytochemical, pharmacology and clinical research on Panax classical prescriptions for the establishment of modern medication guidelines. Exploring the molecular basis of herbal synergistic actions may provide a new understanding of the complex disease mechanisms and accelerate the process of pharmaceutical development.
Collapse
Affiliation(s)
- Lu Liu
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China; College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Fu-Rong Xu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China.
| | - Yuan-Zhong Wang
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China.
| |
Collapse
|
7
|
Miranda AS, Cardozo PL, Silva FR, de Souza JM, Olmo IG, Cruz JS, Gomez MV, Ribeiro FM, Vieira LB. Alterations of Calcium Channels in a Mouse Model of Huntington's Disease and Neuroprotection by Blockage of Ca V1 Channels. ASN Neuro 2020; 11:1759091419856811. [PMID: 31216184 PMCID: PMC6585245 DOI: 10.1177/1759091419856811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative autosomal dominant disorder, characterized by symptoms of involuntary movement of the body, loss of cognitive function, psychiatric disorder, leading inevitably to death. It has been previously described that higher levels of brain expression of Cav1 channels are involved in major neurodegenerative disorders, such as Alzheimer’s disease and Parkinson’s disease. Our results demonstrate that a bacterial artificial chromosome (BAC)-mediated transgenic mouse model (BACHD mice) at the age of 3 and 12 months exhibits significantly increased Cav1.2 protein levels in the cortex, as compared with wild-type littermates. Importantly, electrophysiological analyses confirm a significant increase in L-type Ca2+ currents and total Ca2+ current density in cortical neurons from BACHD mice. By using an in vitro assay to measure neuronal cell death, we were able to observe neuronal protection against glutamate toxicity after treatment with Cav1 blockers, in wild-type and, more importantly, in BACHD neurons. According to our data, Cav1 blockers may offer an interesting strategy for the treatment of HD. Altogether, our results show that mutant huntingtin (mHtt) expression may cause a dysregulation of Cav1.2 channels and we hypothesize that this contributes to neurodegeneration during HD.
Collapse
Affiliation(s)
- Artur S Miranda
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo Leal Cardozo
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flavia R Silva
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jessica M de Souza
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella G Olmo
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jader S Cruz
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Fabiola M Ribeiro
- 1 Department of Biochemistry and Immunology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciene B Vieira
- 3 Department of Pharmacology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
8
|
Abstract
Epilepsy is considered a major serious chronic neurological disorder, characterized by recurrent seizures. It is usually associated with a history of a lesion in the nervous system. Irregular activation of inflammatory molecules in the injured tissue is an important factor in the development of epilepsy. It is unclear how the imbalanced regulation of inflammatory mediators contributes to epilepsy. A recent research goal is to identify interconnected inflammation pathways which may be involved in the development of epilepsy. The clinical use of available antiepileptic drugs is often restricted by their limitations, incidence of several side effects, and drug interactions. So development of new drugs, which modulate epilepsy through novel mechanisms, is necessary. Alternative therapies and diet have recently reported positive treatment outcomes in epilepsy. Vitamin D (Vit D) has shown prophylactic and therapeutic potential in different neurological disorders. So, the aim of current study was to review the associations between different brain inflammatory mediators and epileptogenesis, to strengthen the idea that targeting inflammatory pathway may be an effective therapeutic strategy to prevent or treat epilepsy. In addition, neuroprotective effects and mechanisms of Vit D in clinical and preclinical studies of epilepsy were reviewed.
Collapse
|
9
|
Razgonova MP, Veselov VV, Zakharenko AM, Golokhvast KS, Nosyrev AE, Cravotto G, Tsatsakis A, Spandidos DA. Panax ginseng components and the pathogenesis of Alzheimer's disease (Review). Mol Med Rep 2019; 19:2975-2998. [PMID: 30816465 PMCID: PMC6423617 DOI: 10.3892/mmr.2019.9972] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/15/2019] [Indexed: 12/02/2022] Open
Abstract
Ginseng is one of the main representatives of traditional Chinese medicine and presents a wide range of pharmacological actions. Ginsenosides are the main class of active compounds found in ginseng. They demonstrate unique biological activity and medicinal value, namely anti-tumour, anti-inflammatory and antioxidant properties, as well as anti-apoptotic properties. Increasing levels of stress in life are responsible for the increased incidence of nervous system diseases. Neurological diseases create a huge burden on the lives and health of individuals. In recent years, studies have indicated that ginsenosides play a pronounced positive role in the prevention and treatment of neurological diseases. Nevertheless, research is still at an early stage of development, and the complex mechanisms of action involved remain largely unknown. This review aimed to shed light into what is currently known about the mechanisms of action of ginsenosides in relation to Alzheimer's disease. Scientific material and theoretical bases for the treatment of nervous system diseases with purified Panax ginseng extracts are also discussed.
Collapse
Affiliation(s)
| | - Valery Vyacheslavovich Veselov
- Center of Bioanalytical Investigation and Molecular Design, I.M. Sechenov First Moscow State Medical University, Moscow 119048, Russia
| | | | | | - Alexander Evgenyevich Nosyrev
- Center of Bioanalytical Investigation and Molecular Design, I.M. Sechenov First Moscow State Medical University, Moscow 119048, Russia
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, University of Turin, Turin 10125, Italy
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion 71003, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71003, Greece
| |
Collapse
|
10
|
Zeng X, Hu K, Chen L, Zhou L, Luo W, Li C, Zong W, Chen S, Gao Q, Zeng G, Jiang D, Li X, Zhou H, Ouyang DS. The Effects of Ginsenoside Compound K Against Epilepsy by Enhancing the γ-Aminobutyric Acid Signaling Pathway. Front Pharmacol 2018; 9:1020. [PMID: 30254585 PMCID: PMC6142013 DOI: 10.3389/fphar.2018.01020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/22/2018] [Indexed: 01/15/2023] Open
Abstract
The imbalance between the GABA-mediated inhibition and the glutamate-mediated excitation is the primary pathological mechanism of epilepsy. GABAergic and glutamatergic neurotransmission have become the most important targets for controlling epilepsy. Ginsenoside compound K (GCK) is a main metabolic production of the ginsenoside Rb1, Rb2, and Rc in the intestinal microbiota. Previous studies show that GCK promoted the release of GABA from the hippocampal neurons and enhanced the activity of GABAA receptors. GCK is shown to reduce the expression of NMDAR and to attenuate the function of the NMDA receptors in the brain. The anti-seizure effects of GCK have not been reported so far. Therefore, this study aimed to investigate the effects of GCK on epilepsy and its potential mechanism. The rat model of seizure or status epilepticus (SE) was established with either Pentylenetetrazole or Lithium chloride-pilocarpine. The Racine's scale was used to evaluate seizure activity. The levels of the amino acid neurotransmitters were detected in the pilocarpine-induced epileptic rats. The expression levels of GABAARα1, NMDAR1, KCC2, and NKCC1 protein in the hippocampus were determined via western blot or immunohistochemistry after SE. We found that GCK had deceased seizure intensity and prolonged the latency of seizures. GCK increased the contents of GABA, while the contents of glutamate remained unchanged. GCK enhanced the expression of GABAARα1 in the brain and exhibited a tendency to decrease the expression of NMDAR1 protein in the hippocampus. The expression of KCC2 protein was elevated by the treatment of GCK after SE, while the expression of NKCC1 protein was reversely down-regulated. These findings suggested that GCK exerted anti-epileptic effects by promoting the hippocampal GABA release and enhancing the GABAAR-mediated inhibitory synaptic transmission.
Collapse
Affiliation(s)
- Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Kai Hu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lulu Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Luping Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Wei Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Chaopeng Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Wenjing Zong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Siyu Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Qing Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Guirong Zeng
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Dejian Jiang
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Dong-Sheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| |
Collapse
|
11
|
Zheng M, Xin Y, Li Y, Xu F, Xi X, Guo H, Cui X, Cao H, Zhang X, Han C. Ginsenosides: A Potential Neuroprotective Agent. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8174345. [PMID: 29854792 PMCID: PMC5964429 DOI: 10.1155/2018/8174345] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/06/2018] [Accepted: 04/02/2018] [Indexed: 01/03/2023]
Abstract
Ginseng is a traditional Chinese medicine with a wide range of pharmacological activities. Ginsenosides are the major constituents of ginseng. Ginsenosides have the unique biological activity and medicinal value, such as antitumor, anti-inflammatory, antioxidation, and inhibition of cell apoptosis. With the increase of stress in life, the incidence of nervous system diseases is also increasing. Neurological diseases pose a huge burden on people's life and health. In recent years, some studies have shown that ginsenosides have a certain role in the prevention and treatment of neurological diseases. However, the research is still in its infancy, and the relevant mechanisms are complex. In the paper, we review the effects and mechanisms of ginsenosides on epilepsy, depression, cerebral ischemia reperfusion injury, Alzheimer's disease, and Parkinson's disease. We hope to provide a theoretical basis for the treatment of nervous system diseases by ginsenosides.
Collapse
Affiliation(s)
- Mengmeng Zheng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yizhou Xin
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Yujuan Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Fangxue Xu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiaozhi Xi
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hong Guo
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiaowei Cui
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hui Cao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xi Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chunchao Han
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
12
|
Bjørklund G, Dadar M, Martins N, Chirumbolo S, Goh BH, Smetanina K, Lysiuk R. Brief Challenges on Medicinal Plants: An Eye-Opening Look at Ageing-Related Disorders. Basic Clin Pharmacol Toxicol 2018; 122:539-558. [DOI: 10.1111/bcpt.12972] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine; Mo i Rana Norway
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute; Agricultural Research, Education and Extension Organization (AREEO); Karaj Iran
| | - Natália Martins
- Mountain Research Centre (CIMO), ESA; Polytechnic Institute of Bragança, Campus de Santa Apolónia; Bragança Portugal
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences; University of Verona; Verona Italy
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group (BMEX); School of Pharmacy; Monash University Malaysia; Bandar Sunway Malaysia
- Novel Bacteria and Drug Discovery Research Group (NBDD); School of Pharmacy; Monash University Malaysia; Bandar Sunway Malaysia
- Center of Health Outcomes Research and Therapeutic Safety; School of Pharmaceutical Sciences; University of Phayao; Phayao Thailand
- Asian Centre for Evidence Synthesis in Population; Implementation and Clinical Outcomes; Health and Well-Being Cluster; Global Asia in the 21st Century Platform; Monash University Malaysia; Bandar Sunway Malaysia
| | - Kateryna Smetanina
- Department of Management and Economy of Pharmacy; Postgraduate Faculty; Drug Technology and Pharmacoeconomics; Danylo Halytsky Lviv National Medical University; Lviv Ukraine
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany; Danylo Halytsky Lviv National Medical University; Lviv Ukraine
| |
Collapse
|
13
|
Moon JH, Lee JH, Lee YJ, Park SY. Autophagy flux induced by ginsenoside-Rg3 attenuates human prion protein-mediated neurotoxicity and mitochondrial dysfunction. Oncotarget 2016; 7:85697-85708. [PMID: 27911875 PMCID: PMC5349867 DOI: 10.18632/oncotarget.13730] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/11/2016] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial quality control is a process by which mitochondria undergo successive rounds of fusion and fission with dynamic exchange of components to segregate functional and damaged elements. Removal of mitochondrion that contains damaged components is accomplished via autophagy. In this study, we investigated whether ginsenoside Rg3, an active ingredient of the herbal medicine ginseng that is used as a tonic and restorative agent, could attenuate prion peptide, PrP (106-126)-induced neurotoxicity and mitochondrial damage. To this end, western blot and GFP-LC3B puncta assay were performed to monitor autophagy flux in neuronal cells; LC3B-II protein level was found to increase after Rg3 treatment. In addition, electron microscopy analysis showed that Rg3 enhanced autophagic vacuoles in neuronal cells. By using autophagy inhibitors wortmannin and 3-methyladenine (3MA) or autophagy protein 5 (Atg5) small interfering RNA (siRNA), we demonstrated that Rg3 could protect neurons against PrP (106-126)-induced cytotoxicity via autophagy flux. We found that Rg3 could also attenuate PrP (106-126)-induced mitochondrial damage via autophagy flux. Taken together, our results suggest that Rg3 is a possible therapeutic agent in neurodegenerative disorders, including prion diseases.
Collapse
Affiliation(s)
- Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Ju-Hee Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - You-Jin Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| |
Collapse
|
14
|
Singh D, Goel RK. Anticonvulsant mechanism of saponins fraction from adventitious roots of Ficus religiosa: possible modulation of GABAergic, calcium and sodium channel functions. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2016. [DOI: 10.1016/j.bjp.2015.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Feng L, Yue XF, Chen YX, Liu XM, Wang LS, Cao FR, Wang Q, Liao YH, Pan RL, Chang Q. LC/MS-based metabolomics strategy to assess the amelioration effects of ginseng total saponins on memory deficiency induced by simulated microgravity. J Pharm Biomed Anal 2016; 125:329-38. [DOI: 10.1016/j.jpba.2016.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 01/24/2023]
|
16
|
Yamanaka R, Shindo Y, Karube T, Hotta K, Suzuki K, Oka K. Neural depolarization triggers Mg2+ influx in rat hippocampal neurons. Neuroscience 2015; 310:731-41. [DOI: 10.1016/j.neuroscience.2015.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/26/2015] [Accepted: 10/02/2015] [Indexed: 12/14/2022]
|
17
|
Martinc B, Grabnar I, Vovk T. Antioxidants as a preventive treatment for epileptic process: a review of the current status. Curr Neuropharmacol 2014; 12:527-50. [PMID: 25977679 PMCID: PMC4428026 DOI: 10.2174/1570159x12666140923205715] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/29/2014] [Accepted: 09/22/2014] [Indexed: 11/25/2022] Open
Abstract
Epilepsy is known as one of the most frequent neurological diseases, characterized by an enduring predisposition to generate epileptic seizures. Oxidative stress is believed to directly participate in pathways leading to neurodegeneration, which serves as the most important propagating factor, leading to the epileptic condition and cognitive decline. Moreover, there is also a growing body of evidence showing the disturbance of antioxidant system balance and consequently increased production of reactive species in patients with epilepsy. A meta-analysis, conducted in the present review confirms an association between epilepsy and increased lipid peroxidation. Furthermore, it was also shown that some of the antiepileptic drugs could potentially be responsible for additionally increased lipid peroxidation. Therefore, it is reasonable to propose that during the epileptic process neuroprotective treatment with antioxidants could lead to less sever structural damages, reduced epileptogenesis and milder cognitive deterioration. To evaluate this hypothesis studies investigating the neuroprotective therapeutic potential of various antioxidants in cells, animal seizure models and patients with epilepsy have been reviewed. Numerous beneficial effects of antioxidants on oxidative stress markers and in some cases also neuroprotective effects were observed in animal seizure models. However, despite these encouraging results, till now only a few antioxidants have been further applied to patients with epilepsy as an add-on therapy. Based on the several positive findings in animal models, a strong need for more carefully planned, randomized, double-blind, cross-over, placebo-controlled clinical trials for the evaluation of antioxidants efficacy in patients with epilepsy is warranted.
Collapse
Affiliation(s)
| | | | - Tomaž Vovk
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
18
|
Sun A, Xu X, Lin J, Cui X, Xu R. Neuroprotection by saponins. Phytother Res 2014; 29:187-200. [PMID: 25408503 DOI: 10.1002/ptr.5246] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 08/09/2014] [Accepted: 09/23/2014] [Indexed: 12/17/2022]
Abstract
Saponins, an important group of bioactive plant natural products, are glycosides of triterpenoid or steroidal aglycones. Their diverse biological activities are ascribed to their different structures. Saponins have long been recognized as key ingredients in traditional Chinese medicine. Accumulated evidence suggests that saponins have significant neuroprotective effects on attenuation of central nervous system disorders, such as stroke, Alzheimer's disease, Parkinson's disease, and Huntington's disease. However, our understanding of the mechanisms underlying the observed effects remains incomplete. Based on recently reported data from basic and clinical studies, this review highlights the proposed mechanisms of their neuroprotective function including antioxidant, modulation of neurotransmitters, anti-apoptosis, anti-inflammation, attenuating Ca(2+) influx, modulating neurotrophic factors, inhibiting tau phosphorylation, and regeneration of neural networks.
Collapse
Affiliation(s)
- Aijing Sun
- Institute of Molecular Medicine, Huaqiao University and Engineering Research Center of Molecular Medicine, Ministry of Education, Quanzhou, China
| | | | | | | | | |
Collapse
|
19
|
Histological study on the possible protective action of ginseng on the injurious effect induced by acrylamide on the midbrain in adult male albino rat. ACTA ACUST UNITED AC 2014. [DOI: 10.1097/01.ehx.0000446587.04196.3b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Smith I, Williamson EM, Putnam S, Farrimond J, Whalley BJ. Effects and mechanisms of ginseng and ginsenosides on cognition. Nutr Rev 2014; 72:319-33. [DOI: 10.1111/nure.12099] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Imogen Smith
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | - Elizabeth M Williamson
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | | | | | - Benjamin J Whalley
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| |
Collapse
|
21
|
Nah SY. Ginseng ginsenoside pharmacology in the nervous system: involvement in the regulation of ion channels and receptors. Front Physiol 2014; 5:98. [PMID: 24678300 PMCID: PMC3958645 DOI: 10.3389/fphys.2014.00098] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 02/27/2014] [Indexed: 01/05/2023] Open
Abstract
Ginseng, the root of Panax ginseng C.A. Meyer, is one of the oldest traditional medicines and is thought to be a tonic. It has been claimed that ginseng may improve vitality and health. Recent studies have advanced ginseng pharmacology and shown that ginseng has various pharmacological effects in the nervous system. Ginsenosides, steroid glycosides extracted from ginseng, were one of the first class of biologically active plant glycosides identified. The diverse pharmacological effects of ginsenosides have been investigated through the regulation of various types of ion channels and receptors in neuronal cells and heterologous expression systems. Ginsenoside Rg3 regulates voltage-gated ion channels such as Ca(2+), K(+), and Na(+) channels, and ligand-gated ion channels such as GABAA, 5-HT3, nicotinic acetylcholine, and N-methyl-D-aspartate (NMDA) receptors through interactions with various sites including channel blocker binding sites, toxin-binding sites, channel gating regions, and allosteric channel regulator binding sites when the respective ion channels or receptors are stimulated with depolarization or ligand treatment. Treatment with ginsenoside Rg3 has been found to stabilize excitable cells by blocking influxes of cations such as Ca(2+) and Na(+), or by enhancing Cl(-) influx. The aim of this review is to present recent findings on the pharmacological functions of the ginsenosides through the interactions with ion channels and receptors. This review will detail the pharmacological applications of ginsenosides as neuroprotective drugs that target ion channels and ligand-gated ion channels.
Collapse
Affiliation(s)
- Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University Seoul, South Korea
| |
Collapse
|
22
|
Ginseng extract attenuates early MRI changes after status epilepticus and decreases subsequent reduction of hippocampal volume in the rat brain. Epilepsy Res 2014; 108:223-31. [DOI: 10.1016/j.eplepsyres.2013.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 09/15/2013] [Accepted: 11/21/2013] [Indexed: 11/21/2022]
|
23
|
Peña ID, Yoon SY, Kim HJ, Park S, Hong EY, Ryu JH, Park IH, Cheong JH. Effects of ginseol k-g3, an Rg3-enriched fraction, on scopolamine-induced memory impairment and learning deficit in mice. J Ginseng Res 2013; 38:1-7. [PMID: 24558303 PMCID: PMC3915335 DOI: 10.1016/j.jgr.2013.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/26/2013] [Accepted: 08/28/2013] [Indexed: 12/11/2022] Open
Abstract
Background Although ginsenosides such as Rg1, Rb1 and Rg3 have shown promise as potential nutraceuticals for cognitive impairment, their use has been limited due to high production cost and low potency. In particular, the process of extracting pure Rg3 from ginseng is laborious and expensive. Methods We described the methods in preparing ginseol k-g3, an Rg3-enriched fraction, and evaluated its effects on scopolamine-induced memory impairment in mice. Results Ginseol k-g3 (25–200 mg/kg) significantly reversed scopolamine-induced cognitive impairment in the passive avoidance, but not in Y-maze testing. Ginseol k-g3 (50 and 200 mg/kg) improved escape latency in training trials and increased swimming times within the target zone of the Morris water maze. The effect of ginseol k-g3 on the water maze task was more potent than that of Rg3 or Red ginseng. Acute or subchronic (6 d) treatment of ginseol k-g3 did not alter normal locomotor activity of mice in an open field. Ginseol k-g3 did not inhibit acetylcholinesterase activity, unlike donezepil, an acetylcholinesterase inhibitor. Rg3 enrichment through the ginseol k-g3 fraction enhanced the efficacy of Rg3 in scopolamine-induced memory impairment in mice as demonstrated in the Morris water maze task. Conclusion The effects of ginseol k-g3 in ameliorating scopolamine-induced memory impairment in the passive avoidance and Morris water maze tests indicate its specific influence on reference or long-term memory. The mechanism underlying the reversal of scopolamine-induced amnesia by ginseol k-g3 is not yet known, but is not related to anticholinesterase-like activity.
Collapse
Affiliation(s)
- Ike dela Peña
- Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Korea
| | - Seo Young Yoon
- Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Korea
| | - Sejin Park
- Department of Oriental Pharmaceutical Science, Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Eun Young Hong
- Nutraceuticals and Functional Food Research & Development, Cheil Jedang (CJ) Corporation, Seoul, Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Il Ho Park
- Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Korea
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Korea
- Corresponding author. Uimyung Research Institute for Neuroscience, Sahmyook University, 26-21 Kongreung-2-dong, Hwarangro-815, Nowon-gu, Seoul 139-742, Korea.
| |
Collapse
|
24
|
Kim HJ, Kim P, Shin CY. A comprehensive review of the therapeutic and pharmacological effects of ginseng and ginsenosides in central nervous system. J Ginseng Res 2013; 37:8-29. [PMID: 23717153 PMCID: PMC3659622 DOI: 10.5142/jgr.2013.37.8] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
Ginseng is one of the most widely used herbal medicines in human. Central nervous system (CNS) diseases are most widely investigated diseases among all others in respect to the ginseng’s therapeutic effects. These include Alzheimer’s disease, Parkinson’s disease, cerebral ischemia, depression, and many other neurological disorders including neurodevelopmental disorders. Not only the various types of diseases but also the diverse array of target pathways or molecules ginseng exerts its effect on. These range, for example, from neuroprotection to the regulation of synaptic plasticity and from regulation of neuroinflammatory processes to the regulation of neurotransmitter release, too many to mention. In general, ginseng and even a single compound of ginsenoside produce its effects on multiple sites of action, which make it an ideal candidate to develop multi-target drugs. This is most important in CNS diseases where multiple of etiological and pathological targets working together to regulate the final pathophysiology of diseases. In this review, we tried to provide comprehensive information on the pharmacological and therapeutic effects of ginseng and ginsenosides on neurodegenerative and other neurological diseases. Side by side comparison of the therapeutic effects in various neurological disorders may widen our understanding of the therapeutic potential of ginseng in CNS diseases and the possibility to develop not only symptomatic drugs but also disease modifying reagents based on ginseng.
Collapse
Affiliation(s)
- Hee Jin Kim
- Department of Pharmacology, School of Medicine and Advanced Institute of Biomedical Science and Technology, Konkuk University, Seoul 143-701, Korea
| | | | | |
Collapse
|
25
|
Poly(ADP-ribose) polymerase inhibition protects epileptic hippocampal neurons from apoptosis via suppressing Akt-mediated apoptosis-inducing factor translocation in vitro. Neuroscience 2013. [DOI: 10.1016/j.neuroscience.2012.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
Kim S, Rhim H. Effects of amyloid-β peptides on voltage-gated L-type Ca(V)1.2 and Ca(V)1.3 Ca(2+) channels. Mol Cells 2011; 32:289-94. [PMID: 21822937 PMCID: PMC3887631 DOI: 10.1007/s10059-011-0075-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/18/2011] [Accepted: 06/24/2011] [Indexed: 01/01/2023] Open
Abstract
Overload of intracellular Ca(2+) has been implicated in the pathogenesis of neuronal disorders, such as Alzheimer's disease. Various mechanisms produce abnormalities in intracellular Ca(2+) homeostasis systems. L-type Ca(2+) channels have been known to be closely involved in the mechanisms underlying the neurodegenerative properties of amyloid-β (Aβ) peptides. However, most studies of L-type Ca(2+) channels in Aβ-related mechanisms have been limited to Ca(V)1.2, and surprisingly little is known about the involvement of Ca(V)1.3 in Aβ-induced neuronal toxicity. In the present study, we examined the expression patterns of Ca(V)1.3 after Aβ(25-35) exposure for 24 h and compared them with the expression patterns of Ca(V)1.2. The expression levels of Ca(V)1.3 were not significantly changed by Aβ(25-35) at both the mRNA levels and the total protein level in cultured hippocampal neurons. However, surface protein levels of Ca(V)1.3 were significantly increased by Aβ(25-35), but not by Aβ(35-25). We next found that acute treatment with Aβ(25-35) increased Ca(V)1.3 channel activities in HEK293 cells using whole-cell patch-clamp recordings. Furthermore, using GTP pulldown and co-immunoprecipitation assays in HEK293 cell lysates, we found that amyloid precursor protein interacts with β(3) subunits of Ca(2+) channels instead of Ca(V)1.2 or Ca(V)1.3 α(1) subunits. These results show that Aβ(25-35) chronically or acutely upregulates Ca(V)1.3 in the rat hippocampal and human kidney cells (HEK293). This suggests that Ca(V)1.3 has a potential role along with Ca(V)1.2 in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Sunoh Kim
- Jeollanamdo Institute of Natural Resources Research, Jangheung 529-851, Korea
| | - Hyewhon Rhim
- Biomedical Research Center, Korea Institute of Science and Technology, Seoul 136-791, Korea
| |
Collapse
|
27
|
Shin EJ, Jeong JH, Chung YH, Kim WK, Ko KH, Bach JH, Hong JS, Yoneda Y, Kim HC. Role of oxidative stress in epileptic seizures. Neurochem Int 2011; 59:122-37. [PMID: 21672578 PMCID: PMC3606551 DOI: 10.1016/j.neuint.2011.03.025] [Citation(s) in RCA: 294] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 03/27/2011] [Accepted: 03/28/2011] [Indexed: 11/16/2022]
Abstract
Oxidative stress resulting from excessive free-radical release is likely implicated in the initiation and progression of epilepsy. Therefore, antioxidant therapies aimed at reducing oxidative stress have received considerable attention in epilepsy treatment. However, much evidence suggests that oxidative stress does not always have the same pattern in all seizures models. Thus, this review provides an overview aimed at achieving a better understanding of this issue. We summarize work regarding seizure models (i.e., genetic rat models, kainic acid, pilocarpine, pentylenetetrazol, and trimethyltin), oxidative stress as an etiologic factor in epileptic seizures (i.e., impairment of antioxidant systems, mitochondrial dysfunction, involvement of redox-active metals, arachidonic acid pathway activation, and aging), and antioxidant strategies for seizure treatment. Combined, this review highlights pharmacological mechanisms associated with oxidative stress in epileptic seizures and the potential for neuroprotection in epilepsy that targets oxidative stress and is supported by effective antioxidant treatment.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul 136-705, South Korea
| | - Kwang-Ho Ko
- Pharmacology Laboratory, College of Pharmacy, Seoul National University, Seoul 143-701, South Korea
| | - Jae-Hyung Bach
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Ishikawa 920-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| |
Collapse
|
28
|
Roh YS, Kim HB, Kang CW, Kim BS, Nah SY, Kim JH. Neuroprotective Effects of Ginsenoside Rg3against 24-OH-cholesterol-induced Cytotoxicity in Cortical Neurons. J Ginseng Res 2010. [DOI: 10.5142/jgr.2010.34.3.246] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Ho YS, So KF, Chang RCC. Anti-aging herbal medicine--how and why can they be used in aging-associated neurodegenerative diseases? Ageing Res Rev 2010; 9:354-62. [PMID: 19833234 DOI: 10.1016/j.arr.2009.10.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 10/05/2009] [Accepted: 10/06/2009] [Indexed: 12/30/2022]
Abstract
Aging is a universal biological process that leads to progressive and deleterious changes in organisms. From ancient time, mankind has already interested in preventing and keeping ourselves young. Anti-aging study is certainly not a new research area. Nowadays, the meaning of anti-aging has been changed from simply prolonging lifespan to increasing health span, which emphasizes more on the quality of life. This is the concept of healthy aging and prevention of pathological aging, which is associated with diseases. Keeping our brain functions as in young age is an important task for neuroscientists to prevent aging-associated neurological disorders, such as Alzheimer's diseases (AD) and Parkinson's disease (PD). The causes of these diseases are not fully understood, but it is believed that these diseases are affected by multiple factors. Neurodegenerative diseases can be cross-linked with a number of aging-associated conditions. Based on this, a holistic approach in anti-aging research seems to be more reasonable. Herbal medicine has a long history in Asian countries. It is believed that many of the medicinal herbs have anti-aging properties. Recent studies have shown that some medicinal herbs are effective in intervention or prevention of aging-associated neurological disorders. In this review, we use wolfberry and ginseng as examples to elaborate the properties of anti-aging herbs. The characteristics of medicinal herbs, especially their applications in different disease stages (prevention and intervention) and multi-targets properties, allow them to be potential anti-aging intervention in prevention and treatment of the aging-associated neurological disorders.
Collapse
|
30
|
|
31
|
Peng LL, Shen HM, Jiang ZL, Li X, Wang GH, Zhang YF, Ke KF. Inhibition of NMDA receptors underlies the neuroprotective effect of ginsenoside Rb3. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2009; 37:759-70. [PMID: 19655413 DOI: 10.1142/s0192415x09007223] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In order to investigate the mechanisms underlying the neuroprotective effect of ginsenoside Rb3, rat hippocampal neurons were primarily cultured, and exposed to 1 mM N-methyl-D-aspartate (NMDA), cell viability and lactate dehydrogenase leakage were measured. Ca2+ influx was determined by calcium imaging with a laser confocal microscopy. The influences of ginsenoside Rb3 on these variables were examined. Patch-clamp technique was used to observe the effects of ginsenoside Rb3 on NMDA-evoked current. The results show that treatment of Rb3 raised the neuronal viability, reduced the leakage of lactate dehydrogenase, and inhibited NMDA-elicited Ca2+ influx in a dose-dependent manner. In the presence of Rb3, NMDA-evoked peak current was inhibited, and Ca2+-induced desensitization of NMDA current was facilitated. It is suggested that ginsenoside Rb3 could exert a neuroprotective role on hippocampal neurons, a role which was partly mediated by the facilitation of Ca2+-dependent deactivation of NMDA receptors, and the resultant reduction of intracellular free Ca2+ level.
Collapse
Affiliation(s)
- Liang-Liang Peng
- Department of Neuropharmacology, Institute of Nautical Medicine, Nantong University, Nantong, Jiangsu 226001, China
| | | | | | | | | | | | | |
Collapse
|
32
|
NP04634 prevents cell damage caused by calcium overload and mitochondrial disruption in bovine chromaffin cells. Eur J Pharmacol 2009; 607:47-53. [DOI: 10.1016/j.ejphar.2009.02.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 01/29/2009] [Accepted: 02/09/2009] [Indexed: 11/23/2022]
|
33
|
Nah SY, Kim DH, Rhim H. Ginsenosides: are any of them candidates for drugs acting on the central nervous system? CNS DRUG REVIEWS 2008; 13:381-404. [PMID: 18078425 DOI: 10.1111/j.1527-3458.2007.00023.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The last two decades have shown a marked expansion in the number of publications regarding the effects of Panax ginseng. Ginsenosides, which are unique saponins isolated from Panax ginseng, are the pharmacologically active ingredients in ginseng, responsible for its effects on the central nervous system (CNS) and the peripheral nervous system. Recent studies have shown that ginsenosides regulate various types of ion channels, such as voltage-dependent and ligand-gated ion channels, in neuronal and heterologously expressed cells. Ginsenosides inhibit voltage-dependent Ca(2+), K(+), and Na(+) channel activities in a stereospecific manner. Ginsenosides also inhibit ligand-gated ion channels such as N-methyl-d-aspartate, some subtypes of nicotinic acetylcholine, and 5-hydroxytryptamine type 3 receptors. Competition and site-directed mutagenesis experiments revealed that ginsenosides interact with ligand-binding sites or channel pore sites and inhibit open states of ion channels. This review will introduce recent findings and advances on ginsenoside-induced regulation of ion channel activities in the CNS, and will further expand the possibilities that ginsenosides may be useful and potentially therapeutic choices in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | | | | |
Collapse
|
34
|
Zhang YF, Fan XJ, Li X, Peng LL, Wang GH, Ke KF, Jiang ZL. Ginsenoside Rg1 protects neurons from hypoxic-ischemic injury possibly by inhibiting Ca2+ influx through NMDA receptors and L-type voltage-dependent Ca2+ channels. Eur J Pharmacol 2008; 586:90-9. [PMID: 18430419 DOI: 10.1016/j.ejphar.2007.12.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 12/02/2007] [Accepted: 12/16/2007] [Indexed: 10/22/2022]
Abstract
The purpose of this study is to assess the neuroprotective effect of Rg1, a ginsenoside. We measured cell viability and lactate dehydrogenase (LDH) release from primary culture of rat hippocampal neurons and electrical activities in hippocampal slices of rats, before and after the neurons were deprived of oxygen and glucose. In addition, cerebral damage was evaluated with magnetic resonance imaging after middle cerebral artery was occluded transiently. Nissl staining was used for histological observation and immunohistochemistry analysis for activated caspase-3 expression of the brain. Furthermore, calcium influx was measured with laser confocal microscopy in neurons perfused with KCl (50 mM) or N-methyl-d-aspartate (NMDA, 1 mM), or deprived of oxygen and glucose. The influences of ginsenoside Rg1 on these parameters were determined simultaneously. We found that treatment of Rg1: 1) increased the neuronal viability; 2) promoted the recovery of electrical activity in hippocampal slices; 3) reduced the release of LDH, cerebral damage area, neuronal loss and expression of caspase-3; and 4) inhibited calcium influx induced by NMDA, KCl or oxygen/glucose deprivation. However, the protective effect of Rg1 was blocked by mifepristone, an antagonist of glucocorticoid receptors. Taken together, these results suggest that ginsenoside Rg1 can reduce neuronal death, including apoptotic cell death, induced by hypoxic-ischemic insults. This neuroprotective effect is probably mediated by the activation of glucocorticoid receptors, and by the inhibition of calcium influx through NMDA receptors and L-type voltage-dependent Ca2+ channels and the resultant reduction of intracellular free Ca2+.
Collapse
Affiliation(s)
- Yun-Feng Zhang
- Department of Neuropharmacology, Institute of Nautical Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Ginsenoside Rd attenuates neuroinflammation of dopaminergic cells in culture. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2007:105-12. [PMID: 17982883 DOI: 10.1007/978-3-211-73574-9_13] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In Parkinson's disease clinical and experimental evidence suggest that neuroinflammatory changes in cytokines caused by microglial activation contribute to neuronal death. Experimentally, neuroinflammation of dopaminergic neurons can be evoked by lipopolysaccharide (LPS) exposure. In mesencephalic primary cultures LPS (100 microg/ml) resulted in 30-50% loss of dendritic processes, changes in the perikarya, cellular atrophy and neuronal cell loss of TH-immunoreactive (TH+) cells. iNOS activity was increased dose dependently as well as prostaglandin E2 concentrations. Ginsenosides, as the active compounds responsible for ginseng action, are reported to have antioxidant and anti-inflammatory effects. Here ginsenoside Rd was used to counteract LPS neurodegeneration. Partial reduction of LPS neurotoxic action was seen in dopaminergic neurons. Cell death by LPS as well as neuroprotective action by ginsenoside Rd was not selective for dopaminergic neurons. Neuronal losses as well as cytoprotective effects were similar when counting NeuN identified neurons. The anti-inflammatory effect of ginsenoside Rd could equally be demonstrated by a reduction of NO-formation and PGE2 synthesis. Thus, protective mechanisms of ginsenoside Rd may involve interference with iNOS and COX-2 expression.
Collapse
|
36
|
Kim S, Nah SY, Rhim H. Neuroprotective effects of ginseng saponins against L-type Ca2+ channel-mediated cell death in rat cortical neurons. Biochem Biophys Res Commun 2007; 365:399-405. [PMID: 18023426 DOI: 10.1016/j.bbrc.2007.10.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2007] [Accepted: 10/02/2007] [Indexed: 11/17/2022]
Abstract
In the present study, we have examined any possible involvement of L-type Ca(2+) channels in ginseng-mediated neuroprotective actions. Exposure to a 50mM KCl (high-K) produced neuronal cell death, which was blocked by a selective L-type Ca(2+) channel blocker in cultured cortical neurons. When cultured cells were co-treated with ginseng total saponin (GTS) and high-K, GTS reduced high-K-induced neuronal death. Using Ca(2+) imaging techniques, we found that GTS inhibited high-K-mediated acute and long-term [Ca(2+)](i) changes. These GTS-mediated [Ca(2+)](i) changes were diminished by nifedipine. Furthermore, GTS-mediated effects were also diminished by a saturating concentration of Bay K (10muM). After confirming the protective effect of GTS using a TUNEL assay, we found that ginsenosides Rf and Rg(3) are active components in ginseng-mediated neuroprotection. These results suggest that inhibition of L-type Ca(2+) channels by ginseng could be one of the mechanisms for ginseng-mediated neuroprotection in cultured rat cortical neurons.
Collapse
Affiliation(s)
- Sunoh Kim
- Life Sciences Division, Korea Institute of Science and Technology, 39-1 Hawholgok-dong Sungbuk-gu, Seoul 136-791, Republic of Korea
| | | | | |
Collapse
|
37
|
The Inhibition of Epileptogenesis During Status Epilepticus by Ginsenosides of Korean Red Ginseng and Ginseng Cell Culture (Dan25). J Ginseng Res 2007. [DOI: 10.5142/jgr.2007.31.3.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Kim S, Yun HM, Baik JH, Chung KC, Nah SY, Rhim H. Functional interaction of neuronal Cav1.3 L-type calcium channel with ryanodine receptor type 2 in the rat hippocampus. J Biol Chem 2007; 282:32877-89. [PMID: 17823125 DOI: 10.1074/jbc.m701418200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal L-type Ca(2+) channels do not support synaptic transmission, but they play an essential role in synaptic activity-dependent gene expression. Ca(v)1.2 and Ca(v)1.3 are the two most widely expressed L-type Ca(2+) channels in neurons and have different biophysical and subcellular distributions. The function of the Ca(v) 1.3 L-type Ca(2+) channel and its cellular mechanisms in the central nervous system are poorly understood. In this study, using a yeast two-hybrid assay, we found that the N terminus of the rat Ca(v)1.3 alpha(1) subunit interacts with a partial N-terminal amino acid sequence of ryanodine receptor type 2 (RyR2). Reverse transcription-PCR and Western blot assays revealed high expression of both Ca(v)1.3 and RyR2 in the rat hippocampus. We also demonstrate a physical association of Ca(v)1.3 with RyR2 using co-immunoprecipitation assays. Moreover, immunocytochemistry revealed prominent co-localization between Ca(v)1.3 and RyR2 in hippocampal neurons. Depolarizing cells by an acute treatment of a high concentration of KCl (high-K, 60 mm) showed that the activation of L-type Ca(2+) channels induced RyR opening and led to RyR-dependent Ca(2+) release, even in the absence of extracellular Ca(2+). Furthermore, we found that RyR2 mRNA itself is increased by long term treatment of high-K via activation of L-type Ca(2+) channels. These acute and long term effects of high-K on RyRs were selectively blocked by small interfering RNA-mediated silencing of Ca(v)1.3. These results suggest a physical and functional interaction between Ca(v)1.3 and RyR2 and important implications of Ca(v)1.3/RyR2 clusters in translating synaptic activity into alterations in gene expression.
Collapse
Affiliation(s)
- Sunoh Kim
- Life Sciences Division, Korea Institute of Science and Technology, 39-1 Hawholgok-dong, Sungbuk-gu, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
39
|
Kim JH, Cho SY, Lee JH, Jeong SM, Yoon IS, Lee BH, Lee JH, Pyo MK, Lee SM, Chung JM, Kim S, Rhim H, Oh JW, Nah SY. Neuroprotective effects of ginsenoside Rg3 against homocysteine-induced excitotoxicity in rat hippocampus. Brain Res 2006; 1136:190-9. [PMID: 17239831 DOI: 10.1016/j.brainres.2006.12.047] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Revised: 12/04/2006] [Accepted: 12/05/2006] [Indexed: 10/23/2022]
Abstract
We previously demonstrated that ginsenoside Rg(3) (Rg(3)), one of the active ingredients in Panax ginseng, attenuates NMDA receptor-mediated currents and NMDA-induced neurotoxicity (Kim, S., Kim, T., Ahn, K., Park, W.K., Nah, S.Y., Rhim, H., 2004. Ginsenoside Rg(3) antagonizes NMDA receptors through a glycine modulatory site in rat cultured hippocampal neurons. Biochem. Biophys. Res. Commun. 323, 416-424). Accumulating evidence suggests that homocysteine (HC), a metabolite of methionine, exerts its excitotoxicity through NMDA receptor activation. In the present study, we examined the neuroprotective effects of Rg(3) on HC-induced hippocampal excitotoxicity in vitro and in vivo. Our in vitro studies using rat cultured hippocampal neurons revealed that Rg(3) treatment significantly and dose-dependently inhibited HC-induced hippocampal cell death, with an EC(50) value of 28.7+/-7.5 muM. Rg(3) treatment not only significantly reduced HC-induced DNA damage, but also dose-dependently attenuated HC-induced caspase-3 activity in vitro. Our in vivo studies revealed that intracerebroventricular (i.c.v.) pre-administration of Rg(3) significantly and dose-dependently reduced i.c.v. HC-induced hippocampal damage in rats. To examine the mechanisms underlying the in vitro and in vivo neuroprotective effects of Rg(3) against HC-induced hippocampal excitotoxicity, we examined the effect of Rg(3) on HC-induced intracellular Ca(2+) elevations in cultured hippocampal cells and found that Rg(3) treatment dose-dependently inhibited HC-induced intracellular Ca(2+) elevation, with an IC(50) value of 41.5+/-17.5 muM. In addition, Rg(3) treatment dose-dependently inhibited HC-induced currents in Xenopus oocytes expressing the NMDA receptor, with an IC(50) of 47.3+/-14.2 muM. These results collectively indicate that Rg(3)-induced neuroprotection against HC in rat hippocampus might be achieved via inhibition of HC-mediated NMDA receptor activation.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Jeonju, Korea 561-756
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Radad K, Gille G, Liu L, Rausch WD. Use of ginseng in medicine with emphasis on neurodegenerative disorders. J Pharmacol Sci 2006; 100:175-86. [PMID: 16518078 DOI: 10.1254/jphs.crj05010x] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Ginseng, the root of Panax species, is a well-known herbal medicine. It has been used as a traditional medicine in China, Korea, and Japan for thousands of years and is now a popular and worldwide used natural medicine. The active ingredients of ginseng are ginsenosides which are also called ginseng saponins. Recently, there is increasing evidence in the literature on the pharmacological and physiological actions of ginseng. However, ginseng has been used primarily as a tonic to invigorate weak bodies and help the restoration of homeostasis. Current in vivo and in vitro studies have shown its beneficial effects in a wide range of pathological conditions such as cardiovascular diseases, cancer, immune deficiency, and hepatotoxicity. Moreover, recent research has suggested that some of ginseng's active ingredients also exert beneficial effects on aging, central nervous system (CNS) disorders, and neurodegenerative diseases. In general, antioxidant, anti-inflammatory, anti-apoptotic, and immune-stimulatory activities are mostly underlying the possible ginseng-mediated protective mechanisms. Next to animal studies, data from neural cell cultures contribute to the understanding of these mechanisms that involve decreasing nitric oxide (NO), scavenging of free radicals, and counteracting excitotoxicity. In this review, we focus on recently reported medicinal effects of ginseng and summarize the current knowledge of its effects on CNS disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Khaled Radad
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Egypt.
| | | | | | | |
Collapse
|
41
|
Blair RE, Deshpande LS, Sombati S, Falenski KW, Martin BR, DeLorenzo RJ. Activation of the cannabinoid type-1 receptor mediates the anticonvulsant properties of cannabinoids in the hippocampal neuronal culture models of acquired epilepsy and status epilepticus. J Pharmacol Exp Ther 2006; 317:1072-8. [PMID: 16469864 DOI: 10.1124/jpet.105.100354] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cannabinoids have been shown to have anticonvulsant properties, but no studies have evaluated the effects of cannabinoids in the hippocampal neuronal culture models of acquired epilepsy (AE) and status epilepticus (SE). This study investigated the anticonvulsant properties of the cannabinoid receptor agonist R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrrolol[1,2,3 de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone (WIN 55,212-2) in primary hippocampal neuronal culture models of both AE and SE. WIN 55,212-2 produced dose-dependent anticonvulsant effects against both spontaneous recurrent epileptiform discharges (SRED) (EC50 = 0.85 microM) and SE (EC50 = 1.51 microM), with total suppression of seizure activity at 3 microM and of SE activity at 5 microM. The anticonvulsant properties of WIN 55,212-2 in these preparations were both stereospecific and blocked by the cannabinoid type-1 (CB1) receptor antagonist N-(piperidin-1-yl-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride (SR141716A; 1 microM), showing a CB1 receptor-dependent pathway. The inhibitory effect of WIN 55,212-2 against low Mg2+-induced SE is the first observation in this model of total suppression of SE by a selective pharmacological agent. The clinically used anticonvulsants phenytoin and phenobarbital were not able to abolish low Mg2+-induced SE at concentrations up to 150 microM. The results from this study show CB1 receptor-mediated anticonvulsant effects of the cannabimimetic WIN 55,212-2 against both SRED and low Mg2+-induced SE in primary hippocampal neuronal cultures and show that these in vitro models of AE and SE may represent powerful tools to investigate the molecular mechanisms mediating the effects of cannabinoids on neuronal excitability.
Collapse
Affiliation(s)
- Robert E Blair
- Department of Neurology, Virginia Commonwealth University, School of Medicine, P.O. Box 980599, Richmond, VA 23298, USA
| | | | | | | | | | | |
Collapse
|