1
|
Li Z, Zhang Z, Yu B. Correction to "Unlocking the Therapeutic Potential of Natural Products for Alzheimer's Disease". J Med Chem 2025; 68:9018-9024. [PMID: 40214661 DOI: 10.1021/acs.jmedchem.5c00926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
|
2
|
Quan Q, Ma X, Feng J, Li W, Li X. Ginsenoside Rg1 improves autophagy dysfunction to ameliorate Alzheimer's disease via targeting FGR proto-oncogene. Neuropeptides 2025; 111:102514. [PMID: 40073763 DOI: 10.1016/j.npep.2025.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
Alzheimer's disease (AD) is a neurodegeneration driven by beta-amyloid (Aβ) deposits in the brain involving autophagy dysfunction. Ginsenoside Rg1, a pharmacologically active compound found in ginseng, has possible therapeutic effects for AD. This study discovered that FGR proto-oncogene (FGR) was a therapeutic target of Rg1 in AD and it was possibly involved in autophagy. C57BL/6 J mice were injected with 5 μL (1 μg/mL) Aβ1-42 in the right lateral ventricle to establish an AD model. AD mouse hippocampus had high FGR expression. Intragastrically administered Rg1 (40 mg/kg) decreased FGR protein levels in AD mice's hippocampus and improved memory function in AD mice. Both sides of the mice hippocampal fissure were administered with 2 μL lentiviral particles (1 × 107 TU) containing FGR overexpression plasmids. FGR overexpression rendered Rg1 ineffectual in restoring memory function and reducing hippocampal neuron damage. We injected 2 μL lentiviral particles (1 × 107 TU) containing short hairpin RNA plasmids targeting FGR to the mice hippocampal fissures. FGR knockdown improved spatial memory function of AD mice, reduced hippocampal neuron apoptosis, and prevented Aβ accumulation. HT22 cells were transfected with small interfering RNA targeting FGR. FGR knockdown increased the viability of Aβ1-42 treated HT22 cells. BACE1 and LC3II/I protein levels were decreased and p62 and SIRT1 were increased in AD mice and cells with FGR knockdown. LC3 was down-regulated after inhibiting FGR expression in Aβ1-42 treated hippocampal neurons. In conclusion, Rg1 exerts anti-AD functions by targeting FGR and downregulating its expression.
Collapse
Affiliation(s)
- Qiankun Quan
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinxin Ma
- Department of Psychology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - JianJun Feng
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanni Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
Li Z, Zhang Z, Yu B. Unlocking the Therapeutic Potential of Natural Products for Alzheimer's Disease. J Med Chem 2025; 68:2377-2402. [PMID: 39865664 DOI: 10.1021/acs.jmedchem.4c03049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by memory loss and cognitive decline. With current treatments offering limited effectiveness, researchers are turning to natural products that can target various aspects of AD pathology. Clinically approved natural products, such as galantamine and huperzine A, have shown success in AD treatments. Furthermore, compounds such as epigallocatechin gallate, quercetin, and resveratrol are in clinical trials. This Perspective examines nearly 100 natural compounds with promising neuroprotective effects in preclinical and clinical studies. These compounds exhibit diverse pharmacological actions that help to prevent neurodegeneration while improving cognitive functions. Their unique structures further enhance their biological activities, making them promising candidates for drug discovery. This Perspective stresses the importance of further clinical research to maximize the medical benefits of these compounds and highlights their potential as innovative remedies for AD. Continued exploration of these compounds is crucial to fully leverage their capabilities in combating AD.
Collapse
Affiliation(s)
- Zhonghua Li
- Academy of Chinese Medical Sciences, Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Bin Yu
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450000, China
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
4
|
Jiang M, Chi J, Qiao Y, Wang J, Zhang Z, Liu J, Sheng X, Yuan L. Ginsenosides Rg1, Rb1 and rare ginsenosides: Promising candidate agents for Parkinson's disease and Alzheimer's disease and network pharmacology analysis. Pharmacol Res 2025; 212:107578. [PMID: 39756554 DOI: 10.1016/j.phrs.2025.107578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Ginseng has been commonly used as a traditional Chinese medicine in Asian countries for thousands of years. Ginsenosides are the main pharmacologically active ingredients isolated from ginseng and have neuroprotective effects in the treatment of neurodegenerative disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To summarise and investigate the protective roles of ginsenosides and their underlying mechanisms in PD and AD, we used ''Ginsenoside", ''Parkinson's disease", ''Alzheimer's disease", ''anti-inflammatory", ''antioxidant", and ''apoptosis" as keywords to search and extract relevant literature information from scientific databases such as Elsevier, PubMed, and Google Scholar databases. In particular, we used network pharmacology to identify the potential targets of ginsenosides Rg1 and Rb1 in PD and AD. By analysing the existing research advances and network pharmacology results, we found that the neuroprotective effects of ginsenosides, primarily mediated through anti-inflammation, anti-apoptosis and anti-oxidative stress, etc, may be associated with the PI3K/Akt, BDNF/TrkB, MAPKs, NF-κB, Nrf2 and Wnt/β-catenin signalling pathways. This review systematically summarises the different roles and mechanisms of ginsenosides Rg1, Rb1, and rare ginsenosides in PD and AD and provides new strategies for the treatment of neurodegenerative disorders. Network pharmacology provides a new research paradigm for the treatment of PD and AD using Rg1 and Rb1.
Collapse
Affiliation(s)
- Mingchun Jiang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Jiaxin Chi
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Yifan Qiao
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jinpeng Wang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Zhixin Zhang
- School of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jia Liu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xinhao Sheng
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Liangjie Yuan
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.
| |
Collapse
|
5
|
Xing XX, Wu JJ, Qu J, Ma J, Xu R, Zhu Y, Zheng MX, Hua XY, Xu JG. Rewiring the disordered connectome with circuit-based paired stimulation after stroke-a randomized, double-blind and controlled Phase II trial. Brain Commun 2024; 6:fcae437. [PMID: 39697832 PMCID: PMC11653076 DOI: 10.1093/braincomms/fcae437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 07/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
The cortico-cortical paired associative stimulation, a combined stimulation based on two brain regions, may be an effective strategy for stroke rehabilitation. Our aim was to confirm that the cortico-cortical paired associative stimulation strengthens the connection between brain regions in the motor circuit and promotes improvements in motor function. This was a randomized double-blind, controlled Phase II trial. 44 Stroke patients were treated in a rehabilitation hospital from October 2020 to January 2021 and were randomly assigned to the sham stimulation group and the cortico-cortical paired associative stimulation group. Patients in both groups received 12 days of rehabilitation therapy. Cortico-cortical paired associative stimulation group received one treatment of cortico-cortical paired associative stimulation invention. Both groups received behavioural assessments such as the Fugl-Meyer upper-extremity scale and resting-state functional MRI scans prior to the intervention and on Day 14. 40 patients completed the intervention session. The results of Fugl-Meyer upper-extremity scale showed a more significant improvement in motor function in the cortico-cortical paired associative stimulation group (6.33 ± 1.29) than in the sham stimulation group (3.16 ± 1.38) (P < 0.001). The functional connectivity showed that cortico-cortical paired associative stimulation strengthens connections between brain regions. Correlation analysis confirmed that the enhancement of functional connectivity was positively correlated with the recovery of Fugl-Meyer upper-extremity scale (r2 = 0.146, P = 0.034; r2 = 0.211, P = 0.0093). The results of functional connectivity suggest that cortico-cortical paired associative stimulation strengthens connections between brain regions. It is expected that this study will provide a positive viewpoint for the neurorehabilitation of stroke patients based on the circuit-level plasticity. (Chinese Clinical Trial Registry: ChiCTR2000036685).
Collapse
Affiliation(s)
- Xiang-Xin Xing
- Rehabilitation Center, Qilu Hospital of Shandong University, Qilu Hospital of Shandong University, Jinan 250012, China
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Jia Wu
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiao Qu
- Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Jie Ma
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong Xu
- YangZhi Rehabilitation Hospital, TongJi University, Shanghai 201600, China
| | - Yu Zhu
- Department of Physical Medicine and Rehabilitation, State University of New York Upstate Medical University, Syracuse 13290, USA
| | - Mou-Xiong Zheng
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopedics, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xu-Yun Hua
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopedics, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian-Guang Xu
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
Han D, Zhao Z, Mao T, Gao M, Yang X, Gao Y. Ginsenoside Rg1: A Neuroprotective Natural Dammarane-Type Triterpenoid Saponin With Anti-Depressive Properties. CNS Neurosci Ther 2024; 30:e70150. [PMID: 39639753 PMCID: PMC11621566 DOI: 10.1111/cns.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Depression, a widespread mental disorder, presents significant risks to both physical and mental health due to its high rates of recurrence and suicide. Currently, single-target antidepressants typically alleviate depressive symptoms or delay the progression of depression rather than cure it. Ginsenoside Rg1 is one of the main ginsenosides found in Panax ginseng roots. It improves depressive symptoms through various mechanisms, suggesting its potential as a treatment for depression. MATERIALS AND METHODS We evaluated preclinical studies to comprehensively discuss the antidepressant mechanism of ginsenoside Rg1 and review its toxicity and medicinal value. Additionally, pharmacological network and molecular docking analyses were performed to further validate the antidepressant effects of ginsenoside Rg1. RESULTS The antidepressant mechanism of ginsenoside Rg1 may involve various pharmacological mechanisms and pathways, such as inhibiting neuroinflammation and over-activation of microglia, preserving nerve synapse structure, promoting neurogenesis, regulating monoamine neurotransmitter levels, inhibiting hyperfunction of the hypothalamic-pituitary-adrenal axis, and combatting antioxidative stress. Moreover, ginsenoside Rg1 preserves astrocyte gap junction function by regulating connexin43 protein biosynthesis and degradation, contributing to its antidepressant effect. Pharmacological network and molecular docking studies identified five targets (AKT1, STAT3, EGFR, PPARG, and HSP90AA1) as potential molecular regulatory sites of ginsenoside Rg1. CONCLUSIONS Ginsenoside Rg1 may exert its antidepressant effects via various pharmacological mechanisms. In addition, multicenter clinical case-control and molecular targeted studies are required to confirm both the clinical efficacy of ginsenoside Rg1 and its potential direct targets.
Collapse
Affiliation(s)
- Dong Han
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zheng Zhao
- Department of Emergency MedicineShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Tinghui Mao
- Department of Organ Transplantation and Hepatobiliary SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningChina
| | - Man Gao
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xue Yang
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yan Gao
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
7
|
Elawad MA, Ayaz M, Mosa OF, Usman A, Hamdoon AAE, Almawash S, Salim LHM, Ahmed A, Elkhalifa MEM. Polyphenols and Their Biogenic Nano-Formulations Targeting BACE1 as Anti-Amyloid Therapies; Meeting the Challenges of Bioavailability, Safety, and Specificity for the Treatment of Alzheimer's Disease. Mol Nutr Food Res 2024; 68:e2400525. [PMID: 39628325 DOI: 10.1002/mnfr.202400525] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/06/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), a progressiveneurodegenerative condition is marked by extensive damage in the brain and dementia. Among the pathological hallmarks of AD is beta-amyloid (Aβ). Production of toxic Aβ oligomers production and accumulation in the brain is among the characteristic features of the disease. The abnormal accumulation Aβ is initiated by the catalytic degradation of Amyloid Precursor Proteins (APP) by Beta Amyloid Cleaving Enzyme 1 (BACE1) to generate insoluble amyloid plaques. The abnormal proteins are mitochondrial poison which disrupt the energy production and liberate excessive free radicals causing neuronal damage and mutations. Consequently, targeting Aβ-associated pathways has become a focus in the pursuit of developing effective AD treatments. An obstacle faced by many medications used to treat neurodegenerative diseases (NDs) is the restricted permeability across the blood-brain barrier (BBB). Unfortunately, no anti-amyloid drug is clinically approved till now. Recent advancements in nanotechnology have provided a possible solution for delivering medications to specific targets. By integrating natural products with nano-medicinal approaches, it is possible to develop novel and highly efficient therapeutic strategies for the treatment of AD.
Collapse
Affiliation(s)
- Mohammed Ahmed Elawad
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, 18800 Dir (L), Chakdara, KP, Pakistan
| | - Osama F Mosa
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| | - Assad Usman
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, 18800 Dir (L), Chakdara, KP, Pakistan
| | - Alashary Adam Eisa Hamdoon
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| | - Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra, 11911, Kingdom of Saudi Arabia
| | - Liga Hasan Mohammed Salim
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| | - Alshebli Ahmed
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| | - Modawy Elnour Modawy Elkhalifa
- Public health Department Health Sciences College at Lieth, Umm Al Qura University, Makkah, 21955, Kingdom of Saudi Arabia
| |
Collapse
|
8
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [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: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
9
|
Liu S, Wang M, Xiao H, Ye J, Cao L, Li W, Sun G. Advancements in research on the effects of panax notoginseng saponin constituents in ameliorating learning and memory disorders. Heliyon 2024; 10:e28581. [PMID: 38586351 PMCID: PMC10998096 DOI: 10.1016/j.heliyon.2024.e28581] [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: 09/01/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Learning and memory disorder is a cluster of symptoms caused by neuronal aging and other diseases of the central nervous system (CNS). Panax notoginseng saponins (PNS) are a series of saponins derived from the natural active ingredients of traditional Chinese medicine (TCM) that have neuroprotective effects on the central nervous system. In this paper, we review the ameliorative effects and mechanisms of Panax notoginseng saponin-like components on learning and memory disorders to provide valuable references and insights for the development of new drugs for the treatment of learning and memory disorders. Our summary results suggest that Panax ginseng saponins have significant effects on improving learning and memory disorders, and these effects and potential mechanisms are mediated by their anti-inflammatory, anti-apoptotic, antioxidant, β-amyloid lowering, mitochondrial homeostasis in vivo, neuronal structure and function improving, neurogenesis promoting, neurotransmitter release regulating, and probiotic homeostasis in vivo activities. These findings suggest the potential of Panax notoginseng saponin-like constituents as drug candidates for improving learning and memory disorders.
Collapse
Affiliation(s)
- Shusen Liu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Haiyan Xiao
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Jingxue Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Li Cao
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Wenlan Li
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
10
|
Castillo-Ordoñez WO, Cajas-Salazar N, Velasco-Reyes MA. Genetic and epigenetic targets of natural dietary compounds as anti-Alzheimer's agents. Neural Regen Res 2024; 19:846-854. [PMID: 37843220 PMCID: PMC10664119 DOI: 10.4103/1673-5374.382232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/15/2023] [Accepted: 07/18/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder and the most common cause of dementia that principally affects older adults. Pathogenic factors, such as oxidative stress, an increase in acetylcholinesterase activity, mitochondrial dysfunction, genotoxicity, and neuroinflammation are present in this syndrome, which leads to neurodegeneration. Neurodegenerative pathologies such as Alzheimer's disease are considered late-onset diseases caused by the complex combination of genetic, epigenetic, and environmental factors. There are two main types of Alzheimer's disease, known as familial Alzheimer's disease (onset < 65 years) and late-onset or sporadic Alzheimer's disease (onset ≥ 65 years). Patients with familial Alzheimer's disease inherit the disease due to rare mutations on the amyloid precursor protein (APP), presenilin 1 and 2 (PSEN1 and PSEN2) genes in an autosomal-dominantly fashion with closely 100% penetrance. In contrast, a different picture seems to emerge for sporadic Alzheimer's disease, which exhibits numerous non-Mendelian anomalies suggesting an epigenetic component in its etiology. Importantly, the fundamental pathophysiological mechanisms driving Alzheimer's disease are interfaced with epigenetic dysregulation. However, the dynamic nature of epigenetics seems to open up new avenues and hope in regenerative neurogenesis to improve brain repair in Alzheimer's disease or following injury or stroke in humans. In recent years, there has been an increase in interest in using natural products for the treatment of neurodegenerative illnesses such as Alzheimer's disease. Through epigenetic mechanisms, such as DNA methylation, non-coding RNAs, histone modification, and chromatin conformation regulation, natural compounds appear to exert neuroprotective effects. While we do not purport to cover every in this work, we do attempt to illustrate how various phytochemical compounds regulate the epigenetic effects of a few Alzheimer's disease-related genes.
Collapse
Affiliation(s)
- Willian Orlando Castillo-Ordoñez
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
- Departamento de Estudios Psicológicos, Universidad Icesi, Cali, Colombia
| | - Nohelia Cajas-Salazar
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
| | - Mayra Alejandra Velasco-Reyes
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
| |
Collapse
|
11
|
Jiang YY, Wei RY, Tang K, Wang Z, Tan NH. Ginsenoside Rg1 promotes neurite growth of retinal ganglion cells through cAMP/PKA/CREB pathways. J Ginseng Res 2024; 48:163-170. [PMID: 38465221 PMCID: PMC10920000 DOI: 10.1016/j.jgr.2022.05.002] [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: 02/23/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background Mechanisms of synaptic plasticity in retinal ganglion cells (RGCs) are complex and the current knowledge cannot explain. Growth and regeneration of dendrites together with synaptic formation are the most important parameters for evaluating the cellular protective effects of various molecules. The effect of ginsenoside Rg1 (Rg1) on the growth of retinal ganglion cell processes has been poorly understood. Therefore, we investigated the effect of ginsenoside Rg1 on the neurite growth of RGCs. Methods Expression of proteins and mRNA were detected by Western blot and qPCR. cAMP levels were determined by ELISA. In vivo effects of Rg1 on RGCs were evaluated by hematoxylin and eosin, and immunohistochemistry staining. Results This study found that Rg1 promoted the growth and synaptic plasticity of RGCs neurite by activating the cAMP/PKA/CREB pathways. Meanwhile, Rg1 upregulated the expression of GAP43, Rac1 and PAX6, which are closely related to the growth of neurons. Meantime, H89, an antagonist of PKA, could block this effect of Rg1. In addition, we preliminarily explored the effect of Rg1 on enhancing the glycolysis of RGCs, which could be one of the mechanisms for its neuroprotective effects. Conclusion Rg1 promoted neurite growth of RGCs through cAMP/PKA/CREB pathways. This study may lay a foundation for its clinical use of optic nerve diseases in the future.
Collapse
Affiliation(s)
| | | | - Kai Tang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhen Wang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ning-hua Tan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
12
|
Li L, Wang L, Zhang L. Therapeutic Potential of Natural Compounds from Herbs and Nutraceuticals in Alleviating Neurological Disorders: Targeting the Wnt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2411-2433. [PMID: 38284360 DOI: 10.1021/acs.jafc.3c07536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
As an important signaling pathway in multicellular eukaryotes, the Wnt signaling pathway participates in a variety of physiological processes. Recent studies have confirmed that the Wnt signaling pathway plays an important role in neurological disorders such as stroke, Alzheimer's disease, and Parkinson's disease. The regulation of Wnt signaling by natural compounds in herbal medicines and nutraceuticals has emerged as a potential strategy for the development of new drugs for neurological disorders. Purpose: The aim of this review is to evaluate the latest research results on the efficacy of natural compounds derived from herbs and nutraceuticals in the prevention and treatment of neurological disorders by regulating the Wnt pathway in vivo and in vitro. A manual and electronic search was performed for English articles available from PubMed, Web of Science, and ScienceDirect from the January 2010 to February 2023. Keywords used for the search engines were "natural products,″ "plant derived products,″ "Wnt+ clinical trials,″ and "Wnt+,″ and/or paired with "natural products″/″plant derived products", and "neurological disorders." A total of 22 articles were enrolled in this review, and a variety of natural compounds from herbal medicine and nutritional foods have been shown to exert therapeutic effects on neurological disorders through the Wnt pathway, including curcumin, resveratrol, and querctrin, etc. These natural products possess antioxidant, anti-inflammatory, and angiogenic properties, confer neurovascular unit and blood-brain barrier integrity protection, and affect neural stem cell differentiation, synaptic formation, and neurogenesis, to play a therapeutic role in neurological disorders. In various in vivo and in vitro studies and clinical trials, these natural compounds have been shown to be safe and tolerable with few adverse effects. Natural compounds may serve a therapeutic role in neurological disorders by regulating the Wnt pathway. This summary of the research progress of natural compounds targeting the Wnt pathway may provide new insights for the treatment of neurological disorders and potential targets for the development of new drugs.
Collapse
Affiliation(s)
- Lei Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning PR China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning PR China
| | - Lijuan Zhang
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning PR China
| |
Collapse
|
13
|
Wang X, Li H, Sheng Y, He B, Liu Z, Li W, Yu S, Wang J, Zhang Y, Chen J, Qin L, Meng X. The function of sphingolipids in different pathogenesis of Alzheimer's disease: A comprehensive review. Biomed Pharmacother 2024; 171:116071. [PMID: 38183741 DOI: 10.1016/j.biopha.2023.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Sphingolipids (SPLs) represent a highly diverse and structurally complex lipid class. The discussion of SPL metabolism-related issues is of importance in understanding the neuropathological progression of Alzheimer's disease (AD). AD is characterized by the accumulation of extracellular deposits of the amyloid β-peptide (Aβ) and intraneuronal aggregates of the microtubule-associated protein tau. Critical roles of Aβ oligomer deposited and ganglioside GM1 could be formed as "seed" from insoluble GAβ polymer in initiating the pathogenic process, while tau might also mediate SPLs and their toxicity. The interaction between ceramide and α-Synuclein (α-Syn) accelerates the aggregation of ferroptosis and exacerbates the pathogenesis of AD. For instance, reducing the levels of SPLs can mitigate α-Syn accumulation and inhibit AD progression. Meanwhile, loss of SPLs may inhibit the expression of APOE4 and confer protection against AD, while the loss of APOE4 expression also disrupts SPLs homeostasis. Moreover, the heightened activation of sphingomyelinase promotes the ferroptosis signaling pathway, leading to exacerbated AD symptoms. Ferroptosis plays a vital role in the pathological progression of AD by influencing Aβ, tau, APOE, and α-Syn. Conversely, the development of AD also exacerbates the manifestation of ferroptosis and SPLs. We are compiling the emerging techniques (Derivatization and IM-MS) of sphingolipidomics, to overcome the challenges of AD diagnosis and treatment. In this review, we examined the intricate neuro-mechanistic interactions between SPLs and Aβ, tau, α-Syn, APOE, and ferroptosis, mediating the onset of AD. Furthermore, our findings highlight the potential of targeting SPLs as underexplored avenue for devising innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Huaqiang Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Zeying Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Wanli Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Shujie Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jiajing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian 350122, PR China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| |
Collapse
|
14
|
Wu JJ, Zhang L, Liu D, Xia J, Yang Y, Tang F, Chen L, Ao H, Peng C. Ginsenoside Rg1, lights up the way for the potential prevention of Alzheimer's disease due to its therapeutic effects on the drug-controllable risk factors of Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116955. [PMID: 37536646 DOI: 10.1016/j.jep.2023.116955] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In traditional Chinese medicine, Shen Nong, BenCao Jing, and Compendium of Materia Medica (Bencao Gangmu), Panax ginseng, and its prescriptions have been used for the treatment of dementia, depression, weight loss, Xiaoke disease (similar to diabetes), and vertigo. All these diseases are associated with the drug-controllable risk factors for Alzheimer's disease (AD), including depression, obesity, diabetes, and hypertension. Ginsenoside Rg1, one of the main active ingredients of P. ginseng and its congener Panax notoginseng, possesses therapeutic potentials against AD and associated diseases. This suggests that ginsenoside Rg1 might have the potential for AD prevention and treatment. Although the anti-AD effects of ginsenoside Rg1 have received more attention, a systematic review of its effects on depression, obesity, diabetes, and hypertension is not available. AIM OF THE REVIEW This systematic literature review comprehensively summarized existing literature on the therapeutic potentials of ginsenoside Rg1 in AD prevention for the propose of providing a foundation of future research aimed at enabling the use of such drugs in clinical practice. METHODS Information on ginsenoside Rg1 was collected from relevant published articles identified through a literature search in electronic scientific databases (PubMed, Science Direct, and Google Scholar). The keywords used were "Ginsenoside Rg1," "Panax ginseng," "Source," "Alzheimer's disease," "Brain disorders," "Depression," "Obesity," "Diabetes," and "Hypertension." RESULTS The monomer ginsenoside Rg1 can be relatively easily obtained and has therapeutic potentials against AD. In vitro and in vivo experiments have demonstrated the therapeutic potentials of ginsenoside Rg1 against the drug-controllable risk factors of AD including depression, obesity, diabetes, and hypertension. Thus, ginsenoside Rg1 alleviates diseases resulting from AD risk factors by regulating multiple targets and pathways. CONCLUSIONS Ginsenoside Rg1 has the potentials to prevent AD by alleviating depression, obesity, diabetes, and hypertension.
Collapse
Affiliation(s)
- Jiao-Jiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jia Xia
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
15
|
Liu G, Yang C, Wang X, Chen X, Wang Y, Le W. Oxygen metabolism abnormality and Alzheimer's disease: An update. Redox Biol 2023; 68:102955. [PMID: 37956598 PMCID: PMC10665957 DOI: 10.1016/j.redox.2023.102955] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/13/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Oxygen metabolism abnormality plays a crucial role in the pathogenesis of Alzheimer's disease (AD) via several mechanisms, including hypoxia, oxidative stress, and mitochondrial dysfunction. Hypoxia condition usually results from living in a high-altitude habitat, cardiovascular and cerebrovascular diseases, and chronic obstructive sleep apnea. Chronic hypoxia has been identified as a significant risk factor for AD, showing an aggravation of various pathological components of AD, such as amyloid β-protein (Aβ) metabolism, tau phosphorylation, mitochondrial dysfunction, and neuroinflammation. It is known that hypoxia and excessive hyperoxia can both result in oxidative stress and mitochondrial dysfunction. Oxidative stress and mitochondrial dysfunction can increase Aβ and tau phosphorylation, and Aβ and tau proteins can lead to redox imbalance, thus forming a vicious cycle and exacerbating AD pathology. Hyperbaric oxygen therapy (HBOT) is a non-invasive intervention known for its capacity to significantly enhance cerebral oxygenation levels, which can significantly attenuate Aβ aggregation, tau phosphorylation, and neuroinflammation. However, further investigation is imperative to determine the optimal oxygen pressure, duration of exposure, and frequency of HBOT sessions. In this review, we explore the prospects of oxygen metabolism in AD, with the aim of enhancing our understanding of the underlying molecular mechanisms in AD. Current research aimed at attenuating abnormalities in oxygen metabolism holds promise for providing novel therapeutic approaches for AD.
Collapse
Affiliation(s)
- Guangdong Liu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Cui Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xin Wang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yanjiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
| |
Collapse
|
16
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
17
|
Kim JH, Kim JM, Lee HL, Go MJ, Kim TY, Joo SG, Lee HS, Heo HJ. Korean Red Ginseng Prevents the Deterioration of Lung and Brain Function in Chronic PM 2.5-Exposed Mice by Regulating Systemic Inflammation. Int J Mol Sci 2023; 24:13266. [PMID: 37686071 PMCID: PMC10488300 DOI: 10.3390/ijms241713266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
This study was conducted to confirm the effects of Korean red ginseng on lung and brain dysfunction in a BALB/c mice model exposed to particulate matter (PM)2.5 for 12 weeks. Learning and cognitive abilities were assessed with Y-maze, passive avoidance, and Morris water maze tests. To evaluate the ameliorating effect of red ginseng extract (RGE), the antioxidant system and mitochondrial function were investigated. The administration of RGE protected lung and brain impairment by regulating the antioxidant system and mitochondrial functions damaged by PM2.5-induced toxicity. Moreover, RGE prevented pulmonary fibrosis by regulating the transforming growth factor beta 1 (TGF-β1) pathway. RGE attenuated PM2.5-induced pulmonary and cognitive dysfunction by regulating systemic inflammation and apoptosis via the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/c-Jun N-terminal kinases (JNK) pathway. In conclusion, RGE might be a potential material that can regulate chronic PM2.5-induced lung and brain cognitive dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.H.K.); (J.M.K.); (H.L.L.); (M.J.G.); (T.Y.K.); (S.G.J.); (H.S.L.)
| |
Collapse
|
18
|
Zhang R, Zeng M, Zhang X, Zheng Y, Lv N, Wang L, Gan J, Li Y, Jiang X, Yang L. Therapeutic Candidates for Alzheimer's Disease: Saponins. Int J Mol Sci 2023; 24:10505. [PMID: 37445682 DOI: 10.3390/ijms241310505] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Drug development for Alzheimer's disease, the leading cause of dementia, has been a long-standing challenge. Saponins, which are steroid or triterpenoid glycosides with various pharmacological activities, have displayed therapeutic potential in treating Alzheimer's disease. In a comprehensive review of the literature from May 2007 to May 2023, we identified 63 references involving 40 different types of saponins that have been studied for their effects on Alzheimer's disease. These studies suggest that saponins have the potential to ameliorate Alzheimer's disease by reducing amyloid beta peptide deposition, inhibiting tau phosphorylation, modulating oxidative stress, reducing inflammation, and antiapoptosis. Most intriguingly, ginsenoside Rg1 and pseudoginsenoside-F11 possess these important pharmacological properties and show the best promise for the treatment of Alzheimer's disease. This review provides a summary and classification of common saponins that have been studied for their therapeutic potential in Alzheimer's disease, showcasing their underlying mechanisms. This highlights the promising potential of saponins for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yujia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Nuan Lv
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Luming Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yawen Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
19
|
Nakayama H, Ihara D, Fukuchi M, Toume K, Yuri C, Tsuda M, Shibahara N, Tabuchi A. The extract based on the Kampo formula daikenchuto (Da Jian Zhong Tang) induces Bdnf expression and has neurotrophic effects in cultured cortical neurons. J Nat Med 2023; 77:584-595. [PMID: 37148454 DOI: 10.1007/s11418-023-01703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
Reductions in brain-derived neurotrophic factor (BDNF) expression levels have been reported in the brains of patients with neurological disorders such as Alzheimer's disease. Therefore, upregulating BDNF and preventing its decline in the diseased brain could help ameliorate neurological dysfunctions. Accordingly, we sought to discover agents that increase Bdnf expression in neurons. Here, we screened a library of 42 Kampo extracts to identify those with the ability to induce Bdnf expression in cultured cortical neurons. Among the active extracts identified in the screen, we focused on the extract based on the Kampo formula daikenchuto. The extract of daikenchuto in the library used in this study was prepared using the mixture of Zingiberis Rhizoma Processum (ZIN), Zanthoxyli Piperiti Pericarpium (ZAN), and Ginseng Radix (GIN) without Koi. In this study, we defined DKT as the mixture of ZIN, ZAN, and GIN without Koi (DKT extract means the extract prepared from the mixture of ZIN, ZAN, and GIN without Koi). DKT extract significantly increased endogenous Bdnf expression by mediated, at least in part, via Ca2+ signaling involving L-type voltage-dependent Ca2+ channels in cultured cortical neurons. Furthermore, DKT extract significantly improved the survival of cultured cortical neurons and increased neurite complexity in immature neurons. Taken together, our findings suggest that DKT extract induces Bdnf expression and has a neurotrophic effect in neurons. Because BDNF inducers are expected to have therapeutic potential for neurological disorders, re-positioning of Kampo formulations such as daikenchuto may lead to clinical application in diseases associated with reduced BDNF in the brain.
Collapse
Affiliation(s)
- Hironori Nakayama
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Mamoru Fukuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan.
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan.
| | - Kazufumi Toume
- Department of Medicinal Resources Management, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chisato Yuri
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Masaaki Tsuda
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Naotoshi Shibahara
- Kampo Education and Training Center, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
20
|
Zhou J, Zhang J, Cao L, Liu Y, Liu L, Liu C, Li X. Ginsenoside Rg 1 modulates vesicular dopamine storage and release during exocytosis revealed with single-vesicle electrochemistry. Chem Commun (Camb) 2023; 59:3087-3090. [PMID: 36804575 DOI: 10.1039/d2cc06950d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Ginsenoside Rg1, a tetracyclic triterpenoid derivative extracted from the roots of Panax ginseng C. A. Meyer, can enhance learning and memory and improve cognitive impairment. However, whether or how it affects vesicular dopamine storage and its release during exocytosis remains unknown. By using single-vesicle electrochemistry, we for the first time find out that Rg1 not only upregulates vesicular dopamine content but also increases exocytosis frequency and modulates dopamine release during exocytosis in PC12 cells, which may relate to the activation of protein kinases, causing a series of biological cascades. This finding offers the possible link between Rg1 and vesicular chemical storage and exocytotic release, which is of significance for understanding the nootropic role of Rg1 from the perspective of neurotransmission.
Collapse
Affiliation(s)
- Junlan Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Jing Zhang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Lijiao Cao
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Yuying Liu
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China.,State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Luyao Liu
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Chunlan Liu
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Xianchan Li
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China. .,Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China.,State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
21
|
Jiang N, Lv J, Zhang Y, Sun X, Yao C, Wang Q, He Q, Liu X. Protective effects of ginsenosides Rg1 and Rb1 against cognitive impairment induced by simulated microgravity in rats. Front Pharmacol 2023; 14:1167398. [PMID: 37168997 PMCID: PMC10164943 DOI: 10.3389/fphar.2023.1167398] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Microgravity experienced during space flight is known to exert several negative effects on the learning ability and memory of astronauts. Few effective strategies are currently available to counteract these effects. Rg1 and Rb1, the major steroidal components of ginseng, have shown potent neuroprotective effects with a high safety profile. The present study aimed to investigate the effects of Rg1 and Rb1 on simulated microgravity-induced learning and memory dysfunction and its underlying mechanism in the hindlimb suspension (HLS) rat model. Administration of Rg1 (30 and 60 μmol/kg) and Rb1 (30 and 60 μmol/kg) for 2 weeks resulted in a significant amelioration of impaired spatial and associative learning and memory caused by 4-week HLS exposure, measured using the Morris water maze and Reward operating conditioning reflex (ROCR) tests, respectively. Furthermore, Rg1 and Rb1 administration alleviated reactive oxygen species production and enhanced antioxidant enzyme activities in the prefrontal cortex (PFC). Rg1 and Rb1 also assisted in the recovery of mitochondrial complex I (NADH dehydrogenase) activities, increased the expression of Mfn2 and decreased the fission marker dynamin-related protein (Drp)-1expression. Additionally, Rg1 and Rb1 treatment increased the SYN, and PSD95 protein expressions and decreased the ratio of Bax:Bcl-2 and reduced the expression of cleaved caspase-3 and cytochrome C. Besides these, the BDNF-TrkB/PI3K-Akt pathway was also activated by Rg1 and Rb1 treatment. Altogether, Rg1 and Rb1 treatment attenuated cognitive deficits induced by HLS, mitigated mitochondrial dysfunction, attenuated oxidative stress, inhibited apoptosis, increased synaptic plasticity, and restored BDNF-TrkB/PI3K-Akt signaling.
Collapse
Affiliation(s)
- Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingwei Lv
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinran Sun
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caihong Yao
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Wang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinghu He
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinmin Liu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xinmin Liu,
| |
Collapse
|
22
|
Wu Z, Huang J, Bai X, Wang Q, Wang F, Xu J, Tang H, Yin C, Wang Y, Yu F, Zhang H. Ginsenoside-Rg1 mitigates cardiac arrest-induced cognitive damage by modulating neuroinflammation and hippocampal plasticity. Eur J Pharmacol 2022; 938:175431. [PMID: 36463944 DOI: 10.1016/j.ejphar.2022.175431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Ginsenoside-Rg1 can effectively ameliorate mental disorders, but whether ginsenoside-Rg1 plays a neuroprotective role in cardiac arrest and cardiopulmonary resuscitation (CA/CPR)-induced cognitive impairment remains unclear. In this study, a 5-min asphyxia-based CA/CPR rat model was established to explore the mechanisms underlying the effects of ginsenoside-Rg1 (40 mg·kg-1·d-1, ip, 14 days) on its cognitive alterations. These CA/CPR rats displayed spatial learning and memory impairment in the Morris water maze, as reflected in the compromised basal synaptic transmission and long-term potentiation (LTP) at the Schaffer collateral of hippocampal CA1 area in vivo electrophysiology, whereas the ginsenoside-Rg1 remarkably mitigated these alterations. Next, we found that ginsenoside-Rg1 inhibited hippocampal neuroinflammation by alleviating the CA/CPR-induced hippocampal activation of microglia and astrocytes and the overexpression of related proinflammatory cytokines interleukin-1β (IL-1β) and tumour necrosis factor-α (TNF-α). In addition, ginsenoside-Rg1 improved CA/CPR-induced hippocampal neuronal apoptosis, dendritic spines and synaptic ultrastructure defects as associated with the upregulation of the key synaptic regulatory proteins. Furthermore, ginsenoside-Rg1 could ameliorate CA/CPR-induced aberrant expression of the key regulators of hippocampal glutamate signaling pathways, excitatory amino acid transporter 2 (EAAT2), excitatory amino acid transporter 1 (EAAT1), Glutamine Synthetase (GS), GluN2B, and glutamate. In conclusion, ginsenoside-Rg1 exerts its neuroprotective effects by ameliorating hippocampus-dependent neuroglia activation-mediated neuroinflammation and neuroplasticity deficits, shedding new light on the therapeutic intervention of CA/CPR-related cognitive disorders.
Collapse
Affiliation(s)
- Zhangbi Wu
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jialin Huang
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiaojie Bai
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qunan Wang
- School of Public Health, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230032, China
| | - Fen Wang
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jun Xu
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Huiping Tang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China; Auditory Research Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Chunying Yin
- Cryo-EM Center, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Wang
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Feng Yu
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Hong Zhang
- Department of Emergency Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
23
|
Feng H, Xue M, Deng H, Cheng S, Hu Y, Zhou C. Ginsenoside and Its Therapeutic Potential for Cognitive Impairment. Biomolecules 2022; 12:1310. [PMID: 36139149 PMCID: PMC9496100 DOI: 10.3390/biom12091310] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment (CI) is one of the major clinical features of many neurodegenerative diseases. It can be aging-related or even appear in non-central nerve system (CNS) diseases. CI has a wide spectrum that ranges from the cognitive complaint with normal screening tests to mild CI and, at its end, dementia. Ginsenosides, agents extracted from a key Chinese herbal medicine (ginseng), show great promise as a new therapeutic option for treating CI. This review covered both clinical trials and preclinical studies to summarize the possible mechanisms of how ginsenosides affect CI in different diseases. It shows that ginsenosides can modulate signaling pathways associated with oxidative stress, apoptosis, inflammation, synaptic plasticity, and neurogenesis. The involved signaling pathways mainly include the PI3K/Akt, CREB/BDNF, Keap1/Nrf2 signaling, and NF-κB/NLRP3 inflammasome pathways. We hope to provide a theoretical basis for the treatment of CI for related diseases by ginsenosides.
Collapse
Affiliation(s)
- Hui Feng
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Mei Xue
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Hao Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300073, China
| | - Shiqi Cheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330008, China
| | - Yue Hu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| |
Collapse
|
24
|
New insights into the role and mechanisms of ginsenoside Rg1 in the management of Alzheimer's disease. Biomed Pharmacother 2022; 152:113207. [PMID: 35667236 DOI: 10.1016/j.biopha.2022.113207] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder in the elderly characterized by memory loss and cognitive dysfunction. The pathogenesis of AD is complex. One-targeted anti-AD drugs usually fail to delay AD progression. Traditional Chinese medicine records have documented the use of the roots of Panax ginseng (ginseng roots) and its prescriptions to treat dementia. Ginsenoside Rg1, the main ginsenoside component of ginseng roots, exhibits a certain therapeutic effect in the abovementioned diseases, suggesting its potential in the management of AD. Therefore, we combed the pathogenesis of AD and currently used anti-AD drugs, and reviewed the availability, pharmacokinetics, and pharmaceutic studies of ginsenoside Rg1. This review summarizes the therapeutic effects and mechanisms of ginsenoside Rg1 and its deglycosylated derivatives in AD in vivo and in vitro. The main mechanisms include improvement in Aβ and Tau pathologies, regulation of synaptic function and intestinal microflora, and reduction of inflammation, oxidative stress, and apoptosis. The underlying mechanisms mainly involve the regulation of PKC, MAPK, PI3K/Akt, CDK5, GSK-3β, BDNF/TrkB, PKA/CREB, FGF2/Akt, p21WAF1/CIP1, NF-κB, NLRP1, TLR3, and TLR4 signaling pathways. As the effects and underlying mechanisms of ginsenoside Rg1 on AD have not been systematically reviewed, we have provided a comprehensive review and shed light on the future directions in the utilization of ginsenoside Rg1 and ginseng roots as well as the development of anti-AD drugs.
Collapse
|
25
|
Shi LS, Ji CH, Liu Y, Gu JH, Tang WQ, Zhang W, Guan W. Ginsenoside Rh2 administration produces crucial antidepressant-like effects in a CUMS-induced mice model of depression. Brain Behav 2022; 12:e2705. [PMID: 35848938 PMCID: PMC9392527 DOI: 10.1002/brb3.2705] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION The most striking feature of depression is sadness and a loss of interest in activities, which represents a major cause of disability globally. Therefore, it is necessary to identify novel antidepressants for clinical practice. Ginsenoside Rh2 (Rh2) is one of the major bioactive ginsenosides that can be extracted from Panax ginseng and has been demonstrated to improve both memory and learning. The purpose of this study was to provide broad insight into the mechanisms underlying depression and gain greater insights into antidepressant therapy. METHODS In this study, we first established an effective and feasible depression animal model of chronic unpredictable mild stress (CUMS) and behavioral testing was evaluated by the forced swim test (FST), the tail suspension test (TST) and the sucrose preference test. Following pretreatment with Rh2 (10 and 20 mg/kg), the immobility time of mice was reduced without affecting locomotor activity in both the FST and TST. Western blotting and immunofluorescence were used to investigate the activation of the hippocampal BDNF signaling pathway and hippocampal neurogenesis. RESULTS Different concentrations of Rh2 significantly reduced depressive-like symptoms in CUMS-induced mice and downregulated the effects of the BDNF signaling cascade and neurogenesis in the hippocampus. Furthermore, the administration of K252a completely prevented the antidepressant-like activity of Rh2 in mice. CONCLUSION The results indicated that Rh2 possesses the antidepression action via the positive regulation of the BDNF-TrkB pathway.
Collapse
Affiliation(s)
- Lin-Sheng Shi
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Chun-Hui Ji
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Yue Liu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Jiang-Hong Gu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wen-Qian Tang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wei Zhang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,Provincial key laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
26
|
Current Progress on Neuroprotection Induced by Artemisia, Ginseng, Astragalus, and Ginkgo Traditional Chinese Medicines for the Therapy of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3777021. [PMID: 35746960 PMCID: PMC9213169 DOI: 10.1155/2022/3777021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
Aging is associated with the occurrence of diverse degenerative changes in various tissues and organs and with an increased incidence of neurological disorders, especially neurodegenerative diseases such as Alzheimer's disease (AD). In recent years, the search for effective components derived from medicinal plants in delaying aging and preventing and treating neurodegenerative diseases has been increasing and the number of related publications shows a rising trend. Here, we present a concise, updated review on the preclinical and clinical research progress in the assessment of the therapeutic potential of different traditional Chinese medicines and derived active ingredients and their effect on the signaling pathways involved in AD neuroprotection. Recognized by their multitargeting ability, these natural compounds hold great potential in developing novel drugs for AD.
Collapse
|
27
|
Yang Y, Wang L, Zhang C, Guo Y, Li J, Wu C, Jiao J, Zheng H. Ginsenoside Rg1 improves Alzheimer's disease by regulating oxidative stress, apoptosis, and neuroinflammation through Wnt/GSK-3β/β-catenin signaling pathway. Chem Biol Drug Des 2022; 99:884-896. [PMID: 35313087 DOI: 10.1111/cbdd.14041] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/11/2022] [Accepted: 03/05/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that can cause cognitive impairment. Ginsenoside Rg1 (Rg1) has a significant neuroprotective effect on animals with memory impairment. However, the mechanism of how Rg1 mediates the Wnt signaling pathway and improves cognitive function by regulating oxidative stress, apoptosis, and neuroinflammation is still unclear. In this study, the spatial memory ability of tree shrews was tested by Morris water maze, the expression levels of amyloid protein (Aβ1-42), ionized calcium-binding adapter molecule 1 (iba-1), nitrotyrosine (NT), and 8-hydroxyguanine (8-OHG) were detected by immunohistochemistry. Subsequently, the activity of catalase (CAT) and the glutathione peroxidase (GSH-Px) was, respectively, measured by the ammonium molybdate method and the 5,5'-dithiobis (2-nitrobenzoic acid). Furthermore, the malondialdehyde (MDA) concentration was determined by the thiobarbituric acid test. Finally, the expression levels of Beta-secretase (BACE1), superoxide dismutase (SOD), BCL2-Associated X (Bax), B-cell lymphoma-2 (Bcl-2), caspase-anti-apoptotic factor Cleaved-caspase-3 (Caspase-3), microtubule-associated proteins 2 (MAP2), Neuronal nuclear antigen (NeuN), as well as the phosphorylation of GSK-3β and β-catenin were detected by Western blot. This study implied that Rg1 reduced the phosphorylation of Tau protein, the deposition of Aβ1-42, and the expression of BACE1. It also showed that Rg1 increased the antioxidant activity of SOD, CAT, GPx, and instead reduced the oxidation products of NT, 8-OHG, and MDA, as wells as the inflammatory factor interleukin-1 and iba-1. It further showed that Rg1 increased the ratio of Bcl-2 to Bax and expression of neuronal markers MAP2 and NeuN, but instead reduced the expression of Caspase-3, GSK-3β, and β-catenin. In conclusion, by regulating the Wnt/GSK-3β/β-catenin signaling pathway, Rg1 of moderate and high dose could alleviate oxidative stress damage, improve neuroinflammation, protect neurons, finally improve the cognitive impairment of the AD tree shrew. This study provides theoretical basis for the Rg1 clinical application in AD.
Collapse
Affiliation(s)
- Yi Yang
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| | - Limei Wang
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
| | - Caijun Zhang
- Experiment Center of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yuqian Guo
- Affiliated Hospital of Medical Sergeant School, Army Medical University, Shijiazhuang, China
| | - Jintao Li
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| | - Chao Wu
- Department of Pharmacy, Hefei Ion Medical Center, Hefei, China
| | - Jianlin Jiao
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Hong Zheng
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| |
Collapse
|
28
|
Lu J, Wang X, Wu A, Cao Y, Dai X, Liang Y, Li X. Ginsenosides in central nervous system diseases: Pharmacological actions, mechanisms, and therapeutics. Phytother Res 2022; 36:1523-1544. [PMID: 35084783 DOI: 10.1002/ptr.7395] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/11/2022]
Abstract
The nervous system is one of the most complex physiological systems, and central nervous system diseases (CNSDs) are serious diseases that affect human health. Ginseng (Panax L.), the root of Panax species, are famous Chinese herbs that have been used for various diseases in China, Japan, and Korea since ancient times, and remain a popular natural medicine used worldwide in modern times. Ginsenosides are the main active components of ginseng, and increasing evidence has demonstrated that ginsenosides can prevent CNSDs, including neurodegenerative diseases, memory and cognitive impairment, cerebral ischemia injury, depression, brain glioma, multiple sclerosis, which has been confirmed in numerous studies. Therefore, this review summarizes the potential pathways by which ginsenosides affect the pathogenesis of CNSDs mainly including antioxidant effects, anti-inflammatory effects, anti-apoptotic effects, and nerve protection, which provides novel ideas for the treatment of CNSDs.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anxin Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Youdan Liang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
29
|
Zarneshan SN, Fakhri S, Khan H. Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: A mechanistic approach. Pharmacol Res 2022; 177:106099. [DOI: 10.1016/j.phrs.2022.106099] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2022] [Accepted: 01/23/2022] [Indexed: 12/15/2022]
|
30
|
Chen X, Drew J, Berney W, Lei W. Neuroprotective Natural Products for Alzheimer's Disease. Cells 2021; 10:1309. [PMID: 34070275 PMCID: PMC8225186 DOI: 10.3390/cells10061309] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the number one neurovegetative disease, but its treatment options are relatively few and ineffective. In efforts to discover new strategies for AD therapy, natural products have aroused interest in the research community and in the pharmaceutical industry for their neuroprotective activity, targeting different pathological mechanisms associated with AD. A wide variety of natural products from different origins have been evaluated preclinically and clinically for their neuroprotective mechanisms in preventing and attenuating the multifactorial pathologies of AD. This review mainly focuses on the possible neuroprotective mechanisms from natural products that may be beneficial in AD treatment and the natural product mixtures or extracts from different sources that have demonstrated neuroprotective activity in preclinical and/or clinical studies. It is believed that natural product mixtures or extracts containing multiple bioactive compounds that can work additively or synergistically to exhibit multiple neuroprotective mechanisms might be an effective approach in AD drug discovery.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Joshua Drew
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Wren Berney
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Wei Lei
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Presbyterian College, Clinton, SC 29325, USA
| |
Collapse
|
31
|
Liang HY, Zhang PP, Zhang XL, Zheng YY, Huang YR, Zheng GQ, Lin Y. Preclinical systematic review of ginsenoside Rg1 for cognitive impairment in Alzheimer's disease. Aging (Albany NY) 2021; 13:7549-7569. [PMID: 33686024 PMCID: PMC7993717 DOI: 10.18632/aging.202619] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/01/2020] [Indexed: 05/02/2023]
Abstract
Ginseng has been used for the treatment of aging and memory impairment for thousands of years. Several studies have found that ginsenoside Rg1, as one of the main active components of ginseng, could potentially improve cognitive function in several different animal models. A preclinical systematic review to evaluate the efficacy and mechanisms of ginsenoside Rg1 for ameliorating cognitive impairments in Alzheimer's disease is reported here. We searched six databases from their inceptions to January 2019. Thirty-two studies were selected, which included a total of 1,643 animals. According to various cognitive behavioral tests, the results of the meta-analyses showed that ginsenoside Rg1 significantly improved cognitive behavioral impairments in most Alzheimer's disease models (P < 0.05), but there were no significant effects in animals with neuronal degeneration induced by chronic stress or in SAMP8 transgenic mice. The potential mechanisms included antioxidant and anti-inflammatory effects, amelioration of Alzheimer's disease-related pathology, synapse protection, and up-regulation of nerve cells via multiple signaling pathways.
Collapse
Affiliation(s)
- Hai-Yong Liang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Pei-Pei Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xi-Le Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yan-Yan Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yan-Ran Huang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yan Lin
- Department of Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
32
|
Natural Products Targeting Amyloid Beta in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22052341. [PMID: 33652858 PMCID: PMC7956407 DOI: 10.3390/ijms22052341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by severe brain damage and dementia. There are currently few therapeutics to treat this disease, and they can only temporarily alleviate some of the symptoms. The pathogenesis of AD is mainly preceded by accumulation of abnormal amyloid beta (Aβ) aggregates, which are toxic to neurons. Therefore, modulation of the formation of these abnormal aggregates is strongly suggested as the most effective approach to treat AD. In particular, numerous studies on natural products associated with AD, aiming to downregulate Aβ peptides and suppress the formation of abnormal Aβ aggregates, thus reducing neural cell death, are being conducted. Generation of Aβ peptides can be prevented by targeting the secretases involved in Aβ-peptide formation (secretase-dependent). Additionally, blocking the intra- and intermolecular interactions of Aβ peptides can induce conformational changes in abnormal Aβ aggregates, whereby the toxicity can be ameliorated (structure-dependent). In this review, AD-associated natural products which can reduce the accumulation of Aβ peptides via secretase- or structure-dependent pathways, and the current clinical trial states of these products are discussed.
Collapse
|
33
|
Wang L, Lu J, Zeng Y, Guo Y, Wu C, Zhao H, Zheng H, Jiao J. Improving Alzheimer's disease by altering gut microbiota in tree shrews with ginsenoside Rg1. FEMS Microbiol Lett 2021; 367:5708937. [PMID: 31950993 DOI: 10.1093/femsle/fnaa011] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
Ginsenoside Rg1 (GRg1) has neuroprotective effects on Alzheimer's disease (AD). The occurrence and progression of AD are closely related to gut microbiota. Few studies have learned the direct relationship between GRg1 and gut microbiota. In this study, we found an original way to research this relationship by using GRg1 in the AD model of tree shrews. Morris water maze and immunohistochemistry were performed to test the cognition repairing function of GRg1 by tree shrews and 16S ribosomal RNA sequencing was used to explore the composition and abundance of gut microbiota. After GRg1 treatment, the result of Morris water maze showed an improvement in cognitive function, and immunohistochemistry revealed a decrease in tau protein. Moreover, 16SrRNA sequencing results showed the abundances of Proteobacteria and Verrucomicrobia were significantly different, and Lactobacillaceae was significantly increased in the GRg1 treatment group. It also showed that the gut microbiome with middle and high doses of GRg1 was close to the normal group. In conclusion, this study suggests that GRg1 at middle and high doses may change the abundance of gut microbiota to improve AD, and thatProteobacteria and Verrucomicrobia are key microbiota. This is the first report that has ever studied the relationship between GRg1 and gut microbiota in tree shrews.
Collapse
Affiliation(s)
- Limei Wang
- Deparment of Laboratory Animal Science, Kunming Medical University, Kunming, Yunnan, China
| | - Jiangli Lu
- Deparment of Laboratory Animal Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yueqin Zeng
- Institute of Molecular Clinical Medicine, Kunming Medical Univercity, Kunming, Yunnan, China
| | - Yuqian Guo
- Deparment of Laboratory Animal Science, Kunming Medical University, Kunming, Yunnan, China
| | - Chao Wu
- Deparment of Laboratory Animal Science, Kunming Medical University, Kunming, Yunnan, China
| | - Hongbin Zhao
- Department of Emergency Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Zheng
- Deparment of Laboratory Animal Science, Kunming Medical University, Kunming, Yunnan, China
| | - Jianlin Jiao
- Technology Transfer Center, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
34
|
Lv J, Lu C, Jiang N, Wang H, Huang H, Chen Y, Li Y, Liu X. Protective effect of ginsenoside Rh2 on scopolamine-induced memory deficits through regulation of cholinergic transmission, oxidative stress and the ERK-CREB-BDNF signaling pathway. Phytother Res 2020; 35:337-345. [PMID: 32754961 DOI: 10.1002/ptr.6804] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/04/2020] [Accepted: 06/24/2020] [Indexed: 12/25/2022]
Abstract
Rh2 is a rare ginsenoside and there are few reports of its effect on cognition compared with other similar molecules. This study aimed to establish the impact of Rh2 treatment on improving scopolamine (Scop)-induced memory deficits in mice and illuminate the underlying mechanisms. First, memory-related behavior was evaluated using two approaches: object location recognition (OLR), based on spontaneous activity, and a Morris water maze (MWM) task, based on an aversive stimulus. Our results suggested that Rh2 treatment effectively increased the discrimination index of the mice in the OLR test. In addition, Rh2 elevated the crossing numbers and decreased the escape latency during the MWM task. Moreover, Rh2 markedly upregulated the phosphorylation of the extracellular signal-regulated kinase (ERK)-cAMP response element binding (CREB)-brain derived neurotrophic factor (BDNF) pathway in the hippocampus. Meanwhile, the administration of Rh2 significantly promoted the cholinergic system and dramatically suppressed oxidative stress in the hippocampus. Taken together, Rh2 exhibited neuroprotective effects against Scop-induced memory dysfunction in mice. Rh2 activity might be ascribed to several underlying mechanisms, including its effects on modulating the cholinergic transmission, inhibiting oxidative stress and activating the ERK-CREB-BDNF signaling pathway. Consequently, the ginsenoside Rh2 might serve as a promising candidate compound for Alzheimer's disease.
Collapse
Affiliation(s)
- Jingwei Lv
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Lu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Ning Jiang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haixia Wang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Huang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujiao Li
- Affiliated (T.C.M.) Hospital, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| | - Xinmin Liu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Affiliated (T.C.M.) Hospital, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| |
Collapse
|
35
|
Yang Y, Li S, Huang H, Lv J, Chen S, Pires Dias AC, Li Y, Liu X, Wang Q. Comparison of the Protective Effects of Ginsenosides Rb1 and Rg1 on Improving Cognitive Deficits in SAMP8 Mice Based on Anti-Neuroinflammation Mechanism. Front Pharmacol 2020; 11:834. [PMID: 32587516 PMCID: PMC7298198 DOI: 10.3389/fphar.2020.00834] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/20/2020] [Indexed: 12/31/2022] Open
Abstract
This present study was designed to investigate the different effects of ginsenosides Rb1 and Rg1 on improving cognitive deficits in 4-month-old SAMP8 mice. Mice were divided into six groups, including the SAMP8 group, the SAMP8 + Donepezil (1.6 mg/kg) group, the SAMP8 + Rb1 (30 and 60 µmol/kg), and SAMP8 + Rg1 (30 and 60 µmol/kg) groups. SAMR1 mice of the same age were used as the control group. Ginsenosides and donepezil were administrated orally to animals for 8 weeks, then the learning and memory ability of mice were measured by using Morris water maze (MWM) test, object recognition test and passive avoidance experiments. The possible mechanisms were studied including the anti-glial inflammation of Rb1 and Rg1 using HE staining, immunohistochemistry and western blot experiments. Results revealed that Rb1 and Rg1 treatment significantly improved the discrimination index of SAMP8 mice in the object recognition test. Rb1 (60 µmol/kg) and Rg1 (30, 60 µmol/kg) could significantly shorten the escape latency in the acquisition test of the MWM test in SAMP8 mice. Furthermore, Rb1 and Rg1 treatments effectively reduced the number of errors in the passive avoidance task in SAMP8 mice. Western blot experiments revealed that Rb1 showed higher effect than Rg1 in decreasing protein expression levels of ASC, caspase-1 and Aβ in the hippocampus of SAMP8 mice, while Rg1 was more effective than Rb1 in decreasing the protein levels of iNOS. In addition, although Rb1 and Rg1 treatments showed significant protective effects in repairing neuronal cells loss and inhibiting the activation of astrocyte and microglia in hippocampus of SAMP8 mice, Rb1 was more effective than Rg1. These results suggest that Rb1 and Rg1 could improve the cognitive impairment in SAMP8 mice, and they have different mechanisms for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yujie Yang
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| | - Shanshan Li
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| | - Hong Huang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingwei Lv
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shanguang Chen
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Alberto Carlos Pires Dias
- Centre of Molecular and Environmental Biology (CBMA), SINO-PT Research Center, Department of Biology, University of Minho, Braga, Portugal
| | - Yujiao Li
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| | - Xinmin Liu
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China.,Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Wang
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China.,Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.,National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
36
|
Abstract
Alzheimer's disease is a chronic neurodegenerative devastating disorder affecting a high percentage of the population over 65 years of age and causing a relevant emotional, social, and economic burden. Clinically, it is characterized by a prominent cognitive deficit associated with language and behavioral impairments. The molecular pathogenesis of Alzheimer's disease is multifaceted and involves changes in neurotransmitter levels together with alterations of inflammatory, oxidative, hormonal, and synaptic pathways, which may represent a drug target for both prevention and treatment; however, an effective treatment for Alzheimer's disease still represents an unmet goal. As neurotrophic factors participate in the modulation of the above-mentioned pathways, they have been highlighted as critical contributors of Alzheimer's disease etiology, whose modulation might be beneficial for Alzheimer's disease. We focused on the neurotrophin brain-derived neurotrophic factor, providing several lines of evidence pointing to brain-derived neurotrophic factor as a plausible endophenotype of cognitive deficits in Alzheimer's disease, illustrating some of the most recent possibilities to modulate the expression of this neurotrophin in the brain in an attempt to ameliorate cognition and delay the progression of Alzheimer's disease. This review shows that otherwise disparate pharmacologic or non-pharmacologic approaches converge on brain-derived neurotrophic factor, providing a means whereby apparently unrelated medical approaches may nevertheless produce similar synaptic and cognitive outcomes in Alzheimer's disease pathogenesis, suggesting that brain-derived neurotrophic factor-based synaptic repair may represent a modifying strategy to ameliorate cognition in Alzheimer's disease.
Collapse
|
37
|
Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21041327. [PMID: 32079110 PMCID: PMC7072986 DOI: 10.3390/ijms21041327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles and associates with progressive declines in memory and cognition. Several genes play important roles and regulate enzymes that produce a pathological accumulation of β-amyloid in the brain, such as gamma secretase (γ-secretase). Induced pluripotent stem cells from patients with Alzheimer’s disease with different underlying genetic mechanisms may help model different phenotypes of Alzheimer’s disease and facilitate personalized drug screening platforms for the identification of small molecules. We also discuss recent developments by γ-secretase inhibitors and modulators in the treatment of AD. In addition, small-molecule drugs isolated from Chinese herbal medicines have been shown effective in treating Alzheimer’s disease. We propose a mechanism of small-molecule drugs in treating Alzheimer’s disease. Combining therapy with different small-molecule drugs may increase the chance of symptomatic treatment. A customized strategy tailored to individuals and in combination with therapy may be a more suitable treatment option for Alzheimer’s disease in the future.
Collapse
|
38
|
Wang K, Sun W, Zhang L, Guo W, Xu J, Liu S, Zhou Z, Zhang Y. Oleanolic Acid Ameliorates Aβ25-35 Injection-induced Memory Deficit in Alzheimer's Disease Model Rats by Maintaining Synaptic Plasticity. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 17:389-399. [PMID: 29793416 PMCID: PMC6327117 DOI: 10.2174/1871527317666180525113109] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022]
Abstract
Background: Abnormal amyloid β (Aβ) accumulation and deposition in the hippocampus is an essential process in Alzheimer’s disease (AD). Objective: To investigate whether Oleanolic acid (OA) could improve memory deficit in AD model and its possible mechanism. Methods: Forty-five SD rats were randomly divided into sham operation group, model group, and OA group. AD models by injection of Aβ25-35 were built. Morris water maze (MWM) was applied to inves-tigate learning and memory, transmission electron microscope (TEM) to observe the ultrastructure of synapse, western blot to the proteins, electrophysiology for long-term potentiation (LTP), and Ca2+ con-centration in synapse was also measured. Results: The latency time in model group was significantly longer than that in sham operation group (P=0.0001); while it was significantly shorter in the OA group than that in model group (P=0.0001); compared with model group, the times of cross-platform in OA group significantly increased (P=0.0001). TEM results showed OA could alleviate neuron damage and synapses changes induced by Aβ25-35. The expressions of CaMKII, PKC, NMDAR2B, BDNF, TrkB, and CREB protein were signif-icantly improved by OA (P=0.0001, 0.036, 0.041, 0.0001, 0.0001, 0.026, respectively) compared with that in model group; the concentration of Ca2+ was significantly lower in OA group (1.11±0.42) than that in model group (1.68±0.18); and the slope rate (P=0.0001) and amplitude (P=0.0001) of f-EPSP significantly increased in OA group. Conclusion: The present results support that OA could ameliorate Aβ-induced memory loss of AD rats by maintaining synaptic plasticity of the hippocampus
Collapse
Affiliation(s)
- Kai Wang
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Weiming Sun
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Linlin Zhang
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Wei Guo
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Jiachun Xu
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shuang Liu
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Zhen Zhou
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Yulian Zhang
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| |
Collapse
|
39
|
Han J, Oh JP, Yoo M, Cui CH, Jeon BM, Kim SC, Han JH. Minor ginsenoside F1 improves memory in APP/PS1 mice. Mol Brain 2019; 12:77. [PMID: 31488185 PMCID: PMC6728944 DOI: 10.1186/s13041-019-0495-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/21/2019] [Indexed: 11/10/2022] Open
Abstract
Ginseng has been shown to produce a cognitive improvement effect. The key molecular components in ginseng that produce pharmacological effects are ginsenosides. Previous studies reported a memory improvement effect of a few major ginsenosides. However, the identity of specific minor ginsenosides mediating such function remains unknown. Here, we report that a minor ginsenoside F1 improves memory function in APPswe/PSEN1dE9 (APP/PS1) double-transgenic Alzheimer's disease (AD) model mice. After 8-wk oral administration of F1 jelly, we observed that spatial working memory, but not context-dependent fear memory, was restored in AD mice. To search for a possible underlying molecular and cellular mechanism, we investigated the effect of F1 on Aβ plaque. We observed F1 administration reduced the Aβ plaque area and density in the cortex, but not in the hippocampus of AD mice. Next, we tested for the effect of F1 on the expression level of key molecules involved in learning and memory. Results from Western blot assay revealed that an abnormally reduced level of a phosphorylated form of CREB in the hippocampus of AD mice was restored to a normal level by F1 administration. Moreover, in the same animals, BDNF level was augmented in the cortex. Our results, therefore, suggest that minor ginsenoside F1 constitutes a promising target to develop therapeutic agents for AD.
Collapse
Affiliation(s)
- Junho Han
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Jung-Pyo Oh
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Miran Yoo
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Chang-Hao Cui
- Intelligent Synthetic Biology Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Byeong-Min Jeon
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Sun-Chang Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| | - Jin-Hee Han
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| |
Collapse
|
40
|
Fukuchi M, Okuno Y, Nakayama H, Nakano A, Mori H, Mitazaki S, Nakano Y, Toume K, Jo M, Takasaki I, Watanabe K, Shibahara N, Komatsu K, Tabuchi A, Tsuda M. Screening inducers of neuronal BDNF gene transcription using primary cortical cell cultures from BDNF-luciferase transgenic mice. Sci Rep 2019; 9:11833. [PMID: 31413298 PMCID: PMC6694194 DOI: 10.1038/s41598-019-48361-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/01/2019] [Indexed: 01/04/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a key player in synaptic plasticity, and consequently, learning and memory. Because of its fundamental role in numerous neurological functions in the central nervous system, BDNF has utility as a biomarker and drug target for neurodegenerative and neuropsychiatric disorders. Here, we generated a screening assay to mine inducers of Bdnf transcription in neuronal cells, using primary cultures of cortical cells prepared from a transgenic mouse strain, specifically, Bdnf-Luciferase transgenic (Bdnf-Luc) mice. We identified several active extracts from a library consisting of 120 herbal extracts. In particular, we focused on an active extract prepared from Ginseng Radix (GIN), and found that GIN activated endogenous Bdnf expression via cAMP-response element-binding protein-dependent transcription. Taken together, our current screening assay can be used for validating herbal extracts, food-derived agents, and chemical compounds for their ability to induce Bdnf expression in neurons. This method will be beneficial for screening of candidate drugs for ameliorating symptoms of neurological diseases associated with reduced Bdnf expression in the brain, as well as candidate inhibitors of aging-related cognitive decline.
Collapse
Affiliation(s)
- Mamoru Fukuchi
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma, 370-0033, Japan.
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| | - Yui Okuno
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Hironori Nakayama
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Aoi Nakano
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Satoru Mitazaki
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma, 370-0033, Japan
- Laboratory of Forensic Toxicology, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma, 370-0033, Japan
| | - Yuka Nakano
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma, 370-0033, Japan
| | - Kazufumi Toume
- Division of Pharmacognosy, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Michiko Jo
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Ichiro Takasaki
- Department of Pharmacology, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| | - Kazuki Watanabe
- Laboratory of Natural Medicines, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma, 370-0033, Japan
| | - Naotoshi Shibahara
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Katsuko Komatsu
- Division of Pharmacognosy, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Akiko Tabuchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Masaaki Tsuda
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| |
Collapse
|
41
|
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: 44] [Impact Index Per Article: 7.3] [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
|
42
|
Ginsenoside Rg1 Prevents Chemotherapy-Induced Cognitive Impairment: Associations with Microglia-Mediated Cytokines, Neuroinflammation, and Neuroplasticity. Mol Neurobiol 2019; 56:5626-5642. [DOI: 10.1007/s12035-019-1474-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/10/2019] [Indexed: 12/26/2022]
|
43
|
Yu L, Duan Y, Zhao Z, He W, Xia M, Zhang Q, Cao X. Hydroxysafflor Yellow A (HSYA) Improves Learning and Memory in Cerebral Ischemia Reperfusion-Injured Rats via Recovering Synaptic Plasticity in the Hippocampus. Front Cell Neurosci 2018; 12:371. [PMID: 30405354 PMCID: PMC6200869 DOI: 10.3389/fncel.2018.00371] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022] Open
Abstract
Hydroxysafflor yellow A (HSYA) is the major active chemical component of the safflower plant flower, which is widely used in Chinese medicine for cerebrovascular and cardiovascular disease treatment. Recent studies have demonstrated that HSYA exerts neuroprotective effect on cerebral ischemia, such as neuronal anti-apoptosis, antioxidant activity and oxygen free radical-scavenging. However, whether and how HSYA has a protective effect on cognitive impairment induced by cerebral ischemia reperfusion remains elusive. In the present study, by using the middle cerebral artery occlusion (MCAO) model, we found that 8 mg/kg and 16 mg/kg HSYA administration by common carotid artery (CCA) injection improved impaired cognitive function in Morris water maze (MWM) and passive avoidance tasks, but not 4 mg/kg HSYA treatment, suggesting that HSYA treatment in a certain concentration can improve cognitive impairment in MCAO rats. Furthermore, we found that 8 mg/kg HSYA treatment rescued the impaired long-term potentiation (LTP) in hippocampus of MCAO rats. Taken together, these results for the first time demonstrate that HSYA has the capacity to protect cognitive function and synaptic plasticity against cerebral ischemia-reperfusion injury, and provide a new insight that HSYA may be a promising alternative for recovery of cognitive dysfunction after brain ischemic injury.
Collapse
Affiliation(s)
- Lu Yu
- Comprehensive Department of Traditional Chinese Medicine, Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanhong Duan
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zheng Zhao
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wendi He
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ming Xia
- Comprehensive Department of Traditional Chinese Medicine, Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiujuan Zhang
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohua Cao
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
44
|
Cao C, Xiao J, Liu M, Ge Z, Huang R, Qi M, Zhu H, Zhu Y, Duan JA. Active components, derived from Kai-xin-san, a herbal formula, increase the expressions of neurotrophic factor NGF and BDNF on mouse astrocyte primary cultures via cAMP-dependent signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:554-562. [PMID: 29890314 DOI: 10.1016/j.jep.2018.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kai-Xin-San (KXS), an ancient formula composed of Ginseng Radix et Rhizoma, Polygalae Radix, Acori Tatarinowii Rhizoma and Poria, was frequently applied for Alzheimer's disease and major depression disorders for thousands of years. However, its active components and molecular mechanism have not clearly been investigated. AIM OF THE STUDY We aimed to reveal the active components of KXS on regulating neurotrophic factor NGF and BDNF expressions and its mechanisms on mouse astrocyte primary cultures. MATERIALS AND METHODS Extracts of KXS had been prepared by water reflux and chemical standardization was carried out by HPLC-MS/MS. Various ethanol elution components were prepared by eluting ethanol on macro pore resin column and compound identification was carried out by high-resolution mass spectrometry. KXS extract, elution components and identified chemicals were applied on mouse astrocytes and expressions of NGF and BDNF and related metabolic enzymes were analyzed by qPCR and western blotting analysis. RESULTS One compatible ratio of KXS named D-652 exerted the best effect on stimulation of NGF and BDNF expressions on mouse astrocytes. 70% ethanol elution fraction of D-652 exerted the highest increase tendency on expressions of NGF and BDNF by activating cAMP-dependent signaling pathway as well as stimulating enzymes accounting for neurotrophic factor synthesis. Combined with compound identification by high-resolution mass spectrometry, ginsenoside Rg1 and Rb1 might be the active compounds of this fraction on increasing NGF and BDNF expressions. CONCLUSIONS The active compounds of KXS on increasing NGF and BDNF expressions might be the ginsenosides via activating cAMP-dependent signaling pathway as well as stimulating enzymes accounting for neurotrophic factor synthesis, which partly reveal the target of this formulae supported the clinically usage of this decoction.
Collapse
Affiliation(s)
- Cheng Cao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Junyuan Xiao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mengqiu Liu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Zishuo Ge
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Renjie Huang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mingzhu Qi
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Hongliang Zhu
- Suzhou Guangji Hospital, Su Zhou, Jiangsu Province, China.
| | - Yue Zhu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Jin-Ao Duan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| |
Collapse
|
45
|
Gray NE, Zweig JA, Caruso M, Zhu JY, Wright KM, Quinn JF, Soumyanath A. Centella asiatica attenuates hippocampal mitochondrial dysfunction and improves memory and executive function in β-amyloid overexpressing mice. Mol Cell Neurosci 2018; 93:1-9. [PMID: 30253196 DOI: 10.1016/j.mcn.2018.09.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/21/2022] Open
Abstract
Centella asiatica is a medicinal plant used to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) attenuates β-amyloid (Aβ)-induced spatial memory deficits in mice and improves neuronal health. Yet the effect of CAW on other cognitive domains remains unexplored as does its in vivo mechanism of improving Aβ-related cognitive impairment. This study investigates the effects of CAW on learning, memory and executive function as well as mitochondrial function and antioxidant response in the 5xFAD model of Aβ accumulation. Seven month old 5xFAD female mice were treated with CAW (2 mg/mL) in their drinking water for two weeks prior to behavioral testing. Learning, memory and executive function were assessed using the object location memory task (OLM), conditioned fear response (CFR) and odor discrimination reversal learning (ODRL) test. Mitochondrial function was profiled using the Seahorse XF platform in hippocampal mitochondria isolated from these animals and tissue was harvested for assessment of mitochondrial, antioxidant and synaptic proteins. CAW improved performance in all behavioral tests in the 5xFAD but had no effect on WT animals. Hippocampal mitochondrial function was improved and hippocampal and cortical expression of mitochondrial genes was increased in CAW-treated 5xFAD mice. Gene expression of the transcription factor NRF2, as well as its antioxidant target enzymes, was also increased with CAW treatment in both WT and 5xFAD mice. CAW treatment also decreased Aβ-plaque burden in the hippocampus of treated 5xFAD mice but had no effect on plaques in the cortex. These data show that CAW can improve many facets of Aβ-related cognitive impairment in 5xFAD mice. Oral treatment with CAW also attenuates hippocampal mitochondrial dysfunction in these animals. Because mitochondrial dysfunction and oxidative stress accompany cognitive impairment in many pathological conditions beyond Alzheimer's disease, this suggests potentially broad therapeutic utility of CAW.
Collapse
Affiliation(s)
- Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kirsten M Wright
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
46
|
Jakaria M, Haque ME, Kim J, Cho DY, Kim IS, Choi DK. Active ginseng components in cognitive impairment: Therapeutic potential and prospects for delivery and clinical study. Oncotarget 2018; 9:33601-33620. [PMID: 30323902 PMCID: PMC6173364 DOI: 10.18632/oncotarget.26035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment is a state that affects thinking, communication, understanding, and memory, and is very common in various neurological disorders. Among many factors, age-related cognitive decline is an important area in mental health research. Research to find therapeutic medications or supplements to treat cognitive deficits and maintain cognitive health has been ongoing. Ginseng and its active components may have played a role in treating chronic disorders. Numerous preclinical studies have confirmed that ginseng and its active components such as ginsenosides, gintonin, and compound K are pharmacologically efficacious in different models of and are linked to cognitive impairment. Among their several roles, they act as an anti-neuroinflammatory and help fight against oxidative stress and modulate the cholinergic signal. These roles may be involved in enhancing cognition and attenuating impairment. There have been some clinical studies on the activity of ginseng in cognitive impairment, but many ginseng species and active compounds remain to be investigated. In addition, new formulations of active ginseng components such as nanoparticles and liposomes could be used for preclinical and clinical models of cognitive impairment. Here, we discuss the therapeutic potential of active ginseng components in cognitive impairment and their chemistry and pharmacokinetics and consider prospects for their delivery and clinical study with respect to cognitive impairment.
Collapse
Affiliation(s)
- Md. Jakaria
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Md. Ezazul Haque
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Joonsoo Kim
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Duk-Yeon Cho
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - In-Su Kim
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease, Konkuk University, Chungju 27478, Republic of Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
47
|
Kim EH, Kim W. An Insight into Ginsenoside Metabolite Compound K as a Potential Tool for Skin Disorder. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:8075870. [PMID: 30046346 PMCID: PMC6036801 DOI: 10.1155/2018/8075870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/02/2018] [Indexed: 02/06/2023]
Abstract
Ginsenosides are the major bioactive natural compounds derived from Panax ginseng. Several studies report the pharmaceutical benefits of several ginsenosides, including antidementia, antitumor, and anti-inflammatory activity. Biotransformations by gut microbiome contribute to the biological function of these ginsenosides. After ingestion ginsenosides are hydrolyzed to Rg2, Rg3, compound K, and others by human gut flora. Compound K is considered the representative active metabolite after oral administration of ginseng or ginsenosides. Various studies report the diverse biological functions of compound K, such as antitumor, antidiabetic, antiallergic, and anti-inflammatory activity. Recent clinical trial and in vitro studies demonstrate the antiaging activities of ginsenosides in human skin. Ginsenosides have been considered as an important natural dermatological agent. In this review, we will cover the modern tools and techniques to understand biotransformation and delivery of compound K. Also the biological function of compound K on skin disorder and its potential dermatological application will be discussed.
Collapse
Affiliation(s)
- En Hyung Kim
- Department of Dermatology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Wonnam Kim
- Division of Pharmacology, College of Korean Medicine, Semyung University, Jecheon, Republic of Korea
| |
Collapse
|
48
|
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: 76] [Impact Index Per Article: 10.9] [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
|
49
|
Jia J, Wei C, Chen S, Li F, Tang Y, Qin W, Shi L, Gong M, Xu H, Li F, He J, Song H, Yang S, Zhou A, Wang F, Zuo X, Chu C, Liang J, Jia L, Gauthier S. Efficacy and safety of the compound Chinese medicine SaiLuoTong in vascular dementia: A randomized clinical trial. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2018; 4:108-117. [PMID: 29955654 PMCID: PMC6021260 DOI: 10.1016/j.trci.2018.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Introduction No licensed medications are available to treat vascular dementia (VaD). Methods Patients were randomly assigned to experimental groups (SaiLuoTong [SLT] 360 or 240 mg for groups A and B for 52 weeks, respectively) or placebo group (SLT 360 mg and 240 mg for group C only from weeks 27 to 52, respectively). Results Three hundred twenty-five patients were included in final analysis. At week 26, the difference in VaD Assessment Scale–cognitive subscale scores was 2.67 (95% confidence interval, 1.54 to 3.81) for groups A versus C, and 2.48 (1.34 to 3.62) for groups B versus C (both P < .0001). However, at week 52, no difference was observed among the groups on the VaD Assessment Scale–cognitive subscale (P = .062) because of the emerging efficacy of SLT in placebo beginning at week 27. Discussion This study suggests that SLT is effective for treatment of VaD, and this compound Chinese medicine may represent a better choice to treat VaD.
Collapse
Affiliation(s)
- Jianping Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Shuoqi Chen
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wei Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Lu Shi
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Min Gong
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Hui Xu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Fang Li
- Department of Gerontology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Jia He
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Haiqing Song
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Shanshan Yang
- Department of Neurology, Daqing Oilfield General Hospital, China
| | - Aihong Zhou
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Fen Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Xiumei Zuo
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Changbiao Chu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Junhua Liang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Longfei Jia
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Serge Gauthier
- Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| |
Collapse
|
50
|
Jakaria M, Kim J, Karthivashan G, Park SY, Ganesan P, Choi DK. Emerging signals modulating potential of ginseng and its active compounds focusing on neurodegenerative diseases. J Ginseng Res 2018; 43:163-171. [PMID: 30976157 PMCID: PMC6437449 DOI: 10.1016/j.jgr.2018.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 02/08/2023] Open
Abstract
Common features of neurodegenerative diseases (NDDs) include progressive dysfunctions and neuronal injuries leading to deterioration in normal brain functions. At present, ginseng is one of the most frequently used natural products. Its use has a long history as a cure for various diseases because its extracts and active compounds exhibit several pharmacological properties against several disorders. However, the pathophysiology of NDDs is not fully clear, but researchers have found that various ion channels and specific signaling pathways might have contributed to the disease pathogenesis. Apart from the different pharmacological potentials, ginseng and its active compounds modulate various ion channels and specific molecular signaling pathways related to the nervous system. Here, we discuss the signal modulating potential of ginseng and its active compounds mainly focusing on those relevant to NDDs.
Collapse
Affiliation(s)
- Md Jakaria
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Joonsoo Kim
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Govindarajan Karthivashan
- Research Institute of Inflammatory Disease, and Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea
| | - Shin-Young Park
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea
| | - Palanivel Ganesan
- Research Institute of Inflammatory Disease, and Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.,Nanotechnology Research Center, Konkuk University, Chungju, Republic of Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, Republic of Korea.,Research Institute of Inflammatory Disease, and Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.,Nanotechnology Research Center, Konkuk University, Chungju, Republic of Korea
| |
Collapse
|