1
|
Xu XW, Zhou XW, Zhang L, Wang Q, Wang XX, Jin YM, Li LL, Jin MF, Wu HY, Ding X, Ni H. Complexin 2 contributes to the protective effect of NAD + on neuronal survival following neonatal hypoxia-ischemia. Acta Pharmacol Sin 2025:10.1038/s41401-025-01555-1. [PMID: 40247039 DOI: 10.1038/s41401-025-01555-1] [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: 11/05/2024] [Accepted: 03/30/2025] [Indexed: 04/19/2025]
Abstract
Nicotinamide adenine dinucleotide (NAD) is a key coenzyme involved in cell metabolism associated with aging, cancer, neurodegenerative diseases and metabolic disorders. We recently showed that NAD+ therapy significantly improved neurobehavioral outcomes in neonatal mice after hypoxia-ischemia (HI), and bioinformatics analysis revealed that the expression of complexin 2 (CPLX2) in the injured cerebral cortex was significantly decreased 24 h after HI injury but could be reversed by NAD+ intervention. In this study we explored the role of CPLX2 in the survival and function of neonatal hypoxic-ischemic cortical neurons. HI models were established by permanent ligation of the left common carotid artery in mice. CPLX2-knockdown lentiviral vector was injected intraventricularly on postnatal day 1 (P1); CPLX2 knockout mice were also used. NAD+ (5 mg·kg-1·d-1, i.p.) was administered before HI surgery, thereafter once a day until sampling. We showed that NAD+ administration significantly ameliorated the morphological damages and neurobehavioral defects, and elevated the seizure thresholds in HI mice. All the beneficial effects of NAD+ were abolished by CPLX2 knockdown or knockout. In HT22 neuronal cells subjected to OGD/R, pretreated with NAD+ (100 μM) for 12 h significantly increased the cell viability, decreased the LDH levels, and inhibited the ferroptosis evidenced by the changes in redox-related parameters including concentrations of Fe2+, GSH, MDA, H2O2 as well as the expression of GPX4 and SLC7A11. CPLX2 knockdown in HT22 neuronal cells blocked the protective effects of NAD+ as in HI mice, whereas CPLX2 overexpression enhanced the inhibitory effects of NAD+ on ferroptosis in HT22 neuronal cells.
Collapse
Affiliation(s)
- Xiao-Wen Xu
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xiu-Wen Zhou
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li Zhang
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Qing Wang
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xin-Xin Wang
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Yi-Ming Jin
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li-Li Li
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mei-Fang Jin
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Hai-Ying Wu
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Xin Ding
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Hong Ni
- Department of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
2
|
Ma X, Xue S, Ma H, Saeed S, Zhang Y, Meng Y, Chen H, Yu H, Wang H, Hu S, Cai M. Esketamine alleviates LPS-induced depression-like behavior by activating Nrf2-mediated anti-inflammatory response in adolescent mice. Neuroscience 2025; 567:294-307. [PMID: 39755229 DOI: 10.1016/j.neuroscience.2024.12.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND The mechanisms underlying esketamine's therapeutic effects remain elusive. The study aimed to explore the impact of single esketamine treatment on LPS-induced adolescent depressive-like behaviors and the role of Nrf2 regulated neuroinflammatory response in esketamine-produced rapid antidepressant efficacy. METHODS Adolescent male C57BL/6J mice were randomly assigned to three groups: control, LPS, and LPS + esketamine (15 mg/kg, i.p.). Depressive-like behaviors were evaluated via the OFT, NFST, and TST. Protein expression of Nrf2 and inflammatory cytokines, including TNF-α, IL-1β, and iNOS in the hippocampus and mPFC, were measured by western blot. Moreover, the Nrf2 inhibitor, ML385, was also applied in the current study. The depressive-like behaviors and the protein expression of Nrf2, TNF-α, IL-1β, and iNOS in mPFC and hippocampus were also measured. Additionally, the plasma's pro-inflammatory cytokines and anti-inflammatory cytokines were assessed using ELISA methods with or without ML385. RESULTS A single administration of esketamine treatment alleviated the LPS-induced depressive-like behaviors. Esketamine increased the expression of Nrf2 and reduced the expression of the inflammatory cytokines, including TNF-α, IL-1β, and iNOS, in the mPFC and hippocampus. Notably, pharmacological inhibition of Nrf2 via ML385 administration abrogated the antidepressive-like behaviors and anti-inflammatory effects induced by esketamine. In the periphery, esketamine mitigated the LPS-induced elevation of pro-inflammatory cytokines, and the reduction of anti-inflammatory cytokines, and this effect was reversed by Nrf2 inhibition. CONCLUSION Esketamine treatment exerts rapid antidepressant effects and attenuates neuroinflammation in LPS-induced adolescent depressive-like behaviors, potentially through the activation of Nrf2-mediated anti-inflammatory signaling.
Collapse
Affiliation(s)
- Xinxu Ma
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shanshan Xue
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongzhe Ma
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Saboor Saeed
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yunyun Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Meng
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haixia Chen
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huan Yu
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huaizhi Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; The Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou 310003, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310003, China; Brain Research Institute of Zhejiang University, Hangzhou, 310003, China; The State Key Lab of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China; Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China.
| | - Min Cai
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Qin Y, Zhang Z, Jiang R. Ginsenoside Rg1 Promotes Wound Healing in Mice with Superficial Second-Degree Burns Through Energy Metabolism, Cell Migration, and Cell Adhesion Pathways. J Med Food 2025; 28:165-173. [PMID: 39469786 DOI: 10.1089/jmf.2024.k.0146] [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] [Indexed: 10/30/2024] Open
Abstract
Natural products are known to have distinct roles in the treatment of various diseases. However, the potential role of ginsenoside Rg1 (GRg1) in the context of scald injuries remains unclear. This study aimed to elucidate the effects of GRg1 on scald wound healing by utilizing a mouse scald wound model and administering varying concentrations of GRg1 orally. RNA sequencing (RNA-seq) was employed to identify the signaling pathways and key genes influenced by GRg1 in the wound healing process. Our findings indicate that mice treated with a low concentration of GRg1 exhibited a significantly higher wound healing rate compared with the model group and other treatment groups. Through RNA-seq, we observed that the gene expression profile in the wound tissues of the low-concentration-treated group was consistent with that of the normal control group. Furthermore, a low concentration of GRg1 was found to maintain cellular energy metabolism homeostasis by enhancing mitochondrial aerobic respiration and the tricarboxylic acid cycle. In addition, GRg1 facilitated wound healing by restoring the expression of genes associated with cell migration and adhesion. Confirming the appropriate concentration of GRg1 that accelerates tissue healing at scald sites and enhances our understanding of the efficacy and molecular mechanisms underlying the therapeutic effects of natural products in disease treatment.
Collapse
Affiliation(s)
- Yunna Qin
- Department of Pathology, Jiangxi Maternal and Child Health Hospital, Nanchang, P.R. China
| | - Ziyu Zhang
- Department of Pathology, Jiangxi Maternal and Child Health Hospital, Nanchang, P.R. China
| | - Ru Jiang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
4
|
Yang C, Jiang W, Su D, Yang C, Yuan Q, Kang C, Xiao C, Wang L, Peng C, Zhou T, Zhang J. Contamination of the traditional medicine Radix Dipsaci with aflatoxin B1 impairs hippocampal neurogenesis and cognitive function in a mouse model of osteoporosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116831. [PMID: 39151374 DOI: 10.1016/j.ecoenv.2024.116831] [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: 02/27/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Aflatoxin B1, which can penetrate the blood-brain barrier and kill neural cells, can contaminate traditional herbal medicines, posing a significant risk to human health. The present study examined cellular, cognitive and behavioral consequences of aflatoxin B1 contamination of the anti-osteoporotic medicine Radix Dipsaci. METHODS A mouse model of osteoporosis was created by treating the animals with all-trans-retinoic acid. Then the animals were treated intragastically with water decoctions of Radix Dipsaci that contained detectable aflatoxin B1 or not. The animals were compared in terms of mineral density and mineral salt content of bone, production of pro-inflammatory factors, neurogenesis and microglial activation in hippocampus, as well as behavior and cognitive function. RESULTS Contamination of Radix Dipsaci with aflatoxin B1 significantly reduced the medicine's content of bioactive saponins. It destroyed the ability of the herbal decoction to improve mineral density and mineral salt content in the bones of diseased mice, and it induced the production of the oxidative stress marker malondialdehyde as well as the pro-inflammatory cytokines interleukin-1β and tumor necrosis factor-α. Aflatoxin B1 contamination inhibited formation of new neurons and increased the proportion of activated microglia in the hippocampus. These neurological changes were associated with anhedonia, behavioral despair, and deficits in short-term memory and social memory. CONCLUSION Contamination of Radix Dipsaci with aflatoxin B1 not only eliminates the herbal decoction's anti-osteoporotic effects, but it also induces neurotoxicity that can lead to cognitive decline and behavioral abnormalities. Such contamination should be avoided through tightly regulated production and quality control of medicinal herbs.
Collapse
Affiliation(s)
- Chengyan Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Weike Jiang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Dapeng Su
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Changgui Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Qingsong Yuan
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chuanzhi Kang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Lulu Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Cheng Peng
- Chengdu University of Traditional Chinese Medicine.
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
5
|
Rivero-Segura NA, Zepeda-Arzate EA, Castillo-Vazquez SK, Fleischmann-delaParra P, Hernández-Pineda J, Flores-Soto E, García-delaTorre P, Estrella-Parra EA, Gomez-Verjan JC. Exploring the Geroprotective Potential of Nutraceuticals. Nutrients 2024; 16:2835. [PMID: 39275153 PMCID: PMC11396943 DOI: 10.3390/nu16172835] [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: 07/06/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Aging is the result of the accumulation of a wide variety of molecular and cellular damages over time, meaning that "the more damage we accumulate, the higher the possibility to develop age-related diseases". Therefore, to reduce the incidence of such diseases and improve human health, it becomes important to find ways to combat such damage. In this sense, geroprotectors have been suggested as molecules that could slow down or prevent age-related diseases. On the other hand, nutraceuticals are another set of compounds that align with the need to prevent diseases and promote health since they are biologically active molecules (occurring naturally in food) that, apart from having a nutritional role, have preventive properties, such as antioxidant, anti-inflammatory and antitumoral, just to mention a few. Therefore, in the present review using the specialized databases Scopus and PubMed we collected information from articles published from 2010 to 2023 in order to describe the role of nutraceuticals during the aging process and, given their role in targeting the hallmarks of aging, we suggest that they are potential geroprotectors that could be consumed as part of our regular diet or administered additionally as nutritional supplements.
Collapse
Affiliation(s)
| | | | - Selma Karime Castillo-Vazquez
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), Mexico City 10200, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Jessica Hernández-Pineda
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, SSA, Mexico City 11000, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad No. 3000, Alcaldía de Coyoacán, Mexico City 04510, Mexico
| | - Paola García-delaTorre
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Edgar Antonio Estrella-Parra
- Laboratorio de Fitoquímica, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz 54090, Mexico
| | | |
Collapse
|
6
|
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
|
7
|
Chen Z, Wang X, Du S, Liu Q, Xu Z, Guo Y, Lin X. A review on traditional Chinese medicine natural products and acupuncture intervention for Alzheimer's disease based on the neuroinflammatory. Chin Med 2024; 19:35. [PMID: 38419106 PMCID: PMC10900670 DOI: 10.1186/s13020-024-00900-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with insidious onset and progressive development. It is clinically characterized by cognitive impairment, memory impairment and behavioral change. Chinese herbal medicine and acupuncture are important components of traditional Chinese medicine (TCM), and are commonly used in clinical treatment of AD. This paper systematically summarizes the research progress of traditional Chinese medicine natural products and acupuncture treatment of AD, which combined with existing clinical and preclinical evidence, based on a comprehensive review of neuroinflammation, and discusses the efficacy and potential mechanisms of traditional Chinese medicine natural products and acupuncture treatment of AD. Resveratrol, curcumin, kaempferol and other Chinese herbal medicine components can significantly inhibit the neuroinflammation of AD in vivo and in vitro, and are candidates for the treatment of AD. Acupuncture can alleviate the memory and cognitive impairment of AD by improving neuroinflammation, synaptic plasticity, nerve cell apoptosis and reducing the production and aggregation of amyloid β protein (Aβ) in the brain. It has the characteristics of early, safe, effective and benign bidirectional adjustment. The purpose of this paper is to provide a basis for improving the clinical strategies of TCM for the treatment of AD.
Collapse
Affiliation(s)
- Zhihan Chen
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Xinrui Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Simin Du
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Qi Liu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Zhifang Xu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China.
| | - Xiaowei Lin
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
8
|
Abo Mansour HE, Elberri AI, Ghoneim MES, Samman WA, Alhaddad AA, Abdallah MS, El-Berri EI, Salem MA, Mosalam EM. The Potential Neuroprotective Effect of Thymoquinone on Scopolamine-Induced In Vivo Alzheimer's Disease-like Condition: Mechanistic Insights. Molecules 2023; 28:6566. [PMID: 37764343 PMCID: PMC10534545 DOI: 10.3390/molecules28186566] [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: 07/20/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common neurodegenerative disorder without effective treatment. Thymoquinone (TQ) has demonstrated potential in exhibiting anti-inflammatory, anti-cancer, and antioxidant characteristics. Despite TQ's neuroprotection effect, there is a scarcity of information regarding its application in AD research, and its molecular trajectories remain ambiguous. Thus, the objective of the current investigation was to examine the potential beneficial effects and underlying mechanisms of TQ in scopolamine (SCOP)-induced neuronal injury to mimic AD in vivo model. METHODS Thirty mice were divided into normal, SCOP, and TQ groups. The Y-maze and pole climbing tests were performed to measure memory and motor performance. Afterwards, histopathological and immunohistochemical examinations were carried out. Furthermore, peroxisome proliferator-activated receptor gamma (PPAR-γ) signaling pathway-related proteins and genes were detected with an emphasis on the role of miR-9. RESULTS TQ has the potential to ameliorate cognitive deficits observed in SCOP-induced AD-like model, as evidenced by the improvement in behavioral outcomes, histopathological changes, modulation of the expression pattern of PPAR-γ downstream targets with a significant decrease in the deposition of amyloid beta (Aβ). CONCLUSIONS TQ provided meaningful multilevel neuroprotection through its anti-inflammatory and its PPAR-γ agonist activity. Consequently, TQ may possess a potential beneficial role against AD development.
Collapse
Affiliation(s)
- Hend E. Abo Mansour
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, Shibin El-Kom 32511, Egypt;
| | - Aya Ibrahim Elberri
- Genetic Engineering and Molecular Biology Division, Department of Zoology, Faculty of Science, Menoufia University, Shibin El-Kom 32511, Egypt;
| | - Mai El-Sayed Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Sadat City 32897, Egypt;
| | - Waad A. Samman
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina 42353, Saudi Arabia; (W.A.S.); (A.A.A.)
| | - Aisha A. Alhaddad
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina 42353, Saudi Arabia; (W.A.S.); (A.A.A.)
| | - Mahmoud S. Abdallah
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Sadat City (USC), Sadat City 32897, Egypt;
| | - Eman I. El-Berri
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| | - Mohamed A. Salem
- Department of Pharmacognosy, Faculty of Pharmacy, Menoufia University, Shibin El-Kom 32511, Egypt;
| | - Esraa M. Mosalam
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, Shibin El-Kom 32511, Egypt;
| |
Collapse
|
9
|
Temporal Appearance of Enhanced Innate Anxiety in Alzheimer Model Mice. Biomedicines 2023; 11:biomedicines11020262. [PMID: 36830799 PMCID: PMC9953677 DOI: 10.3390/biomedicines11020262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
The prevalence of Alzheimer's disorder (AD) is increasing worldwide, and the co-morbid anxiety is an important, albeit often neglected problem, which might appear early during disease development. Animal models can be used to study this question. Mice, as prey animals, show an innate defensive response against a predator odor, providing a valuable tool for anxiety research. Our aim was to test whether the triple-transgenic mice model of AD shows signs of innate anxiety, with specific focus on the temporal appearance of the symptoms. We compared 3xTg-AD mice bearing human mutations of amyloid precursor protein, presenilin 1, and tau with age-matched controls. First, separate age-groups (between 2 and 18 months) were tested for the avoidance of 2-methyl-2-thiazoline, a fox odor component. To test whether hypolocomotion is a general sign of innate anxiety, open-field behavior was subsequently followed monthly in both sexes. The 3xTg-AD mice showed more immobility, approached the fox odor container less often, and spent more time in the avoidance zone. This effect was detectable already in two-month-old animals irrespective of sex, not visible around six months of age, and was more pronounced in aged females than males. The 3xTg-AD animals moved generally less. They also spent less time in the center of the open-field, which was detectable mainly in females older than five months. In contrast to controls, the aged 3xTg-AD was not able to habituate to the arena during a 30-min observation period irrespective of their sex. Amyloid beta and phospho-Tau accumulated gradually in the hippocampus, amygdala, olfactory bulb, and piriform cortex. In conclusion, the early appearance of predator odor- and open space-induced innate anxiety detected already in two-month-old 3xTg-AD mice make this genetically predisposed strain a good model for testing anxiety both before the onset of AD-related symptoms as well as during the later phase. Synaptic dysfunction by protein deposits might contribute to these disturbances.
Collapse
|
10
|
Dong X, Kong L, Huang L, Su Y, Li X, Yang L, Ji P. Ginsenoside Rg1 treatment protects against cognitive dysfunction via inhibiting PLC–CN–NFAT1 signaling in T2DM mice. J Ginseng Res 2022; 47:458-468. [DOI: 10.1016/j.jgr.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
|
11
|
Liu L, Du X, Yang Q, Li M, Ran Q, Liu Q, Yang L, Sun L, Guo Y, Li Y, Chen Y, Zhu X, Li Q. Ginsenoside Rg1 promotes remyelination and functional recovery in demyelinating disease by enhancing oligodendrocyte precursor cells-mediated myelin repair. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154309. [PMID: 35994846 DOI: 10.1016/j.phymed.2022.154309] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inefficient differentiation of oligodendrocyte precursor cells (OPCs) is one of the significant pathological obstacles of myelin repair and provides an essential therapeutic target against behavioral dysfunction in various neurodegenerative diseases, especially in secondary progressive multiple sclerosis (SPMS). Ginsenoside Rg1 (Rg1) has traditionally been recognized as a protector of neuronal damages, preventing its degeneration. PURPOSE We investigated the effects of Rg1 on myelin regeneration-mediated by OPCs and its therapeutic significance in SPMS. METHODS A cuprizone (CPZ) model was established and then administered with Rg1 specific for evaluations of functional recovery and remyelination. In vitro, the primary mouse OPCs were isolated and cultured for examining their ability of myelin repair. Furthermore, a chronic experimental autoimmune encephalomyelitis (EAE) model was utilized to assess the therapeutic value on SPMS. RESULTS We found that Rg1 promoted functional recovery of the demyelinated mice, including spatial memory, motor function, and anxiety-like behavior. Histologically, Rg1 enhanced myelin-genesis as proven by myelin staining and microstructures of myelin observed by transmission electron microscope. Furthermore, Rg1 significantly increased Olig2+ oligodendrocyte lineage cells in callosum, implying that the pro-remyelination effect of Rg1 was closely correlated to the enhanced differentiation of OPCs. We further demonstrated that Rg1 increased the survival and proliferation of OPCs as well as induced maturation in oligodendrocytes (OLs). Molecular analysis showed that Rg1 transduced the pro-differentiation signaling programmed by the GSK3β/β-Catenin pathway. Notably, relying on its pro-remyelination effects, Rg1 ameliorated severity and histopathology of EAE disease. CONCLUSION By paving the way for OPCs differentiation, Rg1 could maintain the integrity of myelin and is a promising candidate for functional recovery in demyelinating diseases.
Collapse
Affiliation(s)
- Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xinke Du
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Manjing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingsen Ran
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingwu Liu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lina Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lisong Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuxuan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
12
|
Li RL, Wang LY, Duan HX, Zhang Q, Guo X, Wu C, Peng W. Regulation of mitochondrial dysfunction induced cell apoptosis is a potential therapeutic strategy for herbal medicine to treat neurodegenerative diseases. Front Pharmacol 2022; 13:937289. [PMID: 36210852 PMCID: PMC9535092 DOI: 10.3389/fphar.2022.937289] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Neurodegenerative disease is a progressive neurodegeneration caused by genetic and environmental factors. Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD) are the three most common neurodegenerative diseases clinically. Unfortunately, the incidence of neurodegenerative diseases is increasing year by year. However, the current available drugs have poor efficacy and large side effects, which brings a great burden to the patients and the society. Increasing evidence suggests that occurrence and development of the neurodegenerative diseases is closely related to the mitochondrial dysfunction, which can affect mitochondrial biogenesis, mitochondrial dynamics, as well as mitochondrial mitophagy. Through the disruption of mitochondrial homeostasis, nerve cells undergo varying degrees of apoptosis. Interestingly, it has been shown in recent years that the natural agents derived from herbal medicines are beneficial for prevention/treatment of neurodegenerative diseases via regulation of mitochondrial dysfunction. Therefore, in this review, we will focus on the potential therapeutic agents from herbal medicines for treating neurodegenerative diseases via suppressing apoptosis through regulation of mitochondrial dysfunction, in order to provide a foundation for the development of more candidate drugs for neurodegenerative diseases from herbal medicine.
Collapse
Affiliation(s)
- Ruo-Lan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling-Yu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hu-Xinyue Duan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohui Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaohui Guo, ; Chunjie Wu, ; Wei Peng,
| | - Chunjie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaohui Guo, ; Chunjie Wu, ; Wei Peng,
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaohui Guo, ; Chunjie Wu, ; Wei Peng,
| |
Collapse
|
13
|
Wu Q, Li X, Jiang XW, Yao D, Zhou LJ, Xu ZH, Wang N, Zhao QC, Zhang Z. Yuan-Zhi decoction in the treatment of Alzheimer’s disease: An integrated approach based on chemical profiling, network pharmacology, molecular docking and experimental evaluation. Front Pharmacol 2022; 13:893244. [PMID: 36091836 PMCID: PMC9451491 DOI: 10.3389/fphar.2022.893244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
Yuan-Zhi Decoction (YZD) is a traditional Chinese medical formulation with demonstrated clinical benefits in Alzheimer’s disease (AD). We used liquid chromatography coupled with mass spectrometry to identify 27 unique chemical components of YZD. Analyzing these using network pharmacology and molecular docking models identified 34 potential interacting molecular targets involved in 26 biochemical pathways. When tested in an animal model of AD, the APP/PS1 transgenic mice showed measurable improvements in spatial orientation and memory after the administration of YZD. These improvements coincided with significantly reduced deposition of Aβ plaques and tau protein in the hippocampi in the treated animals. In addition, a decreased BACE1 and beta-amyloid levels, a downregulation of the p-GSK-3β/GSK-3β, and an upregulation of the PI3K and p-AKT/AKT pathway was seen in YZD treated animals. These in vivo changes validated the involvement of molecular targets and pathways predicted in silico analysis of the chemical components of YZD. This study provides scientific support for the clinical use of YZD and justifies further investigations into its effects in AD. Furthermore, it demonstrates the utility of network pharmacology in elucidating the biochemical mechanisms underlying the beneficial effects of traditional Chinese medicines (TCM).
Collapse
Affiliation(s)
- Qiong Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiao-Wen Jiang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Dong Yao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Li-Jun Zhou
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Zi-Hua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Nan Wang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Qing-Chun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Zhou Zhang, ; Qing-Chun Zhao,
| | - Zhou Zhang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
- *Correspondence: Zhou Zhang, ; Qing-Chun Zhao,
| |
Collapse
|
14
|
New Therapeutic Approaches to and Mechanisms of Ginsenoside Rg1 against Neurological Diseases. Cells 2022; 11:cells11162529. [PMID: 36010610 PMCID: PMC9406801 DOI: 10.3390/cells11162529] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Neurological diseases, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), stroke, cerebral infarction, ischemia-reperfusion injury, depression and, stress, have high incidence and morbidity and often lead to disability. However, there is no particularly effective medication against them. Therefore, finding drugs with a suitable efficacy, low toxicity and manageable effects to improve the quality of life of patients is an urgent problem. Ginsenoside Rg1 (Rg1) is the main active component of ginseng and has a variety of pharmacological effects. In this review, we focused on the therapeutic potential of Rg1 for improving neurological diseases. We introduce the mechanisms of Ginsenoside Rg1 in neurological diseases, including apoptosis, neuroinflammation, the microRNA (miRNA) family, the mitogen-activated protein kinase (MAPK) family, oxidative stress, nuclear factor-κB (NF-κB), and learning and memory of Rg1 in neurological diseases. In addition, Rg1 can also improve neurological diseases through the interaction of different signal pathways. The purpose of this review is to explore more in-depth ideas for the clinical treatment of neurological diseases (including PD, AD, HD, stroke, cerebral infarction, ischemia–reperfusion injury, depression, and stress). Therefore, Rg1 is expected to become a new therapeutic method for the clinical treatment of neurological diseases.
Collapse
|
15
|
Sahebnasagh A, Eghbali S, Saghafi F, Sureda A, Avan R. Neurohormetic phytochemicals in the pathogenesis of neurodegenerative diseases. Immun Ageing 2022; 19:36. [PMID: 35953850 PMCID: PMC9367062 DOI: 10.1186/s12979-022-00292-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/24/2022] [Indexed: 12/02/2022]
Abstract
The world population is progressively ageing, assuming an enormous social and health challenge. As the world ages, neurodegenerative diseases are on the rise. Regarding the progressive nature of these diseases, none of the neurodegenerative diseases are curable at date, and the existing treatments can only help relieve the symptoms or slow the progression. Recently, hormesis has increased attention in the treatment of age-related neurodegenerative diseases. The concept of hormesis refers to a biphasic dose-response phenomenon, where low levels of the drug or stress exert protective of beneficial effects and high doses deleterious or toxic effects. Neurohormesis, as the adaptive aspect of hormetic dose responses in neurons, has been shown to slow the onset of neurodegenerative diseases and reduce the damages caused by aging, stroke, and traumatic brain injury. Hormesis was also observed to modulate anxiety, stress, pain, and the severity of seizure. Thus, neurohormesis can be considered as a potentially innovative approach in the treatment of neurodegenerative and other neurologic disorders. Herbal medicinal products and supplements are often considered health resources with many applications. The hormesis phenomenon in medicinal plants is valuable and several studies have shown that hormetic mechanisms of bioactive compounds can prevent or ameliorate the neurodegenerative pathogenesis in animal models of Alzheimer’s and Parkinson’s diseases. Moreover, the hormesis activity of phytochemicals has been evaluated in other neurological disorders such as Autism and Huntington’s disease. In this review, the neurohormetic dose–response concept and the possible underlying neuroprotection mechanisms are discussed. Different neurohormetic phytochemicals used for the better management of neurodegenerative diseases, the rationale for using them, and the key findings of their studies are also reviewed.
Collapse
|
16
|
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
|
17
|
HDAC4 Inhibitors as Antivascular Senescence Therapeutics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3087916. [PMID: 35814270 PMCID: PMC9259336 DOI: 10.1155/2022/3087916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022]
Abstract
Aging is an inevitable consequence of life, and during this process, the epigenetic landscape changes and reactive oxygen species (ROS) accumulation increases. Inevitably, these changes are common in many age-related diseases, including neurodegeneration, hypertension, and cardiovascular diseases. In the current research, histone deacetylation 4 (HDAC4) was studied as a potential therapeutic target in vascular senescence. HDAC4 is a specific class II histone deacetylation protein that participates in epigenetic modifications and deacetylation of heat shock proteins and various transcription factors. There is increasing evidence to support that HDAC4 is a potential therapeutic target, and developments in the synthesis and testing of HDAC4 inhibitors are now gaining interest from academia and the pharmaceutical industry.
Collapse
|
18
|
Transcriptional Profiling of Hippocampus Identifies Network Alterations in Alzheimer’s Disease. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12105035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by rapid brain cell degeneration affecting different areas of the brain. Hippocampus is one of the earliest involved brain regions in the disease. Modern technologies based on high-throughput data have identified transcriptional profiling of several neurological diseases, including AD, for a better comprehension of genetic mechanisms of the disease. In this study, we investigated differentially expressed genes (DEGs) from six Gene Expression Omnibus (GEO) datasets of hippocampus of AD patients. The identified DEGs were submitted to Weighted correlation network analysis (WGCNA) and ClueGo to explore genes with a higher degree centrality and to comprehend their biological role. Subsequently, MCODE was used to identify subnetworks of interconnected DEGs. Our study found 40 down-regulated genes and 36 up-regulated genes as consensus DEGs. Analysis of the co-expression network revealed ACOT7, ATP8A2, CDC42, GAD1, GOT1, INA, NCALD, and WWTR1 to be genes with a higher degree centrality. ClueGO revealed the pathways that were mainly enriched, such as clathrin coat assembly, synaptic vesicle endocytosis, and DNA damage response signal transduction by p53 class mediator. In addition, we found a subnetwork of 12 interconnected genes (AMPH, CA10, CALY, NEFL, SNAP25, SNAP91, SNCB, STMN2, SV2B, SYN2, SYT1, and SYT13). Only CA10 and CALY are targets of known drugs while the others could be potential novel drug targets.
Collapse
|
19
|
Network Pharmacology and Molecular Docking-Based Strategy to Investigate the Multitarget Mechanisms of Shenqi Yizhi Granule on Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8032036. [PMID: 35535155 PMCID: PMC9078761 DOI: 10.1155/2022/8032036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/13/2022] [Indexed: 01/28/2023]
Abstract
Background Traditional Chinese herbal medicine draws more attention to explore an effective therapeutic strategy for Alzheimer's disease (AD). Shenqi Yizhi granule (SQYG), a Chinese herbal recipe, has been applied to ameliorate cognitive impairment in mild-to-moderate AD patients. However, the overall molecular mechanism of SQYG in treating AD has not been clarified. Objective This study aimed to investigate the molecular mechanism of SQYG on AD using an integration strategy of network pharmacology and molecular docking. Methods The active compounds of SQYG and common targets between SQYG and AD were screened from databases. The herb-compound network, compound-target network, and protein-protein interaction network were constructed. The enrichment analysis of common targets and molecular docking were performed. Results 816 compounds and 307 common targets between SQYG and AD were screened. KEGG analysis revealed that common targets were mainly enriched in lipid metabolism, metal ion metabolism, IL-17 signaling pathway, GABA receptor signaling, and neuroactive ligand-receptor interaction. Molecular docking analysis showed high binding affinity between ginsenoside Rg1 and Aβ 1-42, tanshinone IIA and BACE1, baicalin, and AchE. Conclusions The therapeutic mechanisms of SQYG on AD were associated with regulating lipid metabolism, metal ion metabolism, IL-17 signaling pathway, and GABA receptor signaling. Ginsenoside Rg1, tanshinone IIA, baicalin, astragaloside IV, and folic acid may play an important role in AD treatment.
Collapse
|
20
|
Peng Q, Wang L, Wang S, Wang C, Xue Z. MicoRNA-214-3p: a key player in CPLX2-mediated inhibition on temozolomide resistance in glioma. Neurol Res 2022; 44:879-887. [DOI: 10.1080/01616412.2022.2064699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Qian Peng
- Medical Department Hunan Provincial People’s Hospital, (the First Affiliated Hospital of Hunan Normal University), Changsha P.R. China
| | - Lijiao Wang
- Department of Nosocomial Infection, Hunan Provincial People’s Hospital, (the First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Shuling Wang
- Institute of Geriatrics, Hunan Provincial People’s Hospital, (the First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Chenxu Wang
- Institute of Geriatrics, Hunan Provincial People’s Hospital, (the First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Zhi Xue
- Department of Neurosurgery, Hunan Provincial People’s Hospital, (the First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| |
Collapse
|
21
|
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
|
22
|
Sng KS, Li G, Zhou LY, Song YJ, Chen XQ, Wang YJ, Yao M, Cui XJ. Ginseng extract and ginsenosides improve neurological function and promote antioxidant effects in rats with spinal cord injury: A meta-analysis and systematic review. J Ginseng Res 2022; 46:11-22. [PMID: 35058723 PMCID: PMC8753526 DOI: 10.1016/j.jgr.2021.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/26/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is defined as damage to the spinal cord that temporarily or permanently changes its function. There is no definite treatment established for neurological complete injury patients. This study investigated the effect of ginseng extract and ginsenosides on neurological recovery and antioxidant efficacies in rat models following SCI and explore the appropriate dosage. Searches were done on PubMed, Embase, and Chinese databases, and animal studies matches the inclusion criteria were selected. Pair-wise meta-analysis and subgroup analysis were performed. Ten studies were included, and the overall methodological qualities were low quality. The result showed ginseng extract and ginsenosides significantly improve neurological function, through the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale (pooled MD = 4.40; 95% CI = 3.92 to 4.88; p < 0.00001), significantly decrease malondialdehyde (MDA) (n = 290; pooled MD = −2.19; 95% CI = −3.16 to −1.22; p < 0.0001) and increase superoxide dismutase (SOD) levels (n = 290; pooled MD = 2.14; 95% CI = 1.45 to 2.83; p < 0.00001). Both low (<25 mg/kg) and high dosage (≥25 mg/kg) showed significant improvement in the motor function recovery in SCI rats. Collectively, this review suggests ginseng extract and ginsenosides has a protective effect on SCI, with good safety and a clear mechanism of action and may be suitable for future clinical trials and applications.
Collapse
|
23
|
Li L, Li T, Tian X, Zhao L. Ginsenoside Rd Attenuates Tau Phosphorylation in Olfactory Bulb, Spinal Cord, and Telencephalon by Regulating Glycogen Synthase Kinase 3 β and Cyclin-Dependent Kinase 5. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:4485957. [PMID: 34987593 PMCID: PMC8720614 DOI: 10.1155/2021/4485957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Ginseng is a plant of the family Acanthopanaceae. It has been used for thousands of years in China. It is known as the king of hundred herbs. It was recorded first in Shennong Baicao Jing. It has been found that ginsenoside Rd is a neuroprotective agent. This article aims to explore the protective roles of ginsenoside Rd in Alzheimer's disease. Rd, a Chinese herb, may be a promising treatment drug for Alzheimer's disease (AD) and is also reported to be related to several pathological changes, including the deposition of Aβ and tau hyperphosphorylation in AD as it decreases the deposition of tau hyperphosphorylation in APP transgenic mice. METHODS In this study, APP transgenic mice were pretreated with 10 mg/kg Rd for six months, and the effect of Rd on neuropathological deficits in the olfactory bulb, spinal cord, and telencephalon of APP transgenic mice was investigated. The phosphorylation levels of tau (S199/202, S396, S404, and Tau5) and the activities of the proteins glycogen synthase kinase 3β (Tyr216) and cyclin-dependent kinase 5 (P25/P35) were measured. RESULTS The pretreatment of Rd effectively decreased the production and deposition of hyperphosphorylated tau (S199/202, S396, and S404) protein by depressing the expression of glycogen synthase kinase 3β (GSK-3β/Tyr216) and cyclin-dependent kinase 5 (CDK5/P25). CONCLUSION These findings suggest that ginsenoside Rd could improve the pathological changes of AD in the olfactory bulb, spinal cord, and telencephalon, which further demonstrated the potential therapeutic effect of Rd in early AD.
Collapse
Affiliation(s)
- Ling Li
- Department of Geriatrics, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an 710003, China
| | - Tian Li
- Department of Geriatrics, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an 710003, China
| | - Xin Tian
- Department of Cardiology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an 710003, China
| | - Ling Zhao
- Department of Neurology, Xinchang Hospital Affiliated to Wenzhou Medical University, 117 Gushan Middle Rd, Xinchang 312500, China
| |
Collapse
|
24
|
Nie L, He K, Xie F, Xiao S, Li S, Xu J, Zhang K, Yang C, Zhou L, Liu J, Zou L, Yang X. Loganin substantially ameliorates molecular deficits, pathologies and cognitive impairment in a mouse model of Alzheimer's disease. Aging (Albany NY) 2021; 13:23739-23756. [PMID: 34689137 PMCID: PMC8580356 DOI: 10.18632/aging.203646] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disease threatening the health of the elderly, but the available therapeutic and preventive drugs remain suboptimal. Loganin, an iridoid glycoside extracted from Cornus officinalis, is reported to have anti-inflammatory and memory-enhancing properties. This study is aimed to explore the influence of loganin on cognitive function in 3xTg-AD mice and the underlying mechanism associated with its neuroprotection. According to the results of behavioral tests, we found that administration of loganin could significantly alleviate anxiety behavior and improve memory deficits of 3xTg-AD mice. Furthermore, immunohistochemical analysis displayed that there were decreased Aβ deposition in the hippocampus and cortex of 3xTg-AD mice treated with loganin compared with the control mice. Importantly, the Aβ-related pathological change was mainly involved in altering APP expression and processing. And loganin was also found to reduce the levels of phosphorylated tau (i.e. pTauS396 and pTauS262) in 3xTg-AD mice. By performing 2D-DIGE combined with MALDI-TOF-MS/MS, we revealed 28 differentially expressed proteins in the 3xTg-AD mice treated with loganin compared with the control mice. Notably, 10 proteins largely involved in energy metabolism, synaptic proteins, inflammatory response, and ATP binding were simultaneously detected in 3xTg-AD mice compared to WT mice. The abnormal changes of energy metabolism (PAGM1 and ENO1), synaptic proteins (SYN2 and Cplx2), inflammatory response (1433Z) were verified by western blot. Overall, our study suggested that loganin could be used as a feasible candidate drug to ameliorate molecular deficits, pathologies and cognitive impairment for prevention and treatment of AD.
Collapse
Affiliation(s)
- Lulin Nie
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Kaiwu He
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.,School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Fengzhu Xie
- Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, China
| | - Shifeng Xiao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Jia Xu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.,Department of Pathophysiology, Guangzhou Medical University, Guangzhou 510182, China
| | - Kaiqin Zhang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.,College of Public Health, University of South China, Hengyang 421001, China
| | - Chen Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Li Zhou
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jianjun Liu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Liangyu Zou
- Department of Neurology, Shenzhen People's Hospital, Second Clinical College, Jinan University, Shenzhen 518020, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
25
|
Kron NS, Fieber LA. Aplysia Neurons as a Model of Alzheimer's Disease: Shared Genes and Differential Expression. J Mol Neurosci 2021; 72:287-302. [PMID: 34664226 PMCID: PMC8840921 DOI: 10.1007/s12031-021-01918-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/18/2021] [Indexed: 11/19/2022]
Abstract
Although Alzheimer’s disease (AD) is the most common form of dementia in the United States, development of therapeutics has proven difficult. Invertebrate alternatives to current mammalian AD models have been successfully employed to study the etiology of the molecular hallmarks of AD. The marine snail Aplysia californica offers a unique and underutilized system in which to study the physiological, behavioral, and molecular impacts of AD. Mapping of the Aplysia proteome to humans and cross-referencing with two databases of genes of interest in AD research identified 898 potential orthologs of interest in Aplysia. Included among these orthologs were alpha, beta and gamma secretases, amyloid-beta, and tau. Comparison of age-associated differential expression in Aplysia sensory neurons with that of late-onset AD in the frontal lobe identified 59 ortholog with concordant differential expression across data sets. The 21 concordantly upregulated genes suggested increased cellular stress and protein dyshomeostasis. The 47 concordantly downregulated genes included important components of diverse neuronal processes, including energy metabolism, mitochondrial homeostasis, synaptic signaling, Ca++ regulation, and cellular cargo transport. Compromised functions in these processes are known hallmarks of both human aging and AD, the ramifications of which are suggested to underpin cognitive declines in aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Nicholas S Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA.
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| |
Collapse
|
26
|
Hu Y, Wu L, Jiang L, Liang N, Zhu X, He Q, Qin H, Chen W. Notoginsenoside R2 reduces A β25-35-induced neuronal apoptosis and inflammation via miR-27a/SOX8/ β-catenin axis. Hum Exp Toxicol 2021; 40:S347-S358. [PMID: 34533063 DOI: 10.1177/09603271211041996] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Alzheimer's disease (AD) has affected numerous elderly individuals worldwide. Panax notoginseng has been shown to ameliorate AD symptoms, and notoginsenoside R2 is a key saponin identified in this plant. Purpose: In the current study, we aimed to explore whether notoginsenoside R2 could improve the prognosis of AD. Methods: Herein, primary rat cortical neurons were isolated and they were treated with amyloid beta-peptide (Aβ) 25-35 oligomers. Cellular apoptosis was examined via flow cytometry and Western blotting. miR-27a and SOX8 mRNA expression levels were quantified by quantitative reverse transcription-polymerase chain reaction. Furthermore, the interaction between miR-27a and SOX8 was investigated by utilizing a dual-luciferase reporter assay. Finally, an AD mouse model was established to validate the in vitro findings. Results: Notoginsenoside R2 alleviated Aβ25-35-triggered neuronal apoptosis and inflammation. During this process, miR-27a expression was decreased by notoginsenoside R2, and miR-27a negatively modulated SOX8 expression. Furthermore, activation of SOX8 upregulated β-catenin expression, thus suppressing apoptosis and neuroinflammation. Conclusions: Our animal experiments revealed that notoginsenoside R2 enhanced the cognitive function of AD mice and inhibited neuronal apoptosis. Notoginsenoside R2 ameliorated AD symptoms by reducing neuronal apoptosis and inflammation, thus suggesting a novel direction for AD pharmacotherapy.
Collapse
Affiliation(s)
- Yueqiang Hu
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Wu
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, China.,Scientific Laboratorial Centre, Guangxi University of Chinese Medicine, Nanning, China
| | - Lingfei Jiang
- Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Ni Liang
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaomin Zhu
- Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Qianchao He
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Hongling Qin
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Wei Chen
- Department of Neurology, 118330The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
27
|
Yang X, Chu SF, Wang ZZ, Li FF, Yuan YH, Chen NH. Ginsenoside Rg1 exerts neuroprotective effects in 3-nitropronpionic acid-induced mouse model of Huntington's disease via suppressing MAPKs and NF-κB pathways in the striatum. Acta Pharmacol Sin 2021; 42:1409-1421. [PMID: 33214696 PMCID: PMC8379213 DOI: 10.1038/s41401-020-00558-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022]
Abstract
Huntington's disease (HD) is one of main neurodegenerative diseases, characterized by striatal atrophy, involuntary movements, and motor incoordination. Ginsenoside Rg1 (Rg1), an active ingredient in ginseng, possesses a variety of neuroprotective effects with low toxicity and side effects. In this study, we investigated the potential therapeutic effects of Rg1 in a mouse model of HD and explored the underlying mechanisms. HD was induced in mice by injection of 3-nitropropionic acid (3-NP, i.p.) for 4 days. From the first day of 3-NP injection, the mice were administered Rg1 (10, 20, 40 mg·kg-1, p.o.) for 5 days. We showed that oral pretreatment with Rg1 alleviated 3-NP-induced body weight loss and behavioral defects. Furthermore, pretreatment with Rg1 ameliorated 3-NP-induced neuronal loss and ultrastructural morphological damage in the striatum. Moreover, pretreatment with Rg1 reduced 3-NP-induced apoptosis and inhibited the activation of microglia, inflammatory mediators in the striatum. We revealed that Rg1 exerted neuroprotective effects by suppressing 3-NP-induced activation of the MAPKs and NF-κΒ signaling pathways in the striatum. Thus, our results suggest that Rg1 exerts therapeutic effects on 3-NP-induced HD mouse model via suppressing MAPKs and NF-κΒ signaling pathways. Rg1 may be served as a novel therapeutic option for HD.
Collapse
|
28
|
Li J, Huang Q, Chen J, Qi H, Liu J, Chen Z, Zhao D, Wang Z, Li X. Neuroprotective Potentials of Panax Ginseng Against Alzheimer's Disease: A Review of Preclinical and Clinical Evidences. Front Pharmacol 2021; 12:688490. [PMID: 34149431 PMCID: PMC8206566 DOI: 10.3389/fphar.2021.688490] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is a major health concern in the increasingly aged population worldwide. Currently, no clinically effective drug can halt the progression of AD. Panax ginseng C.A. Mey. is a well-known medicinal plant that contains ginsenosides, gintonin, and other components and has neuroprotective effects against a series of pathological cascades in AD, including beta-amyloid formation, neuroinflammation, oxidative stress, and mitochondrial dysfunction. In this review, we summarize the effects and mechanisms of these major components and formulas containing P. ginseng in neuronal cells and animal models. Moreover, clinical findings regarding the prevention and treatment of AD with P. ginseng or its formulas are discussed. This review can provide new insights into the possible use of ginseng in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Jing Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Qingxia Huang
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jinjin Chen
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Qi
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jiaqi Liu
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zhaoqiang Chen
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
29
|
Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD, Gao LC. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer's Disease and Parkinson's Disease. Front Pharmacol 2021; 12:648636. [PMID: 33935751 PMCID: PMC8082498 DOI: 10.3389/fphar.2021.648636] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two typical neurodegenerative diseases that increased with aging. With the emergence of aging population, the health problem and economic burden caused by the two diseases also increase. Phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) signaling pathway regulates signal transduction and biological processes such as cell proliferation, apoptosis and metabolism. According to reports, it regulates neurotoxicity and mediates the survival of neurons through different substrates such as forkhead box protein Os (FoxOs), glycogen synthase kinase-3β (GSK-3β), and caspase-9. Accumulating evidences indicate that some natural products can play a neuroprotective role by activating PI3K/AKT pathway, providing an effective resource for the discovery of potential therapeutic drugs. This article reviews the relationship between AKT signaling pathway and AD and PD, and discusses the potential natural products based on the PI3K/AKT signaling pathway to treat two diseases in recent years, hoping to provide guidance and reference for this field. Further development of Chinese herbal medicine is needed to treat these two diseases.
Collapse
Affiliation(s)
- Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
30
|
Zhang Z, Song Z, Shen F, Xie P, Wang J, Zhu AS, Zhu G. Ginsenoside Rg1 Prevents PTSD-Like Behaviors in Mice Through Promoting Synaptic Proteins, Reducing Kir4.1 and TNF-α in the Hippocampus. Mol Neurobiol 2021; 58:1550-1563. [PMID: 33215390 PMCID: PMC7676862 DOI: 10.1007/s12035-020-02213-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022]
Abstract
Ginsenoside Rg1 is efficient to prevent or treat mental disorders. However, the mechanisms underlying the effects of ginsenoside Rg1 on post-traumatic stress disorder (PTSD) are still not known. In this study, single-prolonged stress (SPS) regime, as well as injection of lipopolysaccharide (LPS), was used to produce PTSD-like behaviors in C57 mice, and the effects of ginsenoside Rg1 (10, 20, 40 mg/kg/d, ip, for 14 days) on PTSD-like behaviors were evaluated. Our results showed that ginsenoside Rg1 promoted fear extinction and prevented depression-like behaviors in both LPS and SPS models. Importantly, ginsenoside Rg1 alleviated LPS- or SPS-stimulated expression of pro-inflammatory cytokines (IL-1β and TNF-α), activation of astrocytes and microglia, and reduction of hippocampal synaptic proteins (PSD95, Arc, and GluA1). Ginsenoside Rg1 also reduced the increase of hippocampal Kir4.1 and GluN2A induced by PTSD regime. Importantly, reducing hippocampal astroglial Kir4.1 expression promoted fear extinction and improved depression-like behaviors in LPS-treated mice. Additionally, intracerebroventricular injection of TNF-α caused an impairment of fear extinction and promoted Kir4.1 expression in the hippocampus. Together, our study reveals novel protective effects of ginsenoside Rg1 against PTSD-like behaviors in mice, likely via promoting synaptic proteins, reducing Kir4.1 and TNF-α in the hippocampus.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road 103, Hefei, 230038, China
| | - Zhujin Song
- Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Fengming Shen
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road 103, Hefei, 230038, China
| | - Pan Xie
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road 103, Hefei, 230038, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road 103, Hefei, 230038, China
| | - Ai-Song Zhu
- Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road 103, Hefei, 230038, China.
| |
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
|
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
|
33
|
Lye S, Aust CE, Griffiths LR, Fernandez F. Exploring new avenues for modifying course of progression of Alzheimer's disease: The rise of natural medicine. J Neurol Sci 2021; 422:117332. [PMID: 33607542 DOI: 10.1016/j.jns.2021.117332] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/01/2022]
Abstract
With a constantly growing elderly population worldwide, a focus on developing efficient prevention and therapy for Alzheimer's disease (AD) seems timely and topical. Emphasis on natural medicine is increasingly popular in the search for drug candidates that are capable of preventing and treating AD related pathology, particularly where suppression of amyloid accumulation, neurofibrillary tangle formation, neuroinflammation and oxidative stress are equally significant. A number of phytochemical compounds have been shown to collectively reduce these AD hallmarks with the progression of natural drug candidates into human clinical trials. This review focuses on current research surrounding the therapies emerging within natural medicines and their related therapeutic potential for AD treatment.
Collapse
Affiliation(s)
- Sarah Lye
- School of Health and Behavioural Science, Faculty of Health Sciences, 1100 Nudgee Road, Australian Catholic University, Brisbane, QLD, Australia
| | - Caitlin E Aust
- School of Health and Behavioural Science, Faculty of Health Sciences, 1100 Nudgee Road, Australian Catholic University, Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Lyn R Griffiths
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Francesca Fernandez
- School of Health and Behavioural Science, Faculty of Health Sciences, 1100 Nudgee Road, Australian Catholic University, Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.
| |
Collapse
|
34
|
Ren J, Wei D, An H, Zhang J, Zhang Z. Shenqi Yizhi granules protect hippocampus of AD transgenic mice by modulating on multiple pathological processes. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:112869. [PMID: 32315734 DOI: 10.1016/j.jep.2020.112869] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine (CHM) draws more attention to explore effective therapeutic strategy for Alzheimer's disease (AD). CHM usually uses combinations of herbs or herbal ingredients to treat diseases, with the components targeting different disease processes. CHM might improve cognition in AD and MCI patients by optimizing network activity, promoting neural plasticity and repairing damaged neurons. Shenqi Yizhi granules (SQYG), a CHM prescription, are mainly consists of Panax ginseng C.A.Mey, Astragalus membranaceus (Fisch.) Bunge, and Scutellaria baicalensis Georgi and have been used to ameliorate cognitive impairment in mild-to-moderate dementia patients. AIM OF THE STUDY To investigate the neuroprotection effect and pharmacological mechanism of SQYG in the hippocampus of 5XFAD transgenic mice. MATERIALS AND METHODS The immunofluorescence detection, 2DE-gels, mass spectrum identification, biological information analysis and Western blot were performed after SQYG treatment. RESULTS SQYG treatment significantly decreased the fluorescence intensities of anti-GFAP and anti-Iba1 in the hippocampus of 5XFAD mice. The expression levels of 31 proteins in the hippocampus were significantly influenced by SQYG, approximately 65% of these proteins are related to energy metabolism, stress response and cytoskeleton, whereas others are related to synaptic transmission, signal transduction, antioxidation, amino acid metabolism, and DNA repair. The expression of these proteins were increased. The changes in the expression levels of malate dehydrogenase (cytoplasmic) and pyruvate kinase M were confirmed by Western blot. CONCLUSIONS The pharmacological mechanism of SQYG on the hippocampus may be related to modulation of multiple pathological processes, including energy metabolism, stress response, cytoskeleton, synaptic transmission, signal transduction, and amino acid metabolism in 5XFAD mice.
Collapse
Affiliation(s)
- Jianting Ren
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China; BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Dongfeng Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haiting An
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China; BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Junying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China; BABRI Centre, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
35
|
Shen Y, Hua L, Yeh CK, Shen L, Ying M, Zhang Z, Liu G, Li S, Chen S, Chen X, Yang X. Ultrasound with microbubbles improves memory, ameliorates pathology and modulates hippocampal proteomic changes in a triple transgenic mouse model of Alzheimer's disease. Am J Cancer Res 2020; 10:11794-11819. [PMID: 33052247 PMCID: PMC7546002 DOI: 10.7150/thno.44152] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease manifested by cognitive impairment. As a unique approach to open the blood-brain barrier (BBB) noninvasively and temporarily, a growing number of studies showed that low-intensity focused ultrasound in combination with microbubbles (FUS/MB), in the absence of therapeutic agents, is capable of ameliorating amyloid or tau pathology, concurrent with improving memory deficits of AD animal models. However, the effects of FUS/MB on both the two pathologies simultaneously, as well as the memory behaviors, have not been reported so far. Methods: In this study, female triple transgenic AD (3×Tg-AD) mice at eight months of age with both amyloid-β (Aβ) deposits and tau phosphorylation were treated by repeated FUS/MB in the unilateral hippocampus twice per week for six weeks. The memory behaviors were investigated by the Y maze, the Morris water maze and the step-down passive avoidance test following repeated FUS/MB treatments. Afterwards, the involvement of Aβ and tau pathology were assessed by immunohistochemical analysis. Neuronal health and phagocytosis of Aβ deposits by microglia in the hippocampus were examined by confocal microscopy. Further, hippocampal proteomic alterations were analyzed by employing two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with mass spectrometry. Results: The three independent memory tasks were indicative of evident learning and memory impairments in eight-month-old 3×Tg-AD mice, which developed intraneuronal Aβ, extracellular diffuse Aβ deposits and phosphorylated tau in the hippocampus and amygdala. Following repeated FUS/MB treatments, significant improvement in learning and memory ability of the 3×Tg-AD mice was achieved. Amelioration in both Aβ deposits and phosphorylated tau in the sonicated hemisphere was induced in FUS/MB-treated 3×Tg-AD mice. Albeit without increase in neuron density, enhancement in axonal neurofilaments emerged from the FUS/MB treatment. Confocal microscopy revealed activated microglia engulfing Aβ deposits in the FUS/MB-treated hippocampus. Further, proteomic analysis revealed 20 differentially expressed proteins, associated with glycolysis, neuron projection, mitochondrial pathways, metabolic process and ubiquitin binding etc., in the hippocampus between FUS/MB-treated and sham-treated 3×Tg-AD mice. Conclusions: Our findings reinforce the positive therapeutic effects on AD models with both Aβ and tau pathology induced by FUS/MB-mediated BBB opening, further supporting the potential of this treatment regime for clinical applications.
Collapse
|
36
|
Quan Q, Li X, Feng J, Hou J, Li M, Zhang B. Ginsenoside Rg1 reduces β‑amyloid levels by inhibiting CDΚ5‑induced PPARγ phosphorylation in a neuron model of Alzheimer's disease. Mol Med Rep 2020; 22:3277-3288. [PMID: 32945455 PMCID: PMC7453505 DOI: 10.3892/mmr.2020.11424] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/16/2020] [Indexed: 12/25/2022] Open
Abstract
The accumulation of β-amyloid peptides (Aβ) in the brain is a hallmark of Alzheimer's disease (AD). Studies have indicated that ginsenoside Rg1, a primary component of ginseng (Panax ginseng), reduces brain Aβ levels in an AD model through peroxisome proliferator-activated receptor γ (PPARγ), thereby regulating the expression of insulin-degrading enzyme (Ide) and β-amyloid cleavage enzyme 1 (Bace1), which are PPARγ target genes. However, the effects of ginsenoside Rg1 on PPARγ remain unclear. Since cyclin-dependent kinase 5 (CDK5) mediates PPARγ phosphorylation in adipose tissue, this study aimed to investigate whether ginsenoside Rg1 regulates PPARγ target genes and reduces Aβ levels by inhibiting PPARγ phosphorylation through the CDK5 pathway. In the present study, a model of AD was established by treating primary cultured rat hippocampal neurons with Aβ1-42. The cells were pretreatment with ginsenoside Rg1 and roscovitine, a CDK5-inhibitor, prior to the treatment with Aβ1-42. Neuronal apoptosis was detected using TUNEL staining. PPARγ phosphorylation and protein expression levels of PPARγ, CDK5, IDE, BACE1, amyloid precursor protein (APP) and Aβ1-42 were measured by western blotting. The mRNA expression levels of PPARγ, CDK5, IDE, BACE1 and APP were assessed using reverse transcription-quantitative PCR. The results of the present study demonstrated that in an AD model induced by Aβ1-42, ginsenoside Rg1 significantly decreased CDK5 expression, inhibited PPARγ phosphorylation at serine 273, elevated IDE expression, downregulated BACE1 and APP expression, decreased Aβ1-42 levels and attenuated neuronal apoptosis. The CDK5 inhibitor, roscovitine, demonstrated similar effects. These results suggest that ginsenoside Rg1 has neuroprotective properties and has potential for use in the treatment of AD.
Collapse
Affiliation(s)
- Qiankun Quan
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jianjun Feng
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jixing Hou
- Department of Psychiatry, Xi'an Mental Health Center, Xi'an, Shaanxi 710061, P.R. China
| | - Ming Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bingwei Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
37
|
Lin K, Sze SCW, Liu B, Zhang Z, Zhang Z, Zhu P, Wang Y, Deng Q, Yung KKL, Zhang S. 20( S)-protopanaxadiol and oleanolic acid ameliorate cognitive deficits in APP/PS1 transgenic mice by enhancing hippocampal neurogenesis. J Ginseng Res 2020; 45:325-333. [PMID: 33841013 PMCID: PMC8020272 DOI: 10.1016/j.jgr.2020.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders. Enhancing hippocampal neurogenesis by promoting proliferation and differentiation of neural stem cells (NSCs) is a promising therapeutic strategy for AD. 20(S)-protopanaxadiol (PPD) and oleanolic acid (OA) are small, bioactive compounds found in ginseng that can promote NSC proliferation and neural differentiation in vitro. However, it is currently unknown whether PPD or OA can attenuate cognitive deficits by enhancing hippocampal neurogenesis in vivo in a transgenic APP/PS1 AD mouse model. Here, we administered PPD or OA to APP/PS1 mice and monitored the effects on cognition and hippocampal neurogenesis. Methods We used the Morris water maze, Y maze, and open field tests to compare the cognitive capacities of treated and untreated APP/PS1 mice. We investigated hippocampal neurogenesis using Nissl staining and BrdU/NeuN double labeling. NSC proliferation was quantified by Sox2 labeling of the hippocampal dentate gyrus. We used western blotting to determine the effects of PPD and OA on Wnt/GSK3β/β-catenin pathway activation in the hippocampus. Results Both PPD and OA significantly ameliorated the cognitive impairments observed in untreated APP/PS1 mice. Furthermore, PPD and OA significantly promoted hippocampal neurogenesis and NSC proliferation. At the mechanistic level, PPD and OA treatments resulted in Wnt/GSK-3β/β-catenin pathway activation in the hippocampus. Conclusion PPD and OA ameliorate cognitive deficits in APP/PS1 mice by enhancing hippocampal neurogenesis, achieved by stimulating the Wnt/GSK-3β/β-catenin pathway. As such, PPD and OA are promising novel therapeutic agents for the treatment of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Kaili Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, China.,Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Stephen Cho-Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhang Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Zhu Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Peili Zhu
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Ying Wang
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Qiudi Deng
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ken Kin-Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| | - Shiqing Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong Special Administrative Region (HKSAR), China.,HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Golden Meditech Center for NeuroRegeneration Sciences, HKBU, Kowloon Tong, HKSAR, China
| |
Collapse
|
38
|
Calabrese EJ. Hormesis and Ginseng: Ginseng Mixtures and Individual Constituents Commonly Display Hormesis Dose Responses, Especially for Neuroprotective Effects. Molecules 2020; 25:E2719. [PMID: 32545419 PMCID: PMC7321326 DOI: 10.3390/molecules25112719] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
This paper demonstrates that ginseng mixtures and individual ginseng chemical constituents commonly induce hormetic dose responses in numerous biological models for endpoints of biomedical and clinical relevance, typically providing a mechanistic framework. The principal focus of ginseng hormesis-related research has been directed toward enhancing neuroprotection against conditions such as Alzheimer's and Parkinson's Diseases, stroke damage, as well as enhancing spinal cord and peripheral neuronal damage repair and reducing pain. Ginseng was also shown to reduce symptoms of diabetes, prevent cardiovascular system damage, protect the kidney from toxicities due to immune suppressant drugs, and prevent corneal damage, amongst other examples. These findings complement similar hormetic-based chemoprotective reports for other widely used dietary-type supplements such as curcumin, ginkgo biloba, and green tea. These findings, which provide further support for the generality of the hormetic dose response in the biomedical literature, have potentially important public health and clinical implications.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
39
|
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
|
40
|
Zhong SJ, Wang L, Gu RZ, Zhang WH, Lan R, Qin XY. Ginsenoside Rg1 ameliorates the cognitive deficits in D-galactose and AlCl 3-induced aging mice by restoring FGF2-Akt and BDNF-TrkB signaling axis to inhibit apoptosis. Int J Med Sci 2020; 17:1048-1055. [PMID: 32410834 PMCID: PMC7211162 DOI: 10.7150/ijms.43979] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Ginsenoside Rg1 is the main active ingredient of Panax ginseng with the activity of neuroprotective, antioxidant and strengthening the immune system. Therefore, we hypothesized that Rg1 may afford anti-aging effects although the mechanism remains to be elucidated. In this study, chemically induced aging mice were established by consecutive administration of D-galactose and AlCl3. We found that Rg1 effectively ameliorates spatial learning and memory deficits in aging mice demonstrated by their improved performance in step down avoidance tests and Morris water maze experiments. Rg1 restored aging-induced decline of FGF2 and BDNF, reactivated TrkB/Akt signaling pathways in the hippocampus and prefrontal cortex to inhibit apoptosis, for the expression of anti-apoptotic protein Bcl-2 and apoptosis promoting enzyme cleaved-Caspase3 were antagonistically restored. Therefore, these results established the anti-aging effects of Rg1, and FGF2, BDNF and associated signaling pathways might be promising targets. Our data may provide a new avenue to the pharmacological research and diet therapeutic role of ethnic products such as Rg1 in anti-aging and aging associated diseases.
Collapse
Affiliation(s)
- Si-Jia Zhong
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China.,College of Economics and management, North China Electric Power University, Beijing 102206, China
| | - Lin Wang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Run-Ze Gu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Wen-Hao Zhang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Rongfeng Lan
- Department of Cell Biology & Medical Genetics, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xiao-Yan Qin
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| |
Collapse
|
41
|
Ning J, Jing-Wei L, Hai-Xia W, Hong H, Qiong W, Shan-Guang C, Li-Na Q, Dias ACP, Xin-Min L. Antidepressant-like Effects of Ginsenoside Rg1 in the Chronic Restraint Stress-induced Rat Model. DIGITAL CHINESE MEDICINE 2019. [DOI: 10.1016/j.dcmed.2020.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
42
|
Baranger K, van Gijsel-Bonnello M, Stephan D, Carpentier W, Rivera S, Khrestchatisky M, Gharib B, De Reggi M, Benech P. Long-Term Pantethine Treatment Counteracts Pathologic Gene Dysregulation and Decreases Alzheimer's Disease Pathogenesis in a Transgenic Mouse Model. Neurotherapeutics 2019; 16:1237-1254. [PMID: 31267473 PMCID: PMC6985318 DOI: 10.1007/s13311-019-00754-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The low-molecular weight thiol pantethine, known as a hypolipidemic and hypocholesterolemic agent, is the major precursor of co-enzyme A. We have previously shown that pantethine treatment reduces amyloid-β (Aβ)-induced IL-1β release and alleviates pathological metabolic changes in primary astrocyte cultures. These properties of pantethine prompted us to investigate its potential benefits in vivo in the 5XFAD (Tg) mouse model of Alzheimer's disease (AD).1.5-month-old Tg and wild-type (WT) male mice were submitted to intraperitoneal administration of pantethine or saline control solution for 5.5 months. The effects of such treatments were investigated by performing behavioral tests and evaluating astrogliosis, microgliosis, Αβ deposition, and whole genome expression arrays, using RNAs extracted from the mice hippocampi. We observed that long-term pantethine treatment significantly reduced glial reactivity and Αβ deposition, and abrogated behavioral alteration in Tg mice. Moreover, the transcriptomic profiles revealed that after pantethine treatment, the expression of genes differentially expressed in Tg mice, and in particular those known to be related to AD, were significantly alleviated. Most of the genes overexpressed in Tg compared to WT were involved in inflammation, complement activation, and phagocytosis and were found repressed upon pantethine treatment. In contrast, pantethine restored the expression of a significant number of genes involved in the regulation of Αβ processing and synaptic activities, which were downregulated in Tg mice. Altogether, our data support a beneficial role for long-term pantethine treatment in preserving CNS crucial functions altered by Aβ pathogenesis in Tg mice and highlight the potential efficiency of pantethine to alleviate AD pathology.
Collapse
Affiliation(s)
- Kevin Baranger
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Manuel van Gijsel-Bonnello
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
- Present Address: MRC Protein Phosphorylation & Ubiquitylation Unit, Sir James Black Centre and School of Life Science - Division of Cell Signalling and Immunology, Welcome Trust Building, University of Dundee, Dundee, DD1 5EH UK
| | - Delphine Stephan
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Wassila Carpentier
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMS Omique, Plateforme Post-génomique de la Pitié-Salpêtrière (P3S), F-75013 Paris, France
| | - Santiago Rivera
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | | | - Bouchra Gharib
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Max De Reggi
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| | - Philippe Benech
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
43
|
Yu L, Wei F, Liang J, Ren G, Liu X, Wang CZ, Yuan J, Zeng J, Luo Y, Bi Y, Yuan CS. Target Molecular-Based Neuroactivity Screening and Analysis of Panax ginseng by Affinity Ultrafiltration, UPLC-QTOF-MS and Molecular Docking. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1345-1363. [PMID: 31495181 DOI: 10.1142/s0192415x19500691] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Panax ginseng exerts good neuroprotective activity at the cell and animal level, but the specific bioactive compounds and action mechanism are needed to be investigated, verified, and confirmed. In this work, affinity ultrafiltration (AUF), UPLC-QTOF-MS, and molecular docking were integrated into one strategy to screen, identify, and evaluate the bioactive compounds in ginseng at the molecular level. Three biological macromolecules (AChE, MAO-B, and NMDA receptor) were selected as the target protein for AUF-MS screening for the first time, and 16 potential neuroactive compounds were found with suitable binding degree. Then, the bioactivity of ginseng and its components were evaluated by AChE-inhibitory test and DPPH assay, and the data indicate that ginseng extract and the screened compounds have good neuroactivity. The interaction between the three targets and the screened compounds was further analyzed by molecular docking, and the results were consistent with a few discrepancies in comparison with the AUF results. Finally, according to the corresponding relation between component-target-pathway, the action mechanism of ginseng elucidated that ginseng exerts a therapeutic effect on AD through multiple relations of components, targets, and pathways, which is in good accordance with the TCM theory.
Collapse
Affiliation(s)
- Lide Yu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Feiting Wei
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Jian Liang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Gang Ren
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Xiaofei Liu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, The University of Chicago, Chicago, IL 60637, USA
| | - Jinbin Yuan
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Jinxiang Zeng
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Yun Luo
- Key Laboratory of Modern Preparation of TCM, Ministry of Education and School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai 264005, P. R. China
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
44
|
Wu X, Tang B, Liao X, Su Z, Lee SMY, Cai Y, Li C. Suppressive effects of the supercritical-carbon dioxide fluid extract of Chrysanthemum indicum on chronic unpredictable mild stress-induced depressive-like behavior in mice. Food Funct 2019; 10:1212-1224. [PMID: 30741293 DOI: 10.1039/c8fo02474j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The aim of the present study was to explore whether the supercritical-carbon dioxide fluid extract from flowers and buds of Chrysanthemum indicum (SEC) exhibits antidepressant-like effects in a chronic unpredictable mild stress (CUMS)-induced mice model. Firstly, SEC was found to reverse a CUMS-induced decrease in the body weight gain in mice. Next, SEC was found to alleviate CUMS-induced depressive-like behavior, evidenced by the reversal of the decrease in the sucrose consumption in the sucrose preference test (SPT), the increase in the locomotor activity in the open field test (OPF), and the alleviation of immobility duration in both the forced swimming test (FST), and tail-suspension test (TST). SEC also attenuated CUMS-induced hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis by decreasing the levels of serum corticosterone and (CORT) and adrenocorticotropic hormone (ACTH), and hypothalamus corticotrophin-releasing hormone (CRH). In addition, SEC was found to suppress the expression of pro-inflammatory cytokines, including the tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in the hippocampal of CUMS mice. Interestingly, further investigations demonstrated that SEC inhibited CUMS-induced activation of the nuclear factor kappa B (NF-κB) and NOD-like receptor protein 3 (NLRP3) inflammasomes pathways but upregulated brain-derived neurotrophic factor (BDNF) expression and promoted phosphorylation of extracellular signal-regulated kinase (ERK) and cAMP-response element-binding protein (CREB) in hippocampal. In summary, SEC was able to alleviate depressive-like behavior in a CUMS-induced mice model, accompanied by inhibitory roles in the hyperactivity of the HPA axis and pro-inflammatory cytokine expression. Modulating the NF-κB/NLRP3 and BDNF/CREB/ERK pathways contributed to SEC-mediated antidepressant-like effects.
Collapse
Affiliation(s)
- Xiaoli Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Park S, Ko E, Lee JH, Song Y, Cui CH, Hou J, Jeon BM, Kim HS, Kim SC. Gypenoside LXXV Promotes Cutaneous Wound Healing In Vivo by Enhancing Connective Tissue Growth Factor Levels Via the Glucocorticoid Receptor Pathway. Molecules 2019; 24:molecules24081595. [PMID: 31018484 PMCID: PMC6515290 DOI: 10.3390/molecules24081595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
Cutaneous wound healing is a well-orchestrated event in which many types of cells and growth factors are involved in restoring the barrier function of skin. In order to identify whether ginsenosides, the main active components of Panax ginseng, promote wound healing, the proliferation and migration activities of 15 different ginsenosides were tested by MTT assay and scratched wound closure assay. Among ginsenosides, gypenoside LXXV (G75) showed the most potent wound healing effects. Thus, this study aimed to investigate the effects of G75 on wound healing in vivo and characterize associated molecular changes. G75 significantly increased proliferation and migration of keratinocytes and fibroblasts, and promoted wound closure in an excision wound mouse model compared with madecassoside (MA), which has been used to treat wounds. Additionally, RNA sequencing data revealed G75-mediated significant upregulation of connective tissue growth factor (CTGF), which is known to be produced via the glucocorticoid receptor (GR) pathway. Consistently, the increase in production of CTGF was confirmed by western blot and ELISA. In addition, GR-competitive binding assay and GR translocation assay results demonstrated that G75 can be bound to GR and translocated into the nucleus. These results demonstrated that G75 is a newly identified effective component in wound healing.
Collapse
Affiliation(s)
- Sungjoo Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Eunsu Ko
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Jun Hyoung Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Yoseb Song
- Department of Biological Sciences and KI for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Chang-Hao Cui
- Intelligent Synthetic Biology Center, Daejeon 34141, Korea.
| | - Jingang Hou
- Intelligent Synthetic Biology Center, Daejeon 34141, Korea.
| | - Byeong Min Jeon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Hun Sik Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea.
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
- Intelligent Synthetic Biology Center, Daejeon 34141, Korea.
| |
Collapse
|
46
|
Ovais M, Zia N, Ahmad I, Khalil AT, Raza A, Ayaz M, Sadiq A, Ullah F, Shinwari ZK. Phyto-Therapeutic and Nanomedicinal Approaches to Cure Alzheimer's Disease: Present Status and Future Opportunities. Front Aging Neurosci 2018; 10:284. [PMID: 30405389 PMCID: PMC6205985 DOI: 10.3389/fnagi.2018.00284] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/30/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive inability manifested due to the accumulation of β-amyloid, formation of hyper phosphorylated neurofibrillary tangles, and a malfunctioned cholinergic system. The degeneration integrity of the neuronal network can appear long after the onset of the disease. Nanotechnology-based interventions have opened an exciting area via theranostics of AD in terms of tailored nanomedicine, which are able to target and deliver drugs across the blood-brain barrier (BBB). The exciting interface existing between medicinal plants and nanotechnology is an emerging marvel in medicine, which has delivered promising results in the treatment of AD. In order to assess the potential applications of the medicinal plants, their derived components, and various nanomedicinal approaches, a review of literature was deemed as necessary. In the present review, numerous phytochemicals and various feats in nanomedicine for the treatment of AD have been discussed mechanistically for the first time. Furthermore, recent trends in nanotechnology such as green synthesis of metal nanoparticles with reference to the treatment of AD have been elaborated. Foreseeing the recent progress, we hope that the interface of medicinal plants and nanotechnology will lead to highly effective theranostic strategies for the treatment of AD in the near future.
Collapse
Affiliation(s)
- Muhammad Ovais
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Nashmia Zia
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Irshad Ahmad
- Department of Life Sciences, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Ali Talha Khalil
- Department of Eastern Medicine and Surgery, Qarshi University, Lahore, Pakistan
| | - Abida Raza
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
- Department of Life Sciences and Chemistry, Faculty of Health, Jacobs University Bremen, Bremen, Germany
| | - Farhat Ullah
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Zabta Khan Shinwari
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Eastern Medicine and Surgery, Qarshi University, Lahore, Pakistan
- Pakistan Academy of Sciences, Islamabad, Pakistan
| |
Collapse
|
47
|
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
|
48
|
Ginsenosides: the need to move forward from bench to clinical trials. J Ginseng Res 2018; 43:361-367. [PMID: 31308807 PMCID: PMC6606839 DOI: 10.1016/j.jgr.2018.09.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/02/2022] Open
Abstract
Panax ginseng, known as Koran ginseng, one of the most commonly used traditional plants, has been demonstrated to show a wide range of pharmacological applications. Ginsenosides are the major active ingredients found in ginseng and are responsible for the biological and pharmacological activities, such as antioxidation, antiinflammation, vasorelaxation, and anticancer actions. Existing studies have mostly focused on identifying and purifying single ginsenosides and investigating pharmacological activities and molecular mechanisms in cells and animal models. However, ginsenoside studies based on clinical trials have been very limited. Therefore, this review aimed to discuss the currently available clinical trials on ginsenosides and provide insights and future directions for developing ginsenosides as efficacious and safe drugs for human disease.
Collapse
|
49
|
Yu H, Jiang X, Lin X, Zhang Z, Wu D, Zhou L, Liu J, Yang X. Hippocampal Subcellular Organelle Proteomic Alteration of Copper-Treated Mice. Toxicol Sci 2018; 164:250-263. [PMID: 29617964 DOI: 10.1093/toxsci/kfy082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Copper neurotoxicity has been implicated in multiple neurological diseases. However, there is a lack of deep understanding on copper neurotoxicity, especially for low-dose copper exposure. In this study, we investigated the effects of chronic, low-dose copper treatment (0.13 ppm copper chloride in drinking water) on hippocampal mitochondrial and nuclear proteome in mice by 2-dimensional fluorescence difference gel electrophoresis coupled with MALDI-TOF-MS/MS. Behavioral tests revealed that low-dose copper caused spatial memory impairment, DNA oxidative damage as well as loss of synaptic proteins. Proteomic analysis revealed modulation of 31 hippocampal mitochondrial proteins (15 increased and 16 decreased), and 46 hippocampal nuclear proteins (18 increased and 28 decreased) in copper-treated versus untreated mice. Bioinformatic analysis indicated that these differentially expressed proteins are mainly involved energy metabolism (NDUV1, COX5B, IDH3A, and PGAM1), synapses (complexin-2, synapsin-2), DNA damage (PDIA3), apoptosis (GRP75), and oxidative stress (SODC, PRDX3). Among these differentially expressed proteins, synapsin-2, a synaptic-related protein, was found to be significantly decreased as confirmed by Western-blot analysis. In addition, we found that superoxide dismutase [Cu-Zn] (SODC), a copper ion target protein, was identified to be decreased in copper-treated mice versus untreated mice. We also found that stathmin (STMN1), a microtubule-destabilizing neuroprotein, was significantly decreased in hippocampal nuclei of copper-treated mice versus untreated mice. Taken together, we conclude that low-dose copper exposure causes spatial memory impairment and perturbs multiple biological/pathogenic processes by dysregulating the mitochondrial and nuclear proteome, particularly the proteins related to respiratory chain, synaptic vesicle fusion, axonal/neurtic integrity, and oxidative stress. The change of STMN1 and SODC may represent early novel biomarkers of copper neurotoxicity.
Collapse
Affiliation(s)
- Haitao Yu
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xin Jiang
- Department of Geriatrics, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Guangdong, China
| | - Xuemei Lin
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Desheng Wu
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Li Zhou
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|