1
|
Yang P, Fan M, Chen Y, Yang D, Zhai L, Fu B, Zhang L, Wang Y, Ma R, Sun L. A novel strategy for the protective effect of ginsenoside Rg1 against ovarian reserve decline by the PINK1 pathway. PHARMACEUTICAL BIOLOGY 2025; 63:68-81. [PMID: 39862058 PMCID: PMC11770866 DOI: 10.1080/13880209.2025.2453699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
CONTEXT The decline in ovarian reserve is a major concern in female reproductive health, often associated with oxidative stress and mitochondrial dysfunction. Although ginsenoside Rg1 is known to modulate mitophagy, its effectiveness in mitigating ovarian reserve decline remains unclear. OBJECTIVE To investigate the role of ginsenoside Rg1 in promoting mitophagy to preserve ovarian reserve. MATERIALS AND METHODS Ovarian reserve function, reproductive capacity, oxidative stress levels, and mitochondrial function were compared between ginsenoside Rg1-treated and untreated naturally aged female Drosophila using behavioral, histological, and molecular biological techniques. The protective effects of ginsenoside Rg1 were analyzed in a Drosophila model of oxidative damage induced by tert-butyl hydroperoxide. Protein expression levels in the PINK1/Parkin pathway were assessed, and molecular docking and PINK1 mutant analyses were conducted to identify potential targets. RESULTS Ginsenoside Rg1 significantly mitigated ovarian reserve decline, enhancing offspring quantity and quality, increasing the levels of ecdysteroids, preventing ovarian atrophy, and elevating germline stem cell numbers in aged Drosophila. Ginsenoside Rg1 improved superoxide dismutase, catalase activity, and gene expression while reducing reactive oxygen species levels. Ginsenoside Rg1 activated the mitophagy pathway by upregulating PINK1, Parkin, and Atg8a and downregulating Ref(2)P. Knockdown of PINK1 in the ovary by RNAi attenuated the protective effects of ginsenoside Rg1. Molecular docking analysis revealed that the ginsenoside Rg1 could bind to the active site of the PINK1 kinase domain. DISCUSSION AND CONCLUSIONS Ginsenoside Rg1 targets PINK1 to regulate mitophagy, preserving ovarian reserve. These findings suggest the potential of ginsenoside Rg1 as a therapeutic strategy to prevent ovarian reserve decline.
Collapse
Affiliation(s)
- Pengdi Yang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Meiling Fan
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Chen
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Dan Yang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Lu Zhai
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Baoyu Fu
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Zhang
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Yanping Wang
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Rui Ma
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Wang Q, Wei L, Chen G, Chen Q. Ginsenoside Rg1 in Parkinson's disease: from basic research to clinical applications. Front Pharmacol 2025; 16:1490480. [PMID: 40308780 PMCID: PMC12040930 DOI: 10.3389/fphar.2025.1490480] [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: 09/03/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
This review provides an in-depth exploration of the potential of Ginsenoside Rg1 in the treatment of Parkinson's disease (PD). The emphasis of this article was the therapeutic mechanisms of Rg1, which involved the reduction of inflammation, antioxidant properties, support for neuronal survival and regeneration, regulation of cellular energy processes, and enhancement of autophagic pathways. Rg1 may protect neurons and improve both motor and cognitive impairments associated with PD through multiple mechanisms. However, challenges exist in the clinical application of Rg1, such as low bioavailability as well as a lack of comprehensive long-term safety and efficacy data. This article also reviewed network pharmacology analyses published previously to identify and explore the potential molecular targets of Rg1 in PD treatment, while evaluating strategies such as drug delivery technologies, optimizing administration routes, and combination therapies. Ultimately, this review highlights the necessity for large-scale clinical trials to validate the clinical efficacy of Rg1 and discusses its potential for PD treatment clinically.
Collapse
Affiliation(s)
- Qianyan Wang
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Wei
- Department of Pharmacy, Tianmen Hospital of Traditional Chinese Medicine Affiliated to Hubei University of Chinese Medicine, Tianmen, China
| | - Guanghui Chen
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, China
| | - Qiang Chen
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Mehrnoosh F, Rezaei D, Pakmehr SA, Nataj PG, Sattar M, Shadi M, Ali-Khiavi P, Zare F, Hjazi A, Al-Aouadi RFA, Sapayev V, Zargari F, Alkhathami AG, Ahmadzadeh R, Khedmatgozar M, Hamzehzadeh S. The role of Panax ginseng in neurodegenerative disorders: mechanisms, benefits, and future directions. Metab Brain Dis 2025; 40:183. [PMID: 40232582 DOI: 10.1007/s11011-025-01610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Multiple sclerosis (MS), and Huntington's disease (HD) represent a growing global health challenge, especially with aging populations. Characterized by progressive neuronal loss, these diseases lead to cognitive, motor, and behavioral impairments, significantly impacting patients' quality of life. Current therapies largely address symptoms without halting disease progression, underscoring the need for innovative, disease-modifying treatments. Ginseng, a traditional herbal medicine with well-known adaptogenic and neuroprotective properties, has gained attention as a potential therapeutic agent for neurodegeneration. Rich in bioactive compounds called ginsenosides, ginseng exhibits antioxidant, anti-inflammatory, and anti-apoptotic effects, making it a promising candidate for addressing the complex pathology of neurodegenerative diseases. Recent studies demonstrate that ginsenosides modulate disease-related processes such as oxidative stress, protein aggregation, mitochondrial dysfunction, and inflammation. In AD models, ginsenosides have been shown to reduce amyloid-beta accumulation and tau hyperphosphorylation, while in PD, they help protect dopaminergic neurons and mitigate motor symptoms. Ginseng's effects in ALS, MS, and HD models include improving motor function, extending neuronal survival, and reducing cellular toxicity. This review provides a comprehensive overview of the neuroprotective mechanisms of ginseng, emphasizing its therapeutic potential across various neurodegenerative diseases and discussing future research directions for its integration into clinical practice.
Collapse
Affiliation(s)
- Faranak Mehrnoosh
- Department of Food Science and Technology, Faculty of Agriculture and Natural Resources, Urmia University, Urmia, Iran
| | | | | | | | - Mustafa Sattar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Melina Shadi
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| | - Payam Ali-Khiavi
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farshad Zare
- Student Research Committee, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | | | - Valisher Sapayev
- General Professional Science Department, Mamun University, Khiva, Uzbekistan
| | - Faranak Zargari
- Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, West Azerbaijan, Iran
| | - Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha, 9088, Saudi Arabia
| | - Roya Ahmadzadeh
- Medicine Faculty, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | | | |
Collapse
|
4
|
Hushmandi K, Reiter RJ, Farahani N, Cho WC, Alimohammadi M, Khoshnazar SM. Pyroptosis; igniting neuropsychiatric disorders from mild depression to aging-related neurodegeneration. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111325. [PMID: 40081561 DOI: 10.1016/j.pnpbp.2025.111325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Neuropsychiatric disorders significantly impact global health and socioeconomic well-being, highlighting the urgent need for effective treatments. Chronic inflammation, often driven by the innate immune system, is a key feature of many neuropsychiatric conditions. NOD-like receptors (NLRs), which are intracellular sensors, detect danger signals and trigger inflammation. Among these, NLR protein (NLRP) inflammasomes play a crucial role by releasing pro-inflammatory cytokines and inducing a particular cell death process known as pyroptosis. Pyroptosis is defined as a proinflammatory form of programmed cell death executed by cysteine-aspartic proteases, also known as caspases. Currently, the role of pyroptotic flux has emerged as a critical factor in innate immunity and the pathogenesis of multiple diseases. Emerging evidence suggests that the induction of pyroptosis, primarily due to NLRP inflammasome activation, is involved in the pathophysiology of various neuropsychiatric disorders, including depression, stress-related issues, schizophrenia, autism spectrum disorders, and neurodegenerative diseases. Within this framework, the current review explores the complex relationship between pyroptosis and neuropsychiatric diseases, aiming to identify potential therapeutic targets for these challenging conditions.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
5
|
Jiang M, Chi J, Qiao Y, Wang J, Zhang Z, Liu J, Sheng X, Yuan L. Ginsenosides Rg1, Rb1 and rare ginsenosides: Promising candidate agents for Parkinson's disease and Alzheimer's disease and network pharmacology analysis. Pharmacol Res 2025; 212:107578. [PMID: 39756554 DOI: 10.1016/j.phrs.2025.107578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Ginseng has been commonly used as a traditional Chinese medicine in Asian countries for thousands of years. Ginsenosides are the main pharmacologically active ingredients isolated from ginseng and have neuroprotective effects in the treatment of neurodegenerative disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To summarise and investigate the protective roles of ginsenosides and their underlying mechanisms in PD and AD, we used ''Ginsenoside", ''Parkinson's disease", ''Alzheimer's disease", ''anti-inflammatory", ''antioxidant", and ''apoptosis" as keywords to search and extract relevant literature information from scientific databases such as Elsevier, PubMed, and Google Scholar databases. In particular, we used network pharmacology to identify the potential targets of ginsenosides Rg1 and Rb1 in PD and AD. By analysing the existing research advances and network pharmacology results, we found that the neuroprotective effects of ginsenosides, primarily mediated through anti-inflammation, anti-apoptosis and anti-oxidative stress, etc, may be associated with the PI3K/Akt, BDNF/TrkB, MAPKs, NF-κB, Nrf2 and Wnt/β-catenin signalling pathways. This review systematically summarises the different roles and mechanisms of ginsenosides Rg1, Rb1, and rare ginsenosides in PD and AD and provides new strategies for the treatment of neurodegenerative disorders. Network pharmacology provides a new research paradigm for the treatment of PD and AD using Rg1 and Rb1.
Collapse
Affiliation(s)
- Mingchun Jiang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Jiaxin Chi
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Yifan Qiao
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jinpeng Wang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Zhixin Zhang
- School of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jia Liu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xinhao Sheng
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Liangjie Yuan
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.
| |
Collapse
|
6
|
Zhang Y, Liu S, Cao D, Zhao M, Lu H, Wang P. Rg1 improves Alzheimer's disease by regulating mitochondrial dynamics mediated by the AMPK/Drp1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119285. [PMID: 39733799 DOI: 10.1016/j.jep.2024.119285] [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: 10/05/2024] [Revised: 12/05/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is the most prevalent form of dementia, characterized by a complex pathogenesis that includes Aβ deposition, abnormal phosphorylation of tau protein, chronic neuroinflammation, and mitochondrial dysfunction. In traditional medicine, ginseng is revered as the 'king of herbs'. Ginseng has the effects of greatly tonifying vital energy, strengthening the spleen and benefiting the lungs, generating fluids and nourishing the blood, and calming the mind while enhancing intelligence. Ginsenoside Rg1 (Rg1) is a well-defined major active component found in ginseng, known for its relatively high content. It has been demonstrated to exhibit neuroprotective effects in both in vivo and in vitro models, capable of ameliorating Aβ and tau pathology, regulating synaptic function, and reducing inflammation, oxidative stress, and apoptosis. However, the potential of Rg1 to improve AD pathology through the regulation of mitochondrial dynamics is still uncertain. AIM OF THE STUDY Despite the active research efforts on drugs for AD, the currently available anti-AD medications can only slow disease progression and manage symptoms, yet unable to provide a cure for AD. Furthermore, some anti-AD drugs failed phase III and IV clinical trials due to significant side effects. Therefore, there is an urgent need to further investigate the pathogenesis of AD, to identify new therapeutic targets, and to explore more effective therapies. The aim of this study is to evaluate the potential therapeutic effects of Rg1 on APP/PS1 double transgenic mice and Aβ42-induced HT22 cell models, and to investigate the potential mechanisms through which it provides neuroprotective effects. MATERIALS AND METHODS This study investigates the effects of Rg1 in treating AD on APP/PS1 double transgenic mice and Aβ42-induced HT22 cells. In the in vivo experiments, APP/PS1 mice were divided into a model group, Rg1-L group, Rg1-H group, and donepezil group, with C57BL/6 mice serving as the control group (n = 12 per group). The Rg1-L and Rg1-H groups were administered Rg1 at doses of 5 mg/kg/d and 10 mg/kg/d, respectively, while the donepezil group received donepezil at a dose of 1.3 mg/kg/d. Both the control and model groups received an equal volume of physiological saline daily for 28 days. Learning and spatial memory were assessed by the Morris water maze (MWM) and novel object recognition (NOR) tests, and neuronal damage by Nissl staining. Aβ deposition was analyzed through immunohistochemistry and Western blot, while the expression levels of synaptic proteins PSD95 and SYN were evaluated via immunofluorescence staining and Western blot. The dendritic spines of neurons was observed by Golgi staining.The ultrastructure of neuronal mitochondria and synapses was examined by transmission electron microscopy (TEM). Mitochondrial function was assessed through measurements of Reactive oxygen species (ROS), Superoxide Dismutase (SOD), and Adenosine Triphosphate (ATP), and Western blot analysis was performed to detect the expression levels of AMPK, p-AMPK, Drp1, p-Drp1, OPA1, Mfn1, and Mfn2, thereby investigating the protective effects of Rg1 on mitochondrial dysfunction and cognitive impairment in APP/PS1 double transgenic mice. In vitro experiments, HT22 cells were treated with Aβ42 of 10 μM for 24 h to verify the therapeutic effects of Rg1. Flow cytometry was used to detect ROS and JC-1, biochemical methods were employed to measure SOD and ATP, immunofluorescence staining was used to detect the expression levels of PSD95 and SYN, and Western blot analysis was conducted to elucidate its potential mechanisms of action. RESULTS The findings suggest that after 28 days of Rg1 treatment, cognitive dysfunction in APP/PS1 mice was improved. Pathological and immunohistochemical analyses demonstrated that Rg1 treatment significantly reduced Aβ deposition and neuronal loss. Rg1 can improve synaptic dysfunction and mitochondrial function in APP/PS1 mice. Rg1 activated AMPK, enhanced p-AMPK expression, inhibited Drp1, and reduced p-Drp1 levels, which led to increased expression of OPA1, Mfn1, and Mfn2, thereby inhibiting mitochondrial fission and facilitating mitochondrial fusion. Additionally, Rg1 effectively reversed the decrease in mitochondrial membrane potential (MMP) and the increase in ROS production induced by Aβ42 in HT22 cells, restoring SOD and ATP levels. Furthermore, Rg1 regulated mitochondrial fission mediated by the AMPK/Drp1 signaling pathway, promoting mitochondrial fusion and improving synaptic dysfunction. CONCLUSION Our research provides evidence for the neuroprotective mechanisms of Rg1 in AD models. Rg1 modulates mitochondrial dynamics through the AMPK/Drp1 signaling pathway, thereby reducing synaptic and mitochondrial dysfunction in APP/PS1 mice and AD cell models.
Collapse
Affiliation(s)
- Yini Zhang
- Hubei University of Chinese Medicine, Basic Medical College, Wuhan, Hubei, 430070, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430070, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430070, China.
| | - Shangzhi Liu
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430070, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430070, China; Hubei University of Chinese Medicine, College of Chinese Medicine, Wuhan, Hubei, 430065, China.
| | - Di Cao
- Hubei University of Chinese Medicine, Basic Medical College, Wuhan, Hubei, 430070, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430070, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430070, China.
| | - Min Zhao
- Hubei University of Chinese Medicine, Basic Medical College, Wuhan, Hubei, 430070, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430070, China.
| | - Haifei Lu
- Huanggang Hospital of Chinese Medicine, Affiliated to Hubei University of Chinese Medicine, Huanggang, Hubei, 438000, China.
| | - Ping Wang
- Hubei University of Chinese Medicine, Basic Medical College, Wuhan, Hubei, 430070, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430070, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430070, China.
| |
Collapse
|
7
|
Ju IG, Lee JH, Lee JM, Im H, Eo H, Moon M, Song MK, Kim YS, Oh MS, Kim YJ. NXP031 restores memory function by dual effects degrading Aβ accumulation and facilitating antioxidant pathway in Alzheimer's disease models. Free Radic Biol Med 2025; 226:158-170. [PMID: 39521153 DOI: 10.1016/j.freeradbiomed.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Alzheimer's disease (AD) is a representative neurodegenerative disease that is characterized by the overaccumulation of amyloid beta (Aβ) proteins. Since AD is accompanied by excessive oxidative stress, which aggravates neurological pathologies, the use of antioxidants has been considered to prevent disease development. NXP031, a combination of vitamin C (VitC) and an optimized aptamer that binds to VitC and stabilizes the reactivity of VitC, was designed. This study aimed to evaluate the effects of NXP031 on AD pathology, including Aβ accumulation, Aβ-induced oxidative stress, neuronal damage, and neuroinflammation. When NXP031 was administered to 5xFAD transgenic mice, NXP031 exerted a strong inhibitory action on Aβ accumulation, superior to that of VitC, by inducing an increase in Aβ-degrading endopeptidase expression. NXP031 diminished lipid peroxidation levels, activated Nrf2-mediated antioxidant pathways, and suppressed overactivated neuroinflammation. An in vitro study using Neuro2a cells revealed that NXP031 protects the cells against oxidative stress by regulating the MAPK signaling pathway-mediated apoptosis. Additionally, the neuroprotective effects of NXP031 were confirmed in a dose-dependent manner when administered to intrahippocampal Aβ-injected mice, as NXP031 attenuated memory decline, neuronal apoptosis, synaptic degeneration, and excessive glial activation, and reduced NOX-2 expression in the hippocampus. Taken together, NXP031 reduced the Aβ burden by regulating Aβ-degeneration and attenuated memory impairment, neuronal death, synaptic degeneration, and neuroinflammation induced by Aβ toxicity. These results suggest the potential of NXP031 as a therapeutic agent for AD.
Collapse
Affiliation(s)
- In Gyoung Ju
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Joo Hee Lee
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Jae-Min Lee
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hyeri Im
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hyeyoon Eo
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 32992, Republic of Korea.
| | - Min Kyung Song
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Yoon-Seong Kim
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, 08854, USA.
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Youn-Jung Kim
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
8
|
Zhou X, Du K, Mao T, Wang N, Zhang L, Tian Y, Liu T, Wang L, Wang X. BMAL1 upregulates STX17 levels to promote autophagosome-lysosome fusion in hippocampal neurons to ameliorate Alzheimer's disease. iScience 2024; 27:111413. [PMID: 39687016 PMCID: PMC11647228 DOI: 10.1016/j.isci.2024.111413] [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: 05/06/2024] [Revised: 08/02/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
We aim to investigate muscle ARNT-like protein 1 (BMAL1) regulation of syntaxin17 (STX17) in mouse hippocampal neurons, focusing on autophagy and amyloid-β (Aβ) deposition. Autophagosome-lysosome fusion in APP/PS1 hippocampal tissues was observed using transmission electron microscopy, while mRNA levels of LC3II and P62 were measured via reverse-transcription PCR (RT-PCR) after Amyloid precursor protein (APP) overexpression. STX17, linked to autophagy and differentially expressed in Alzheimer's disease (AD) brains, was knocked down or overexpressed to assess its effects. The results showed that reduced STX17 impairs autophagosome-lysosome fusion, leading to abnormal Aβ deposition. Coimmunoprecipitation (Co-IP) and immunofluorescence confirmed STX17 interaction with SNAP29 and VAMP8 to form SNARE complexes. Furthermore, BMAL1 binding to STX17 was examined using luciferase assays. Circadian rhythm disturbances and decreased BMAL1 expression in APP/PS1 mice were noted, while BMAL1 overexpression upregulated STX17 expression and promoted autophagy to reduce Aβ deposition. Thus, the BMAL1 protein can promote STX17 transcription to induce STX17-SNAP29-VAMP8 complex formation to clear intracellular Aβ through autophagy.
Collapse
Affiliation(s)
- Xiuya Zhou
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kaili Du
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Tian Mao
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ning Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Lifei Zhang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Yuan Tian
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ting Liu
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Li Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Xiaohui Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
9
|
Han D, Zhao Z, Mao T, Gao M, Yang X, Gao Y. Ginsenoside Rg1: A Neuroprotective Natural Dammarane-Type Triterpenoid Saponin With Anti-Depressive Properties. CNS Neurosci Ther 2024; 30:e70150. [PMID: 39639753 PMCID: PMC11621566 DOI: 10.1111/cns.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Depression, a widespread mental disorder, presents significant risks to both physical and mental health due to its high rates of recurrence and suicide. Currently, single-target antidepressants typically alleviate depressive symptoms or delay the progression of depression rather than cure it. Ginsenoside Rg1 is one of the main ginsenosides found in Panax ginseng roots. It improves depressive symptoms through various mechanisms, suggesting its potential as a treatment for depression. MATERIALS AND METHODS We evaluated preclinical studies to comprehensively discuss the antidepressant mechanism of ginsenoside Rg1 and review its toxicity and medicinal value. Additionally, pharmacological network and molecular docking analyses were performed to further validate the antidepressant effects of ginsenoside Rg1. RESULTS The antidepressant mechanism of ginsenoside Rg1 may involve various pharmacological mechanisms and pathways, such as inhibiting neuroinflammation and over-activation of microglia, preserving nerve synapse structure, promoting neurogenesis, regulating monoamine neurotransmitter levels, inhibiting hyperfunction of the hypothalamic-pituitary-adrenal axis, and combatting antioxidative stress. Moreover, ginsenoside Rg1 preserves astrocyte gap junction function by regulating connexin43 protein biosynthesis and degradation, contributing to its antidepressant effect. Pharmacological network and molecular docking studies identified five targets (AKT1, STAT3, EGFR, PPARG, and HSP90AA1) as potential molecular regulatory sites of ginsenoside Rg1. CONCLUSIONS Ginsenoside Rg1 may exert its antidepressant effects via various pharmacological mechanisms. In addition, multicenter clinical case-control and molecular targeted studies are required to confirm both the clinical efficacy of ginsenoside Rg1 and its potential direct targets.
Collapse
Affiliation(s)
- Dong Han
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zheng Zhao
- Department of Emergency MedicineShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Tinghui Mao
- Department of Organ Transplantation and Hepatobiliary SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningChina
| | - Man Gao
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xue Yang
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yan Gao
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
10
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
11
|
Kim Y, Cho IH, Cho SH. Effect of ginseng and ginsenosides on attention deficit hyperactivity disorder: A systematic review. J Ginseng Res 2024; 48:437-448. [PMID: 39263306 PMCID: PMC11385392 DOI: 10.1016/j.jgr.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 09/13/2024] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a rapidly increasing neurodevelopmental disorder but currently available treatments are associated with abuse risk, side effects, and incomplete symptom relief. There is growing interest in exploring complementary options, and ginseng has gained attention for its therapeutic potential. This systematic review aimed to assess current evidence on the efficacy of ginseng and its active components, ginsenosides, for ADHD. Eligible studies were identified through searches of PubMed, Embase, Cochrane Library, and Web of Science, up to June 2023. The inclusion criteria included both human and animal studies that investigated the effects of ginseng or ginsenosides on ADHD. The risk of bias was assessed according to study type. Six human studies and three animal studies met the inclusion criteria. The results suggest that ginseng and ginsenosides may have beneficial effects on ADHD symptoms, particularly inattention, through dopaminergic/norepinephrinergicmodulation and BDNF/TrkB signaling. Ginseng and ginsenosides have promising potential for ADHD treatment. Due to limitations in evidence quality, such as the risk of bias and variability in study designs, larger controlled studies are essential. Integrating ginseng into ADHD management may have valuable implications for individuals seeking well-tolerated alternatives or adjunctive therapies.
Collapse
Affiliation(s)
- Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seung-Hun Cho
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Kong L, Liu Y, Li J, Wang Y, Ji P, Shi Q, Han M, Xu H, Li W, Li W. Ginsenoside Rg1 alleviates chronic inflammation-induced neuronal ferroptosis and cognitive impairments via regulation of AIM2 - Nrf2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118205. [PMID: 38641079 DOI: 10.1016/j.jep.2024.118205] [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: 03/04/2024] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is a valuable herb in traditional Chinese medicine. Modern research has shown that it has various benefits, including tonifying vital energy, nourishing and strengthening the body, calming the mind, improving cognitive function, regulating fluids, and returning blood pressure, etc. Rg1 is a primary active component of ginseng. It protects hippocampal neurons, improves synaptic plasticity, enhances cognitive function, and boosts immunity. Furthermore, it exhibits anti-aging and anti-fatigue properties and holds great potential for preventing and managing neurodegenerative diseases (NDDs). AIM OF THE STUDY The objective of this study was to examine the role of Rg1 in treating chronic inflammatory NDDs and its molecular mechanisms. MATERIALS AND METHODS In vivo, we investigated the protective effects of Rg1 against chronic neuroinflammation and cognitive deficits in mice induced by 200 μg/kg lipopolysaccharide (LPS) for 21 days using behavioral tests, pathological sections, Western blot, qPCR and immunostaining. In vitro experiments involved the stimulation of HT22 cells with 10 μg/ml of LPS, verification of the therapeutic effect of Rg1, and elucidation of its potential mechanism of action using H2DCFDA staining, BODIPY™ 581/591 C11, JC-1 staining, Western blot, and immunostaining. RESULTS Firstly, it was found that Rg1 significantly improved chronic LPS-induced behavioral and cognitive dysfunction in mice. Further studies showed that Rg1 significantly attenuated LPS-induced neuronal damage by reducing levels of IL-6, IL-1β and ROS, and inhibiting AIM2 inflammasome. Furthermore, chronic LPS exposure induced the onset of neuronal ferroptosis by increasing the lipid peroxidation product MDA and regulating the ferroptosis-associated proteins Gpx4, xCT, FSP1, DMT1 and TfR, which were reversed by Rg1 treatment. Additionally, Rg1 was found to activate Nrf2 and its downstream antioxidant enzymes, such as HO1 and NQO1, both in vivo and in vitro. In vitro studies also showed that the Nrf2 inhibitor ML385 could inhibit the anti-inflammatory, antioxidant, and anti-ferroptosis effects of Rg1. CONCLUSIONS This study demonstrated that Rg1 administration ameliorated chronic LPS-induced cognitive deficits and neuronal ferroptosis in mice by inhibiting neuroinflammation and oxidative stress. The underlying mechanisms may be related to the inhibition of AIM2 inflammasome and activation of Nrf2 signaling. These findings provide valuable insights into the treatment of chronic neuroinflammation and associated NDDs.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Jingwei Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yanyan Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Pengmin Ji
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Qifeng Shi
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Min Han
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Hanyang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weiping Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Weizu Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Zhuang D, Yu N, Han S, Zhang X, Ju C. The Kv7 channel opener Retigabine reduces neuropathology and alleviates behavioral deficits in APP/PS1 transgenic mice. Behav Brain Res 2024; 471:115137. [PMID: 38971432 DOI: 10.1016/j.bbr.2024.115137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Hyperexcitability of neuronal networks is central to the pathogenesis of Alzheimer's disease (AD). Pharmacological activation of Kv7 channels is an effective way to reduce neuronal firing. Our results showed that that pharmacologically activating the Kv7 channel with Retigabine (RTG) can alleviate cognitive impairment in mice without affecting spontaneous activity. RTG could also ameliorate damage to the Nissl bodies in cortex and hippocampal CA and DG regions in 9-month-old APP/PS1 mice. Additionally, RTG could reduce the Aβ plaque number in the hippocampus and cortex of both 6-month-old and 9-month-old mice. By recordings of electroencephalogram, we showed that a decrease in the number of abnormal discharges in the brains of the AD model mice when the Kv7 channel was opened. Moreover, Western blot analysis revealed a reduction in the expression of the p-Tau protein in both the hippocampus and cortex upon Kv7 channel opening. These findings suggest that Kv7 channel opener RTG may ameliorate cognitive impairment in AD, most likely by reducing brain excitability.
Collapse
Affiliation(s)
- Dongpei Zhuang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Nan Yu
- Department of Pharmacy, Qingdao Eighth People's Hospital, China.
| | - Shuo Han
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Xinyao Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| |
Collapse
|
14
|
Tian L, Tang P, Liu J, Liu Y, Hou L, Zhao J, Wang Q. Microglial gp91phox-mediated neuroinflammation and ferroptosis contributes to learning and memory deficits in rotenone-treated mice. Free Radic Biol Med 2024; 220:56-66. [PMID: 38697489 DOI: 10.1016/j.freeradbiomed.2024.04.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/26/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Apart from dopaminergic neurotoxicity, exposure to rotenone, a commonly used insecticide in agriculture, also adversely affects hippocampal and cortical neurons, resulting in cognitive impairments in mice. We recently established a role of microglia-mediated neuroinflammation in rotenone-elicited deficits of cognition, yet the mechanisms remain elusive. Here, we investigated the involvement of NADPH oxidase 2 (NOX2) catalytic subunit gp91phox in rotenone-induced cognitive deficits and the associated mechanisms. Our study demonstrated that rotenone exposure elevated expression of gp91phox and phosphorylation of the NOX2 cytosolic subunit p47phox, along with NADPH depletion in the hippocampus and cortex of mice, indicating NOX2 activation. Specific knockdown of gp91phox in microglia via adeno-associated virus delivery resulted in reduced microglial activation, proinflammatory gene expression and improved learning and memory capacity in rotenone-intoxicated mice. Genetic deletion of gp91phox also reversed rotenone-elicited cognitive dysfunction in mice. Furthermore, microglial gp91phox knockdown attenuated neuronal damage and synaptic loss in mice. This intervention also suppressed iron accumulation, disruption of iron-metabolism proteins and iron-dependent lipid peroxidation and restored the balance of ferroptosis-related parameters, including GPX4, SLC711, PTGS2, and ACSL4 in rotenone-lesioned mice. Intriguingly, pharmacological inhibition of ferroptosis with liproxstatin-1 conferred protection against rotenone-induced neurodegeneration and cognitive dysfunction in mice. In summary, our findings underscored the contribution of microglial gp91phox-dependent neuroinflammation and ferroptosis to learning and memory dysfunction in rotenone-lesioned mice. These results provided valuable insights into the pathogenesis of cognitive deficits associated with pesticide-induced Parkinsonism, suggesting potential therapeutic avenues for intervention.
Collapse
Affiliation(s)
- Lu Tian
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; Chaoyang Center for Disease Control and Prevention, Beijing, China
| | - Peiyan Tang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Jianing Liu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Yiyang Liu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Liyan Hou
- Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; School of Public Health, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
15
|
Ye C, Wang S, Niu L, Yang F, Wang G, Wang S, Xie J, Chen Y, Qi J, Shen H, Dou Y, Wang J. Unlocking potential of oxytocin: improving intracranial lymphatic drainage for Alzheimer's disease treatment. Theranostics 2024; 14:4331-4351. [PMID: 39113801 PMCID: PMC11303076 DOI: 10.7150/thno.98587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
Background: The impediment to β-amyloid (Aβ) clearance caused by the invalid intracranial lymphatic drainage in Alzheimer's disease is pivotal to its pathogenesis, and finding reliable clinical available solutions to address this challenge remains elusive. Methods: The potential role and underlying mechanisms of intranasal oxytocin administration, an approved clinical intervention, in improving intracranial lymphatic drainage in middle-old-aged APP/PS1 mice were investigated by live mouse imaging, ASL/CEST-MRI scanning, in vivo two-photon imaging, immunofluorescence staining, ELISA, RT-qPCR, Western blotting, RNA-seq analysis, and cognitive behavioral tests. Results: Benefiting from multifaceted modulation of cerebral hemodynamics, aquaporin-4 polarization, meningeal lymphangiogenesis and transcriptional profiles, oxytocin administration normalized the structure and function of both the glymphatic and meningeal lymphatic systems severely impaired in middle-old-aged APP/PS1 mice. Consequently, this intervention facilitated the efficient drainage of Aβ from the brain parenchyma to the cerebrospinal fluid and then to the deep cervical lymph nodes for efficient clearance, as well as improvements in cognitive deficits. Conclusion: This work broadens the underlying neuroprotective mechanisms and clinical applications of oxytocin medication, showcasing its promising therapeutic prospects in central nervous system diseases with intracranial lymphatic dysfunction.
Collapse
Affiliation(s)
- Caihua Ye
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Shengnan Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Lin Niu
- Department of Cellular Biology, School of Basic Science, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Fan Yang
- School of Life Sciences, Tianjin University, Tianjin300072, P. R. China
| | - Guohe Wang
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Siqi Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Jiamei Xie
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Yihan Chen
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Jinbo Qi
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Hui Shen
- Department of Cellular Biology, School of Basic Science, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Yan Dou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Junping Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
16
|
Kumar Nelson V, Jha NK, Nuli MV, Gupta S, Kanna S, Gahtani RM, Hani U, Singh AK, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Al Othaim A, Begum SS, Ahmad F, Mishra PC, Jha SK, Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: Factors and therapeutic implications. Ageing Res Rev 2024; 98:102224. [PMID: 38346505 DOI: 10.1016/j.arr.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that has devastating effects on individuals, often resulting in dementia. AD is primarily defined by the presence of extracellular plaques containing insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein (P-tau). In addition, individuals afflicted by these age-related illnesses experience a diminished state of health, which places significant financial strain on their loved ones. Several risk factors play a significant role in the development of AD. These factors include genetics, diet, smoking, certain diseases (such as cerebrovascular diseases, obesity, hypertension, and dyslipidemia), age, and alcohol consumption. Age-related factors are key contributors to the development of vascular-based neurodegenerative diseases such as AD. In general, the process of aging can lead to changes in the immune system's responses and can also initiate inflammation in the brain. The chronic inflammation and the inflammatory mediators found in the brain play a crucial role in the dysfunction of the blood-brain barrier (BBB). Furthermore, maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. Therefore, in this review, we discussed the role of age and its related factors in the breakdown of the blood-brain barrier and the development of AD. We also discussed the importance of different compounds, such as those with anti-aging properties, and other compounds that can help maintain the integrity of the blood-brain barrier in the prevention of AD. This review builds a strong correlation between age-related factors, degradation of the BBB, and its impact on AD.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sandeep Kanna
- Department of pharmaceutics, Chalapathi Institute of Pharmaceutical Sciences, Chalapathi Nagar, Guntur 522034, India
| | - Reem M Gahtani
- Departement of Clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Arun Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Science, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - S Sabarunisha Begum
- Department of Biotechnology, P.S.R. Engineering College, Sivakasi 626140, India
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh, 13713, Saudi Arabia
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
17
|
Manju, Bharadvaja N. Exploring the Potential Therapeutic Approach Using Ginsenosides for the Management of Neurodegenerative Disorders. Mol Biotechnol 2024; 66:1520-1536. [PMID: 37330923 DOI: 10.1007/s12033-023-00783-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023]
Abstract
There is a need for an efficient and long-lasting treatment due to the population's increasing prevalence of neurodegenerative disorders. In an effort to generate fresh ideas and create novel therapeutic medications, scientists have recently started to investigate the biological functions of compounds derived from plants and herbs. Ginseng, famous Chinese herbal medicine, has therapeutic value by virtue of its compounds ginsenosides or panaxosides, which are triterpene saponins and steroid glycosides. Research revealed positive impacts on ameliorating various disease conditions and found it as a possible drug candidate. Several neuroprotection mechanisms followed by this compound are inhibition of cell apoptosis, oxidative stress, inflammatory, and tumor activity. It has been demonstrated that controlling these mechanisms enhances cognitive performance and safeguards the brain against neurodegenerative disorders. The main objective of this review is to give a description of the most recent studies on ginsenoside's possible therapeutic application in the treatment of neurodegenerative diseases. Using organic compounds like ginseng and its various components may create new avenues for innovative treatment approaches development for neurological diseases. However, further research is necessary to confirm the stability and effectiveness of ginsenosides for neurodegenerative disease.
Collapse
Affiliation(s)
- Manju
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042, India
| | - Navneeta Bharadvaja
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042, India.
| |
Collapse
|
18
|
Zhang H, Cai W, Dong L, Yang Q, Li Q, Ran Q, Liu L, Wang Y, Li Y, Weng X, Zhu X, Chen Y. Jiaohong pills attenuate neuroinflammation and amyloid-β protein-induced cognitive deficits by modulating the mitogen-activated protein kinase/nuclear factor kappa-B pathway. Animal Model Exp Med 2024; 7:222-233. [PMID: 38177948 PMCID: PMC11228096 DOI: 10.1002/ame2.12369] [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: 12/11/2022] [Accepted: 11/15/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Jiaohong pills (JHP) consist of Pericarpium Zanthoxyli (PZ) and Radix Rehmanniae, two herbs that have been extensively investigated over many years due to their potential protective effects against cognitive decline and memory impairment. However, the precise mechanisms underlying the beneficial effects remain elusive. Here, research studies were conducted to investigate and validate the therapeutic effects of JHP on Alzheimer's disease. METHODS BV-2 cell inflammation was induced by lipopolysaccharide. AD mice were administered amyloid-β (Aβ). Behavioral experiments were used to evaluate learning and memory ability. The levels of nitric oxide (NO), tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were detected using enzyme-linked immunosorbent assay (ELISA). The protein expressions of inducible nitric oxide synthase (iNOS) and the phosphorylation level of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) were detected using Western blot. Nissl staining was used to detect neuronal degeneration. RESULTS The results demonstrated that an alcoholic extract of PZ significantly decreased the levels of NO, IL-1β, TNF-α, and iNOS; increased the expression level of IL-10; and significantly decreased the phosphorylation levels of MAPK and NF-κB. These inhibitory effects were further confirmed in the AD mouse model. Meanwhile, JHP improved learning and memory function in AD mice, reduced neuronal damage, and enriched the Nissl bodies in the hippocampus. Moreover, IL-1β and TNF-α in the cortex were significantly downregulated after JHP administration, whereas IL-10 showed increased expression. CONCLUSIONS It was found that JHP reduced neuroinflammatory response in AD mice by targeting the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lijinchuan Dong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingsen Ran
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
19
|
She L, Tang H, Zeng Y, Li L, Xiong L, Sun J, Chen F, Ren J, Zhang J, Wang W, Zhao X, Liang G. Ginsenoside RK3 promotes neurogenesis in Alzheimer's disease through activation of the CREB/BDNF pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117462. [PMID: 37981117 DOI: 10.1016/j.jep.2023.117462] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the ancient book "Shen Nong's Herbal Classic," Panax ginseng CA Mey was believed to have multiple benefits, including calming nerves, improving cognitive function, and promoting longevity. Ginsenosides are the main active ingredients of ginseng. Ginsenoside RK3 (RK3), a rare ginsenoside extracted from ginseng, displays strong pharmacological potential. However, its effect on neurogenesis remains insufficiently investigated. AIM OF THE STUDY This study aims to investigate whether RK3 improves learning and memory by promoting neurogenesis, and to explore the mechanism of RK3 action. MATERIALS AND METHODS The therapeutic effect of RK3 on learning and memory was determined by the Morris water maze (MWM) and novel object recognition test (NORT). The pathogenesis and protective effect of RK3 on primary neurons and animal models were detected by immunofluorescence and western blotting. Protein expression of cAMP response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathway was detected by western blotting. RESULTS Our results showed that RK3 treatment significantly improved cognitive function in APPswe/PSEN1dE9 (APP/PS1) mice and C57BL/6 (C57) mice. RK3 promotes neurogenesis and synaptogenesis in the mouse hippocampus. In vitro, RK3 prevents Aβ-induced injury in primary cultured neurons and promotes the proliferation of PC12 as well as the expression of synapse-associated proteins. Mechanically, the positve role of RK3 on neurogenesis was combined with the activation of CREB/BDNF pathway. Inhibition of CREB/BDNF pathway attenuated the effect of RK3. CONCLUSION In conclusion, this study demonstrated that RK3 promotes learning and cognition in APP/PS1 and C57 mice by promoting neurogenesis and synaptogenesis through the CREB/BDNF signaling pathway. Therefore, RK3 is expected to be further developed into a potential drug candidate for the treatment of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Lingyu She
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China; Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
| | - Hao Tang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuqing Zeng
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Liwei Li
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Li Xiong
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jinfeng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Fan Chen
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Juan Ren
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jing Zhang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wei Wang
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China.
| | - Xia Zhao
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
20
|
Chen Y, Mateski J, Gerace L, Wheeler J, Burl J, Prakash B, Svedin C, Amrick R, Adams BD. Non-coding RNAs and neuroinflammation: implications for neurological disorders. Exp Biol Med (Maywood) 2024; 249:10120. [PMID: 38463392 PMCID: PMC10911137 DOI: 10.3389/ebm.2024.10120] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 03/12/2024] Open
Abstract
Neuroinflammation is considered a balanced inflammatory response important in the intrinsic repair process after injury or infection. Under chronic states of disease, injury, or infection, persistent neuroinflammation results in a heightened presence of cytokines, chemokines, and reactive oxygen species that result in tissue damage. In the CNS, the surrounding microglia normally contain macrophages and other innate immune cells that perform active immune surveillance. The resulting cytokines produced by these macrophages affect the growth, development, and responsiveness of the microglia present in both white and gray matter regions of the CNS. Controlling the levels of these cytokines ultimately improves neurocognitive function and results in the repair of lesions associated with neurologic disease. MicroRNAs (miRNAs) are master regulators of the genome and subsequently control the activity of inflammatory responses crucial in sustaining a robust and acute immunological response towards an acute infection while dampening pathways that result in heightened levels of cytokines and chemokines associated with chronic neuroinflammation. Numerous reports have directly implicated miRNAs in controlling the abundance and activity of interleukins, TGF-B, NF-kB, and toll-like receptor-signaling intrinsically linked with the development of neurological disorders such as Parkinson's, ALS, epilepsy, Alzheimer's, and neuromuscular degeneration. This review is focused on discussing the role miRNAs play in regulating or initiating these chronic neurological states, many of which maintain the level and/or activity of neuron-specific secondary messengers. Dysregulated miRNAs present in the microglia, astrocytes, oligodendrocytes, and epididymal cells, contribute to an overall glial-specific inflammatory niche that impacts the activity of neuronal conductivity, signaling action potentials, neurotransmitter robustness, neuron-neuron specific communication, and neuron-muscular connections. Understanding which miRNAs regulate microglial activation is a crucial step forward in developing non-coding RNA-based therapeutics to treat and potentially correct the behavioral and cognitive deficits typically found in patients suffering from chronic neuroinflammation.
Collapse
Affiliation(s)
- Yvonne Chen
- Department of Biology, Brandeis University, Waltham, MA, United States
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
| | - Julia Mateski
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Biological Sciences, Gustavus Adolphus College, St. Peter, MN, United States
| | - Linda Gerace
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Missouri State University, Springfield, MO, United States
| | - Jonathan Wheeler
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Electrical and Computer Engineering Tech, New York Institute of Tech, Old Westbury, NY, United States
| | - Jan Burl
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Southern New Hampshire University, Manchester, NH, United States
| | - Bhavna Prakash
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Medicine, Tufts Medical Center, Medford, MA, United States
| | - Cherie Svedin
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of Biology, Utah Tech University, St. George, UT, United States
| | - Rebecca Amrick
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
- Department of English, Villanova University, Villanova, PA, United States
| | - Brian D Adams
- Department of RNA Sciences, The Brain Institute of America, New Haven, CT, United States
| |
Collapse
|
21
|
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
|
22
|
Bian Z, Yu H, Hu X, Bian Y, Sun H, Tadokoro K, Takemoto M, Yunoki T, Nakano Y, Fukui Y, Morihara R, Abe K, Yamashita T. Tocovid Attenuated Oxidative Stress and Cognitive Decline by Inhibiting Amyloid-β-Induced NOX2 Activation in Alzheimer's Disease Mice. J Alzheimers Dis 2024; 99:S23-S33. [PMID: 36565115 DOI: 10.3233/jad-220761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background NADPH oxidase 2 (NOX2) is an important source of reactive oxygen species (ROS). Activated NOX2 may contribute to Alzheimer's disease (AD). Our previous studies showed that a novel vitamin E mixture, Tocovid, had potential neuroprotective effects in a stroke mice model and an AD cell model. Objective The aim of this study was two-fold: to assess whether long-term Tocovid treatment can regulate NOX2, and the therapeutic effects of long-term administration of Tocovid to an AD mice model. Methods Therapeutic effects of long-term administration of Tocovid (200 mg/kg /day) on an Aβ-overexpressed transgenic AD mice model (APP23, n = 8) was investigated. The therapeutic effect of Tocovid in 16-month-old mice compared with the no-treatment APP23 group (n = 9) was assessed. Results Tocovid treatment strongly improved motor and memory deficits of APP23 mice by attenuating NOX2 expression, oxidative stress, neuroinflammation, neurovascular unit dysfunction, synaptic alteration, and Aβ deposition after 16 months. Conclusion These findings suggest that NOX2 is a potential target in AD pathology. Long-term administration of Tocovid may be a promising candidate for AD treatment.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Koji Abe
- National Center Hospital, National Center of Neurology and Psychiatry, Kodaira-shi, Tokyo, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
23
|
Lee MY, Kim M. Effects of Red ginseng on neuroinflammation in neurodegenerative diseases. J Ginseng Res 2024; 48:20-30. [PMID: 38223824 PMCID: PMC10785270 DOI: 10.1016/j.jgr.2023.08.003] [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: 03/29/2023] [Revised: 07/17/2023] [Accepted: 08/25/2023] [Indexed: 01/16/2024] Open
Abstract
Red ginseng (RG) is widely used as a herbal medicine. As the human lifespan has increased, numerous diseases have developed, and RG has also been used to treat various diseases. Neurodegenerative diseases are major problems that modern people face through their lives. Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are featured by progressive nerve system damage. Recently, neuroinflammation has emerged as a degenerative factor and is an immune response in which cytokines with nerve cells that constitute the nervous system. RG, a natural herbal medicine with fewer side effects than chemically synthesized drugs, is currently in the spotlight. Therefore, we reviewed studies reporting the roles of RG in treating neuroinflammation and neurodegenerative diseases and found that RG might help alleviate neurodegenerative diseases by regulating neuroinflammation.
Collapse
Affiliation(s)
- Min Yeong Lee
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| |
Collapse
|
24
|
Zhang M, Niu H, Li Q, Jiao L, Li H, Wu W. Active Compounds of Panax ginseng in the Improvement of Alzheimer's Disease and Application of Spatial Metabolomics. Pharmaceuticals (Basel) 2023; 17:38. [PMID: 38256872 PMCID: PMC10818864 DOI: 10.3390/ph17010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
Panax ginseng C.A. Meyer (P. ginseng) is one of the more common traditional Chinese medicines (TCMs). It contains numerous chemical components and exhibits a range of pharmacological effects. An enormous burden is placed on people's health and life by Alzheimer's disease (AD), a neurodegenerative condition. Recent research has shown that P. ginseng's chemical constituents, particularly ginsenosides, have a significant beneficial impact on the prevention and management of neurological disorders. To understand the current status of research on P. ginseng to improve AD, this paper discusses the composition of P. ginseng, its mechanism of action, and its clinical application. The pathogenesis of AD includes amyloid beta protein (Aβ) generation and aggregation, tau protein hyperphosphorylation, oxidant stress, neuroinflammation, mitochondrial damage, and neurotransmitter and gut microbiota disorders. This review presents the key molecular mechanisms and signaling pathways of the active ingredients in P. ginseng involved in improving AD from the perspective of AD pathogenesis. A P. ginseng-related signaling pathway network was constructed to provide effective targets for the treatment of AD. In addition, the application of spatial metabolomics techniques in studying P. ginseng and AD is discussed. In summary, this paper discusses research perspectives for the study of P. ginseng in the treatment of AD, including a systematic and in-depth review of the mechanisms of action of the active substances in P. ginseng, and evaluates the feasibility of applying spatial metabolomics in the study of AD pathogenesis and pharmacological treatment.
Collapse
Affiliation(s)
| | | | | | | | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.Z.); (H.N.); (Q.L.); (L.J.)
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.Z.); (H.N.); (Q.L.); (L.J.)
| |
Collapse
|
25
|
Gong Q, Wang Y, Wang X, Pan H, Yan C. Baicalein promotes the microglia M2 polarization and suppresses apoptosis by targeting HMOX1/PDE4D to alleviate Alzheimer's disease. Immunobiology 2023; 228:152761. [PMID: 38006681 DOI: 10.1016/j.imbio.2023.152761] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/28/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that has quickly becoming one of the most expensive, lethal, and burdening diseases of this century. In the past twenty years, hundreds of drugs have been tested while only several have been authorized by FDA for AD treatment, hence, searching for candidate agent with therapeutic potential for AD is imminent. Controlling polarization direction of microglia is crucial in AD therapy. Recent research suggests that baicalein has potential to reduce neuroinflammation and prevent neurodegenerative diseases by affecting microglia, while the specific molecular mechanism of baicalein in regulating microglia in the treatment of AD is still unclear. In this study, we investigated how baicalein affected microglial polarization in AD and potential biological mechanisms. In cell experiments, it was verified that baicalein significantly shifted the BV-2 microglia phenotype from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype, inhibited the microglial apoptosis and pro-inflammatory factors, promoted the microglial Aβ uptake and anti-inflammatory factors after LPS stimulated. In APP/PS1 mice, it was found that baicalein decreased the Aβ plaque deposition in brain, attenuated NLRP3 inflammasome activation and neuronal apoptosis in APP/PS1 mice. Furthermore, bioinformatics analysis and experiment validated that HMOX1 is a target of baicalein, and we elucidated that baicalein modulated the microglial polarization to inhibit neuroinflammation and neural injury through targeting on the HMOX1/PDE4D axis in AD. In conclusion, our findings indicate the therapeutic effect of baicalein for AD, and baicalein might serve a potential agent for AD treatment.
Collapse
Affiliation(s)
- Qingmei Gong
- Department of Neurology, the Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province 310000, China
| | - Yanbo Wang
- Department of Neurology, the Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province 310000, China
| | - Xiaowei Wang
- Department of Respiratory, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province 310000, China
| | - Haiyan Pan
- Department of Endocrinology, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province 310000, China
| | - Ci Yan
- Departments of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310000, China.
| |
Collapse
|
26
|
Liu J, Li T, Zhong G, Pan Y, Gao M, Su S, Liang Y, Ma C, Liu Y, Wang Q, Shi Q. Exploring the therapeutic potential of natural compounds for Alzheimer's disease: Mechanisms of action and pharmacological properties. Biomed Pharmacother 2023; 166:115406. [PMID: 37659206 DOI: 10.1016/j.biopha.2023.115406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023] Open
Abstract
Alzheimer's Disease (AD) is a global public health priority characterized by high mortality rates in adults and an increasing prevalence in aging populations worldwide. Despite significant advancements in comprehending the pathogenesis of AD since its initial report in 1907, there remains a lack of effective curative or preventive measures for the disease. In recent years, natural compounds sourced from diverse origins have garnered considerable attention as potential therapeutic agents for AD, owing to their anti-inflammatory, antioxidant, and neuroprotective properties. This review aims to consolidate the therapeutic effects of natural compounds on AD, specifically targeting the reduction of β-amyloid (Aβ) overproduction, anti-apoptosis, autophagy, neuroinflammation, oxidative stress, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction. Notably, the identified compounds exhibiting these effects predominantly originate from plants. This review provides valuable insights into the potential of natural compounds as a reservoir of novel therapeutic agents for AD, thereby stimulating further research and contributing to the development of efficacious treatments for this devastating disease.
Collapse
Affiliation(s)
- Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Minghuang Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cuiru Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanyue Liu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210017, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qing Shi
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China.
| |
Collapse
|
27
|
Liu J, Zuo X, Huang M, Fang J, Li W, Shi Q, Wang Q, Liang Y. Multifunctional Gomisin B enhances cognitive function in APP/PS1 transgenic mice by regulating Aβ clearance and neuronal apoptosis. Biomed Pharmacother 2023; 166:115423. [PMID: 37673021 DOI: 10.1016/j.biopha.2023.115423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023] Open
Abstract
This study aimed to investigate the potential effects of Gomisin B, a natural compound known for its inhibition of CYP3A4, on cognitive dysfunction in APP/PS1 transgenic mice with Alzheimer's disease (AD). Additionally, the study explored the combined effects of Gomisin B and Osthole (OST). The research involved male wild-type (WT) mice and 7-month-old APP/PS1 transgenic AD mice. The assessment of behavioral changes included the use of the open field test (OFT) and the Morris water maze (MWM). OST levels in brain tissue were quantified using LC-MS/MS, while levels of oxidative stress were measured through an assay kit. Neuronal apoptosis was studied using Nissl staining, RT-qPCR, and immunofluorescence. Amyloid plaque clearance was assessed using thioflavine-S (Th-S) staining, RT-qPCR, and ELISA. The results of the study revealed that Gomisin B led to a significant improvement in cognitive dysfunction in APP/PS1 mice. Moreover, the simultaneous administration of OST and Gomisin B demonstrated enhanced therapeutic effects. These effects were attributed to the inhibition of β-site APP-Cleaving Enzyme 1 (BACE1) and oxidative stress by Gomisin B, along with its anti-apoptotic properties. The combined use of OST and Gomisin B exhibited a synergistic impact, resulting in more pronounced anti-oxidant and anti-apoptotic effects. In summary, this study pioneers the exploration of Gomisin B's multifunctional anti-AD properties in APP/PS1 mice. The findings provide a solid groundwork for the development of anti-Alzheimer's drugs based on natural active ingredients.
Collapse
Affiliation(s)
- Jinman Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Xue Zuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Mingjun Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing Shi
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
28
|
Wang X, Shi YJ, Niu TY, Chen TT, Li HB, Wu SH, Li GL. Neuroprotective effect of 20 (S) - Protopanaxadiol (PPD) attenuates NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia rats. Neurosci Lett 2023; 814:137439. [PMID: 37579868 DOI: 10.1016/j.neulet.2023.137439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
20(S)-protopanaxadiol (PPD), one of the ginsenosides from Panax ginseng, has been reported to improve performance with dementia. This study aimed to investigate the neuroprotective effect of PPD attenuating NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia (VD) rats induced by bilateral common carotid artery ligation (2-VO). Male Sprague-Dawley rats (SPF, 150-180 g, n = 10/group) were randomly divided into PPD (20, 10, 5 mg/kg, subcutaneous injection once per day for 3 weeks), model, and vehicle-sham group. It was found that PPD significantly reversed 2-VO-induced cognitive impairment by decreasing escape latency and spontaneous alternation and increasing the number of crossing platforms, showing memory-improving effects. PPD improved the pathological morphology of brain tissue in VD rats. PPD significantly reduced the cerebral infarction area and the activation of microglia in the cortex and hippocampal DG, CA1, and CA3 area. Moreover, PPD could attenuate NLRP3 inflammasome-mediated microglial pyroptosis, inhibit the positive expression of NLRP3, decrease IL-1β, and IL-18 levels, and increase IL-10 levels in the brain cortex. PPD also significantly alleviated the neurotoxicity by decreasing the Aβ and p-Tau in hippocampal DG, CA1, and CA3 areas. In addition, the levels of NLRP3, ASC, and IL-1β in the cortex, APP, BACE1, and p-Tau in the hippocampus were significantly reduced by PPD. These results suggested that PPD hinders microglial activation to alleviate neuroinflammation of NLRP3 inflammasome and inhibits neurotoxicity of Aβ deposition and Tau phosphorylation in 2-VO-induced VD rats.
Collapse
Affiliation(s)
- Xue Wang
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ya-Jin Shi
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ting-Yuan Niu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Ting-Ting Chen
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Han-Bing Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Su-Hui Wu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| | - Gen-Lin Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| |
Collapse
|
29
|
Zhou L, Huang X, Li H, Wang J, Lu Z. Triptolide improves Alzheimer's disease by regulating the NF‑κB signaling pathway through the lncRNA NEAT1/microRNA 361‑3p/TRAF2 axis. Exp Ther Med 2023; 26:440. [PMID: 37614428 PMCID: PMC10443046 DOI: 10.3892/etm.2023.12139] [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: 02/01/2023] [Accepted: 07/03/2023] [Indexed: 08/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and is a serious social and medical problem threatening human health. The present study investigated the effect and underlying action mechanism of triptolide (Tri) on AD progression. Reverse transcription-quantitative PCR and western blotting analysis were used to determine the changes in RNA expression and levels of NF-κB signaling pathway proteins before and after lipopolysaccharide (LPS) induction. Nucleocytoplasmic separation experiments determined the intracellular localization of long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1). A dual-luciferase assay was used to analyze the binding between NEAT1 and microRNA (miRNA/miR)-361 or tumor necrosis factor receptor-associated factor 2 (TRAF2) and miR-361-3p and RNA pull-down was used to analyze the binding between NEAT1 and miR-361-3p. Cell Counting Kit-8, flow cytometry and ELISA were used to detect the effects of interaction between Tri and NEAT1/miR-361-3p/TRAF2 on cell viability, apoptosis and inflammatory factor levels, respectively. The results showed that LPS-mediated human microglial clone 3 cell line (HMC3) viability decreased and apoptosis and inflammatory factors (IL-1β, IL-6, IL-18 and TNF-α) increased. Tri inhibited LPS-mediated effects in a dose-dependent manner by downregulating NEAT1 expression. NEAT1 is highly expressed in the cytoplasm and reduces the transcription and translation of downstream TRAF2 by acting as a competitive endogenous RNA that adsorbs miR-361-3p. LPS-mediated HMC3 cell injury, inflammation and activation of NF-κB signaling were partially reversed in presence of Tri. The miR-361-3p mimic promoted the Tri effect and overexpression of (ov)-NEAT1 partially reversed the Tri-miR-361-3p combined effect. The effects of ov-NEAT1 were partially attenuated by small interfering (si)-TRAF2. Overall, Tri inhibited the LPS-induced decrease in viability, increase in apoptosis and inflammation and activation of NF-κB signaling in HMC3 cells. Tri regulation affected the NEAT1/miR-361-3p/TRAF2 axis. These findings suggested a potential therapeutic role for Tri in the clinical management of AD by modulating this molecular axis.
Collapse
Affiliation(s)
- Li Zhou
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuming Huang
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Haiyan Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Jihui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
30
|
Ma Y, Fan C, Wang Y, Li W, Jiang H, Yang W. Comprehensive analysis of mRNAs in the cerebral cortex in APP/PS1 double-transgenic mice with Alzheimer's disease based on high-throughput sequencing of N4-acetylcytidine. Funct Integr Genomics 2023; 23:267. [PMID: 37548859 DOI: 10.1007/s10142-023-01192-z] [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: 02/11/2023] [Revised: 07/12/2023] [Accepted: 07/29/2023] [Indexed: 08/08/2023]
Abstract
N4-acetylcytidine (ac4C), a significant modified nucleoside, participates in the development of many diseases. Messenger RNAs (mRNAs) contain most of the information of the genome and are the molecules that transmit information from genes to proteins. Alzheimer's disease (AD) is a progressive neurodegenerative disease in which fibrillar amyloid plaques are present. However, it remains unknown how mRNA ac4C modification affects the development of AD. In the current study, ac4C-modified mRNAs were comprehensively analyzed in AD mice by ac4C-RIP-seq and RNA-seq. Next, a protein-protein interaction (PPI) network was constructed to examine the relationships between the genes with differential ac4C modification levels and their RNA expression levels. The differentially expressed genes (DEGs) acquired above were subjected to Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to further analyze the molecular mechanisms in AD. In total, 3312 significant ac4C peaks were found on 2512 mRNAs, 1241 of which were hyperacetylated and 1271 of which were hypoacetylated. In addition, 956 mRNAs with differential expression were found, including 520 upregulated mRNAs and 436 downregulated mRNAs. Overall, 134 mRNAs with simultaneous changes at the ac4C levels as well as RNA expression levels were identified via joint analysis. Then, through PPI network construction and functional enrichment analysis, 37 key mRNAs were screened, which were predominantly enriched in GABAergic synapses and the PI3K/AKT signaling pathway. The significant difference in the abundance of mRNA ac4C modification indicates that this modification is associated with AD progression, which may provide insight for more investigations of the potential mechanisms.
Collapse
Affiliation(s)
- Yanzhen Ma
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui, China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui, China
| | - Yongzhong Wang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui, China.
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Wenming Yang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui, China.
| |
Collapse
|
31
|
Cheng M, Ye C, Tian C, Zhao D, Li H, Sun Z, Miao Y, Zhang Q, Wang J, Dou Y. Engineered macrophage-biomimetic versatile nanoantidotes for inflammation-targeted therapy against Alzheimer's disease by neurotoxin neutralization and immune recognition suppression. Bioact Mater 2023; 26:337-352. [PMID: 36950153 PMCID: PMC10027514 DOI: 10.1016/j.bioactmat.2023.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Immune recognition of excessive neurotoxins by microglia is a trigger for the onset of neuroinflammation in the brain, leading to neurodegeneration in Alzheimer's disease (AD). Blocking active recognition of microglia while removing neurotoxins holds promise for fundamentally alleviating neurotoxin-induced immune responses, but is very challenging. Herein, an engineered macrophage-biomimetic versatile nanoantidote (OT-Lipo@M) is developed for inflammation-targeted therapy against AD by neurotoxin neutralization and immune recognition suppression. Coating macrophage membranes can not only endow OT-Lipo@M with anti-phagocytic and inflammation-tropism capabilities to target inflammatory lesions in AD brain, but also efficiently reduce neurotoxin levels to prevent them from activating microglia. The loaded oxytocin (OT) can be slowly released to downregulate the expression of immune recognition site Toll-like receptor 4 (TLR4) on microglia, inhibiting TLR4-mediated pro-inflammatory signalling cascade. Benefiting from this two-pronged immunosuppressive strategy, OT-Lipo@M exhibits outstanding therapeutic effects on ameliorating cognitive deficits, inhibiting neuronal apoptosis, and enhancing synaptic plasticity in AD mice, accompanied by the delayed hippocampal atrophy and brain microstructural disruption by in vivo 9.4T MR imaging. This work provides new insights into potential AD therapeutics targeting microglia-mediated neuroinflammation at the source.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chunxiao Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, PR China
| | - Dongju Zhao
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Haonan Li
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
- Corresponding author.
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| |
Collapse
|
32
|
Shan M, Bai Y, Fang X, Lan X, Zhang Y, Cao Y, Zhu D, Luo H. American Ginseng for the Treatment of Alzheimer's Disease: A Review. Molecules 2023; 28:5716. [PMID: 37570686 PMCID: PMC10420665 DOI: 10.3390/molecules28155716] [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: 06/25/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent degenerative condition that is increasingly affecting populations globally. American ginseng (AG) has anti-AD bioactivity, and ginsenosides, as the main active components of AG, have shown strong anti-AD effects in both in vitro and in vivo studies. It has been reported that ginsenosides can inhibit amyloid β-protein (Aβ) production and deposition, tau phosphorylation, apoptosis and cytotoxicity, as well as possess anti-oxidant and anti-inflammatory properties, thus suppressing the progression of AD. In this review, we aim to provide a comprehensive overview of the pathogenesis of AD, the potential anti-AD effects of ginsenosides found in AG, and the underlying molecular mechanisms associated with these effects. Additionally, we will discuss the potential use of AG in the treatment of AD, and how ginsenosides in AG may exert more potent anti-AD effects in vivo may be a direction for further research.
Collapse
Affiliation(s)
- Mengyao Shan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yunfan Bai
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xiaoxue Fang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xintian Lan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yegang Zhang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yiming Cao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Difu Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Biopharmaceutical and Health Food, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
33
|
Kong L, Sun R, Zhou H, Shi Q, Liu Y, Han M, Li W, Qun S, Li W. Trpc6 knockout improves behavioral dysfunction and reduces Aβ production by inhibiting CN-NFAT1 signaling in T2DM mice. Exp Neurol 2023; 363:114350. [PMID: 36791875 DOI: 10.1016/j.expneurol.2023.114350] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/17/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023]
Abstract
As the prevalence of diabetes and health awareness increase, type 2 diabetes mellitus -associated cognitive dysfunction is receiving increasing attention. However, the pathogenesis is not entirely understood. Transient receptor potential cation channel 6 (TRPC6) is highly correlated with intracellular Ca2+ concentrations, and neuronal calcium overload is an important cause of cognitive dysfunction. In the present study, we investigated the effect and mechanism of Trpc6 knockout in high-fat diet and streptozotocin-induced T2DM mice. The body weight and fasting blood glucose were recorded during the experiment. Behavioral dysfunction was detected using the open field test (OFT), elevated plus maze (EPM), hole-board test (HBT), Morris water maze (MWM) test and contextual fear conditioning (CFC) test. Nissl and H&E staining were used to examine neuronal damage. Western blot, quantitative real-time polymerase chain reaction (q-PCR), and immunofluorescence were performed to detect amyloid beta protein (Aβ) deposition and related indicators of neurological impairments in the cerebral cortex and hippocampus. The results indicated that Trpc6 knockout inhibited body weight loss and fasting blood glucose increase, improved spontaneous activity, learning and memory dysfunction, and alleviated neuroinflammation and neuronal damage in T2DM mice. The further results demonstrated that Trpc6 knockout decreased Aβ generation and deposition, and reduced the expressions of inflammasome-related proteins in T2DM mice. In addition, Trpc6 knockout inhibited intracellular calcium overload in diabetic mice and primary cultured hippocampal neurons, which in turn suppressed CN and NFAT1 expression. These data suggest that Trpc6 knockout may inhibit the CN-NFAT1 signaling pathway by decreasing intracellular calcium overload in the brain of T2DM mice, which consequently reduce Aβ deposition and neuroinflammation, and ultimately delay the development of T2DM-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Ran Sun
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Huimsin Zhou
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Qifeng Shi
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Yan Liu
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Min Han
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Weiping Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Sen Qun
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
34
|
Yang SJ, Wang JJ, Cheng P, Chen LX, Hu JM, Zhu GQ. Ginsenoside Rg1 in neurological diseases: From bench to bedside. Acta Pharmacol Sin 2023; 44:913-930. [PMID: 36380226 PMCID: PMC10104881 DOI: 10.1038/s41401-022-01022-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Ginseng has been used in China as a superior medicinal material for thousands of years that can nourish the five internal organs, calm the mind and benefit wisdom. Due to its anti-inflammatory, antioxidant and neuroprotective activities, one of the active components of ginseng, ginsenoside Rg1, has been extensively investigated in the remedy of brain disorders, especially dementia and depression. In this review, we summarized the research progress on the action mechanisms of Rg1 ameliorating depression-like behaviors, including inhibition of hyperfunction of hypothalamic-pituitary-adrenal (HPA) axis, regulation of synaptic plasticity and gut flora. Rg1 may alleviate Alzheimer's disease in the early phase, as well as in the middle-late phases through repairing dendrite, axon and microglia- and astrocyte-related inflammations. We also proposed that Rg1 could regulate memory state (the imbalance of working and aversive memory) caused by distinct stimuli. These laboratory studies would further the clinical trials on Rg1. From the prospective of drug development, we discussed the limitations of the present investigations and proposed our ideas to increase permeability and bioavailability of Rg1. Taken together, Rg1 has the potential to treat neuropsychiatric disorders, but a future in-depth investigation of the mechanisms is still required. In addition, drug development will benefit from the clinical trials in one specific neuropsychiatric disorder.
Collapse
Affiliation(s)
- Shao-Jie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jing-Ji Wang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, 230061, China.
| | - Ping Cheng
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Li-Xia Chen
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jia-Min Hu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Guo-Qi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
35
|
Zhang ZY, Yuan X, He HY, He WB, Zhang Z, Chu SF. Upregulation of HBXIP contributed to the anti-DND by ginsenoside Rg1 after global cerebral ischemia-reperfusion. J Pharm Pharmacol 2023; 75:437-444. [PMID: 36734364 DOI: 10.1093/jpp/rgac097] [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: 05/03/2022] [Accepted: 12/05/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Ginsenoside Rg1 (Rg1) has been well-documented to be effective against ischemic/reperfusion (I/R) injury. However, whether it has therapeutic effect on delayed neuronal death is still unclear. The aim of this study is to investigate the effect of Rg1 on delayed neuronal death and elucidate its underlying mechanism. METHODS Delayed neuronal death model was prepared by global cerebral ischemia-reperfusion in rats, Rg1 was intravenously administered once a day. Nissl and Fluoro Jade B staining were carried out to evaluate the effect of Rg1 on delayed neuronal death. Western blot and qPCR were used to investigate the levels of HBXIP and Survivin. HBXIP/Survivin complex was observed by co-immunoprecipitation. AAV-CMV-shRNA (HBXIP) was used to observe the role of HBXIP on delayed neuronal death improved by Rg1. KEY FINDINGS Rg1 attenuated delayed neuronal death at the dose of 20 mg/kg, which also improved the mRNA and protein levels of HBXIP, as well as Survivin. Moreover, administration of Rg1 promoted the formation of HBXIP/Survivin complex, which contributed to the reduction of caspases signaling pathway. Knockdown of HBXIP abolished the alleviation of DND and inhibition of caspase cascade induced by Rg1. CONCLUSIONS Rg1 alleviated delayed neuronal death by promoting anti-apoptosis effect by HBXIP/Survivin complex.
Collapse
Affiliation(s)
- Zhi-Yong Zhang
- Department of Neurology, Beijing Geriatric Hospital, Beijing 100095, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing100191, China
| | - Hong-Yuan He
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Shanxi University of Chinese Medicine, Taiyuan 030024, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shi-Feng Chu
- Department of Neurology, Beijing Geriatric Hospital, Beijing 100095, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
36
|
Wang Z, Zhang Z, Liu J, Guo M, Li H. Panax Ginseng in the treatment of Alzheimer's disease and vascular dementia. J Ginseng Res 2023. [DOI: 10.1016/j.jgr.2023.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
|
37
|
Structure, Activation, and Regulation of NOX2: At the Crossroad between the Innate Immunity and Oxidative Stress-Mediated Pathologies. Antioxidants (Basel) 2023; 12:antiox12020429. [PMID: 36829988 PMCID: PMC9952346 DOI: 10.3390/antiox12020429] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) is a multisubunit enzyme complex that participates in the generation of superoxide or hydrogen peroxide (H2O2) and plays a key role in several biological functions. Among seven known NOX isoforms, NOX2 was the first identified in phagocytes but is also expressed in several other cell types including endothelial cells, platelets, microglia, neurons, and muscle cells. NOX2 has been assigned multiple roles in regulating many aspects of innate and adaptive immunity, and human and mouse models of NOX2 genetic deletion highlighted this key role. On the other side, NOX2 hyperactivation is involved in the pathogenesis of several diseases with different etiologies but all are characterized by an increase in oxidative stress and inflammatory process. From this point of view, the modulation of NOX2 represents an important therapeutic strategy aimed at reducing the damage associated with its hyperactivation. Although pharmacological strategies to selectively modulate NOX2 are implemented thanks to new biotechnologies, this field of research remains to be explored. Therefore, in this review, we analyzed the role of NOX2 at the crossroads between immunity and pathologies mediated by its hyperactivation. We described (1) the mechanisms of activation and regulation, (2) human, mouse, and cellular models studied to understand the role of NOX2 as an enzyme of innate immunity, (3) some of the pathologies associated with its hyperactivation, and (4) the inhibitory strategies, with reference to the most recent discoveries.
Collapse
|
38
|
She L, Xiong L, Li L, Zhang J, Sun J, Wu H, Ren J, Wang W, Zhao X, Liang G. Ginsenoside Rk3 ameliorates Aβ-induced neurotoxicity in APP/PS1 model mice via AMPK signaling pathway. Biomed Pharmacother 2023; 158:114192. [PMID: 36587558 DOI: 10.1016/j.biopha.2022.114192] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) has become a major public health problem affecting the elderly population, and there is currently no effective treatment. Although the pathogenesis of AD is unclear, neurotoxicity induced by oxidative stress plays an important role in the progression of AD. Ginseng, the root and rhizome of Panax ginseng C. A. Meyer, is used not only as an herbal medicine but also as a functional food to support bodily functions. Ginsenoside Rk3 (Rk3), the main bioactive component in ginseng, has a strong antioxidant effect and has not been reported in AD. In this study, we showed that Rk3 improved neuronal apoptosis, decreased intracellular reactive oxygen species (ROS) production and restored mitochondrial membrane potential in PC12 and primary neuronal cells. In vivo, we found that Rk3 improved spatial learning and memory deficit in precursor protein (APP)/presenilin 1 (PS1) double transgenic mouse model of AD. Additionally, Rk3 increases glutathione reductase (GSH) and superoxide dismutase (SOD) levels while inhibits malondialdehyde (MDA) production, apoptosis and activation of glial cells in APP/PS1 mice. Mechanistically, we found that the protective effect of Rk3 is in correlation with the activation of AMPK/Nrf2 signaling pathway. In conclusion, the findings of this study provide support for Rk3 as a new strategy for the treatment of AD.
Collapse
Affiliation(s)
- Lingyu She
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China; Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Li Xiong
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Liwei Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Jing Zhang
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China
| | - Jinfeng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Haibin Wu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Juan Ren
- School of Laboratory Medicine and Bioengineer, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Wei Wang
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China.
| | - Xia Zhao
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| | - Guang Liang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China; Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, Zhejiang 321399, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
39
|
Campos HC, Ribeiro DE, Hashiguchi D, Glaser T, Milanis MDS, Gimenes C, Suchecki D, Arida RM, Ulrich H, Monteiro Longo B. Neuroprotective effects of resistance physical exercise on the APP/PS1 mouse model of Alzheimer's disease. Front Neurosci 2023; 17:1132825. [PMID: 37090809 PMCID: PMC10116002 DOI: 10.3389/fnins.2023.1132825] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/16/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction Physical exercise has beneficial effects by providing neuroprotective and anti-inflammatory responses to AD. Most studies, however, have been conducted with aerobic exercises, and few have investigated the effects of other modalities that also show positive effects on AD, such as resistance exercise (RE). In addition to its benefits in developing muscle strength, balance and muscular endurance favoring improvements in the quality of life of the elderly, RE reduces amyloid load and local inflammation, promotes memory and cognitive improvements, and protects the cortex and hippocampus from the degeneration that occurs in AD. Similar to AD patients, double-transgenic APPswe/PS1dE9 (APP/PS1) mice exhibit Αβ plaques in the cortex and hippocampus, hyperlocomotion, memory deficits, and exacerbated inflammatory response. Therefore, the aim of this study was to investigate the effects of 4 weeks of RE intermittent training on the prevention and recovery from these AD-related neuropathological conditions in APP/PS1 mice. Methods For this purpose, 6-7-month-old male APP/PS1 transgenic mice and their littermates, negative for the mutations (CTRL), were distributed into three groups: CTRL, APP/PS1, APP/PS1+RE. RE training lasted four weeks and, at the end of the program, the animals were tested in the open field test for locomotor activity and in the object recognition test for recognition memory evaluation. The brains were collected for immunohistochemical analysis of Aβ plaques and microglia, and blood was collected for plasma corticosterone by ELISA assay. Results APP/PS1 transgenic sedentary mice showed increased hippocampal Aβ plaques and higher plasma corticosterone levels, as well as hyperlocomotion and reduced central crossings in the open field test, compared to APP/PS1 exercised and control animals. The intermittent program of RE was able to recover the behavioral, corticosterone and Aβ alterations to the CTRL levels. In addition, the RE protocol increased the number of microglial cells in the hippocampus of APP/PS1 mice. Despite these alterations, no memory impairment was observed in APP/PS1 mice in the novel object recognition test. Discussion Altogether, the present results suggest that RE plays a role in alleviating AD symptoms, and highlight the beneficial effects of RE training as a complementary treatment for AD.
Collapse
Affiliation(s)
- Henrique Correia Campos
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Debora Hashiguchi
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto do Cérebro - ICe, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Milena da Silva Milanis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Christiane Gimenes
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ricardo Mario Arida
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| | - Beatriz Monteiro Longo
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| |
Collapse
|
40
|
Ma Y, Li W, Fan C, Wang Y, Jiang H, Yang W. Comprehensive Analysis of Long Non-Coding RNAs N4-Acetylcytidine in Alzheimer's Disease Mice Model Using High-Throughput Sequencing. J Alzheimers Dis 2022; 90:1659-1675. [PMID: 36314201 DOI: 10.3233/jad-220564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND N4-acetylcytidine (ac4C), an important posttranscriptional modification, is involved in various disease processes. Long noncoding RNAs (lncRNAs) regulate gene expression mainly through epigenetic modification, transcription, and posttranscriptional modification. Alzheimer's disease (AD) is a neurodegenerative disease characterized by amyloidosis of the brain. However, the role of lncRNA ac4C modification in AD remains unclear. OBJECTIVE In this study, we investigated the association between ac4C modification and AD, and the underlying mechanisms of ac4C modification in AD. METHODS The male 9-month-old APP/PS1 double transgenic mice, age- and sex-matched wild type (WT) mice were used in this study. Then, ac4C-RIP-seq and RNA-seq were used to comprehensively analyze lncRNA ac4C modification in AD mice. The lncRNA-miRNA-mRNA regulatory networks using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses showed the regulatory relationships among these three lncRNAs and AD. RESULTS The results showed that there were 120 significantly different ac4C peaks located on 102 lncRNAs in AD, of which 55 were hyperacetylated and 47 were hypoacetylated. Simultaneously, 231 differentially expressed lncRNAs were identified, including 138 upregulated lncRNAs and 93 downregulated lncRNAs. Moreover, 3 lncRNAs, lncRNA Gm26508, lncRNA A430046D13Rik, and lncRNA 9530059O14Rik, showed significant changes in both the ac4C and RNA levels using conjoint analysis. CONCLUSION The abundance of lncRNA ac4C modification is significantly different in AD and indicates that lncRNA ac4C is associated with the occurrence and development of AD, which could provide a basis for further exploration of the related regulatory mechanisms.
Collapse
Affiliation(s)
- Yanzhen Ma
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yongzhong Wang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.,Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wenming Yang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
41
|
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
|
42
|
Shi Z, Chen H, Zhou X, Yang W, Lin Y. Pharmacological effects of natural medicine ginsenosides against Alzheimer's disease. Front Pharmacol 2022; 13:952332. [PMID: 36467099 PMCID: PMC9708891 DOI: 10.3389/fphar.2022.952332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/08/2022] [Indexed: 08/04/2023] Open
Abstract
Ginsenosides are the most important pharmacological active ingredient of ginseng, with multiple biological therapeutic targets, mild action and no side effects. It is having shown beneficial effects in vitro and in vivo models of AD. In this review, we analyze large literature, summarize the inhibition of ginsenosides fibrous extracellular deposition of β-amyloid (Aβ) and neurofibrillary tangles (NFTs) of possible mechanisms, and explain the effects of ginsenosides on AD neuroprotection from the aspects of antioxidant, anti-inflammatory, and anti-apoptosis, prove the potential of ginsenosides as a new class of drugs for the treatment of AD. In addition, according to the current clinical application status of natural drugs, this paper analysis the delivery route and delivery mode of ginsenosides from the perspective of pharmacokinetics, providing a deeper insight into the clinical application of ginsenosides in the treatment of AD.
Collapse
Affiliation(s)
- Zhikun Shi
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, China
| | - Hongyu Chen
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xu Zhou
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yang Lin
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Yan YH, Huang ZH, Xiong QP, Song YW, Li SY, Yang BW, Sun L, Zhang MY, Ji Y. Effects of Broussonetia papyrifera (L.) L'Hér. ex Vent. fruits water extract on hippocampal neurogenesis in the treatment of APP/PS1 transgenic mice. Front Pharmacol 2022; 13:1056614. [DOI: 10.3389/fphar.2022.1056614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Adult neurogenesis plays an important role in repairing damaged neurons and improving cognitive impairment in Alzheimer’s disease (AD). B. Papyrifera (L.) L'Hér. ex Vent. fruits (BL), a traditional Chinese medicine for tonifying the kidney, has been reported to improve cognitive function in AD mice, but the underlying mechanisms have not been clearly illuminated. This study aimed to provide an overview of the differential compounds in the brain of APP/PS1 mice after BL water extract (BLWE) treatment through metabolomics technology and to elucidate whether the therapeutic effect and mechanism are through the enhancement of neurogenesis.Methods: APP/PS1 transgenic mice were treated with different doses of BLWE. After 6 weeks of intragastric injection, the therapeutic effects of BLWE on APP/PS1 transgenic mice were determined by the Morris water maze test, immunohistochemistry, hematoxylin & eosin and Nissl staining, enzyme-linked immunosorbent assay and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Subsequently, metabolomics technology was used to analyze the regulatory effect of BLWE on differential compounds in the brain of APP/PS1 mice, and on this basis, its molecular mechanism of BLWE was screened. Finally, the protein expression of the Wnt/β-catenin signaling pathway was detected by Western blotting.Results: After BLWE treatment, the learning and memory function of APP/PS1 mice were significantly improved, which was related to the increase in the number of Nestin+/BrdU+ and NeuN+/BrdU+ cells, and the decrease in the number of apoptotic cells in the hippocampus. BLWE treatment could also up-regulate the expression of synapse-associated proteins. Moreover, BLWE could modulate endogenous metabolic compounds in the brains of AD mice, including N-acetyl-aspartate, glutamine, etc. Furthermore, BLWE inhibited the phosphorylation of Tyr216-GSK-3β and β-catenin protein while increased CyclinD1 protein expression.Conclusion: We demonstrated that BLWE can enhance neural stem cells proliferation and improve neurogenesis, thereby efficiently repairing damaged neurons in the hippocampus and ameliorating cognitive impairment in APP/PS1 transgenic mice. The mechanism is at least partly through activating the Wnt/β-catenin signaling pathway.
Collapse
|
44
|
Feng H, Xue M, Deng H, Cheng S, Hu Y, Zhou C. Ginsenoside and Its Therapeutic Potential for Cognitive Impairment. Biomolecules 2022; 12:1310. [PMID: 36139149 PMCID: PMC9496100 DOI: 10.3390/biom12091310] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment (CI) is one of the major clinical features of many neurodegenerative diseases. It can be aging-related or even appear in non-central nerve system (CNS) diseases. CI has a wide spectrum that ranges from the cognitive complaint with normal screening tests to mild CI and, at its end, dementia. Ginsenosides, agents extracted from a key Chinese herbal medicine (ginseng), show great promise as a new therapeutic option for treating CI. This review covered both clinical trials and preclinical studies to summarize the possible mechanisms of how ginsenosides affect CI in different diseases. It shows that ginsenosides can modulate signaling pathways associated with oxidative stress, apoptosis, inflammation, synaptic plasticity, and neurogenesis. The involved signaling pathways mainly include the PI3K/Akt, CREB/BDNF, Keap1/Nrf2 signaling, and NF-κB/NLRP3 inflammasome pathways. We hope to provide a theoretical basis for the treatment of CI for related diseases by ginsenosides.
Collapse
Affiliation(s)
- Hui Feng
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Mei Xue
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Hao Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300073, China
| | - Shiqi Cheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330008, China
| | - Yue Hu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| |
Collapse
|
45
|
Liu Y, Huang L, Kim MY, Cho JY. The Role of Thymoquinone in Inflammatory Response in Chronic Diseases. Int J Mol Sci 2022; 23:ijms231810246. [PMID: 36142148 PMCID: PMC9499585 DOI: 10.3390/ijms231810246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Anti-inflammatory therapies have been shown to be effective in the prevention of various cardiovascular diseases, tumors, and cancer complications. Thymoquinone (TQ), the main active constituent of Nigella sativa, has shown promising therapeutic properties in many in vivo and in vitro models. However, TQ has poor bioavailability and is hydrophobic, prohibiting clinical trials with TQ alone. Studies have explored the combination of TQ with biological nanomaterials to improve its bioavailability. The TQ nanoparticle formulation shows better bioavailability than free TQ, and these formulations are ready for clinical trials to determine their potential as therapeutic agents. In this paper, we review current knowledge about the interaction between TQ and the inflammatory response and summarize the research prospects in Korea and abroad. We discuss the different biological activities of TQ and various combination therapies of TQ and nanomaterials in clinical trials.
Collapse
Affiliation(s)
- Yan Liu
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Lei Huang
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi-Yeon Kim
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.:+82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.:+82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
46
|
Shen Y, Qian L, Luo H, Li X, Ruan Y, Fan R, Si Z, Chen Y, Li L, Liu Y. The Significance of NLRP Inflammasome in Neuropsychiatric Disorders. Brain Sci 2022; 12:brainsci12081057. [PMID: 36009120 PMCID: PMC9406040 DOI: 10.3390/brainsci12081057] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/02/2022] Open
Abstract
The NLRP inflammasome is a multi-protein complex which mainly consists of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain. Its activation is linked to microglial-mediated neuroinflammation and partial neuronal degeneration. Many neuropsychiatric illnesses have increased inflammatory responses as both a primary cause and a defining feature. The NLRP inflammasome inhibition delays the progression and alleviates the deteriorating effects of neuroinflammation on several neuropsychiatric disorders. Evidence on the central effects of the NLRP inflammasome potentially provides the scientific base of a promising drug target for the treatment of neuropsychiatric disorders. This review elucidates the classification, composition, and functions of the NLRP inflammasomes. It also explores the underlying mechanisms of NLRP inflammasome activation and its divergent role in neuropsychiatric disorders, including Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, depression, drug use disorders, and anxiety. Furthermore, we explore the treatment potential of the NLRP inflammasome inhibitors against these disorders.
Collapse
Affiliation(s)
- Yao Shen
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Liyin Qian
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Hu Luo
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Xiaofang Li
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Yuer Ruan
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Runyue Fan
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
- Ningbo Yinzhou District Center for Disease Control and Prevention, Ningbo 315199, China
| | - Zizhen Si
- Department of Physiological Pharmacology, School of Medicine, Ningbo University, Ningbo 315021, China
- Department of Pharmacology, Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
| | - Yunpeng Chen
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Longhui Li
- Ningbo Kangning Hospital, Ningbo 315201, China
| | - Yu Liu
- Department of Physiological Pharmacology, School of Medicine, Ningbo University, Ningbo 315021, China
- Correspondence:
| |
Collapse
|
47
|
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
|
48
|
Zhang D, Ji P, Sun R, Zhou H, Huang L, Kong L, Li W, Li W. Ginsenoside Rg1 attenuates LPS-induced chronic renal injury by inhibiting NOX4-NLRP3 signaling in mice. Biomed Pharmacother 2022; 150:112936. [PMID: 35421784 DOI: 10.1016/j.biopha.2022.112936] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic renal injury (CRI) is a common pathological damage in chronic renal disease, and the therapeutic options for preventing its progression are limited at present. Ginsenoside Rg1 (Rg1) is reported to have a protective effect on renal injury by improving oxidative stress and inflammation. Lipopolysaccharide (LPS) plays important roles in inducing inflammatory and high-dose LPS is often used to perform acute renal injury. However, little is known about the effect of low-dose LPS on CRI, and the protective effect of Rg1 against chronic LPS-induced CRI. Here, we reported the protective effect and mechanism of Rg1 against LPS-induced CRI in mice. In this study, the results demonstrated that low-dose LPS (0.25 mg/kg) exposure for 14 days significantly induced renal function impairment and renal injury and fibrosis. Meanwhile, LPS exposure significantly increased reactive oxygen species (ROS) generation, NADPH oxidase 4 (NOX4) and NLRP3 inflammasome expression in renal cortex. However, treatment with Rg1, tempol (a superoxide dismutase mimetic), and apocynin (a NOX inhibitor) significantly improved renal function impairment and renal fibrosis, and significantly decreased the levels of TGF-β, IL-1β, KIM-1, β-Gal, and collagen IV in the kidneys. And Rg1 treatment also significantly reduced ROS generation and inhibited the activation of NOX4 and NLRP3 inflammasome. Overall, these results suggest that Rg1 treatment can ameliorate LPS-induced chronic kidney injury and renal fibrosis, the mechanisms may be involved in reducing NOX2-mediated oxidative stress and inhibiting NLRP1 inflammasome.
Collapse
Affiliation(s)
- Duoduo Zhang
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Pengmin Ji
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Ran Sun
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Huimin Zhou
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Lei Huang
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Liangliang Kong
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Weiping Li
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China; Anqing Medical and Pharmaceutical College, Anqing 246052, Anhui, China.
| | - Weizu Li
- Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, Anhui, China; Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
49
|
Wang J, Kong L, Guo RB, He SY, Liu XZ, Zhang L, Liu Y, Yu Y, Li XT, Cheng L. Multifunctional icariin and tanshinone IIA co-delivery liposomes with potential application for Alzheimer's disease. Drug Deliv 2022; 29:1648-1662. [PMID: 35616263 PMCID: PMC9154764 DOI: 10.1080/10717544.2022.2072543] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The blood-brain barrier (BBB) is a protective barrier for brain safety, but it is also a major obstacle to the delivery of drugs to the cerebral parenchyma such as the hippocampus, hindering the treatment of central nervous system diseases such as Alzheimer's disease (AD). In this work, an anti-AD brain-targeted nanodrug delivery system by co-loading icariin (ICA) and tanshinone IIA (TSIIA) into Aniopep-2-modified long-circulating (Ang2-ICA/TSIIA) liposomes was developed. Low-density lipoprotein receptor-related protein-1 (LRP1) was a receptor overexpressed on the BBB. Angiopep-2, a specific ligand of LRP1, exhibited a high binding efficiency with LRP1. Additionally, ICA and TSIIA, drugs with neuroprotective effects are loaded into the liposomes, so that the liposomes not only have an effective BBB penetration effect, but also have a potential anti-AD effect. The prepared Ang2-ICA/TSIIA liposomes appeared narrow dispersity and good stability with a diameter of 110 nm, and a round morphology. Cell uptake observations, BBB models in vitro, and imaging analysis in vivo showed that Ang2-ICA/TSIIA liposomes not only penetrate the BBB through endocytosis, but also accumulate in N2a cells or brain tissue. The pharmacodynamic analysis in vivo demonstrated that Ang2-ICA/TSIIA liposomes could improve AD-like pathological features in APP/PS1 mice, including inhibiting neuroinflammation and oxidative stress, reducing apoptosis, protecting neurons, and improving cognitive function. Therefore, Ang2-ICA/TSIIA liposomes are considered a potentially effective therapeutic strategy for AD.
Collapse
Affiliation(s)
- Jiao Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China.,Shenyang Medical College, Shenyang, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China.,Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Rui-Bo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Si-Yu He
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xin-Ze Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Lan Cheng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| |
Collapse
|
50
|
Wang C, Zheng C. Using Caenorhabditis elegans to Model Therapeutic Interventions of Neurodegenerative Diseases Targeting Microbe-Host Interactions. Front Pharmacol 2022; 13:875349. [PMID: 35571084 PMCID: PMC9096141 DOI: 10.3389/fphar.2022.875349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 12/02/2022] Open
Abstract
Emerging evidence from both clinical studies and animal models indicates the importance of the interaction between the gut microbiome and the brain in the pathogenesis of neurodegenerative diseases (NDs). Although how microbes modulate neurodegeneration is still mostly unclear, recent studies have started to probe into the mechanisms for the communication between microbes and hosts in NDs. In this review, we highlight the advantages of using Caenorhabditis elegans (C. elegans) to disentangle the microbe-host interaction that regulates neurodegeneration. We summarize the microbial pro- and anti-neurodegenerative factors identified using the C. elegans ND models and the effects of many are confirmed in mouse models. Specifically, we focused on the role of bacterial amyloid proteins, such as curli, in promoting proteotoxicity and neurodegeneration by cross-seeding the aggregation of endogenous ND-related proteins, such as α-synuclein. Targeting bacterial amyloid production may serve as a novel therapeutic strategy for treating NDs, and several compounds, such as epigallocatechin-3-gallate (EGCG), were shown to suppress neurodegeneration at least partly by inhibiting curli production. Because bacterial amyloid fibrils contribute to biofilm formation, inhibition of amyloid production often leads to the disruption of biofilms. Interestingly, from a list of 59 compounds that showed neuroprotective effects in C. elegans and mouse ND models, we found that about half of them are known to inhibit bacterial growth or biofilm formation, suggesting a strong correlation between the neuroprotective and antibiofilm activities. Whether these potential therapeutics indeed protect neurons from proteotoxicity by inhibiting the cross-seeding between bacterial and human amyloid proteins awaits further investigations. Finally, we propose to screen the long list of antibiofilm agents, both FDA-approved drugs and novel compounds, for their neuroprotective effects and develop new pharmaceuticals that target the gut microbiome for the treatment of NDs. To this end, the C. elegans ND models can serve as a platform for fast, high-throughput, and low-cost drug screens that target the microbe-host interaction in NDs.
Collapse
Affiliation(s)
| | - Chaogu Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|