1
|
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
|
2
|
Zheng Q, Lei FP, Hui S, Tong M, Liang LH. Ginsenoside Rb1 Relieves Cellular Senescence and Pulmonary Fibrosis by Promoting NRF2/QKI/SMAD7 Axis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2491-2509. [PMID: 39756830 DOI: 10.1142/s0192415x24500952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Cellular senescence is an adverse factor in the development of pulmonary fibrosis (PF). Ginsenoside Rb1 has been found to inhibit both cellular senescence and PF. This study aimed to elucidate the molecular mechanisms by which ginsenoside Rb1 regulates cellular senescence and PF. A PF mouse model was established by Bleomycin (BLM) administration, and a cell model of senescence was constructed using MRC-5 cells treated with Adriamycin RD (ARD) administration. Hematoxylin and Eosin (HE) staining and Masson staining were employed to evaluate cellular structure and collagen fiber content. RT-qPCR and western blotting were used to detect mRNA and protein expression of the target genes. Enzyme-linked Immunosorbent Assay (ELISA) was applied to measure the protein concentration of IL-1[Formula: see text] and IL-18. SA-[Formula: see text]-gal staining was used to evaluate cellular senescence. Our results show that ginsenoside Rb1 effectively suppressed BLM-induced PF in mice. ARD administration to induce cellular senescence reduced NRF2, QKI, and SMAD7 expression in MRC-5 cells. By inducing NRF2 overexpression, ARD-induced cellular senescence and fibrosis in MRC-5 cells were relieved. Notably, NRF2 knockdown abolished the mitigating effects of ginsenoside Rb1 on ARD-induced cellular senescence and fibrosis in MRC-5 cells. Mechanistically, NRF2 increased SMAD7 mRNA stability through the transcriptional regulation of QKI. As expected, ginsenoside Rb1 alleviated ARD-induced senescence and fibrosis in MRC-5 cells by activating the NRF2/QKI/SMAD7 axis. Therefore, it was found that ginsenoside Rb1 mitigates cellular senescence and fibrosis during PF progression by activating the NRF2/QKI/SMAD7 axis. This study provides a potential therapeutic strategy for the treatment of PF and elucidates its mechanism of action.
Collapse
Affiliation(s)
- Qing Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Feng-Ping Lei
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Shan Hui
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Ming Tong
- Department of Infectious Diseases, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Li-Hui Liang
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| |
Collapse
|
3
|
Zhou B, Sh G, Xie D, Zhao X, Hao B, Liu D, Wang M, Wu L, Lin L, Qian X. Ginsenoside Rb1 prevents age-related endothelial senescence by modulating SIRT1/caveolin-1/enos signaling pathway. Heliyon 2024; 10:e24586. [PMID: 38322899 PMCID: PMC10844051 DOI: 10.1016/j.heliyon.2024.e24586] [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: 08/15/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Background Advancing age is one of the independent risk factors for cardiovascular disorders. The Compendium of Materia Medica, a classic book on traditional Chinese medicine, states that ginseng "harmonizes the five internal organs, calming the spirit and prolonging the years of life." Considered one of the primary bioactive compounds derived from Panax ginseng, ginsenoside Rb1 (g-Rb1) has been scientifically suggested to possess anti-senescence efficacy. More research is needed to explore the vascular pharmacological activity and potential clinical application value of g-Rb1. Aims of the study Our previous study demonstrated that g-Rb1 could mitigate cellular senescence via the SIRT1/eNOS pathway. This study was performed to explore the exact mechanisms by which g-Rb1 modulates the SIRT1/eNOS pathway. Materials and methods We used human primary umbilical vein endothelial cells (HUVECs) to establish a replicative ageing model. Real-time (RT‒PCR), western blotting, small interfering RNA (siRNA), and immunoprecipitation were conducted to detect the effect of g-Rb1 on the SIRT1/caveolin-1/eNOS axis. Results G-Rb1 increased NO production and alleviated replicative senescence of HUVECs. The application of g-Rb1 elevated the mRNA and protein abundance of both SIRT1 and eNOS while concomitantly suppressing the expression of caveolin-1. Inhibition of SIRT1 and eNOS by siRNAs suppressed the anti-senescence function of g-Rb1, while caveolin-1 siRNA could enhance it. G-Rb1 decreased the acetylation level of caveolin-1 and increased NO production, which was suppressed by SIRT1 siRNA. Both g-Rb1 and caveolin-1 siRNA could reduce the acetylation level of eNOS and increase NO production. Conclusion G-Rb1 prevents age-related endothelial senescence by modulating the SIRT1/caveolin-1/eNOS signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | - Baoshun Hao
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dinhui Liu
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Min Wang
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Wu
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangying Lin
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoxian Qian
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Jouabadi SM, Ataabadi EA, Golshiri K, Bos D, Stricker BHC, Danser AHJ, Mattace-Raso F, Roks AJM. Clinical Impact and Mechanisms of Nonatherosclerotic Vascular Aging: The New Kid to Be Blocked. Can J Cardiol 2023; 39:1839-1858. [PMID: 37495207 DOI: 10.1016/j.cjca.2023.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Ischemic cardiovascular disease and stroke remain the leading cause of global morbidity and mortality. During aging, protective mechanisms in the body gradually deteriorate, resulting in functional, structural, and morphologic changes that affect the vascular system. Because atherosclerotic plaques are not always present along with these alterations, we refer to this kind of vascular aging as nonatherosclerotic vascular aging (NAVA). To maintain proper vascular function during NAVA, it is important to preserve intracellular signalling, prevent inflammation, and block the development of senescent cells. Pharmacologic interventions targeting these components are potential therapeutic approaches for NAVA, with a particular emphasis on inflammation and senescence. This review provides an overview of the pathophysiology of vascular aging and explores potential pharmacotherapies that can improve the function of aged vasculature, focusing on NAVA.
Collapse
Affiliation(s)
- Soroush Mohammadi Jouabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bruno H C Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Francesco Mattace-Raso
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
5
|
Tian T, Ko CN, Luo W, Li D, Yang C. The anti-aging mechanism of ginsenosides with medicine and food homology. Food Funct 2023; 14:9123-9136. [PMID: 37766674 DOI: 10.1039/d3fo02580b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
With the acceleration of global aging and the rise in living standards, the achievement of healthy aging is becoming an imperative issue globally. Ginseng, a medicinal plant that has a long history of dietary intake and remarkable medicinal value, has become a research hotspot in the field of food and medicine. Ginsenosides, especially protopanaxadiol-type saponins and protopanaxatriol-type saponins, are among the most important active ingredients in ginseng. Ginsenosides have been found to exhibit powerful and diverse pharmacological activities, such as antiaging, antitumor, antifatigue and immunity enhancement activities. Their effects in antiaging mainly include (1) promotion of metabolism and stem cell proliferation, (2) protection of skin and nerves, (3) modulation of intestinal flora, (4) maintenance of mitochondrial function, and (5) enhancement of telomerase activity. The underlying mechanisms are primarily associated with the intervention of the signaling pathways in apoptosis, inflammation and oxidative stress. In this review, the mechanism of action of ginsenosides in antiaging as well as the potential values of developing ginsenoside-based functional foods and antiaging drugs are discussed.
Collapse
Affiliation(s)
- Tiantian Tian
- Center for Biological Science and Technology, Beijing Normal University, Zhuhai, Guangdong Province, 519087, China
| | - Chung-Nga Ko
- C-MER Dennis Lam and Partners Eye Center, Hong Kong International Eye Care Group, Hong Kong, China
| | - Wenya Luo
- Haikou Orthopedics and Diabetes Hospital, Haikou, Hainan, 570206, China
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, Zhejiang Province, 316022, China.
| |
Collapse
|
6
|
Meng Z, Lu J, Ge G, Wang G, Zhang R, Li Y, Guan S, Lu J. Ginsenoside Rb1 induces autophagic lipid degradation via miR-128 targeting TFEB. Food Funct 2023; 14:240-249. [PMID: 36484324 DOI: 10.1039/d2fo02719d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, the effect of lipid metabolism on health has attracted more and more attention. Ginseng is a traditional Chinese herbal medicine in China and is widely used as food in Asia. Ginsenoside Rb1 (Gs-Rb1) is the most abundant ingredient in ginsenoside, which has a variety of biological activities. In this study, we found that Gs-Rb1 can reduce lipid accumulation in mice and HepG2 cells induced by a high-fat diet (HFD) and palmitic acid (PA). At the same time, we also found that Gs-Rb1 could stimulate the autophagic flux of HFD-fed mice and PA-treated HepG2 cells, and it is further verified by adding the autophagy activator rapamycin (Rapa) and autophagy inhibitor chloroquine (CQ). Furthermore, we found that Gs-Rb1 promoted the nucleus translocation of the transcription factor EB (TFEB) and the target role of miR-128, thus stimulating autophagic flux. Therefore, our results showed that Gs-Rb1 enhanced the transcription of TFEB and its downstream lysosome-related genes by inhibiting miR-128, improved the degradation ability of lysosomes to autophagosomes, and then promoted autophagic lipid degradation.
Collapse
Affiliation(s)
- Zhuoqun Meng
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Jianing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China. .,Health Technology College, Jilin Sport University, Changchun, Jilin 130062, People's Republic of China
| | - Guangcai Ge
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Guang Wang
- Office of Laboratory and Equipment Management, Jilin University, Changchun, Jilin 130000, People's Republic of China
| | - Ran Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Yuhan Li
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China. .,Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China. .,Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
7
|
Cai Z, Chen Y. Synergetic protective effect of berberine and ginsenoside Rb1 against tumor necrosis factor alpha-induced inflammation in adipocytes. Bioengineered 2021; 12:11784-11796. [PMID: 34699329 PMCID: PMC8810088 DOI: 10.1080/21655979.2021.1996508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Obesity significantly impacts living a normal life by increasing morbidity. Additionally, obesity has been shown to be closely associated with severe inflammation in adipocytes. It is widely reported that berberine (BBR) has an anti-inflammatory effect and can reduce glucose and lipid accumulation, whereas ginsenoside Rb1 (Rb1) has been shown to have a significant inhibitory effect on insulin resistance and lipid peroxidation. In this study, we aimed to explore the synergetic effect of BBR and Rb1 on tumor necrosis factor alpha (TNF-α)-treated adipocytes and the mechanisms underlying it. We found that TNF-α reduced cell viability, facilitated the production of inflammatory factors, induced adipogenesis, activated the nuclear factor kappa B (NF-κB) pathway, and increased the expression of peroxisome proliferator-activated receptor gamma, CCAAT enhancer-binding protein alpha, and sterol regulatory element-binding protein-1 c in adipocytes. However, these effects were significantly alleviated by BBR or Rb1. Additionally, a synergetic effect was observed when BBR and Rb1 were used in combination. The effects of BBR in combination with Rb1 on cell proliferation, inflammation, adipogenesis, and the NF-κB pathway in TNF-α-treated adipocytes were significantly abolished by receptor activator of nuclear factor kappa-Β ligand, which is an activator of the NF-κB pathway. Collectively, the results revealed that BBR and Rb1 have a synergetic protective effect against TNF-α-induced inflammation in adipocytes. The mechanism underlying this synergetic effect was found to be inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhixing Cai
- Department of Traditional Chinese Medicine, Tongren Hospital, Shanghai, China
| | - Yue Chen
- Department of Traditional Chinese Medicine, Tongren Hospital, Shanghai, China
| |
Collapse
|
8
|
Kim TH, Kim JY, Bae J, Kim YM, Won MH, Ha KS, Kwon YG, Kim YM. Korean Red ginseng prevents endothelial senescence by downregulating the HO-1/NF-κB/miRNA-155-5p/eNOS pathway. J Ginseng Res 2020; 45:344-353. [PMID: 33841015 PMCID: PMC8020293 DOI: 10.1016/j.jgr.2020.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/05/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022] Open
Abstract
Background Korean Red ginseng extract (KRGE) has beneficial effects on the cardiovascular system by improving endothelial cell function. However, its pharmacological effect on endothelial cell senescence has not been clearly elucidated. Therefore, we examined the effect and molecular mechanism of KRGE on the senescence of human umbilical vein endothelial cells (HUVECs). Methods HUVECs were grown in normal or KRGE-supplemented medium. Furthermore, they were transfected with heme oxygenase-1 (HO-1) gene or treated with its inhibitor, a NF-κB inhibitor, and a miR-155-5p mimic or inhibitor. Senescence-associated characteristics of endothelial cells were determined by biochemical and immunohistochemical analyses. Results Treatment of HUVECs with KRGE resulted in delayed onset and progression of senescence-associated characteristics, such as increased lysosomal acidic β-galactosidase and decreased telomerase activity, angiogenic dysfunction, and abnormal cell morphology. KRGE preserved the levels of anti-senescent factors, such as eNOS-derived NO, MnSOD, and cyclins D and A: however, it decreased the levels of senescence-promoting factors, such as ROS, activated NF-κB, endothelial cell inflammation, and p21 expression. The beneficial effects of KRGE were due to the induction of HO-1 and the inhibition of NF-κB-dependent biogenesis of miR-155-5p that led to the downregulation of eNOS. Moreover, treatment with inhibitors of HO-1, NF-κB, and miR-155-5p abolished the anti-senescence effects of KRGE. Conclusion KRGE delayed or prevented HUVEC senescence through a signaling cascade involving the induction of HO-1, the inhibition of NF-κB-dependent miR-155-5p biogenesis, and the maintenance of the eNOS/NO axis activity, suggesting that it may protect against vascular diseases associated with endothelial senescence.
Collapse
Affiliation(s)
- Tae-Hoon Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Jieun Bae
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Young-Mi Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| |
Collapse
|
9
|
Yu S, Xia H, Guo Y, Qian X, Zou X, Yang H, Yin M, Liu H. Ginsenoside Rb1 retards aging process by regulating cell cycle, apoptotic pathway and metabolism of aging mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 255:112746. [PMID: 32165173 DOI: 10.1016/j.jep.2020.112746] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/26/2020] [Accepted: 03/06/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginsenoside Rb1 (GRb1), an active ingredient of traditional Chinese medicine Panax ginseng C. A. Meyer, has displayed various activities such as antioxidative stress, autophagic regulation and apoptotic inhibition. However, the role of GRb1 in natural aging process remains unclear. AIM OF THE STUDY In this study, we investigated the anti-aging effect and underlying molecular mechanisms of ginsenoside Rb1 in natural aging process. MATERIALS AND METHODS We treated the natural aging C57BL/6J mice by intragastrical administration of GRb1 (100 mg/kg·BW) every other day for 10 months and investigated the effect of GRb1 on aging symptoms. By RT-qPCR and WB analysis, we examined the expression levels of senescence-associated biomarkers and aging-related pathways, including cell cycle, apoptosis and inflammation in aging process. Further, metabolomics analysis was conducted to investigate the changes of aging-related metabolites after GRb1 treatment. RESULTS Treatment with GRb1 significantly attenuated the aging-induced physiological changes, including slowed reduction of body weight, suppression of hair loss, decrease of arterial wall thickness and heart weight. We found that GRb1 treatment remarkably reversed the changed expression of p53-p21-Cdk2 axis in heart tissues of aging mice, which was responsible for the cell cycle repression. And the activations of apoptosis-associated factors (Bax and Caspase-3) were also inhibited by GRb1 treatment. Further, based on the serum metabolomics analysis using HPLC-MS/MS analysis, several metabolites were identified as potential biomarkers related to the anti-aging effect of GRb1, including glycerophospholipids, carboxylic acids and fatty acyls. Especially, the change of glycerophospholipid metabolism pathway was found to be the mostly changed. CONCLUSION Our studies suggest that GRb1 retards the aging process in mice by regulating cell cycle and apoptotic pathway, which were associated with the alleviation of metabolic disorders.
Collapse
Affiliation(s)
- Shujie Yu
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, China.
| | - Hui Xia
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing, 400065, China.
| | - Xiaoxian Qian
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, China.
| | - Xiaojuan Zou
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
| | - Huabing Yang
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
| | - Mingzhu Yin
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Hongtao Liu
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
| |
Collapse
|
10
|
Zheng Z, Wang M, Cheng C, Liu D, Wu L, Zhu J, Qian X. Ginsenoside Rb1 reduces H2O2‑induced HUVEC dysfunction by stimulating the sirtuin‑1/AMP‑activated protein kinase pathway. Mol Med Rep 2020; 22:247-256. [PMID: 32377712 PMCID: PMC7248484 DOI: 10.3892/mmr.2020.11096] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Endothelial dysfunction and senescence are closely associated with cardiovascular diseases including atherosclerosis and hypertension. Ginsenoside Rb1 (Rb1), the major active constituent of ginseng, has been investigated intensively because of its anti-obesity and anti-inflammatory effects. In a previous study, hydrogen peroxide (H2O2) was applied to induce human umbilical vein endothelial cell (HUVEC) aging. It was demonstrated that Sirtuin-1 (SIRT1) was activated by Rb1 to protect HUVECs from H2O2-induced senescence. However, the mechanisms are not fully understood. The present study examined the role of AMP-activated protein kinase (AMPK), an energy sensor of cellular metabolism, in the signaling pathway of SIRT1 during H2O2-stimulated HUVEC aging. It was identified that Rb1 restored the H2O2-induced reduction of SIRT1 expression, which was consistent with our previous study, together with the activation of AMPK phosphorylation. Using compound C, an AMPK inhibitor, the role of AMPK in the protective effect of Rb1 against H2O2-induced HUVEC senescence was examined. It was identified that the induction of phosphorylated AMPK by Rb1 markedly increased endothelial nitric oxide synthase expression and nitric oxide production, and suppressed PAI-1 expression, which were abrogated in HUVECs pretreated with compound C. Further experiments demonstrated that nicotinamide, a SIRT1 inhibitor, downregulated the phosphorylation of AMPK and reduced the protective effects of Rb1 against H2O2-induced endothelial aging. Taken together, these results provide new insights into the possible molecular mechanisms by which Rb1 protects against H2O2-induced HUVEC senescence via the SIRT1/AMPK pathway.
Collapse
Affiliation(s)
- Zhenda Zheng
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Min Wang
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Cailian Cheng
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Dinghui Liu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Lin Wu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jieming Zhu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xiaoxian Qian
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
11
|
Ginsenoside Rb1 Alleviates Oxidative Low-Density Lipoprotein–Induced Vascular Endothelium Senescence via the SIRT1/Beclin-1/Autophagy Axis. J Cardiovasc Pharmacol 2020; 75:155-167. [DOI: 10.1097/fjc.0000000000000775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Gut Flora: Novel Therapeutic Target of Chinese Medicine for the Treatment of Cardiovascular Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3719596. [PMID: 31531111 PMCID: PMC6721502 DOI: 10.1155/2019/3719596] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 01/08/2023]
Abstract
Cardiovascular disease (CVD) is one of the three major threats to human health identified by WHO. Dyslipidemia, hypertension, diabetes, and obesity are well established as common CVD risk factors. However, controversies exist on the effects of gut flora on cardiovascular disease (CVD). Current evidence suggests that gut microbiota is a double-edged sword for CVD risk, and its effects are largely determined by the metabolites of the gut microbiota. Trimethylamine N-oxide (TMAO), as one of the metabolites of gut flora, is consistently associated with higher CVD risk. A few studies have emerged providing early evidence about the safety and efficacy of traditional Chinese medicine (TCM) in treating cardiovascular diseases by regulating gut flora. In this article, we review and interpret the existing evidence as well as explore the potential of intestinal flora as novel therapeutic targets of traditional Chinese medicine for the prevention of cardiovascular disease (CVD).
Collapse
|
13
|
Lee YS, Kim H, Kim J, Seol GH, Lee KW. Lancemaside A, a major triterpene saponin of Codonopsis lanceolata enhances regulation of nitric oxide synthesis via eNOS activation. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:110. [PMID: 31126276 PMCID: PMC6534936 DOI: 10.1186/s12906-019-2516-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/30/2019] [Indexed: 02/08/2023]
Abstract
Background Many studies on the effect of saponin-rich Codonopsis lanceolata as a bioactive source for improving physical health have been performed. C. lanceolata contains triterpenoid saponins, including lancemasides. These saponins are known to be particularly involved in the regulation of blood pressure or hypertension. This study investigated whether lancemaside A (LA), a major triterpenoid saponin from C. lanceolata, regulates nitric oxide (NO) production via the activation of endothelial NO synthase (eNOS) in human umbilical vein endothelial cells. Methods Upon separation with petroleum ether, ethyl acetate, and n-butanol, LA was found to be abundant in the n-butanol-soluble portion. For further purification of LA, HPLC was performed to collect fraction, and LA was identified using analysis of LC/MSMS and 13C-NMR values. In in vitro, the effects of LA on NO release mechanism in HUVECs were investigated by Griess assay, quantitative real-time reverse-transcription PCR, and Western blotting. Results Our results showed that NO production was efficiently improved by treatment with LA in a dose-dependent manner. In addition, the LA treatment resulted in extensive recovery of the NO production suppressed by the eNOS inhibitor, L-NAME, compared with that in the control group. Additionally, the level of eNOS mRNA was increased by this treatment in a dose-dependent manner. These results suggested that LA is an inducer of NO synthesis via eNOS mRNA expression. Also, the study indicated that LA is involved in activating the PI3K/Akt/eNOS signaling pathway. Conclusion These results suggested that LA is an inducer of NO synthesis via eNOS mRNA expression. Also, the study indicated that LA is involved in activating the PI3K/Akt/eNOS signaling pathway. These findings suggest the value of using LA as a component of functional foods and natural pharmaceuticals. Electronic supplementary material The online version of this article (10.1186/s12906-019-2516-6) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Lan Y, Li YJ, Li DJ, Li P, Wang JY, Diao YP, Ye GD, Li YF. Long noncoding RNA MEG3 prevents vascular endothelial cell senescence by impairing miR-128-dependent Girdin downregulation. Am J Physiol Cell Physiol 2018; 316:C830-C843. [PMID: 30576236 DOI: 10.1152/ajpcell.00262.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are commonly associated with various biological functions, in which the function of lncRNA maternally expressed gene 3 (MEG3) has been identified in various cancers. Strikingly, an association between MEG3 with microRNAs (miRNAs), mRNAs, and proteins has been reported. This study investigates the role of MEG3 in vascular endothelial cell (VEC) senescence. Expression of Girdin and miR-128 was monitored in the blood vessel samples of young and old mice/healthy volunteers, along with the measurement of human umbilical vein endothelial cells (HUVECs). The relationship between MEG3/Girdin and miR-128 was determined and verified. Loss- and gain-of-function approaches were applied to analyze the regulatory effects of MEG3 on platelet phagocytosis and lipoprotein oxidation of HUVEC membrane. In addition, the effect of MEG3 on HUVEC senescence was evaluated by detection of the reactive oxygen species, telomerase activity, and telomere length. To further analyze the MEG3-mediated regulatory mechanism, miR-128 upregulation and inhibition were introduced into the HUVECs. Downregulated Girdin and upregulated miR-128 were found in the blood vessels of old individuals and old mice, as well as in senescent HUVECs. MEG3 downregulation was found to be capable of inhibiting Girdin but enhancing miR-128 expression. It was also indicated to inhibit platelet phagocytosis and reduce telomerase activity and telomere length, while enhancing lipoprotein oxidation and reactive oxygen species production, which ultimately contributed in preventing and protecting HUEVCs from senescence. These findings provide evidence supporting that MEG3 leads to miR-128 downregulation and Girdin upregulation, which promotes platelet phagocytosis, thus protecting VECs from senescence.
Collapse
Affiliation(s)
- Yong Lan
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Da-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Peng Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Ji-Yang Wang
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Peng Diao
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Guo-Dong Ye
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yang-Fang Li
- Beijing Neurosurgical Institute, Capital Medical University , Beijing , People's Republic of China
| |
Collapse
|
15
|
Guo Y, Chao L, Chao J. Kallistatin attenuates endothelial senescence by modulating Let-7g-mediated miR-34a-SIRT1-eNOS pathway. J Cell Mol Med 2018; 22:4387-4398. [PMID: 29992759 PMCID: PMC6111868 DOI: 10.1111/jcmm.13734] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/08/2018] [Indexed: 01/18/2023] Open
Abstract
Kallistatin, a plasma protein, protects against vascular and organ injury. This study is aimed to investigate the role and mechanism of kallistatin in endothelial senescence. Kallistatin inhibited H2 O2 -induced senescence in human endothelial cells, as indicated by reduced senescence-associated-β-galactosidase activity, p16INK4a and plasminogen activator inhibitor-1 expression, and elevated telomerase activity. Kallistatin blocked H2 O2 -induced superoxide formation, NADPH oxidase levels and VCAM-1, ICAM-1, IL-6 and miR-34a synthesis. Kallistatin reversed H2 O2 -mediated inhibition of endothelial nitric oxide synthase (eNOS), SIRT1, catalase and superoxide dismutase (SOD)-2 expression, and kallistatin alone stimulated the synthesis of these antioxidant enzymes. Moreover, kallistatin's anti-senescence and anti-oxidant effects were attributed to SIRT1-mediated eNOS pathway. Kallistatin, via interaction with tyrosine kinase, up-regulated Let-7g, whereas Let-7g inhibitor abolished kallistatin's effects on miR-34a and SIRT1/eNOS synthesis, leading to inhibition of senescence, oxidative stress and inflammation. Furthermore, lung endothelial cells isolated from endothelium-specific kallistatin knockout mice displayed marked reduction in mouse kallistatin levels. Kallistatin deficiency in mouse endothelial cells exacerbated senescence, oxidative stress and inflammation compared to wild-type mouse endothelial cells, and H2 O2 treatment further magnified these effects. Kallistatin deficiency caused marked reduction in Let-7g, SIRT1, eNOS, catalase and SOD-1 mRNA levels, and elevated miR-34a synthesis in mouse endothelial cells. These findings indicate that endogenous kallistatin through novel mechanisms protects against endothelial senescence by modulating Let-7g-mediated miR-34a-SIRT1-eNOS pathway.
Collapse
Affiliation(s)
- Youming Guo
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| | - Lee Chao
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| | - Julie Chao
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| |
Collapse
|
16
|
Wang M, Chen Y, Xiong Z, Yu S, Zhou B, Ling Y, Zheng Z, Shi G, Wu Y, Qian X. Ginsenoside Rb1 inhibits free fatty acids‑induced oxidative stress and inflammation in 3T3‑L1 adipocytes. Mol Med Rep 2017; 16:9165-9172. [PMID: 28990058 DOI: 10.3892/mmr.2017.7710] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/14/2017] [Indexed: 11/05/2022] Open
Abstract
Free fatty acids (FFAs) increase in visceral fat and are inferred to be one of the underlying inducers of adipose tissue inflammation. In our previous study, it was demonstrated that ginsenoside Rb1 stimulates endothelial nitric oxide synthase (eNOS) and Sirtuin 1 to protect against endothelial cell senescence. In the present study, 3T3‑L1 adipocytes were exposed to 0.5 mM FFAs with or without Rb1 (10‑40 µM). Monocyte chemotactic protein‑1 (MCP‑1) and interleukin‑6 (IL‑6) secretion was measured using ELISA. Tumor necrosis factor‑α (TNF‑α) expression and nuclear factor‑κB (NF‑κB) p65 phosphorylation were detected using western blot analysis. Oxidative stress was determined via measuring intracellular reactive oxygen species (ROS) and nitric oxide (NO) production. The results demonstrated that MCP‑1 and IL‑6 secretion, as well as TNF‑α expression, were significantly increased following FFA treatment, which was attenuated by Rb1 in a dose‑dependent manner. Furthermore, Rb1 attenuated FFA‑induced NF‑κB phosphorylation, suggesting that the inhibitory effect of Rb1 on inflammatory cytokines was partially mediated through blockade of NF‑κB phosphorylation. Further experiments demonstrated that Rb1 ameliorated FFA‑induced ROS generation and NO reduction through upregulation of superoxide dismutase 2 and eNOS expression. Taken together, these results demonstrate proinflammatory and pro‑oxidant effects of FFA on 3T3‑L1 adipocytes, which are effectively ameliorated by Rb1. Suppression of inflammatory responses and oxidative stress may be a novel mechanism for attenuating the effect of Rb1 on adipocyte dysfunction.
Collapse
Affiliation(s)
- Min Wang
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yanming Chen
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhaojun Xiong
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Shujie Yu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Bin Zhou
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yesheng Ling
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhenda Zheng
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Guangyao Shi
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yongxiang Wu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xiaoxian Qian
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
17
|
MicroRNAs in Hyperglycemia Induced Endothelial Cell Dysfunction. Int J Mol Sci 2016; 17:518. [PMID: 27070575 PMCID: PMC4848974 DOI: 10.3390/ijms17040518] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 01/15/2023] Open
Abstract
Hyperglycemia is closely associated with prediabetes and Type 2 Diabetes Mellitus. Hyperglycemia increases the risk of vascular complications such as diabetic retinopathy, diabetic nephropathy, peripheral vascular disease and cerebro/cardiovascular diseases. Under hyperglycemic conditions, the endothelial cells become dysfunctional. In this study, we investigated the miRNA expression changes in human umbilical vein endothelial cells exposed to different glucose concentrations (5, 10, 25 and 40 mM glucose) and at various time intervals (6, 12, 24 and 48 h). miRNA microarray analyses showed that there is a correlation between hyperglycemia induced endothelial dysfunction and miRNA expression. In silico pathways analyses on the altered miRNA expression showed that the majority of the affected biological pathways appeared to be associated to endothelial cell dysfunction and apoptosis. We found the expression of ten miRNAs (miR-26a-5p, -26b-5p, 29b-3p, -29c-3p, -125b-1-3p, -130b-3p, -140-5p, -192-5p, -221-3p and -320a) to increase gradually with increasing concentration of glucose. These miRNAs were also found to be involved in endothelial dysfunction. At least seven of them, miR-29b-3p, -29c-3p, -125b-1-3p, -130b-3p, -221-3p, -320a and -192-5p, can be correlated to endothelial cell apoptosis.
Collapse
|
18
|
Zhao H, Li X, Li G, Sun BO, Ren L, Zhao C. Protective effects of monosialotetrahexosylganglioside sodium on H 2O 2-induced human vascular endothelial cells. Exp Ther Med 2015; 10:947-953. [PMID: 26622420 DOI: 10.3892/etm.2015.2603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 05/14/2015] [Indexed: 11/05/2022] Open
Abstract
Monosialotetrahexosylganglioside sodium (GM1) is widely used in the treatment of central and peripheral neurological injuries. In addition to its neuroprotective activity, GM1 exerts protective effects on brain microvascular endothelial cells, although the mechanisms underlying these effects remain unclear. The aim of the present study was to clarify the protective effects and underlying mechanisms of GM1 on human umbilical vein endothelial cells (HUVECs). In this study, hydrogen peroxide (H2O2) was applied to induce the HUVEC injury. HUVECs in a logarithmic growth phase were divided into five groups, namely the control, H2O2-treated, 10-mg/l GM1, 5-mg/l GM1 and 1-mg/l GM1 groups. In all the groups, cell proliferation was detected using a Cell Counting Kit-8 assay, a flow cytometric method was applied to analyze the cell cycle and nuclear factor (NF)-κB expression was evaluated using immunofluorescence analysis. In addition, the protein expression levels of NF-κB, phosphatidylinositol 3-kinase (PI3K) and glycogen synthase kinase (GSK)-3 were detected via western blot analysis. The results indicated that GM1 exerted significant protective effects on H2O2-injured cells by increasing the ratio of cells in the S/G2 phase. Furthermore, western blot analysis revealed that PI3K expression levels were markedly increased after 24 h, as a result of the GM1 treatment, while the expression of both GSK-3 markedly decreased. In addition, the ratio of nuclear-to-cytoplasmic NF-κB expression increased in the GM1-treated cells. In summary, GM1 exhibited marked protective effects on the HUVECs, possibly due to the ability of GM1 in maintaining the integrity of the endothelium and increasing the proportion of cells undergoing mitosis, a process in which the PI3K/GSK-3 and NF-κB pathways are crucially involved.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University, Changchun, Jilin 130021, P.R. China ; Department of Neurosurgery, Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guijie Li
- Department of Otorhinolaryngology, Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - B O Sun
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Conghai Zhao
- Department of Neurosurgery, Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
19
|
Cellular senescence: a hitchhiker’s guide. Hum Cell 2015; 28:51-64. [DOI: 10.1007/s13577-015-0110-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/03/2015] [Indexed: 12/21/2022]
|
20
|
Wang Y, Dong J, Liu P, Lau CW, Gao Z, Zhou D, Tang J, Ng CF, Huang Y. Ginsenoside Rb3 attenuates oxidative stress and preserves endothelial function in renal arteries from hypertensive rats. Br J Pharmacol 2015; 171:3171-81. [PMID: 24571453 DOI: 10.1111/bph.12660] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/06/2014] [Accepted: 02/20/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Panax ginseng is commonly used to treat cardiovascular conditions in Oriental countries. This study investigated the mechanisms underlying the vascular benefits of ginsenoside Rb3 (Rb3) in hypertension. EXPERIMENTAL APPROACH Rings of renal arteries were prepared from spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto (WKY) rats and were cultured ex vivo for 8 h. Contractile responses of the rings were assessed with myograph techniques. Expression of NADPH oxidases was assessed by Western blotting and immunohistochemistry. Reactive oxygen species (ROS) were measured using dihydroethidium fluorescence imaging and production of NO was determined using the fluorescent NO indicator DAF-FM diacetate in human umbilical vein endothelial cells. KEY RESULTS Ex vivo treatment with Rb3 concentration-dependently augmented endothelium-dependent relaxations, suppressed endothelium-dependent contractions and reduced ROS production and expressions of NOX-2, NOX-4 and p67(phox) in arterial rings from SHR. Rb3 treatment also normalized angiotensin II (Ang II)-stimulated elevation in ROS and expression of NOX-2 and NOX-4 in arterial rings from WKY rats. Rb3 inhibited Ang II-induced reduction of NO production and phosphorylation of endothelial NOS in cultures of human umbilical vein endothelial cells. Rb3 also inhibited oxidative stress in renal arterial rings from hypertensive patients or in Ang II-treated arterial rings from normotensive subjects. CONCLUSION AND IMPLICATIONS Ex vivo Rb3 treatment restored impaired endothelial function in arterial rings from hypertensives by reversing over-expression of NADPH oxidases and over-production of ROS, and improved NO bioavailability. Our findings suggest that medicinal plants containing Rb3 could decrease oxidative stress and protect endothelial function in hypertension.
Collapse
Affiliation(s)
- Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu D, Huang P, Yu Z, Xing DH, Ouyang S, Xing G. Efficacy and Safety of Panax notoginseng Saponin Therapy for Acute Intracerebral Hemorrhage, Meta-Analysis, and Mini Review of Potential Mechanisms of Action. Front Neurol 2015; 5:274. [PMID: 25620952 PMCID: PMC4288044 DOI: 10.3389/fneur.2014.00274] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022] Open
Abstract
Intracranial/intracerebral hemorrhage (ICH) is a leading cause of death and disability in people with traumatic brain injury (TBI) and stroke. No proven drug is available for ICH. Panax notoginseng (total saponin extraction, PNS) is one of the most valuable herb medicines for stroke and cerebralvascular disorders in China. We searched for randomized controlled clinical trials (RCTs) involving PNS injection to treat cerebral hemorrhage for meta-analysis from various databases including the Chinese Stroke Trials Register, the trials register of the Cochrane Complementary Medicine Field, the Cochrane Central Register of Controlled Trials, MEDLINE, Chinese BioMedical disk, and China Doctorate/Master Dissertations Databases. The quality of the eligible trials was assessed by Jadad’s scale. Twenty (20) of the 24 identified randomized controlled trials matched the inclusive criteria including 984 ICH patients with PNS injection and 907 ICH patients with current treatment (CT). Compared to the CT groups, PNS-treated patients showed better outcomes in the effectiveness rate (ER), neurological deficit score, intracranial hematoma volume, intracerebral edema volume, Barthel index, the number of patients died, and incidence of adverse events. Conclusion: PNS injection is superior to CT for acute ICH. A review of the literature shows that PNS may exert multiple protective mechanisms against ICH-induced brain damage including hemostasis, anti-coagulation, anti-thromboembolism, cerebral vasodilation, invigorated blood dynamics, anti-inflammation, antioxidation, and anti-hyperglycemic effects. Since vitamin C and other brain cell activators (BCA) that are not considered common practice were also used as parts of the CT in several trials, potential PNS and BCA interactions could exist that may have made the effect of PNS therapy less or more impressive than by PNS therapy alone. Future PNS trials with and without the inclusion of such controversial BCAs as part of the CT could clarify the situation. As PNS has a long clinical track record in Asia, it could potentially become a therapy option to treat ICH in the US and Europe. Further clinical trials with better experimental design could determine the long-term effects of PNS treatment for TBI and stroke.
Collapse
Affiliation(s)
- Dongying Xu
- Faculty of Nursing, Guangxi University of Chinese Medicine , Nanning , China
| | - Ping Huang
- Faculty of Nursing, Guangxi University of Chinese Medicine , Nanning , China
| | - Zhaosheng Yu
- Department of Oncology, Huanggang Hospital of Traditional Chinese Medicine , Huanggang , China
| | | | - Shuai Ouyang
- School of Business, University of Alberta , Edmonton, AB , Canada
| | | |
Collapse
|
22
|
The senescent hepatocyte gene signature in chronic liver disease. Exp Gerontol 2014; 60:37-45. [DOI: 10.1016/j.exger.2014.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/10/2014] [Accepted: 09/16/2014] [Indexed: 12/25/2022]
|
23
|
Tongxinluo decreases apoptosis of mesenchymal stem cells concentration-dependently under hypoxia and serum deprivation conditions through the AMPK/eNOS pathway. J Cardiovasc Pharmacol 2014; 63:265-73. [PMID: 24220313 DOI: 10.1097/fjc.0000000000000044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tongxinluo (TXL), a traditional Chinese medicine, is widely used to treat cardiovascular diseases in China. Our previous study has demonstrated the pro-survival role of TXL on mesenchymal stem cells (MSCs) in vivo. But whether TXL could decrease apoptosis of MSCs in vitro, and the underlying mechanism are still unknown. Moreover, AMPK/eNOS pathway is crucial in regulating cell apoptosis. Therefore, we designed the study to investigate whether TXL could decrease MSCs apoptosis under hypoxia and serum deprivation (H/SD) conditions and to determine the role of AMPK/eNOS pathway. To test the hypothesis, MSCs were treated with TXL (50-400 μg/mL) under H/SD for 6 hours. For inhibitor studies, the cells were preincubated with AMPK inhibitor compound C. Results indicated that TXL decreased MSCs apoptosis concentration-dependently evidenced by reduced Annexin V+/PI- cells and increased red/green ratio of JC-1. Further, TXL enhanced the phosphorylation of AMPK and eNOS. Whereas, treatment with compound C decreased the phosphorylation of AMPK and eNOS and was accompanied by attenuated anti-apoptotic effect of TXL. In conclusion, TXL protected MSCs against H/SD-induced injury at least in part through the AMPK/eNOS pathway, which provides a novel explanation for the multi-effect of TXL on cardiovascular system.
Collapse
|
24
|
Xu J, Huang Z, Lin L, Fu M, Song Y, Shen Y, Ren D, Gao Y, Su Y, Zou Y, Chen Y, Zhang D, Hu W, Qian J, Ge J. miRNA-130b is required for the ERK/FOXM1 pathway activation-mediated protective effects of isosorbide dinitrate against mesenchymal stem cell senescence induced by high glucose. Int J Mol Med 2014; 35:59-71. [PMID: 25355277 PMCID: PMC4249746 DOI: 10.3892/ijmm.2014.1985] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022] Open
Abstract
The present study was carried out to investigate the hypothesis that organic nitrates can attenuate the senescence of mesenchymal stem cells (MSCs), a superior cell source involved in the regeneration and repair of damaged tissue. MSCs were treated with high glucose (HG) in order to induce senescence, which was markedly attenuated by pre-treatment with isosorbide dinitrate (ISDN), a commonly used nitrate, as indicated by senescence-associated galactosidase (SA-β-gal) activity, p21 expression, as well as by the mRNA levels of DNA methyltransferase 1 (DNMT1) and differentiated embryo chondrocyte expressed gene 1 (DEC1), which are senescence-related biomarkers. It was also found that the senescent MSCs (induced by HG glucose) exhibited a marked downregulation in ERK activity and forkhead box M1 (FOXM1) expression, which was reversed by ISDN preconditioning. Of note, the inhibition of ERK phosphorylation or the downregulation of FOXM1 statistically abolished the favourable effects of ISDN. In addition, the investigation of the senescence-associated miR-130 family suggested that miR-130b mediates the beneficial effects of ISDN; it was found that the protective effects of ISDN against the senescence of MSCs were prominently reversed by the knockdown of miR-130b. Furthermore, the downregulation of ERK phosphorylation or FOXM1 expression decreased the miR-130b expression level; however, the suppression of miR-130b demonstrated no significant impact on ERK phosphorylation or FOXM1 expression. Taken together, to the best of our knowledge, the present study is the first to demonstrate the favourable effects of ISDN against HG-induced MSC senescence, which are mediated through the activation of the ERK/FOXM1 pathway and the upregulation of miR-130b.
Collapse
Affiliation(s)
- Jianfeng Xu
- Department of Cardiology, Minhang Hospital, Ruijin Hospital Group, Shanghai Jiaotong University School of Medicine, Shanghai 201199, P.R. China
| | - Zheyong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Li Lin
- Department of Cardiology, Eastern Hospital, Tongji University, Shanghai 200120, P.R. China
| | - Mingqiang Fu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yanan Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yunli Shen
- Department of Cardiology, Eastern Hospital, Tongji University, Shanghai 200120, P.R. China
| | - Daoyuan Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yanhua Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yangang Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yueguang Chen
- Department of Cardiology, Minhang Hospital, Ruijin Hospital Group, Shanghai Jiaotong University School of Medicine, Shanghai 201199, P.R. China
| | - Dadong Zhang
- Department of Cardiology, Minhang Hospital, Ruijin Hospital Group, Shanghai Jiaotong University School of Medicine, Shanghai 201199, P.R. China
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Ruijin Hospital Group, Shanghai Jiaotong University School of Medicine, Shanghai 201199, P.R. China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
25
|
Jamal J, Mustafa MR, Wong PF. Paeonol protects against premature senescence in endothelial cells by modulating Sirtuin 1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:428-436. [PMID: 24768807 DOI: 10.1016/j.jep.2014.04.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 04/12/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paeonol is a phenolic compound isolated mainly from Moutan cortex, root bark of Chinese Peony tree. Moutan cortex holds a significant value in traditional Chinese medicine for alleviating various oxidative stress-related diseases mainly atherosclerosis and myocardial infarction. The present study seeks to identify the protective mechanisms of paeonol in oxidative stress-induced premature senescence in endothelial cells. MATERIALS AND METHODS HUVECs were pretreated with paeonol or DMSO control at different doses for 24h prior to an exposure of 200μM of reactive oxygen species (ROS) inducer, hydrogen peroxide (H2O2). The protective effects of paeonol against H2O2-induced senescence were evaluated and the activation of Sirtuin 1 pathway by paeonol pretreatment was investigated in HUVECs. RESULTS Paeonol attenuated H2O2-induced cell growth arrest at G0/G1 phase, reduced the percentage of SA-β-Gal positive cells and increased BrdU incorporation. In addition, enzymatic Sirt1 activation assay indicated that paeonol significantly increased lysyl deactylase activity of Sirt1 enzyme with a fold change of 2.4±0.195 (p<0.05). Furthermore, pretreatment with paeonol significantly decreased the levels of p53, acetyl H3K14 and H4K16 protein expression upregulated by H2O2 stimulation. The changes in the histone protein levels were accompanied with an increase in Sirt1 protein expression level. CONCLUSION These findings suggest that paeonol protects endothelial cells against oxidative stress-induced premature senescence by modulating the expressions of Sirt1 protein and its substrates.
Collapse
Affiliation(s)
- Juliana Jamal
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
26
|
Xie W, Parker JL, Heaps CL. Exercise training-enhanced, endothelium-dependent dilation mediated by altered regulation of BK(Ca) channels in collateral-dependent porcine coronary arterioles. Microcirculation 2013; 20:170-82. [PMID: 23002811 DOI: 10.1111/micc.12016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/19/2012] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Test the hypothesis that exercise training increases the contribution of BK(Ca) channels to endothelium-mediated dilation in coronary arterioles from collateral-dependent myocardial regions of chronically occluded pig hearts and may function downstream of H2O2. METHODS An ameroid constrictor was placed around the proximal left circumflex coronary artery to induce gradual occlusion in Yucatan miniature swine. Eight weeks postoperatively, pigs were randomly assigned to sedentary or exercise training (treadmill; 14 week) regimens. RESULTS Exercise training significantly enhanced bradykinin-mediated dilation in collateral-dependent arterioles (~125 μm diameter) compared with sedentary pigs. The BK(Ca) -channel blocker, iberiotoxin alone or in combination with the H2O2 scavenger, polyethylene glycol catalase, reversed exercise training-enhanced dilation in collateral-dependent arterioles. Iberiotoxin-sensitive whole-cell K+ currents (i.e., BK(Ca)-channel currents) were not different between smooth muscle cells of nonoccluded and collateral-dependent arterioles of sedentary and exercise trained groups. CONCLUSIONS These data provide evidence that BK(Ca)-channel activity contributes to exercise training-enhanced endothelium-dependent dilation in collateral-dependent coronary arterioles despite no change in smooth muscle BK(Ca)-channel current. Taken together, our findings suggest that a component of the bradykinin signaling pathway, which stimulates BK(Ca) channels, is enhanced by exercise training in collateral-dependent arterioles and suggest a potential role for H2O2 as the mediator.
Collapse
Affiliation(s)
- Wei Xie
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas, USA
| | | | | |
Collapse
|
27
|
Cho IH. Effects of Panax ginseng in Neurodegenerative Diseases. J Ginseng Res 2013; 36:342-53. [PMID: 23717136 PMCID: PMC3659610 DOI: 10.5142/jgr.2012.36.4.342] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/21/2012] [Accepted: 05/21/2012] [Indexed: 12/11/2022] Open
Abstract
Ginseng, the root of the Panax ginseng, has been a popular and widely-used traditional herbal medicine in Korea, China, and Japan for thousands of years. Now it has become popular as a functional health food and is used globally as a natural medicine. Evidence is accumulating in the literature on the physiological and pharmacological effects of P. ginseng on neurodegenerative diseases. Possible ginseng- or ginsenosides-mediated neuroprotective mechanisms mainly involve maintaining homeostasis, and anti-inflammatory, anti-oxidant, anti-apoptotic, and immune-stimulatory activities. This review considers publications dealing with the various actions of P. ginseng that are indicative of possible neurotherapeutic efficacies in neurodegenerative diseases and neurological disorders such as Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis and multiple sclerosis.
Collapse
Affiliation(s)
- Ik-Hyun Cho
- Department of Anatomy, College of Oriental Medicine and Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
28
|
Lei J, Gu X, Ye Z, Shi J, Zheng X. Antiaging effects of simvastatin on vascular endothelial cells. Clin Appl Thromb Hemost 2012; 20:212-8. [PMID: 22964779 DOI: 10.1177/1076029612458967] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The anti-inflammatory, antioxidative, and antiarteriosclerosis activities of simvastatin along with its protective effects on the endothelium suggest that it may also have antiaging effects. The aim of this study was to investigate the antiaging effects of simvastatin as well as its effects on sirtuin 1 (SIRT1) expression in endothelial cells. Aged rats and human umbilical vein endothelial cells were treated with simvastatin in the presence and absence of oxidized low-density lipoprotein (OX-LDL). Aortic β-galactosidase staining was undertaken to determine senescence, and SIRT1 protein expression was evaluated using Western blot analysis. After simvastatin therapy, arterial endothelial cell aging was significantly reduced, and SIRT1 expression was significantly increased. The OX-LDL significantly accelerated the senescence of umbilical vein endothelial cells and decreased SIRT1 expression. The OX-LDL-induced downregulation of SIRT1 was blocked by simvastatin. Simvastatin treatment also reduced umbilical vein endothelial cell aging and increased SIRT1 expression.
Collapse
Affiliation(s)
- Junping Lei
- 1Department of Cardiovascular Diseases, Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|