1
|
Wang Y, Li F, Wei S, Li W, Wu J, Li S, Hu X, Tang T, Liu X. Puerarin-Loaded Liposomes Co-Modified by Ischemic Myocardium-Targeting Peptide and Triphenylphosphonium Cations Ameliorate Myocardial Ischemia-Reperfusion Injury. Int J Nanomedicine 2024; 19:7997-8014. [PMID: 39130683 PMCID: PMC11317047 DOI: 10.2147/ijn.s468394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Mitochondrial damage may lead to uncontrolled oxidative stress and massive apoptosis, and thus plays a pivotal role in the pathological processes of myocardial ischemia-reperfusion (I/R) injury. However, it is difficult for the drugs such as puerarin (PUE) to reach the mitochondrial lesion due to lack of targeting ability, which seriously affects the expected efficacy of drug therapy for myocardial I/R injury. Methods We prepared triphenylphosphonium (TPP) cations and ischemic myocardium-targeting peptide (IMTP) co-modified puerarin-loaded liposomes (PUE@T/I-L), which effectively deliver the drug to mitochondria and improve the effectiveness of PUE in reducing myocardial I/R injury. Results In vitro test results showed that PUE@T/I-L had sustained release and excellent hemocompatibility. Fluorescence test results showed that TPP cations and IMTP double-modified liposomes (T/I-L) enhanced the intracellular uptake, escaped lysosomal capture and promoted drug targeting into the mitochondria. Notably, PUE@T/I-L inhibited the opening of the mitochondrial permeability transition pore, reduced intracellular reactive oxygen species (ROS) levels and increased superoxide dismutase (SOD) levels, thereby decreasing the percentage of Hoechst-positive cells and improving the survival of hypoxia-reoxygenated (H/R)-injured H9c2 cells. In a mouse myocardial I/R injury model, PUE@T/I-L showed a significant myocardial protective effect against myocardial I/R injury by protecting mitochondrial integrity, reducing myocardial apoptosis and decreasing infarct size. Conclusion This drug delivery system exhibited excellent mitochondrial targeting and reduction of myocardial apoptosis, which endowed it with good potential extension value in the precise treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
2
|
LIU B, LI J, SI J, CHEN Q, YANG S, JI E. Ginsenoside Rb1 alleviates chronic intermittent hypoxia-induced diabetic cardiomyopathy in db/db mice by regulating the adenosine monophosphate-activated protein kinase/Nrf2/heme oxygenase-1 signaling pathway. J TRADIT CHIN MED 2023; 43:906-914. [PMID: 37679978 PMCID: PMC10465847 DOI: 10.19852/j.cnki.jtcm.20221206.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/08/2022] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To examine the protective effect of ginsenoside Rb1 (Rb1), the main component of Renshen (), on cardiomyopathy in db/db mice exposed to chronic intermittent hypoxia (CIH) and explore the potential underlying mechanism of Rb1 in treating diabetic cardiomyopathy (DCM). METHODS The db/db mice were randomly separated into five groups: normal control group, model group, Rb1 20 mg/kg group, Rb1 40 mg/kg group, and glucagon-like peptide-1 (GLP-1) group. Mice were exposed to air-condition or CIH for 8 weeks, and Rb1 and GLP-1 were administrated before CIH exposure every day. Oral glucose tolerance test (OGTT), intraperitoneal insulin tolerance test (IPITT), total cholesterol (TC), triglyceride (TG), and high-density lipoprotein cholesterol (HDL-C) were detected to evaluate glycolipid metabolism. The level of insulin was detected by a mouse enzyme-linked immunosorbent assay (ELISA). Cardiac function was detected by echocardiography, and myocardial pathology was observed by hematoxylin-eosin and Masson staining. The expression of collagen Ⅰ and collagen Ⅲ was detected by immunohistochemistry. Adenosine monophosphate-activated protein kinase (AMPK)/Nrf2/heme oxygenase-1 (HO-1) signaling pathway was detected by Western blot and immunofluorescence. RESULTS Rb1 treatment could improve glucose tolerance and the level of cardiac function indexes, and inhibit the level of oxidative stress indexes and the expression of collagen Ⅰ and collagen Ⅲ. Moreover, Rb1 treatment enhanced AMPK phosphorylation and increased Nrf2 and HO-1 expression. CONCLUSION Rb1 treatment alleviated CIH-induced diabetic cardiomyopathy and glycolipid metabolism disorders in db/db mice by inhibiting oxidative stress and regulating the AMPK/Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Bingbing LIU
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jieru LI
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- 2 Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Shijiazhuang 050200, China
| | - Jianchao SI
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qi CHEN
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Shengchang YANG
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- 2 Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Shijiazhuang 050200, China
| | - Ensheng JI
- 1 Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- 2 Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Shijiazhuang 050200, China
| |
Collapse
|
3
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
4
|
Hossain MA, Kim JH. Possibility as role of ginseng and ginsenosides on inhibiting the heart disease of COVID-19: A systematic review. J Ginseng Res 2022; 46:321-330. [PMID: 35068945 PMCID: PMC8767974 DOI: 10.1016/j.jgr.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/25/2022] Open
Abstract
Coronavirus has been spreading rapidly around the world since it broke out in China in 2019. Respiratory diseases caused by coronavirus infection cause various diseases ranging from asymptomatic subclinical infections to severe pneumonia and cardiovascular complications, leading to death. In this regard, natural products are being studied to prevent various diseases caused by COVID-19. In current review, we would like to present mechanisms related to the inhibition of heart disease in ginseng and ginsenoside against SARS-CoV-2. In many previous studies, ginseng and ginsenoside are known to have antioxidant, blood flow improvement, improvement of vascular and heart function, blood pressure control, suppression of myocardial infarction and heart failure, and antiarrhythmia. Therefore, ginseng and ginsenoside have a possibility to suppress cardiovascular complications caused by COVID-19. Many of research provide evidence for ginseng and ginsenoside as treatments for the risk of cardiovascular complications. However, in this review, more specific contents on the proposition of the efficacy of ginseng and ginsenoside for COVID-19 should be presented. Therefore, we hope that researches to reduce cardiovascular complications of ginseng and ginsenoside for COVID-19 should be presented to reduce mortality for COVID-19.
Collapse
Affiliation(s)
- Mohammad Amjad Hossain
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| |
Collapse
|
5
|
Zhang X, Wang L, Guo R, Xiao J, Liu X, Dong M, Luan X, Ji X, Lu H. Ginsenoside Rb1 Ameliorates Diabetic Arterial Stiffening via AMPK Pathway. Front Pharmacol 2021; 12:753881. [PMID: 34712140 PMCID: PMC8546248 DOI: 10.3389/fphar.2021.753881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Macrovascular complication of diabetes mellitus, characterized by increased aortic stiffness, is a major cause leading to many adverse clinical outcomes. It has been reported that ginsenoside Rb1 (Rb1) can improve glucose tolerance, enhance insulin activity, and restore the impaired endothelial functions in animal models. The aim of this study was to explore whether Rb1 could alleviate the pathophysiological process of arterial stiffening in diabetes and its potential mechanisms. Experimental Approach: Diabetes was induced in male C57BL/6 mice by administration of streptozotocin. These mice were randomly selected for treatment with Rb1 (10-60 mg/kg, i. p.) once daily for 8 weeks. Aortic stiffness was assessed using ultrasound and measurement of blood pressure and relaxant responses in the aortic rings. Mechanisms of Rb1 treatment were studied in MOVAS-1 VSMCs cultured in a high-glucose medium. Key Results: Rb1 improved DM-induced arterial stiffening and the impaired aortic compliance and endothelium-dependent vasodilation. Rb1 ameliorated DM-induced aortic remodeling characterized by collagen deposition and elastic fibers disorder. MMP2, MMP9, and TGFβ1/Smad2/3 pathways were involved in this process. In addition, Rb1-mediated improvement of arterial stiffness was partly achieved via inhibiting oxidative stress in DM mice, involving regulating NADPH oxidase. Finally, Rb1 could blunt the inhibition effects of DM on AMPK phosphorylation. Conclusion and Implications: Rb1 may represent a novel prevention strategy to alleviate collagen deposition and degradation to prevent diabetic macroangiopathy and diabetes-related complications.
Collapse
Affiliation(s)
- Xinyu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rong Guo
- Department of Cardiology, Ji'an Municipal Center People's Hospital, Ji'an, China
| | - Jie Xiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaorong Luan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoping Ji
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Dr. Gilbert Hung Ginseng Laboratory, Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, Hong Kong, SAR China
| |
Collapse
|
6
|
Taurine with combined aerobic and resistance exercise training alleviates myocardium apoptosis in STZ-induced diabetes rats via Akt signaling pathway. Life Sci 2020; 258:118225. [PMID: 32771557 DOI: 10.1016/j.lfs.2020.118225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 11/21/2022]
Abstract
AIM The aim of this study was considering the effects of taurine supplementation with combined aerobic and resistance training (CARE) on myocardial apoptosis and Protein Kinase B (akt) level changes in diabetic rat. MAIN METHODS Forty male Wistar rats were randomly divided in to 5 groups of 8 animals in each: 1) control, 2) Diabetes Mellitus (DM), 3) DM with taurine supplementation (DM/T), 4) DM with CARE (DM/CARE), and 5) DM with combination of taurine and CARE (DM/T/CARE). DM was induced by injection of streptozotocin (STZ) and nicotine amid (NA) for 2, 3, 4 and 5 groups. Supplement groups received taurine in gavage, 100 mg/kg of body weight, 6 day per weeks, 8 weeks. CARE was performed at maximal speed and 1RM (40-60% of maximum for both). KEY FINDINGS The results of this study showed that DM significantly increased blood glucose and caspase 3, caspase 9 expressions and apoptosis cells in heart tissue and reduced Akt expression (p < 0.001). However, taurine and CARE interventions significantly decreased apoptosis markers (caspase 3 and caspase 9) and significantly increased Akt in heart of diabetic rats compare to DM groups (p < 0.05). The highest improvement observed in DM/T/CARE group (p < 0.05). SIGNIFICANCE Based on these results, it seems that the use of taurine with combined aerobic and exercise training minimize the cardiac damage caused by diabetes (especially apoptosis) trough increasing protein kinase Akt expression. This could improve cardiac remodeling after diabetes. However, more research is needed, especially on the human samples.
Collapse
|
7
|
Wu RM, Jiang B, Li H, Dang WZ, Bao WL, Li HD, Ye G, Shen X. A network pharmacology approach to discover action mechanisms of Yangxinshi Tablet for improving energy metabolism in chronic ischemic heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112227. [PMID: 31509780 DOI: 10.1016/j.jep.2019.112227] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/07/2019] [Accepted: 09/08/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Most cardiovascular diseases ultimately result in heart failure, an intractable problem in modern medicine. Yangxinshi tablet (YXS) is a Chinese medicine formula that is used clinically to treat coronary heart disease. However, the active compounds, potential targets, and pharmacological and molecular mechanism of its anti-heart failure activity remain unclear. Therefore, further investigation is required. AIM OF STUDY Active ingredients and potential targets of YXS for treating heart failure have been reported previously. However, the molecular functions or biological processes of YXS in energy metabolism have not been discovered. To date, no experimental study to validate the potential anti-heart failure mechanism of YXS. The aim of this study was to study the therapeutic effect of YXS on rats with chronic ischemic heart failure by evaluating rat cardiac function and exercise tolerance, and to explore its potential mechanism by network pharmacology, western blotting, quantitative RT-PCR and histological analysis. MATERIALS AND METHODS In this investigation, chronic ischemic heart failure rats were randomly assigned to five groups: control group (sham operation), model group (0.5% CMC-Na), trimetazidine group (positive control) and two YXS groups (low- and high-dose groups). Experimental rats were treated by gavage with 10 mg/kg/d (clinical equivalent dose) trimetazidine (TMZ), 500 mg/kg/d (clinical equivalent dose) YXS and 1000 mg/kg/d YXS, respectively, for 5 weeks. The cardiac functions of rats were detected by High-Resolution In Vivo Imaging System. We elucidated novel understanding of the active compounds of YXS in rat plasma and predicted the energy metabolism related targets and processes for heart failure. Then, we validated experimentally the targets and mechanism of YXS on these pathological processes in vivo. RESULTS It was found that YXS was able to effectively improve cardiac LVIDs, LVEDV, LVESV and EF, decrease myocardial oxygen consumption and reduce myocardial infarct size in rats with chronic ischemic heart failure was similar to that of TMZ. We identified 63 major candidate targets for YXS that are closely to heart failure progression. Enrichment analysis revealed key targets for YXS associated to oxygen delivery, glucose utilization, and mitochondrial biogenesis. Meanwhile, we validated that YXS could promote the expression of downstream HIF-1α, PGC1α and GLUT4 by increasing phosphorylation of PI3K, Akt, mTOR, rpS6 and AMPK. The results show that YXS could activate related PI3K/Akt/mTOR/rpS6/HIF-1α and AMPK/PGC1α/GLUT4 signaling pathways in chronic ischemic heart failure rats. Further experiments demonstrated that YXS increased mitochondrial biogenesis in chronic ischemic heart failure rats and improved exercise tolerance CONCLUSION: YXS treated chronic ischemic heart failure through activating its targets which play pivotal roles in oxygen delivery, glucose utilization and mitochondrial biogenesis to improve energy metabolism through a multi-component, multi-level, multi-target, multi-pathway and multi-mechanism approaches.
Collapse
Affiliation(s)
- Ruo-Ming Wu
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd, Shanghai, China
| | - Bing Jiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hui Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wen-Zhen Dang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei-Lian Bao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hai-Dong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Lim S, Park J, Um JY. Ginsenoside Rb1 Induces Beta 3 Adrenergic Receptor-Dependent Lipolysis and Thermogenesis in 3T3-L1 Adipocytes and db/db Mice. Front Pharmacol 2019; 10:1154. [PMID: 31680950 PMCID: PMC6803469 DOI: 10.3389/fphar.2019.01154] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity is constantly rising into a major health threat worldwide. Activation of brown-like transdifferentiation of white adipocytes (browning) has been proposed as a promising molecular target for obesity treatment. In this study, we investigated the effect of ginsenoside Rb1 (Rb1), a saponin derived from Panax ginseng Meyer, on browning. We used 3T3-L1 murine adipocytes and leptin receptor mutated db/db mice. The lipid accumulation, AMP-activated protein kinase alpha (AMPKα)-related pathways, lipolytic and thermogenic factors were measured after Rb treatment in 3T3-L1 adipocytes. Body weight change and lipolysis-thermogenesis factors were investigated in Rb1-treated db/db mice. Beta 3 adrenergic receptor activation (β3AR) changes were measured in Rb1-treated 3T3-L1 cells with or without β3AR inhibitor L748337 co-treatment. As a result, Rb1 treatment decreased lipid droplet size in 3T3-L1 adipocytes. Rb1 also induced phosphorylations of AMPKα pathway and sirtuins. Moreover, lipases and thermogenic factors such as uncoupling protein 1 were increased by Rb1 treatment. Through these results, we could expect that the non-shivering thermogenesis program can be induced by Rb1. In db/db mice, 6-week injection of Rb1 resulted in decreased inguinal white adipose tissue (iWAT) weight associated with shrunken lipid droplets and increased lipolysis and thermogenesis. The thermogenic effect of Rb1 was possibly due to β3AR, as L748337 pre-treatment abolished the effect of Rb1. In conclusion, we suggest Rb1 as a potential lipolytic and thermogenic therapeutic agent which can be used for obesity treatment.
Collapse
Affiliation(s)
- Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Basic Research Laboratory for Comorbidity Research and Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Basic Research Laboratory for Comorbidity Research and Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
9
|
Liu ZJ, Liu H, Wu C, Xue K. Effect of sepsis on the action potential and cardiac serotonin response in rats. Exp Ther Med 2019; 18:2207-2212. [PMID: 31452710 DOI: 10.3892/etm.2019.7810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/16/2019] [Indexed: 01/22/2023] Open
Abstract
The current study aimed to investigate the effect of sepsis on rat serotonin (5-HT) responses and cardiac action potentials. A total of 20 rats were randomly divided into a sepsis and control group (each, n=10). Rat hearts were harvested and perfused using the Langendorff method 18-h after the induction of sepsis, which was assessed using cecal puncture. Cardiac action potential was subsequently measured using a multichannel electrophysiology instrument. Immunohistochemistry and quantitative analysis were performed to identify the effect of sepsis on myocardial 5-HT expression. The results revealed that mitochondrial changes were present in septic rat hearts. Heart rate (361.10±12.29 bpm vs. 348.60±12.38 bpm; P<0.05) was significantly higher, atrial action potential duration (106.40±2.95 ms vs. 86.60±4.12 ms; P<0.01) was significantly longer and the area (0.62±0.06 µm2 vs. 0.39±0.05 µm2; P<0.05) and number (0.92±0.02/field vs. 0.46±0.01/field; P<0.01) of myocardial cells were significantly higher in the septic compared with the control group. These results demonstrated that 5-HT prolongs the atrial action potential, increases heart rate and aggravates myocardial injury, indicating that it may therefore be one of the factors that leads to cardiac dysfunction in sepsis.
Collapse
Affiliation(s)
- Zheng-Jiang Liu
- Department of Cardiology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Hua Liu
- Department of Cardiology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Changdong Wu
- Intensive Medicine Unit, The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| | - Kedong Xue
- Intensive Medicine Unit, The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| |
Collapse
|
10
|
Dai SN, Hou AJ, Zhao SM, Chen XM, Huang HT, Chen BH, Kong HL. Ginsenoside Rb1 Ameliorates Autophagy of Hypoxia Cardiomyocytes from Neonatal Rats via AMP-Activated Protein Kinase Pathway. Chin J Integr Med 2019; 25:521-528. [PMID: 30088211 DOI: 10.1007/s11655-018-3018-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate whether ginsenoside-Rb1 (Gs-Rb1) improves the CoCl-induced autophagy of cardiomyocytes via upregulation of adenosine 5'-monophosphate-activated protein kinase (AMPK) pathway. METHODS Ventricles from 1- to 3-day-old Wistar rats were sequentially digested, separated and incubated in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum for 3 days followed by synchronization. Neonatal rat cardiomyocytes were randomly divided into 7 groups: control group (normal level oxygen), hypoxia group (500 μmol/L CoCl2), Gs-Rb1 group (200 μmol/L Gs-Rb1 + 500 μmol/L CoCl2), Ara A group (500 μmol/L Ara A + 500 μmol/L CoCl2), Ara A+ Gs-Rb1 group (500 μmol/L Ara A + 200 μmol/L Gs-Rb1 + 500 μmol/L CoCl2), AICAR group [1 mmol/L 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) + 500 μmol/L CoCl2], and AICAR+Gs-Rb1 group (1 mmol/L AICAR + 200 μmol/L Gs-Rb1 + 500 μmol/L CoCl2). Cells were treated for 12 h and cell viability was determined by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and cardiac troponin I (cTnI) levels were detected by enzyme-linked immunosorbent assay (ELISA). AMPK activity was assessed by 2',7'-dichlorofluorescein diacetate (DCFH-DA) ELISA assay. The protein expressions of Atg4B, Atg5, Atg6, Atg7, microtubule-associated protein 1A/1B-light chain 3 (LC3), P62, and active-cathepsin B were measured by Western blot. RESULTS Gs-Rb1 significantly improved the cell viability of hypoxia cardiomyocytes (P<0.01). However, the viability of hypoxia-treated cardiomyocytes was significantly inhibited by Ara A (P<0.01). Gs-Rb1 increased the AMPK activity of hypoxia-treated cardiomyocytes. The AMPK activity of hypoxia-treated cadiomyocytes was inhibited by Ara A (P<0.01) and was not affected by AICAR =0.983). Gs-Rb1 up-regulated Atg4B, Atg5, Beclin-1, Atg7, LC3B II, the LC3B II/I ratio and cathepsin B activity of hypoxia cardiomyocytes (P<0.05), each of these protein levels was significantly enhanced by Ara A (all P<0.01), but was not affected by AICAR (all P>0.05). Gs-Rb1 significantly down-regulated P62 levels of hypoxic cardiomyocytes (P<0.05). The P62 levels of hypoxic cardiomyocytes were inhibited by Ara A (P<0.05) and were not affected by AICAR (P=0.871). CONCLUSION Gs-Rb1 may improve the viability of hypoxia cardiomyocytes by ameliorating cell autophagy via the upregulation of AMPK pathway.
Collapse
Affiliation(s)
- Sheng-Nan Dai
- Department of Cardiology, the People's Hospital of China Medical University, the People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Ai-Jie Hou
- Department of Cardiology, the People's Hospital of China Medical University, the People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Shu-Mei Zhao
- International Education College, Shenyang Normal University, Shenyang, 110034, China
| | - Xiao-Ming Chen
- Department of Cardiology, the People's Hospital of China Medical University, the People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Hua-Ting Huang
- Department of Cardiology, the People's Hospital of China Medical University, the People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Bo-Han Chen
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Hong-Liang Kong
- Department of Cardiology, the People's Hospital of China Medical University, the People's Hospital of Liaoning Province, Shenyang, 110016, China.
| |
Collapse
|
11
|
Ginsenoside Rb1 promotes the growth of mink hair follicle via PI3K/AKT/GSK-3β signaling pathway. Life Sci 2019; 229:210-218. [PMID: 31102746 DOI: 10.1016/j.lfs.2019.05.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/29/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022]
Abstract
AIMS Hair follicles play a critical role in the process of hair growth. The dermal papilla cells (DPCs) are an important component in the hair follicle regeneration and growth. This study investigated the effects of ginsenoside Rb1 on the growth of cultured mink hair follicles and DPCs. MAIN METHODS The mink hair follicles were treated with ginsenoside Rb1 for 9 days and their lengths were measured every three days. Real-time PCR was used to determine the mRNA expression of vascularization endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGF-R2) and TGF-β1. In addition, the levels of proteins were detected by western blot. Cell proliferation was determined by immunofluorescence staining of proliferation marker Ki-67 and cell cycle analysis was performed on flow cytometry. Moreover, cell migration was evaluated by wound healing assay. KEY FINDINGS Ginsenoside Rb1 promoted the growth of hair follicles, and proliferation and migration of DPCs. Ginsenoside Rb1 improved the expression levels of VEGFA and VEGF-R2, while attenuated the TGF-β1 expression both in hair follicles and DPCs. Furthermore, ginsenoside Rb1 facilitated the activation of PI3K/AKT/GSK-3β signaling pathway in hair follicles and DPCs. SIGNIFICANCE The results reveals a crucial role of PI3K/AKT/GSK-3β signaling pathway in ginsenoside Rb1-induced growth of hair follicles and DPCs.
Collapse
|
12
|
Manu TM, Anand T, Pandareesh MD, Kumar PB, Khanum F. Terminalia arjuna extract and arjunic acid mitigate cobalt chloride-induced hypoxia stress-mediated apoptosis in H9c2 cells. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1107-1119. [PMID: 31069430 DOI: 10.1007/s00210-019-01654-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Arjunic acid (AA) is one of the major active component of Terminalia arjuna known for its health benefits. In the present study, we evaluated cardioprotective potential of Terminalia arjuna extract (TAE) and AA against cobalt chloride (CoCl2)-induced hypoxia damage and apoptosis in rat cardiomyocytes. TAE (50 μg/ml) and AA (8 μg/ml) significantly (p < 0.001) protected H9c2 cells as evidenced by cell viability assays against CoCl2 (1.2 mM)-induced cytotoxicity. TAE and AA pretreatments protected the cells from oxidative damage by decreasing the generation of free radicals (ROS, hydroperoxide, and nitrite levels). TAE and AA pretreatments retained mitochondrial membrane potential by alleviating the rate of lipid peroxidation induced by CoCl2 treatment. TAE and AA pretreatments elevated antioxidant status including phase II antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase) and total glutathione levels against CoCl2-induced oxidative stress. Further immunoblotting studies confirmed anti-apoptotic effects of TAE and AA by alleviating the phosphorylation of JNK and c-jun and also by regulating protein expression levels of Bcl2, Bax, caspase 3, heat shock protein-70, and inducible nitric oxide synthase. Overall, our results suggest that both the extract and the active component exhibit antioxidant and anti-apoptotic defense against CoCl2-induced hypoxic injury.
Collapse
Affiliation(s)
- T Mohan Manu
- Nutrition, Biochemistry and Toxicology Division, Defence Food Research Laboratory, Mysuru, 570011, India
| | - T Anand
- Nutrition, Biochemistry and Toxicology Division, Defence Food Research Laboratory, Mysuru, 570011, India.
| | - M D Pandareesh
- Nutrition, Biochemistry and Toxicology Division, Defence Food Research Laboratory, Mysuru, 570011, India
| | - P Bhuvanesh Kumar
- Nutrition, Biochemistry and Toxicology Division, Defence Food Research Laboratory, Mysuru, 570011, India
| | - Farhath Khanum
- Nutrition, Biochemistry and Toxicology Division, Defence Food Research Laboratory, Mysuru, 570011, India
| |
Collapse
|
13
|
Al-Damry NT, Attia HA, Al-Rasheed NM, Al-Rasheed NM, Mohamad RA, Al-Amin MA, Dizmiri N, Atteya M. Sitagliptin attenuates myocardial apoptosis via activating LKB-1/AMPK/Akt pathway and suppressing the activity of GSK-3β and p38α/MAPK in a rat model of diabetic cardiomyopathy. Biomed Pharmacother 2018; 107:347-358. [PMID: 30099338 DOI: 10.1016/j.biopha.2018.07.126] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/07/2018] [Accepted: 07/24/2018] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to investigate the protective effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on diabetic cardiomyopathy (DCM)-associated apoptosis and if this effect is mediated via modulating the activity of the survival kinases; AMP-activated protein kinase (AMPK) and Akt & the apoptotic kinases; glycogen synthase kinase-3 β (GSK-3β) and p38 mitogen-activated protein kinase (p38MAPK). Diabetes was induced by a single intraperitoneal injection of streptozotocin (55 mg/kg). Diabetic rats were treated with sitagliptin (10 mg/kg/day, p.o.) and metformin (200 mg/kg/day, p.o. as positive control) for six weeks. Chronic hyperglycemia resulted in elevation of serum cardiac biomarkers reflecting cardiac damage which was supported by H&E stain. The mRNA levels of collagen types I and III were augmented reflecting cardiac fibrosis and hypertrophy which was supported by Masson trichome stain and enhanced phosphorylation of p38MAPK. Cardiac protein levels of cleaved casapse-3, BAX were elevated, whereas, the levels of Bcl-2 and p-BAD were reduced indicating cardiac apoptosis which could be attributed to the diabetes-induced reduced phosphorylation of Akt and AMPK with concomitant augmented activation of GSK-3β and p38MAPK. Protein levels of liver kinase B-1, the upstream kinase of AMPK were also supressed. Sitagliptin administration alleviated the decreased phosphorylation of AMPK and Akt, inactivated the GSK-3β and p38 AMPK, therefore, attenuating the apoptosis and hypertrophy induced by hyperglycemia in the diabetic heart. In conclusion, sitagliptin exhibits valuable therapeutic potential in the management of DCM by attenuating apoptosis. The underlying mechanism may involve the modulating activity of AMPK, Akt, GSK-3β and p38MAPK.
Collapse
Affiliation(s)
- Nouf T Al-Damry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hala A Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Nawal M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nouf M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raeesa A Mohamad
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha A Al-Amin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nduna Dizmiri
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Muhammad Atteya
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Fu J, Zheng H, Cui Q, Chen C, Bao S, Sun J, Li L, Yang B, Wang H, Hou Y, Xu Y, Xu Y, Zhang Q, Pi J. Nfe2l1-silenced insulinoma cells acquire aggressiveness and chemoresistance. Endocr Relat Cancer 2018; 25:185-200. [PMID: 29203613 DOI: 10.1530/erc-17-0458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022]
Abstract
The transcription factor nuclear factor erythroid 2-like 1 (NFE2L1 or NRF1) is involved in various critical cell processes such as maintenance of ubiquitin-proteasome system and regulation of the cellular antioxidant response. We previously determined that pancreatic β-cell-specific Nfe2l1-knockout mice had hyperinsulinemia and that silencing of Nfe2l1 in mouse islets or MIN6 insulinoma β-cells induced elevated basal insulin release and altered glucose metabolism. Hypoglycemia is a major issue with aggressive insulinomas, although a role of NFE2L1 in this pathology is not defined. In the present work, we studied the tumorigenicity of Nfe2l1-deficient insulinoma MIN6 cells (Nfe2l1-KD) and sensitivity to chemotherapy. Nfe2l1-KD cells grew faster and were more aggressive than Scramble cells in vitro In a mouse allograft transplantation model, insulinomas arising from Nfe2l1-KD cells were more aggressive and chemoresistant. The conclusion was amplified using streptozotocin (STZ) administration in an allograft transplantation model in diabetic Akita background mice. Furthermore, Nfe2l1-KD cells were resistant to damage by the chemotherapeutic drugs STZ and 5-fluorouracil, which was linked to binding of hexokinase 1 with mitochondria, enhanced mitochondrial membrane potential and closed mitochondrial potential transition pore. Overall, both in vitro and in vivo data from Nfe2l1-KD insulinoma cells provided evidence of a previously un-appreciated action of NFE2L1 in suppression of tumorigenesis. Nfe2l1 silencing desensitizes insulinoma cells and derived tumors to chemotherapeutic-induced damage, likely via metabolic reprograming. These data indicate that NFE2L1 could potentially play an important role in the carcinogenic process and impact chemosensitivity, at least within a subset of pancreatic endocrine tumors.
Collapse
Affiliation(s)
- Jingqi Fu
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Hongzhi Zheng
- Department of Geriatric EndocrinologyThe First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Qi Cui
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Chengjie Chen
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Simeng Bao
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Jing Sun
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Lu Li
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Bei Yang
- Department of Histology and EmbryologySchool of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Huihui Wang
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yongyong Hou
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yuanyuan Xu
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yuanhong Xu
- Department of Pancreatic SurgeryThe First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Qiang Zhang
- Department of Environmental HealthRollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Jingbo Pi
- Program of Environmental ToxicologySchool of Public Health, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
15
|
Huang Y, Kwan KKL, Leung KW, Yao P, Wang H, Dong TT, Tsim KWK. Ginseng extracts modulate mitochondrial bioenergetics of live cardiomyoblasts: a functional comparison of different extraction solvents. J Ginseng Res 2018; 43:517-526. [PMID: 31695560 PMCID: PMC6823796 DOI: 10.1016/j.jgr.2018.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/31/2017] [Accepted: 02/06/2018] [Indexed: 01/08/2023] Open
Abstract
Background The root of Panax ginseng, a member of Araliaceae family, has been used as herbal medicine and functional food in Asia for thousands of years. According to Traditional Chinese medicine, ginseng is the most widely used “Qi-invigorating” herbs, which provides tonic and preventive effects by resisting oxidative stress, influencing energy metabolism, and improving mitochondrial function. Very few reports have systematically measured cell mitochondrial bioenergetics after ginseng treatment. Methods Here, H9C2 cell line, a rat cardiomyoblast, was treated with ginseng extracts having extracted using solvents of different polarity, i.e., water, 50% ethanol, and 90% ethanol, and subsequently, the oxygen consumption rate in healthy and tert-butyl hydroperoxide–treated live cultures was determined by Seahorse extracellular flux analyzer. Results The 90% ethanol extracts of ginseng possessed the strongest antioxidative and tonic activities to mitochondrial respiration and therefore provided the best protective effects to H9C2 cardiomyocytes. By increasing the spare respiratory capacity of stressed H9C2 cells up to three-folds of that of healthy cells, the 90% ethanol extracts of ginseng greatly improved the tolerance of myocardial cells to oxidative damage. Conclusion These results demonstrated that the low polarity extracts of ginseng could be the best extract, as compared with others, in regulating the oxygen consumption rate of cultured cardiomyocytes during mitochondrial respiration.
Collapse
Affiliation(s)
- Yun Huang
- HKUST Shenzhen Research Institute, Hi-Tech Park, Guangdong, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Kenneth Kin Leung Kwan
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ka Wing Leung
- HKUST Shenzhen Research Institute, Hi-Tech Park, Guangdong, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ping Yao
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Huaiyou Wang
- HKUST Shenzhen Research Institute, Hi-Tech Park, Guangdong, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Tina Tingxia Dong
- HKUST Shenzhen Research Institute, Hi-Tech Park, Guangdong, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Karl Wah Keung Tsim
- HKUST Shenzhen Research Institute, Hi-Tech Park, Guangdong, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| |
Collapse
|
16
|
González Arbeláez LF, Ciocci Pardo A, Fantinelli JC, Schinella GR, Mosca SM, Ríos JL. Cardioprotection and natural polyphenols: an update of clinical and experimental studies. Food Funct 2018; 9:6129-6145. [DOI: 10.1039/c8fo01307a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mechanisms involved in ischemia–reperfusion injury.
Collapse
Affiliation(s)
- Luisa F. González Arbeláez
- Centro de Investigaciones Cardiovasculares
- CCT-CONICET
- Universidad Nacional de la Plata
- 1900 La Plata
- Argentina
| | - Alejandro Ciocci Pardo
- Centro de Investigaciones Cardiovasculares
- CCT-CONICET
- Universidad Nacional de la Plata
- 1900 La Plata
- Argentina
| | - Juliana C. Fantinelli
- Centro de Investigaciones Cardiovasculares
- CCT-CONICET
- Universidad Nacional de la Plata
- 1900 La Plata
- Argentina
| | - Guillermo R. Schinella
- Cátedra de Farmacología Básica
- Facultad de Ciencias Médicas
- Universidad Nacional de La Plata
- 1900 La Plata
- Argentina
| | - Susana M. Mosca
- Centro de Investigaciones Cardiovasculares
- CCT-CONICET
- Universidad Nacional de la Plata
- 1900 La Plata
- Argentina
| | - José-Luis Ríos
- Departament de Farmacologia
- Facultat de Farmàcia
- Universitat de València
- 46100 Burjassot
- Spain
| |
Collapse
|
17
|
Xiaogang L, Zhe Z, Rui W, Xinxin X, Yonghui L, Lianqing S. Ginsenoside Rb1 prevents high glucose-induced Schwann cell injury through the mitochondrial apoptosis pathway. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(18)30037-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
18
|
Baicalin and ginsenoside Rb1 promote the proliferation and differentiation of neural stem cells in Alzheimer's disease model rats. Brain Res 2017; 1678:187-194. [PMID: 29038007 DOI: 10.1016/j.brainres.2017.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND This study aimed to explore the effects of ginsenoside Rb1 and baicalin on the proliferation and differentiation of neural stem cells (NSC) in Alzheimer's disease model rats. METHOD The healthy Sprague Dawley male rats were randomly divided into 4 groups: control group, model group, ginsenoside Rb1 group and baicalin group. Besides, the animal model of dementia was induced by the injection of Aβ1-40. 2 weeks later, the rats in the baicalin and ginsenoside Rb1 groups were injected with baicalin and ginsenoside Rb1, respectively. The contents, expression sites of Nestin, GFAP and NSE and the percentage of viable cells were detected by immunohistochemistry. In addition, the expression levels of Nestin, GFAP and NSE in hippocampus of rats were detected by western-blot and metrology analysis was performed using quantity. RESULTS Injection of Aβ1-40 significantly reduced the number of neuronal cells (p < .05). In addition, compared with the control group, the percentages of positive cells of NSCs, astrocytes and neuronal were increased. Besides, compared with the model group, the percentage of positive neural cells was improved by ginsenoside Rb1 (p < .05), and the percentages of astrocytes and neuronal were increased by ginsenoside Rb1 and baicalin (p < .05). Moreover, the expressions of Nestin and NSE were enhanced by ginsenoside Rb1 and baicalin (p < .05), while the GFAP level was only affected by ginsenoside Rb1 (p < .05) when compared with the model group. CONCLUSION Ginsenoside Rb1 and baicalin might promote the proliferation and differentiation of endogenous NSCs in AD rat model.
Collapse
|
19
|
Li YH, Li YY, Fan GW, Yu JH, Duan ZZ, Wang LY, Yu B. Cardioprotection of ginsenoside Rb1 against ischemia/reperfusion injury is associated with mitochondrial permeability transition pore opening inhibition. Chin J Integr Med 2016:10.1007/s11655-015-2433-6. [PMID: 26740222 DOI: 10.1007/s11655-015-2433-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the role of ginsenoside Rb1 (Gs-Rb1) in cardioprotection against ischemia/reperfusion (I/R) or hypoxia/reoxygenation (H/R) injury and to explore whether the cardioprotective action is mediated via attenuating the formation of mitochondrial permeability transition pore (mPTP). METHODS A Langendorff-perfused model of rat heart was employed. I/R injury was induced by breaking off perfusion for 40 min then reperfusion for 60 min. Gs-Rb1 (100 μmol/L) were administrated for 10 min before I/R. Infarct size was estimated by the 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Lactate dehydrogenase (LDH) and creatine kinase (CK) released from effluents were measured. Transmission electron microscopy was performed to assess morphological difference between cardiac mitochondrial isolated from I/R rats and Gs-Rb1 pretreated rats. Western blot analysis was used to determine phosphorylation of protein kinase B/Akt, and its downstream target glycogen synthase kinase 3β (GSK-3β). Incubation isolated cardiac mitochondria with Gs-Rb1, Ca2+-induced opening of the mPTP was assessed by the reduction of absorbance at 520 nm (A520). Neonatal rat cardiomyocytes were subjected to hypoxia 9 h followed by reoxygenation 4 h to induce H/R injury. After pretreated with different concentration of Gs-Rb1 (6.25, 25, 100 μmol/L ), cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide (MTT) method. The membrane potential was estimated by Rh123 fluorescence. mPTP opening was measured using the probe calcein-AM. RESULTS Gs-Rb1 100 μmol/L significantly reduced the infarct size of hearts (26.39%±11.67% vs. I/R group 56.68%±5.88%, P<0.01). Compared with the I/R group, Gs-Rb1 pretreatment decreased LDH and CK levels in the coronary effluent (P<0.05 or P<0.01) as well as attenuated destructive ultrastructure induced by I/R. The protective effect of Gs-Rb1 involved in phosphorylating protein kinase B/PKB (Akt) and GSK-3β. In mitochondria isolated from rat hearts, significant inhibition of Ca2+-induced swelling was observed in samples that were pretreated with Gs-Rb1 (6.25, 25, 100, 400 μmol/L) for 10 min. When cardiomyocytes were isolated from neonatal rat and subjected to H/R, cell viability was increased with treatment of Gs-Rb1 (6.25, 25, 100 μmol/L ). Gs-Rb1 inhibited mPTP opening and restored subsequent loss of mitochondrial membrane potential. CONCLUSION Gs-Rb1 presents cardioprotective effect against I/R or H/R injury which involves in activating Akt, phosphorylating GSK-3β and inhibiting mPTP opening.
Collapse
Affiliation(s)
- Yu-Hong Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yan-Yan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Guan-Wei Fan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Jia-Hui Yu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Zhen-Zhen Duan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ling-Yan Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Bin Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
20
|
Mu Q, Fang X, Li X, Zhao D, Mo F, Jiang G, Yu N, Zhang Y, Guo Y, Fu M, Liu JL, Zhang D, Gao S. Ginsenoside Rb1 promotes browning through regulation of PPARγ in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2015; 466:530-5. [PMID: 26381176 DOI: 10.1016/j.bbrc.2015.09.064] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 11/30/2022]
Abstract
Browning of white adipocyte tissue (WAT) has received considerable attention due to its potential implication in preventing obesity and related comorbidities. Ginsenoside Rb1 is reported to improve glycolipid metabolism and reduce body weight in obese animals. However whether the body reducing effect mediates by browning effect remains unclear. For this purpose, 3T3-L1 adipocytes were used to study the effect of ginsenoside Rb1 on browning adipocytes specific genes and oxygen consumptions. The results demonstrate that 10 μM of ginsenoside Rb1 increases basal glucose uptake and promoted browning evidenced by significant increases in mRNA expressions of UCP-1, PGC-1α and PRDM16 in 3T3-L1 mature adipocytes. Further, ginsenoside Rb1 also increases PPARγ activity. And the browning effect is abrogated by GW9692, a PPARγ antagonist. In addition, ginsenoside Rb1 increases basal respiration rate, ATP production and uncoupling capacity in 3T3-L1 adipocytes. Those effects are also blunted by GW9692. The results suggest that ginsenoside Rb1 promote browning of 3T3-L1 adipocytes through induction of PPARγ. Our finding offer a new source to discover browning agonists and also useful to understand and extend the applications of ginseng and its constituents.
Collapse
Affiliation(s)
- Qianqian Mu
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xin Fang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoke Li
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dandan Zhao
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fangfang Mo
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guangjian Jiang
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Na Yu
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi Zhang
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yubo Guo
- Preclinical Medicine School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Min Fu
- The Research Institute of McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Jun-Li Liu
- The Research Institute of McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Dongwei Zhang
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Sihua Gao
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
21
|
Ke L, Guo W, Xu J, Zhang G, Wang W, Huang W. Ginsenoside Rb1 attenuates activated microglia-induced neuronal damage. Neural Regen Res 2014; 9:252-9. [PMID: 25206809 PMCID: PMC4146149 DOI: 10.4103/1673-5374.128217] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2014] [Indexed: 01/05/2023] Open
Abstract
The microglia-mediated inflammatory reaction promotes neuronal damage under cerebral ischemia/hypoxia conditions. We therefore speculated that inhibition of hypoxia-induced microglial activation may alleviate neuronal damage. To test this hypothesis, we co-cultured ginsenoside Rb1, an active component of ginseng, and cortical neurons. Ginsenoside Rb1 protected neuronal morphology and structure in a single hypoxic culture system and in a hypoxic co-culture system with microglia, and reduced neuronal apoptosis and caspase-3 production. The protective effect was observable prior to placing in co-culture. Additionally, ginsenoside Rb1 inhibited levels of tumor necrosis factor-α in a co-culture system containing activated N9 microglial cells. Ginsenoside Rb1 also significantly decreased nitric oxide and superoxide production induced by N9 microglia. Our findings indicate that ginsenoside Rb1 attenuates damage to cerebral cortex neurons by downregulation of nitric oxide, superoxide, and tumor necrosis factor-α expression in hypoxia-activated microglia.
Collapse
Affiliation(s)
- Lining Ke
- Department of Human Anatomy, Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Guo
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jianwen Xu
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Guodong Zhang
- Department of Human Anatomy, Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Wang
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wenhua Huang
- Department of Human Anatomy, Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
22
|
Ginsenoside rb1 protects neonatal rat cardiomyocytes from hypoxia/ischemia induced apoptosis and inhibits activation of the mitochondrial apoptotic pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:149195. [PMID: 25120573 PMCID: PMC4120487 DOI: 10.1155/2014/149195] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/02/2014] [Accepted: 06/23/2014] [Indexed: 12/18/2022]
Abstract
Aim. To investigate the effect of Ginsenoside Rb1 (GS-Rb1) on hypoxia/ischemia (H/I) injury in cardiomyocytes in vitro and the mitochondrial apoptotic pathway mediated mechanism. Methods. Neonatal rat cardiomyocytes (NRCMs) for the H/I groups were kept in DMEM without glucose and serum, and were placed into a hypoxic jar for 24 h. GS-Rb1 at concentrations from 2.5 to 40 µM was given during hypoxic period for 24 h. NRCMs injury was determined by MTT and lactate dehydrogenase (LDH) leakage assay. Cell apoptosis, ROS accumulation, and mitochondrial membrane potential (MMP) were assessed by flow cytometry. Cytosolic translocation of mitochondrial cytochrome c and Bcl-2 family proteins were determined by Western blot. Caspase-3 and caspase-9 activities were determined by the assay kit. Results. GS-Rb1 significantly reduced cell death and LDH leakage induced by H/I. It also reduced H/I induced NRCMs apoptosis induced by H/I, in accordance with a minimal reactive oxygen species (ROS) burst. Moreover, GS-Rb1 markedly decreased the translocation of cytochrome c from the mitochondria to the cytosol, increased the Bcl-2/ Bax ratio, and preserved mitochondrial transmembrane potential (ΔΨm). Its administration also inhibited activities of caspase-9 and caspase-3. Conclusion. Administration of GS-Rb1 during H/I in vitro is involved in cardioprotection by inhibiting apoptosis, which may be due to inhibition of the mitochondrial apoptotic pathway.
Collapse
|
23
|
Apelin-APJ effects of ginsenoside-Rb1 depending on hypoxia-induced factor 1α in hypoxia neonatal cardiomyocytes. Chin J Integr Med 2014; 21:139-46. [PMID: 24893658 DOI: 10.1007/s11655-014-1774-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate whether ginsenoside-Rb1 (Gs-Rb1) inhibits the apoptosis of hypoxia cardiomyocytes by up-regulating apelin-APJ system and whether the system is affected by hypoxia-induced factor 1α (Hif-1α). METHODS Neonatal rat cardiomyocytes were randomly divided into 6 groups: a control group, a simple CoCl group, a simple Gs-Rb1 group, a CoCl and Gs-Rb1 hypoxia group, a CoCl and 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) group, a CoCl and YC-1 group and a Gs-Rb1 group, in which YC-1 inhibits the synthesis and accelerates the degradation of Hif-1a. The concentration of CoCl, Gs-Rb1 and YC-1 was 500 μmol/L, 200 μmol/L and 5 μmol/L, respectively; the apoptosis ratio was analyzed with a flow cytometer; and apelin, APJ and Hif-1α were assayed with immunocytochemistry, Western blot assays and reverse transcription polymerase chain reaction (RT-PCR). RESULTS (1) The anti-apoptosis effect of Gs-Rb1 on hypoxia cardiomyocytes was significantly inhibited by YC-1; (2) Hypoxia significantly up-graded the expression of mRNA and protein of apelin; this effect was further reinforced by Gs-Rb1 and significantly inhibited by YC-1; (3) Gs-Rb1 further strengthened the expression of APJ mRNA and APJ proteins once hypoxia occurred, which was significantly inhibited by YC-1; (4) Gs-Rb1 significantly increased the expression of Hif-1α, which was completely abolished by YC-1; (5) There was a negative relationship between AR and apelin (or APJ, including mRNA and protein), a positive correlation between apelin (or APJ) protein and Hif-1a protein, in hypoxia cardiomyocytes. CONCLUSION The apelin-APJ system plays an important role in the anti-apoptosis effect of Gs-Rb1 on hypoxia neonatal cardiomyocytes, which was partly adjusted by Hif-1α.
Collapse
|
24
|
Xinxing W, Wei L, Lei W, Rui Z, Baoying J, Lingjia Q. A neuroendocrine mechanism of co-morbidity of depression-like behavior and myocardial injury in rats. PLoS One 2014; 9:e88427. [PMID: 24551098 PMCID: PMC3923793 DOI: 10.1371/journal.pone.0088427] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 01/08/2014] [Indexed: 11/19/2022] Open
Abstract
Depression is generally a recurrent psychiatric disorder. Evidence shows that depression and cardiovascular diseases are common comorbid conditions, but the specific pathological mechanisms remain unclear. The purpose of this study is to determine the effects of depression induced by chronic unpredictable mild stress (CUMS) on myocardial injury and to further elucidate the biological mechanism of depression. Rats were used as a model. The CUMS procedure lasted for a total of 8 weeks. After 4 weeks of CUMS, treated rats exhibited a reduced sucrose preference and changes in scores on an open field test, body weight and content of 5-HT in the brain as compared with the values of these variables in controls. These changes indicated depression-like changes in CUMS rats and demonstrated the feasibility of the depression model. In addition, pathological changes in the myocardium and increased cardiomyocyte apoptosis demonstrated that myocardial injury had occurred after 6 weeks of CUMS and had increased significantly by the end of 8 weeks of CUMS. Plasma serotonin (5-HT), norepinephrine (NE) and epinephrine (E), all depression-related neuroendocrine factors, were measured by HPLC-ECD techniques, and the content of plasma corticosterone (GC) was evaluated by an I(125)-cortisol radioactivity immunoassay in control and CUMS rats. The results indicated that 5-HT had decreased, whereas NE, E and GC had increased in CUMS rats, and these factors might be associated with depression-induced myocardial injury. The effects of 5-HT, NE and GC on the survival rate of cultured cardiomyocytes were determined using an orthogonal design. The results showed that 5-HT was a more important factor affecting cell survival than GC or NE. The results suggested that normal blood levels of 5-HT had a cytoprotective effect. The neuroendocrine disorders characterized by decreased 5-HT combined with increased GC and NE mediated the occurrence of depression-induced myocardial injury.
Collapse
Affiliation(s)
- Wang Xinxing
- Beijing Institute of Basic Medical Sciences, Beijing, China
- * E-mail: (WX); (JB); (QL)
| | - Liu Wei
- Institute of Health & Environmental Medicine, Tianjin, China
| | - Wu Lei
- Institute of Health & Environmental Medicine, Tianjin, China
| | - Zhan Rui
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jin Baoying
- Tianjin Occupational Disease Prevention Hospital (Hospital Workers), Tianjin, China
- * E-mail: (WX); (JB); (QL)
| | - Qian Lingjia
- Beijing Institute of Basic Medical Sciences, Beijing, China
- * E-mail: (WX); (JB); (QL)
| |
Collapse
|
25
|
Finsterer J, Ohnsorge P. Influence of mitochondrion-toxic agents on the cardiovascular system. Regul Toxicol Pharmacol 2013; 67:434-45. [DOI: 10.1016/j.yrtph.2013.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 09/01/2013] [Accepted: 09/02/2013] [Indexed: 10/26/2022]
|
26
|
Ginsenoside rb1 reduces isoproterenol-induced cardiomyocytes apoptosis in vitro and in vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:454389. [PMID: 24363767 PMCID: PMC3852087 DOI: 10.1155/2013/454389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 09/30/2013] [Accepted: 10/07/2013] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes apoptosis can lead to heart failure. Conventional and alternative drugs, such as Chinese herbal remedies, have been developed to target cardiomyoblast cells apoptosis. In this study, we investigated the effects of ginsenoside Rb1 (Rb1), an active compound, which is isolated from Notoginseng and Ginseng on isoproterenol-(ISO-) induced apoptosis in rat cardiomyocytes and its mechanism in vivo and in vitro. Rb1 reduced the ISO-induced apoptosis in rat cardiomyocytes and H9c2 cells. The effect of Rb1 was significantly suppressed by H89 (inhibitor for PKA), but not by C-1 (inhibitor for PKC). Based on in-cell blot analysis, the ISO-induced PKA and PKC expressions were decreased by Rb1, which was inhibited by H89, but not by C-1. The expressions of caspase-3 and caspase-9 were decreased after treatment with both ISO and Rb1, but with no change for caspase-8. Our results indicated that Rb1 reducing ISO-induced rat cardiomyocytes apoptosis may be involved in PKA and caspase-9 pathways.
Collapse
|
27
|
Liang J, Yu Y, Wang B, Lu B, Zhang J, Zhang H, Ge P. Ginsenoside Rb1 attenuates oxygen-glucose deprivation-induced apoptosis in SH-SY5Y cells via protection of mitochondria and inhibition of AIF and cytochrome c release. Molecules 2013; 18:12777-92. [PMID: 24135936 PMCID: PMC6270437 DOI: 10.3390/molecules181012777] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/22/2013] [Accepted: 10/14/2013] [Indexed: 11/16/2022] Open
Abstract
To investigate the role of mitochondria in the protective effects of ginsenoside Rb1 on cellular apoptosis caused by oxygen-glucose deprivation, in this study, MTT assay, TUNEL staining, flow cytometry, immunocytochemistry and western blotting were used to examine the cellular viability, apoptosis, ROS level, mitochondrial membrane potential, and the distribution of apoptosis inducing factor, cytochrome c, Bax and Bcl-2 in nucleus, mitochondria and cytoplasm. We found that pretreatment with GRb1 improved the cellular viability damaged by OGD. Moreover, GRb1 inhibited apoptosis in SH-SY5Y cells induced by OGD. Further studies showed that the elevation of cellular reactive oxygen species levels and the reduction of mitochondrial membrane potential caused by OGD were both counteracted by GRb1. Additionally, GRb1 not only suppressed the translocation of apoptosis inducing factor into nucleus and cytochrome c into cytoplasm, but also inhibited the increase of Bax within mitochondria and alleviated the decrease of mitochondrial Bcl-2. Our study indicates that the protection of GRb1 on OGD-induced apoptosis in SH-SY5Y cells is associated with its protection on mitochondrial function and inhibition of release of AIF and cytochrome c.
Collapse
Affiliation(s)
- Jianmin Liang
- Department of Pediatrics, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (J.L.); (H.Z.)
| | - Ying Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (Y.Y.); (B.W.); (B.L.)
| | - Boyu Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (Y.Y.); (B.W.); (B.L.)
| | - Bin Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (Y.Y.); (B.W.); (B.L.)
| | - Jizhou Zhang
- Department of Biochemistry, Bethune Medical School of Jilin University, Changchun 130021, China; E-Mail:
| | - Hongbo Zhang
- Department of Pediatrics, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (J.L.); (H.Z.)
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China; E-Mails: (Y.Y.); (B.W.); (B.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-431-8187-5707; Fax: +86-431-8878-2466
| |
Collapse
|
28
|
Liu Z, Chen J, Huang W, Zeng Z, Yang Y, Zhu B. Ginsenoside Rb1 protects rat retinal ganglion cells against hypoxia and oxidative stress. Mol Med Rep 2013; 8:1397-403. [PMID: 24002069 DOI: 10.3892/mmr.2013.1658] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 08/12/2013] [Indexed: 11/05/2022] Open
Abstract
The current study was designed to investigate the effect of ginsenoside Rb1 (Rb1) on apoptosis induced by hypoxia and oxidative stress in a retinal ganglion cell line (RGC-5). The underlying mechanism was also investigated. RGC-5 cells were pretreated with 10 µmol/l Rb1 for 24 h and exposed to 400 µmol/l cobalt chloride (CoCl2) for 48 h or 600 µmol/l H2O2 for 24 h. The percentage of cells actively undergoing apoptosis was determined by flow cytometry with Annexin V/propidium iodide (PI) double staining. The expression of caspases was determined using western blot analysis. CoCl2 and H2O2 treatments significantly increased the apoptotic percentages to 24.5 and 21.63%, respectively. Pretreatment of Rb1 reduced the total apoptotic percentages to 15.12 and 12.03%, respectively. The expression of cleaved caspase-3, -9 and -8 was increased in the CoCl2-treated group, however, caspase-3 was not increased in the H2O2-treated group. Pretreatment of Rb1 reduced the expression of cleaved caspase-3 and -9 in the CoCl2-treated group, but reduced only cleaved caspase-9 in the H2O2-treated group. These results suggest that Rb1 may prevent RGC-5 cells from apoptosis against hypoxia and oxidative stress via the mitochondrial pathway.
Collapse
Affiliation(s)
- Zhaochun Liu
- Department of Pharmacology, Zhongshan Medical College, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | | | | | | | | | | |
Collapse
|
29
|
Ginsenoside Rb1 inhibits the carotid neointimal hyperplasia induced by balloon injury in rats via suppressing the phenotype modulation of vascular smooth muscle cells. Eur J Pharmacol 2012; 685:126-32. [DOI: 10.1016/j.ejphar.2012.04.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 12/29/2022]
|
30
|
Li HX, Han SY, Ma X, Zhang K, Wang L, Ma ZZ, Tu PF. The saponin of red ginseng protects the cardiac myocytes against ischemic injury in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2012; 19:477-483. [PMID: 22341690 DOI: 10.1016/j.phymed.2012.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 11/28/2011] [Accepted: 01/15/2012] [Indexed: 05/31/2023]
Abstract
Steamed root of Panax ginseng C.A. Mayer, known as "red ginseng", differs from other ginseng preparations in terms of its saponin components and content, as some partly deglycosylated saponins are produced as artifacts during the steaming process. However, whether saponins derived from red ginseng (SRG) can have a protective effect on cardiomyocytes remains unknown. The present study aimed to explore the effect of SRG on myocardial ischemia in vitro and in vivo. MTT assays revealed that SRG pretreatment significantly increased the viability of cardiomyocytes injured by Na(2)S(2)O(4) hypoxia in vitro. This effect was almost completely abolished by glibenclamide, a blocker of the ATP-sensitive potassium channel, but the cardioprotective activity of SRG was not influenced by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. SRG also significantly reduced the Na(2)S(2)O(4)-induced increase in intracellular calcium, as shown by Fluo-3/AM probes with flow cytometry. Adult rat heart ischemia, which was induced by ligation of the left anterior descending coronary artery, was employed for the in vivo analysis. SRG pretreatment reduced infarct size and resulted in a higher left ventricle (LV) developed pressure, LV (+)dP/dt(max) and LV systolic pressure and lower LV (-)dP/dt(max) and LV end diastolic pressure after 24h of ischemia. Moreover, SRG significantly reduced the level of cardiac Troponin I (cTnI) in the serum, which suggests that cTnI, a protein component of the troponin regulatory complex involved in cardiac contractility, contributes to the SRG-mediated recovery of cardiac systolic function. In conclusion, this study is the first to provide evidence and a mechanistic analysis of the cardioprotective effects of SRG. SRG significantly attenuated myocardial ischemic injury by improving cardiac systole function, partly by reducing cTnI secretion and improving cardiac diastolic function. Also, SRG attenuated the Ca(2+) overload in cardiomyocytes and modulated the K(ATP), but not PI3K, signaling pathway; taken together, these mechanisms synergistically reduced infarct size.
Collapse
Affiliation(s)
- Hai-Xia Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | | | | | | | | | | | | |
Collapse
|
31
|
Karmazyn M, Moey M, Gan XT. Therapeutic potential of ginseng in the management of cardiovascular disorders. Drugs 2012; 71:1989-2008. [PMID: 21985167 DOI: 10.2165/11594300-000000000-00000] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Although employed in Asian societies for thousands of years, the use of ginseng as an herbal medication for a variety of disorders has increased tremendously worldwide in recent years. Ginseng belongs to the genus Panax, of which there exists a variety, generally reflecting their geographic origin. North American ginseng (Panax quinquefolius) and Asian ginseng (Panax ginseng) are two such varieties possessing a plethora of pharmacological properties, which are attributed primarily to the presence of different ginsenosides that bestow these ginsengs with distinct pharmacodynamic profiles. The many cardiovascular benefits attributed to ginseng include cardioprotection, antihypertensive effects, and attenuation of myocardial hypertrophy and heart failure. Experimental studies have revealed a number of beneficial properties of ginseng, particularly in the area of cardiac protection, where ginseng and ginsenosides have been shown to protect the ischaemic and reperfused heart in a variety of experimental models. Emerging evidence also suggests that ginseng attenuates myocardial hypertrophy, thus blunting the remodelling and heart failure processes. However, clinical evidence of efficacy is not convincing, likely owing primarily to the paucity of well designed, randomized, controlled clinical trials. Adding to the complexity in understanding the cardiovascular effects of ginseng is the fact that each of the different ginseng varieties possesses distinct cardiovascular properties, as a result of their respective ginsenoside composition, rendering it difficult to assign a general, common cardiovascular effect to ginseng. Additional challenges include the identification of mechanisms (likely multifaceted) that account for the effects of ginseng and determining which ginsenoside(s) mediate these cardiovascular properties. These concerns notwithstanding, the potential cardiovascular benefit of ginseng is worthy of further studies in view of its possible development as a cardiovascular therapeutic agent, particularly as adjunctive therapy to existing medications.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | | | | |
Collapse
|
32
|
Compound K, a metabolite of ginsenosides, induces cardiac protection mediated nitric oxide via Akt/PI3K pathway. Life Sci 2011; 88:725-9. [PMID: 21338613 DOI: 10.1016/j.lfs.2011.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 01/13/2011] [Accepted: 02/02/2011] [Indexed: 11/20/2022]
Abstract
AIMS Compound K (C-K; 20-O-D-glucopyranosyl-20(S)-protopanaxadiol) is a novel ginsenoside metabolite formed by intestinal bacteria and does not occur naturally in ginseng. In this study, we investigated whether administration of C-K has protective effects on myocardial ischemia-reperfusion injury and its potential mechanisms. MAIN METHODS We used in vivo mouse models of ischemia-reperfusion injury and performed biochemical assays in excised hearts. KEY FINDINGS C-K reduced infarct size compared with the control group after ischemia-reperfusion. Immunoblot analysis showed that C-K significantly enhanced protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) activity. Wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, blocked cardiac protection in vivo and attenuated phosphorylation of Akt and eNOS. Additionally, the hearts of C-K pretreated mice showed inhibition of mitochondrial swelling induced by Ca(2+). SIGNIFICANCE This study showed that Compound K pretreatment has protective effects on myocardial ischemia-reperfusion injury, partly by mediating the activation of PI3K pathway and phosphorylation of Akt and eNOS.
Collapse
|