1
|
Hu QR, Hong H, Zhang ZH, Feng H, Luo T, Li J, Deng ZY, Chen F. Methods on improvements of the poor oral bioavailability of ginsenosides: Pre-processing, structural modification, drug combination, and micro- or nano- delivery system. J Ginseng Res 2023; 47:694-705. [PMID: 38107396 PMCID: PMC10721471 DOI: 10.1016/j.jgr.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 12/19/2023] Open
Abstract
Panax ginseng Meyer is a traditional Chinese medicine that is widely used as tonic in Asia. The main pharmacologically active components of ginseng are the dammarane-type ginsenosides, which have been shown to have anti-cancer, anti-inflammatory, immunoregulatory, neuroprotective, and metabolic regulatory activities. Moreover, some of ginsenosides (eg, Rh2 and Rg3) have been developed into nutraceuticals. However, the utilization of ginsenosides in clinic is restrictive due to poor permeability in cells and low bioavailability in human body. Obviously, the dammarane skeleton and glycosyls of ginsenosides are responsible for these limitations. Therefore, improving the oral bioavailability of ginsenosides has become a pressing issue. Here, based on the structures of ginsenosides, we summarized the understanding of the factors affecting the oral bioavailability of ginsenosides, introduced the methods to enhance the oral bioavailability and proposed the future perspectives on improving the oral bioavailability of ginsenosides.
Collapse
Affiliation(s)
- Qi-rui Hu
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Huan Hong
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Zhi-hong Zhang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Hua Feng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Ze-yuan Deng
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Fang Chen
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Zhang W, Wan Y, Sun S, Xie Y, Zhao D, Li B, Li J, Tian Y, Feng S. A Pharmacokinetic Study of Sixteen Major Bioactive Components of Jinshui-Huanxian Granules in Pulmonary Fibrosis Model and Control Rats Using Orbitrap Fusion Mass Spectrometry. Molecules 2023; 28:6492. [PMID: 37764268 PMCID: PMC10534582 DOI: 10.3390/molecules28186492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Jinshui-Huanxian granules (JHGs), a Chinese herbal compound prescription, have shown a therapeutic effect in reducing lung tissue damage, improving the degree of pulmonary fibrosis, replenishing lungs and kidneys, relieving cough and asthma, reducing phlegm, and activating blood circulation. However, these active compounds' pharmacokinetics and metabolic processes were unclear. This study aimed to compare the pharmacokinetics, reveal the metabolic dynamic changes, and obtain the basic pharmacokinetic parameters of 16 main bioactive compounds after intragastric administration of JHGs in control and pulmonary fibrosis (PF) model rats by using Orbitrap Fusion MS. After administration of JHGs, the rat plasma was collected at different times. Pretreating the plasma sample with methanol and internal standard (IS) solution carbamazepine (CBZ), and it was then applied to a C18 column by setting gradient elution with a mobile phase consisting of methanol 0.1% formic acid aqueous solution. Detection was performed on an electrospray ionization source (ESI), and the scanning mode was SIM. Pharmacokinetic parameters were analyzed according to the different analytes' concentrations in plasma. The matrix effect was within the range of 79.01-110.90%, the extraction recovery rate was 80.37-102.72%, the intra-day and inter-day precision relative standard deviation (RSD) was less than 7.76%, and the stability was good, which met the requirements of biological sample testing. The method was validated (r ≥ 0.9955) and applied to compare the pharmacokinetic profiles of the control group and PF model group after intragastric administration of the JHGs. The 16 analytes exhibited different pharmacokinetic behaviors in vivo. In the pathological state of the PF model, most of the components were more favorable for metabolism and absorption, and it was more meaningful to study the pharmacokinetics. Above all, this study provided an essential reference for exploring the mechanism of action of JHGs and guided clinical medication as well.
Collapse
Affiliation(s)
- Weiwei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- Faculty of Chemistry, University of Strasbourg, 67008 Strasbourg, France
| | - Yan Wan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450003, China;
| | - Shuding Sun
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
| | - Yang Xie
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou 450003, China
| | - Di Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
| | - Bing Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450003, China;
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou 450003, China
| | - Yange Tian
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
| | - Suxiang Feng
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450003, China; (W.Z.); (S.S.); (D.Z.); (B.L.)
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou 450046, China; (Y.X.); (J.L.)
| |
Collapse
|
3
|
Balusamy SR, Perumalsamy H, Huq MA, Yoon TH, Mijakovic I, Thangavelu L, Yang DC, Rahimi S. A comprehensive and systemic review of ginseng-based nanomaterials: Synthesis, targeted delivery, and biomedical applications. Med Res Rev 2023; 43:1374-1410. [PMID: 36939049 DOI: 10.1002/med.21953] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 11/22/2022] [Accepted: 02/26/2023] [Indexed: 03/21/2023]
Abstract
Among 17 Panax species identified across the world, Panax ginseng (Korean ginseng), Panax quinquefolius (American ginseng), and Panax notoginseng (Chinese ginseng) are highly recognized for the presence of bioactive compound, ginsenosides and their pharmacological effects. P. ginseng is widely used for synthesis of different types of nanoparticles compared to P. quinquefolius and P. notoginseng. The use of nano-ginseng could increase the oral bioavailability, membrane permeability, and thus provide effective delivery of ginsenosides to the target sites through transport system. In this review, we explore the synthesis of ginseng nanoparticles using plant extracts from various organs, microbes, and polymers, as well as their biomedical applications. Furthermore, we highlight transporters involved in transport of ginsenoside nanoparticles to the target sites. Size, zeta potential, temperature, and pH are also discussed as the critical parameters affecting the quality of ginseng nanoparticles synthesis.
Collapse
Affiliation(s)
- Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Seoul, Gwangjin-gu, Republic of Korea
| | - Haribalan Perumalsamy
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Md Amdadul Huq
- Department of Food and Nutrition, Chung Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Tae Hyun Yoon
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Ivan Mijakovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
| | - Deok Chun Yang
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Shadi Rahimi
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
4
|
Huang AX, Li JM, Yang L, Chen L, Zhou ZH, Li P, Gao W. A mass defect filtering combined background subtraction strategy for rapid screening and identification of metabolites in rat plasma after oral administration of Yindan Xinnaotong soft capsule. J Pharm Biomed Anal 2023; 231:115400. [PMID: 37099978 DOI: 10.1016/j.jpba.2023.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
The absorbed prototypes and metabolites of traditional Chinese medicines (TCMs) serves an important part in pharmacological action and clinical effects. However, the comprehensive characterization of which is facing actual or possible rigorous challenges due to the lack of data mining methods and the complexity of metabolite samples. Yindan Xinnaotong soft capsule (YDXNT), a typical traditional Chinese medicine prescription consisting of extracts from 8 herbal medicines, is widely used for the treatment of angina pectoris and ischemic stroke in the clinic. This study established a systematic data mining strategy based on ultra-high performance liquid chromatography tandem quadrupole-time-of-fight mass spectrometry (UHPLC-Q-TOF MS) for comprehensive metabolite profiling of YDXNT in rat plasma after oral administration. The multi-level feature ion filtration strategy was primarily conducted through the full scan MS data of plasma samples. All potential metabolites were rapidly fileted out from the endogenous background interference based on the background subtract and the chemical type specifically mass defect filter (MDF) windows including flavonoids, ginkgolides, phenolic acids, saponins, and tanshinones. As the MDF windows of certain types were overlapped, the screened-out potential metabolites were deeply characterized and identified according to their retention times (RT), integrating neutral loss filtering (NLF), diagnostic fragment ions filtering (DFIF), and further confirmed by reference standards. Thus, a total of 122 compounds, consisting of 29 prototype components (16 confirmed with reference standards) and 93 metabolites had been identified. This study provides a rapid and robust metabolite profiling method for researching complicated traditional Chinese medicine prescriptions.
Collapse
Affiliation(s)
- An-Xian Huang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jun-Ming Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Liu Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ling Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zi-Han Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Wen Gao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China.
| |
Collapse
|
5
|
Zhou R, He D, Zhang H, Xie J, Zhang S, Tian X, Zeng H, Qin Y, Huang L. Ginsenoside Rb1 protects against diabetes-associated metabolic disorders in Kkay mice by reshaping gut microbiota and fecal metabolic profiles. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115997. [PMID: 36509256 DOI: 10.1016/j.jep.2022.115997] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/14/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax quinquefolius Linn. is one of the most valuable herbal medicine in the world for its broad health benefits, including anti-diabetes. Ginsenoside Rb1, the principal active constituent of Panax quinquefolius Linn., could attenuate insulin resistance and metabolic disorders. The dysfunction of gut microbiota and fecal metabolites plays an important role in the pathogenesis of Type 2 Diabetes mellitus (T2DM). However, whether ginsenoside Rb1's hypoglycemic effect is related to gut microbiota remains elusive. AIM OF THE STUDY Our study aimed to explore the insulin-sensitizing and anti-diabetic effects of ginsenoside Rb1 as well as the underlying mechanisms. MATERIALS AND METHODS The T2DM model were established by high fat diet (HFD)-induced Kkay mice. The anti-diabetic effect of ginsenoside Rb1 (200 mg/kg/day) was evaluated by random blood glucose (RBG), fasting blood glucose (FBG), glucose tolerance test (OGTT), serum insulin level, insulin resistance index (HOMA-IR), pancreatic histology analysis, liver indexes, total triglyceride (TG) and total cholesterol (TC). Subsequently, 16S rRNA sequencing and LC-MS-based untargeted metabolomics were applied to characterize the microbiome and metabolites profile in HFD-induced Kkay mice, respectively. Finally, antibiotic treatment was used to validate the potential mechanism of ginsenoside Rb1 by modulating gut microbiota. RESULTS Our results showed that ginsenoside Rb1 reduced blood glucose, OGTT, serum insulin level, HOMA-IR, liver indexes as well as pancreatic injury. In addition, the ginsenoside Rb1 reversed the gut microbiota dysbiosis in diabetic Kkay mice, as indicated by the elevated abundance of Parasutterella, decreased population of Alistipes, f_Prevotellaceae_unclassified, Odoribacter, Anaeroplasma. Moreover, ginsenoside Rb1 altered free fatty acid (FFA) levels in fecal metabolites, such as decreased the level of α-linolenic acid, 13-OxoODE, oleic acid, 13-HODE, arachidonic acid, palmitic acid, stearic acid, while increased the level of PC (14:0/22:1(13Z)) and PC (16:0/16:0). Notably, ginsenoside Rb1 failed to improve HFD-induced diabetes in Kkay mice with antibiotics intervention. CONCLUSION These findings suggested that ginsenoside Rb1 may serve as a potential prebiotic agent to modulate specific gut microbes and related metabolites, which play essential roles in diabetes-associated metabolic disorders and insulin resistance.
Collapse
Affiliation(s)
- Rongrong Zhou
- The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| | - Dan He
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China; Hunan University of Chinese Medicine, Changsha, PR China
| | - Haichao Zhang
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China; Hunan University of Chinese Medicine, Changsha, PR China
| | - Jing Xie
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China; Hunan University of Chinese Medicine, Changsha, PR China
| | - Shuihan Zhang
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China
| | - Xuefei Tian
- Hunan University of Chinese Medicine, Changsha, PR China
| | - Hongliang Zeng
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China.
| | - Yuhui Qin
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China; Hunan University of Chinese Medicine, Changsha, PR China.
| | - Luqi Huang
- China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Zou H, Zhang M, Zhu X, Zhu L, Chen S, Luo M, Xie Q, Chen Y, Zhang K, Bu Q, Wei Y, Ye T, Li Q, Yan X, Zhou Z, Yang C, Li Y, Zhou H, Zhang C, You X, Zheng G, Zhao G. Ginsenoside Rb1 Improves Metabolic Disorder in High-Fat Diet-Induced Obese Mice Associated With Modulation of Gut Microbiota. Front Microbiol 2022; 13:826487. [PMID: 35516426 PMCID: PMC9062662 DOI: 10.3389/fmicb.2022.826487] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota plays an important role in metabolic homeostasis. Previous studies demonstrated that ginsenoside Rb1 might improve obesity-induced metabolic disorders through regulating glucose and lipid metabolism in the liver and adipose tissues. Due to low bioavailability and enrichment in the intestinal tract of Rb1, we hypothesized that modulation of the gut microbiota might account for its pharmacological effects as well. Here, we show that oral administration of Rb1 significantly decreased serum LDL-c, TG, insulin, and insulin resistance index (HOMA-IR) in mice with a high-fat diet (HFD). Dynamic profiling of the gut microbiota showed that this metabolic improvement was accompanied by restoring of relative abundance of some key bacterial genera. In addition, the free fatty acids profiles in feces were significantly different between the HFD-fed mice with or without Rb1. The content of eight long-chain fatty acids (LCFAs) was significantly increased in mice with Rb1, which was positively correlated with the increase of Akkermansia and Parasuttereller, and negatively correlated with the decrease of Oscillibacter and Intestinimonas. Among these eight increased LCFAs, eicosapentaenoic acid (EPA), octadecenoic acids, and myristic acid were positively correlated with metabolic improvement. Furthermore, the colonic expression of the free fatty acid receptors 4 (Ffar4) gene was significantly upregulated after Rb1 treatment, in response to a notable increase of LCFA in feces. These findings suggested that Rb1 likely modulated the gut microbiota and intestinal free fatty acids profiles, which should be beneficial for the improvement of metabolic disorders in HFD-fed mice. This study provides a novel mechanism of Rb1 for the treatment of metabolic disorders induced by obesity, which may provide a therapeutic avenue for the development of new nutraceutical-based remedies for treating metabolic diseases, such as hyperlipidemia, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Hong Zou
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Immunology, School of Life Sciences, Fudan University, Shanghai, China
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Man Zhang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Xiaoting Zhu
- Zhejiang Hongguan Bio-Pharma Co., Ltd., Jiaxing, China
| | - Liyan Zhu
- Zhejiang Hongguan Bio-Pharma Co., Ltd., Jiaxing, China
| | - Shuo Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mingjing Luo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qinglian Xie
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yue Chen
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Kangxi Zhang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Qingyun Bu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Yuchen Wei
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tao Ye
- Zhejiang Hongguan Bio-Pharma Co., Ltd., Jiaxing, China
| | - Qiang Li
- Suzhou BiomeMatch Therapeutics Co., Ltd., Shanghai, China
| | - Xing Yan
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Zhihua Zhou
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Chen Yang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Yu Li
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Haokui Zhou
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Haokui Zhou,
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Chenhong Zhang,
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Xiaoyan You,
| | - Guangyong Zheng
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Guangyong Zheng,
| | - Guoping Zhao
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Immunology, School of Life Sciences, Fudan University, Shanghai, China
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
- Suzhou BiomeMatch Therapeutics Co., Ltd., Shanghai, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Guoping Zhao,
| |
Collapse
|
7
|
Liang W, Liu Y, Zhou K, Jian P, Zhang Q, Chang Z, Wu L, Chang H, Zhang L. Ginsenoside Rb1 prevents lipopolysaccharide-induced depressive-like behavior by inhibiting inflammation and neural dysfunction and F2 elicits a novel antidepressant-like effect: A metabolite-based network pharmacology study. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114655. [PMID: 34537284 DOI: 10.1016/j.jep.2021.114655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammatory responses are associated wieh the pathophysiology of depression. Ginsenoside Rb1 (Rb1) exerts antidepressant effect, but the relationship between its activity and inflammation remains unclear. AIM OF THE STUDY In this study, the antidepressant-like effect and underlying mechanisms of Rb1 were been investigated. MATERIALS AND METHODS The neuroinflammatory mouse model of lipopolysaccharide (LPS)-induced acute depression-like behavior was employed to detect the action of Rb1. An integrative strategy combining the identification of prototype (Rb1) and its metabolites in vivo with network pharmacology analysis was used to explore therapeutic mechanisms of these ingredients. The putative targets and signalings were experimentally validated. The antidepressant-like effect of F2, the metabolite of Rb1, was firstly evaluated. RESULTS Rb1 significantly ameliorated LPS-induced depressive-like behavior. Rb1 and its metabolites (Rd, F2, compound K, Rh2, Rg3, PPD) were identified and then a disease-component-target network was established. Experimental validation showed that Rb1 inhibited peripheral and hippocampal inflammation via MAPK/NF-κB signaling. In inflammatory-mediated depression state, Rb1 improved impaired glucocorticoid receptor, suppressed indoleamine 2,3-dioxygenase activity, increased 5-HT level and 5-HT1A receptor expression. Additionally, F2 was firstly discovered to exert antidepressant-like effect, and it existed higher activity than Rb1 against depression. CONCLUSION The study highlighted the potential of Rb1 and F2 as healthy supplement or agent for inflammation-induced depression.
Collapse
Affiliation(s)
- Wenyi Liang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; School of Pharmacy, Navy Military Medical University, Shanghai, China.
| | - Yue Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Kun Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Ping Jian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Qiunan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Zihao Chang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Lingfang Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Hebei TCM Formula Granule Engineering and Technology Research Center, Hebei University of Chinese Medicine, Shijiazhuang, China.
| | - Hongsheng Chang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Lanzhen Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
8
|
Zheng D, Chu Y, Li S, Zhou S, Li W, Xie Y, Sun H. Enhancing effect of borneol on pharmacokinetics of ginsenoside Rb 1 , ginsenoside Rg 1 and notoginsenoside R 1 in healthy volunteers after oral administration of compound Danshen dropping pills (CDDP). Biomed Chromatogr 2022; 36:e5311. [PMID: 34981536 DOI: 10.1002/bmc.5311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022]
Abstract
Borneol (Bingpian), a monoterpenoid pharmaceutical ingredient, is commonly used as a main composition in traditional Chinese medicine (TCM) preparations such as compound Danshen dropping pills (CDDP), and has also been approved by the US FDA as a flavoring substance or adjuvant in food. Borneol plays a regulating and guiding role as messenger drug in CDDP. However, the effect of borneol on the pharmacokinetics of the components of CDDP in human plasma is unclear. In this study, we investigate the effects of borneol on the pharmacokinetics of ginsenoside Rb1 (Rb1 ), ginsenoside Rg1 (Rg1 ) and notoginsenoside R1 (NR1 ) in CDDP. We used a double-cycle crossover-administration model in twelve healthy male volunteers, giving CDDP with borneol (drug T) and without borneol (drug R). The selective response monitoring (SRM) mode was used for mass spectrometry quantification in the positive mode. As a result, we found that borneol could significantly affect the pharmacokinetic parameters of notoginsenosides, and increase the absorption and systemic exposure of Rb1 , Rg1 and NR1 in human plasma by about 1.85 to 3.71 times.
Collapse
Affiliation(s)
- Dayong Zheng
- School of Pharmacy, North China University of Science and Technology, Tangshan, China
| | - Yang Chu
- State Key Laboratory of Core Technology in Innovation Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China.,Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| | - Shuming Li
- State Key Laboratory of Core Technology in Innovation Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China.,Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| | - Shuiping Zhou
- State Key Laboratory of Core Technology in Innovation Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China.,Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| | - Wei Li
- School of Pharmacy, North China University of Science and Technology, Tangshan, China
| | - Yuesheng Xie
- School of Pharmacy, North China University of Science and Technology, Tangshan, China
| | - He Sun
- State Key Laboratory of Core Technology in Innovation Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China.,Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| |
Collapse
|
9
|
Zhuang T, Li W, Yang L, Wang Z, Ding L, Zhou M. Gut Microbiota: Novel Therapeutic Target of Ginsenosides for the Treatment of Obesity and Its Complications. Front Pharmacol 2021; 12:731288. [PMID: 34512356 PMCID: PMC8429618 DOI: 10.3389/fphar.2021.731288] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, generally characterized by excessive lipid accumulation, is a metabolic threat worldwide due to its rapid growth in global prevalence. Ginsenosides are crucial components derived from natural plants that can confer metabolic benefits for obese patients. Considering the low bioavailability and degradable properties of ginsenosides in vivo, it should be admitted that the mechanism of ginsenosides on anti-obesity contribution is still obscure. Recently, studies have indicated that ginsenoside intervention has beneficial metabolic effects on obesity and its complications because it allows for the correction of gut microbiota dysbiosis and regulates the secretion of related endogenous metabolites. In this review, we summarize the role of gut microbiota in the pathogenetic process of obesity, and explore the mechanism of ginsenosides for ameliorating obesity, which can modulate the composition of gut microbiota by improving the metabolism of intestinal endogenous substances and alleviating the level of inflammation. Ginsenosides are expected to become a promising anti-obesity medical intervention in the foreseeable clinical settings.
Collapse
Affiliation(s)
- Tongxi Zhuang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Li
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Li Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Zhengtao Wang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Mingmei Zhou
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
Jang SN, Park SY, Lee H, Jeong H, Jeon JH, Song IS, Kwon MJ, Liu KH. In vitro modulatory effects of ginsenoside compound K, 20( S)-protopanaxadiol and 20( S)-protopanaxatriol on uridine 5'-diphospho-glucuronosyltransferase activity and expression. Xenobiotica 2021; 51:1087-1094. [PMID: 34338601 DOI: 10.1080/00498254.2021.1963503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We explored the inhibitory effect of ginsenoside compound K (CK), 20(S)-protopanaxadiol (PPD), and 20(S)-protopanaxatriol (PPT) on six uridine 5'-diphospho-glucuronosyltransferase (UGT) enzyme (UGT1A1, 1A3, 1A4, 1A6, 1A9, and 2B7) activities in human liver microsomes (HLMs) and 10 UGT enzyme (UGT1A1, 1A3, 1A4, 1A6, 1A9, 2B4, 2B7, 2B10, 2B15, and 2B17) activities in recombinant UGT isoforms.PPD was a potent inhibitor of UGT1A3 activity with half-maximal inhibitory concentration values of 5.62 and 3.38 μM in HLMs and recombinant UGT1A3, respectively. UGT1A3 inhibition by CK and PPD was competitive with inhibitory constant (Ki) values of 17.4 and 1.21 μM, respectively, and inhibition by PPT was non-competitive with a Ki value of 8.07 μM in HLMs. PPD exhibited more than 3.4-fold selectivity for UGT1A3 inhibition compared with other UGT isoforms inhibition, while CK and PPT showed more than 2.16- and 2.21-fold selectivity, respectively.PPD did not significantly increase the mRNA expression of UGT1A1, 1A3, 1A4, 1A9, and 2B7 in hepatocytes.Given the low plasma concentrations of PPD in healthy human subjects and the absence of induction potential on UGT isoforms, we conclude that PPD cause no pharmacokinetic interactions with other co-administered drugs metabolised by UGT1A3.
Collapse
Affiliation(s)
- Su-Nyeong Jang
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu, South Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - So-Young Park
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu, South Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Hyunyoung Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Hyojin Jeong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Ji-Hyeon Jeon
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu, South Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Im-Sook Song
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu, South Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Mi Jeong Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Kwang-Hyeon Liu
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu, South Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea.,Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
11
|
Zhu GX, Zuo JL, Xu L, Li SQ. Ginsenosides in vascular remodeling: Cellular and molecular mechanisms of their therapeutic action. Pharmacol Res 2021; 169:105647. [PMID: 33964471 DOI: 10.1016/j.phrs.2021.105647] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Evidence is mounting that abnormal vascular remodeling (VR) is a vital pathological event that precedes many cardiovascular diseases (CVD). This provides us with a new research perspective that VR can be a pivotal target for CVD treatment and prevention. However, the current drugs for treating CVD do not fundamentally reverse VR and repair vascular function. The reason may be that a complicated regulatory network is formed between the various signaling pathways involved in VR. Recently, ginsenoside, the main active substance of ginseng, has become increasingly the focus of many researchers for its multiple targets, multiple pathways, and few side effects. Several data have revealed that ginsenosides can improve VR caused by vasodilation dysfunction, abnormal vascular structure and blood pressure. This review is intended to discuss the therapeutic effects and mechanisms of ginsenosides in some diseases involved in VR. Besides, we herein also give a new and contradictory insight into intracellular and molecular signaling of ginsenosides in all kinds of vascular cells. Most importantly, we also discuss the feasibility of ginsenosides Rb1/Rg1/Rg3 in drug development by combining the pharmacodynamics and pharmacokinetics of ginsenosides, and provide a pharmacological basis for the development of ginsenosides in clinical applications.
Collapse
Affiliation(s)
- Guang-Xuan Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China.
| | - Jian-Li Zuo
- College of Pharmacy, Chongqing Medical University, Chongqing 410016, China
| | - Lin Xu
- College of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Shu-Qing Li
- The Second Xiangya Hospital of Central South University Shenzhen Hospital, Shenzhen, Guangdong 518067, China
| |
Collapse
|
12
|
Zheng Y, Lee EH, Lee JH, In G, Kim J, Lee MH, Lee OH, Kang IJ. Preclinical Research on a Mixture of Red Ginseng and Licorice Extracts in the Treatment and Prevention of Obesity. Nutrients 2020; 12:nu12092744. [PMID: 32916854 PMCID: PMC7551630 DOI: 10.3390/nu12092744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 11/24/2022] Open
Abstract
The anti-obesity effects of RL (a 3:1 mixture of Panax ginseng saponin fractions and Glycyrrhiza glabra L. extracts) on 3T3-L1 adipocytes and C57BL/6J obese mice were evaluated at different concentrations. We investigated the anti-obesity effects of RL through lipid accumulation inhibition rate, serum lipid composition analysis, adipose tissue size, adipogenic transcription factors and AMPK pathway. RL inhibited the lipid accumulation of 3T3-L1 adipocytes in a dose-dependent manner at concentrations of 50–200 μg/mL without cytotoxicity (50–400 μg/mL). Oral administration of RL at the highest concentration (400 mg/kg/day) did not cause significant liver toxicity in high-fat diet-induced obese mice. RL stimulated adiponectin secretion in a dose-dependent manner and primarily mediates the AMPK pathway to inhibit triglyceride synthesis and attenuate adipocyte hypertrophy. RL significantly reduced weight in obese mice, but none of the body weight, adipose tissue weight, serum triglyceride level, and AMPK pathway activation degree showed any difference between dosing concentrations of 200 and 400 mg/kg/day. Therefore, 200 mg/kg/day of RL is the optimal preclinical concentration, which can be a reference concentration for conversion into a human clinical trial dose.
Collapse
Affiliation(s)
- Yulong Zheng
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Korea; (Y.Z.); (E.-H.L.); (J.-H.L.)
- The Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Eun-Hye Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Korea; (Y.Z.); (E.-H.L.); (J.-H.L.)
- The Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Ji-Hyun Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Korea; (Y.Z.); (E.-H.L.); (J.-H.L.)
- The Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Gyo In
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon 34337, Korea; (G.I.); (J.K.); (M.-H.L.)
| | - JongHan Kim
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon 34337, Korea; (G.I.); (J.K.); (M.-H.L.)
| | - Mi-Hyang Lee
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon 34337, Korea; (G.I.); (J.K.); (M.-H.L.)
| | - Ok-Hwan Lee
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea;
| | - Il-Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Korea; (Y.Z.); (E.-H.L.); (J.-H.L.)
- The Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
- Correspondence: ; Tel.: +82-33-248-2135; Fax: +82-33-256-3420
| |
Collapse
|
13
|
Hao DC, Xiao PG. Impact of Drug Metabolism/Pharmacokinetics and their Relevance Upon Traditional Medicine-based Cardiovascular Drug Research. Curr Drug Metab 2020; 20:556-574. [PMID: 31237211 DOI: 10.2174/1389200220666190618101526] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/09/2019] [Accepted: 05/16/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The representative cardiovascular herbs, i.e. Panax, Ligusticum, Carthamus, and Pueraria plants, are traditionally and globally used in the prevention and treatment of various cardiovascular diseases. Modern phytochemical studies have found many medicinal compounds from these plants, and their unique pharmacological activities are being revealed. However, there are few reviews that systematically summarize the current trends of Drug Metabolism/Pharmacokinetic (DMPK) investigations of cardiovascular herbs. METHODS Here, the latest understanding, as well as the knowledge gaps of the DMPK issues in drug development and clinical usage of cardiovascular herbal compounds, was highlighted. RESULTS The complicated herb-herb interactions of cardiovascular Traditional Chinese Medicine (TCM) herb pair/formula significantly impact the PK/pharmacodynamic performance of compounds thereof, which may inspire researchers to develop a novel herbal formula for the optimized outcome of different cardiovascular diseases. While the Absorption, Distribution, Metabolism, Excretion and Toxicity (ADME/T) of some compounds has been deciphered, DMPK studies should be extended to more cardiovascular compounds of different medicinal parts, species (including animals), and formulations, and could be streamlined by versatile omics platforms and computational analyses. CONCLUSION In the context of systems pharmacology, the DMPK knowledge base is expected to translate bench findings to clinical applications, as well as foster cardiovascular drug discovery and development.
Collapse
Affiliation(s)
- Da-Cheng Hao
- Biotechnology Institute, School of Environment and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, China
| | - Pei-Gen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
14
|
An X, Bao Q, Di S, Zhao Y, Zhao S, Zhang H, Lian F, Tong X. The interaction between the gut Microbiota and herbal medicines. Biomed Pharmacother 2019; 118:109252. [DOI: 10.1016/j.biopha.2019.109252] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
|
15
|
Gut microbial transformation, a potential improving factor in the therapeutic activities of four groups of natural compounds isolated from herbal medicines. Fitoterapia 2019; 138:104293. [PMID: 31398447 DOI: 10.1016/j.fitote.2019.104293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022]
Abstract
Herbal medicines (HMs) have attracted widespread attention because of their significant contributions to the prevention and treatment of many human diseases. Recently, gut microbiota has become an important frontier to understand the therapeutic mechanisms of medicines. Gut microbiota-mediated transformation is a microbial metabolic form after oral administrations of HMs compounds. A great number of studies showed that gut microbiota could transform some HMs compounds by the variation of chemical structures into several active metabolites, which exerted better bioavailabilities and therapeutic activities than their parent compounds. Among these HMs compounds, alkaloids, flavonoids, polyphenols and terpenoids were the representative ones. However, there is no systemic review focusing on the potential improved therapeutic activities of these natural compounds caused by gut microbial transformation. Here, this review summarizes the therapeutic activities that are more potent in microbial transformed metabolites than in their parent compounds (alkaloids, flavonoids, polyphenols and terpenoids) from HMs. We hope this review will be conducive to deepening the understanding of the relationship between gut microbial transformation and therapeutic activities of HMs compounds.
Collapse
|
16
|
Ju Z, Li J, Lu Q, Yang Y, Yang L, Wang Z. Identification and quantitative investigation of the effects of intestinal microflora on the metabolism and pharmacokinetics of notoginsenoside Fc assayed by liquid chromatography with electrospray ionization tandem mass spectrometry. J Sep Sci 2019; 42:1740-1749. [PMID: 30805999 DOI: 10.1002/jssc.201801237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 12/13/2022]
Abstract
Notoginsenoside Fc, which is a protopanaxdiol-type saponin isolated from the leaves of Panax notoginseng, exhibits an exceptional antiplatelet aggregatory effect. To study the modulating effect of gastrointestinal contents on the metabolic profile and pharmacokinetics, pseudo germ-free rats were used to study the influence of the bacterial community structure on the metabolic profile. Glycosidase activities were measured using the spectrophotometric method. Biotransformations of notoginsenoside Fc in normal and pseudo germ-free rat intestinal microflora were systematically investigated using ultra high performance liquid chromatography with tandem quadrupole/time-of-flight mass spectrometry. Moreover, a liquid chromatography with tandem mass spectrometry method was established for simultaneous determination of the notoginsenoside Fc prototype and its degradation products. Through an in vivo pharmacokinetic study, the pharmacokinetic characteristics were compared between normal rats and pseudo germ-free rats. During the in vitro biotransformation, seven deglycosylated products were detected and identified after incubation in the intestinal bacteria of normal rats. In pseudo germ-free rats, glycosidase activities were significantly decreased, and no obvious degradation occurred. In an in vivo study, the systemic exposure was significantly increased 40%, as evidenced by the area under the blood concentration-time curve from time zero to infinity value and half-life value, which were prolonged more in the pseudo germ-free group than in normal rats. The results demonstrate that patients who use intestinal bacteria-metabolized herbs, such as panax notoginseng, should understand the profile of intestinal bacteria to ensure therapeutic efficacy.
Collapse
Affiliation(s)
- Zhengcai Ju
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Jia Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China.,Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, P. R. China
| | - Qian Lu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yingbo Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China.,Kanion Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China.,Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, P. R. China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China.,Shanghai R&D Centre for Standardization of Chinese Medicines, Shanghai, P. R. China
| |
Collapse
|
17
|
Zhi H, Deng Y, Yan B, Li Z, Han S, Zhang Y, Hou J, Wu W, Guo D. Study on the herb-herb interaction of Danqi Tongmai Tablet based on the pharmacokinetics of twelve notoginsenoides in acute myocardial ischemia and sham rats. J Pharm Biomed Anal 2019; 166:52-65. [DOI: 10.1016/j.jpba.2018.12.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/22/2018] [Accepted: 12/28/2018] [Indexed: 12/22/2022]
|
18
|
Ginsenoside Rb1 as an Anti-Diabetic Agent and Its Underlying Mechanism Analysis. Cells 2019; 8:cells8030204. [PMID: 30823412 PMCID: PMC6468558 DOI: 10.3390/cells8030204] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/22/2019] [Accepted: 02/23/2019] [Indexed: 12/19/2022] Open
Abstract
Panax ginseng and Panax notoginseng, two well-known medical plants with economic value, have a long history of use for managing various diseases in Asian countries. Accumulating clinical and experimental evidence suggests that notoginsenosides and ginsenosides, which are the major bioactive components of the plants, have a variety of beneficial effects on several types of disease, including metabolic, vascular, and central nervous system disease. Considerable attention has been focused on ginsenoside Rb1 derived from their common ownership as an anti-diabetic agent that can attenuate insulin resistance and various complications. Particularly, in vitro and in vivo models have suggested that ginsenoside Rb1 exerts various pharmacological effects on metabolic disorders, including attenuation of glycemia, hypertension, and hyperlipidemia, which depend on the modulation of oxidative stress, inflammatory response, autophagy, and anti-apoptosis effects. Regulation of these pathophysiological mechanisms can improve blood glucose and insulin resistance and protect against macrovascular/microvascular related complications. This review summarizes the pharmacological effects and mechanisms of action of ginsenoside Rb1 in the management of diabetes or diabetic complications. Moreover, a multi-target effect and mechanism analysis of its antidiabetic actions were performed to provide a theoretical basis for further pharmacological studies and new drug development for clinical treatment of type 2 diabetes. In conclusion, ginsenoside Rb1 exerts significant anti-obesity, anti-hyperglycemic, and anti-diabetic effects by regulating the effects of glycolipid metabolism and improving insulin and leptin sensitivities. All of these findings suggest ginsenoside Rb1 exerts protective effects on diabetes and diabetic complications by the regulation of mitochondrial energy metabolism, improving insulin resistance and alleviating the occurrence complications, which should be further explored. Hence, ginsenoside Rb1 may be developed as a potential anti-obesity, anti-hyperglycemic, and anti-diabetic agent with multi-target effects.
Collapse
|
19
|
Zhou J, Wu J, Wu CY, Long F, Shen H, Zhang W, Li SL. Herb-drug interaction: A case study of effects and involved mechanisms of cisplatin on the pharmacokinetics of ginsenoside Rb1 in tumor-bearing mice. Biomed Pharmacother 2018; 110:95-104. [PMID: 30466007 DOI: 10.1016/j.biopha.2018.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/25/2018] [Accepted: 11/06/2018] [Indexed: 10/27/2022] Open
Abstract
Ginseng is often prescribed together with cisplatin for treatment of cancer, but the interaction between ginseng and cisplatin is still unknown. This study employed ginsenoside Rb1 (Rb1), one of the major components in ginseng, to explore the effects and involved mechanisms of cisplatin on the pharmacokinetics of ginseng. The effects of cisplatin on the pharmacokinetics of Rb1 and its bioactive metabolites Rd, Rg3, and F2 were investigated by using A549-bearing mice with and without cisplatin intervention. Our data showed that cisplatin could significantly decrease the AUC(0-t) and Cmax of Rd, Rg3, and F2, except Rb1. To evaluate the involved mechanisms, feces and intestinal mucosa were collected to explore the effects of cisplatin on the gut metabolism of Rb1 in vitro; meanwhile, Caco-2 cell model and small intestine histological characters were examined to evaluate the effects of cisplatin on the gut absorptive areas and permeability. The mechanisms involved may be mainly related to the comprehensive contributions of inhibited intestinal bacteria and mucosa metabolisms, narrowed intestinal absorptive area, increased efflux ratio of intestinal absorption and enhanced intestinal permeability. All these findings suggested that the dosage of ginseng traditionally used for health protection should be adjusted when it was prescribed together with cisplatin in the treatment of cancer.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, PR China
| | - Jie Wu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Cheng-Ying Wu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Fang Long
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Hong Shen
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, PR China
| | - Wei Zhang
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, PR China.
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, PR China.
| |
Collapse
|
20
|
Chen H, Shen J, Li H, Zheng X, Kang D, Xu Y, Chen C, Guo H, Xie L, Wang G, Liang Y. Ginsenoside Rb1 exerts neuroprotective effects through regulation of Lactobacillus helveticus abundance and GABA A receptor expression. J Ginseng Res 2018; 44:86-95. [PMID: 32095096 PMCID: PMC7033341 DOI: 10.1016/j.jgr.2018.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/02/2018] [Accepted: 09/11/2018] [Indexed: 12/26/2022] Open
Abstract
Background Ginsenoside Rb1 (Rb1), one of the most abundant protopanaxadiol-type ginsenosides, exerts excellent neuroprotective effects even though it has low intracephalic exposure. Purpose The present study aimed to elucidate the apparent contradiction between the pharmacokinetics and pharmacodynamics of Rb1 by studying the mechanisms underlying neuroprotective effects of Rb1 based on regulation of microflora. Methods A pseudo germ-free (PGF) rat model was established, and neuroprotective effects of Rb1 were compared between conventional and PGF rats. The relative abundances of common probiotics were quantified to reveal the authentic probiotics that dominate in the neuroprotection of Rb1. The expressions of the gamma-aminobutyric acid (GABA) receptors, including GABAA receptors (α2, β2, and γ2) and GABAB receptors (1b and 2), in the normal, ischemia/reperfusion (I/R), and I/R+Rb1 rat hippocampus and striatum were assessed to reveal the neuroprotective mechanism of Rb1. Results The results showed that microbiota plays a key role in neuroprotection of Rb1. The relative abundance of Lactobacillus helveticus (Lac.H) increased 15.26 fold after pretreatment with Rb1. I/R surgery induced effects on infarct size, neurological deficit score, and proinflammatory cytokines (IL-1β, IL-6, and TNF-α) were prevented by colonizing the rat gastrointestinal tract with Lac.H (1 × 109 CFU) by gavage 15 d before I/R surgery. Both Rb1 and Lac.H upregulated expression of GABA receptors in I/R rats. Coadministration of a GABAA receptor antagonist significantly attenuated neuroprotective effects of Rb1 and Lac.H. Conclusion In sum, Rb1 exerts neuroprotective effects by regulating Lac.H and GABA receptors rather than through direct distribution to the target sites.
Collapse
Affiliation(s)
- Huimin Chen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiajia Shen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haofeng Li
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiao Zheng
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dian Kang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yangfan Xu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chong Chen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huimin Guo
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
21
|
Wu W, Jiao C, Li H, Ma Y, Jiao L, Liu S. LC-MS based metabolic and metabonomic studies of Panax ginseng. PHYTOCHEMICAL ANALYSIS : PCA 2018; 29:331-340. [PMID: 29460310 DOI: 10.1002/pca.2752] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 05/28/2023]
Abstract
INTRODUCTION Panax ginseng has received much attention as a valuable health supplement with medicinal potential. Its chemical diversity and multiple pharmacological properties call for comprehensive methods to better understand the effects of ginseng and ginsenosides. Liquid chromatography-mass spectrometry (LC-MS) based metabonomic approaches just fit the purpose. OBJECTIVE Aims to give a review of recent progress on LC-MS based pharmacokinetic, metabolic, and phytochemical metabolomic studies of ginseng, and metabonomic studies of ginseng intervention effects. METHODS The review has four sections: the first section discusses metabolic studies of ginsenosides based on LC-MS, the second focuses on ginsenoside-drug interactions and pharmacokinetic interaction between herb compounds based on LC-MS, the third is phytochemical metabolomic studies of ginseng based on LC-MS, and the fourth deals with metabonomic studies of ginseng intervention effects based on LC-MS. RESULTS LC-MS based metabonomic research on ginseng include analysis of single ginsenoside and total ginsenosides. The theory of multi-components and multi-targeted mechanisms helps to explain ginseng effects. CONCLUSION LC-MS based metabonomics is a promising way to comprehensively assess ginseng. It is valuable for quality control and mechanism studies of ginseng.
Collapse
Affiliation(s)
- Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Chuanxi Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Yue Ma
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| |
Collapse
|
22
|
Bai L, Gao J, Wei F, Zhao J, Wang D, Wei J. Therapeutic Potential of Ginsenosides as an Adjuvant Treatment for Diabetes. Front Pharmacol 2018; 9:423. [PMID: 29765322 PMCID: PMC5938666 DOI: 10.3389/fphar.2018.00423] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Ginseng, one of the oldest traditional Chinese medicinal herbs, has been used widely in China and Asia for thousands of years. Ginsenosides extracted from ginseng, which is derived from the roots and rhizomes of Panax ginseng C. A. Meyer, have been used in China as an adjuvant in the treatment of diabetes mellitus. Owing to the technical complexity of ginsenoside production, the total ginsenosides are generally extracted. Accumulating evidence has shown that ginsenosides exert antidiabetic effects. In vivo and in vitro tests revealed the potential of ginsenoside Rg1, Rg3, Rg5, Rb1, Rb2, Rb3, compound K, Rk1, Re, ginseng total saponins, malonyl ginsenosides, Rd, Rh2, F2, protopanaxadiol (PPD) and protopanaxatriol (PPT)-type saponins to treat diabetes and its complications, including type 1 diabetes mellitus, type 2 diabetes mellitus, diabetic nephropathy, diabetic cognitive dysfunction, type 2 diabetes mellitus with fatty liver disease, diabetic cerebral infarction, diabetic cardiomyopathy, and diabetic erectile dysfunction. Many effects are attributed to ginsenosides, including gluconeogenesis reduction, improvement of insulin resistance, glucose transport, insulinotropic action, islet cell protection, hepatoprotective activity, anti-inflammatory effect, myocardial protection, lipid regulation, improvement of glucose tolerance, antioxidation, improvement of erectile dysfunction, regulation of gut flora metabolism, neuroprotection, anti-angiopathy, anti-neurotoxic effects, immunosuppression, and renoprotection effect. The molecular targets of these effects mainly contains GLUTs, SGLT1, GLP-1, FoxO1, TNF-α, IL-6, caspase-3, bcl-2, MDA, SOD, STAT5-PPAR gamma pathway, PI3K/Akt pathway, AMPK-JNK pathway, NF-κB pathway, and endoplasmic reticulum stress. Rg1, Rg3, Rb1, and compound K demonstrated the most promising therapeutic prospects as potential adjuvant medicines for the treatment of diabetes. This paper highlights the underlying pharmacological mechanisms of the anti-diabetic effects of ginsenosides.
Collapse
Affiliation(s)
- Litao Bai
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialiang Gao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Danwei Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Influence of Nutritional Status on the Absorption of Polyphyllin I, an Anticancer Candidate from Paris polyphylla in Rats. Eur J Drug Metab Pharmacokinet 2018; 43:587-597. [DOI: 10.1007/s13318-018-0473-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
24
|
The gastrointestinal behavior of saponins and its significance for their bioavailability and bioactivities. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.11.032] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
25
|
Jang SH, Park J, Kim SH, Choi KM, Ko ES, Cha JD, Lee YR, Jang H, Jang YS. Red ginseng powder fermented with probiotics exerts antidiabetic effects in the streptozotocin-induced mouse diabetes model. PHARMACEUTICAL BIOLOGY 2017; 55:317-323. [PMID: 27927080 PMCID: PMC6130625 DOI: 10.1080/13880209.2016.1237978] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 09/13/2016] [Indexed: 05/28/2023]
Abstract
CONTEXT Red ginseng (heat-processed Panax ginseng) is a well-known alternative medicine with pharmacological antidiabetic activity. It exerts pharmacological effects through the transformation of saponin into metabolites by the intestinal microbiota. Given that intestinal conditions and intestinal microflora vary among individuals, the pharmacological effects of orally administered red ginseng likely may vary among individuals. OBJECTIVE To overcome this variation and produce homogeneously effective red ginseng, we evaluated the antidiabetic effects of probiotic-fermented red ginseng in a mouse model. MATERIALS AND METHODS The antidiabetic efficacy of orally administered probiotic-fermented red ginseng was assessed in ICR mice after induction of diabetes using streptozotocin (170 mg/kg body weight). Samples were given orally for 8 weeks, and indicators involved in diabetic disorders such as body weight change, water intake, blood glucose, glucose tolerance and various biochemical parameters were determined. RESULTS Oral administration of probiotic-fermented red ginseng significantly decreased the level of blood glucose of about 62.5% in the fasting state and induced a significant increase in glucose tolerance of about 10.2% compared to the control diabetic mice. Additionally, various indicators of diabetes and biochemical data (e.g., blood glycosylated haemoglobin level, serum concentrations of insulin, and α-amylase activity) showed a significant improvement in the diabetic conditions of the mice treated with probiotic-fermented red ginseng in comparison with those of control diabetic mice. DISCUSSION AND CONCLUSION Our results demonstrate the antidiabetic effects of probiotic-fermented red ginseng in the streptozotocin-induced mouse diabetes model and suggest that probiotic-fermented red ginseng may be a uniformly effective red ginseng product.
Collapse
Affiliation(s)
- Sun-Hee Jang
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
| | - Jisang Park
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, Korea
| | - Sae-Hae Kim
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, Korea
| | - Kyung-Min Choi
- Department of Efficacy Research, Institute of Jinan Red Ginseng, Jinan, Korea
| | - Eun-Sil Ko
- Department of Efficacy Research, Institute of Jinan Red Ginseng, Jinan, Korea
| | - Jeong-Dan Cha
- Department of Efficacy Research, Institute of Jinan Red Ginseng, Jinan, Korea
| | - Young-Ran Lee
- Department of Mathematics, Sogang University, Seoul, Korea
| | - Hyonseok Jang
- Department of Oral Maxillofacial Surgery, Korea University Ansan Hospital, Ansan, Korea
| | - Yong-Suk Jang
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
- Department of Efficacy Research, Institute of Jinan Red Ginseng, Jinan, Korea
| |
Collapse
|
26
|
Dong WW, Han XZ, Zhao J, Zhong FL, Ma R, Wu S, Li D, Quan LH, Jiang J. Metabolite profiling of ginsenosides in rat plasma, urine and feces by LC-MS/MS and its application to a pharmacokinetic study after oral administration of Panax ginseng
extract. Biomed Chromatogr 2017; 32. [DOI: 10.1002/bmc.4105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/02/2017] [Accepted: 09/24/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Wei-Wei Dong
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Xiong-Zhe Han
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Jinhua Zhao
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Fei-Liang Zhong
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Rui Ma
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Songquan Wu
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Donghao Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Lin-Hu Quan
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| | - Jun Jiang
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education; Yanji City Jilin China
| |
Collapse
|
27
|
Gimenez-Bastida JA, Martinez Carreras L, Moya-Pérez A, Laparra Llopis JM. Pharmacological Efficacy/Toxicity of Drugs: A Comprehensive Update About the Dynamic Interplay of Microbes. J Pharm Sci 2017; 107:778-784. [PMID: 29107046 PMCID: PMC6712421 DOI: 10.1016/j.xphs.2017.10.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
Oral ingestion is a common, easy to access, route for therapeutic drugs to be delivered. The conception of the gastrointestinal tract as a passive physiological compartment has evolved toward a dynamic perspective of the same. Thus, microbiota plays an important role in contributing with additional metabolic capacities to its host as well as to its phenotypic heterogeneity. These adaptations in turn influence the efficacy and toxicity of a broad range of drugs. Notwithstanding, xenobiotics and therapeutic drugs affecting the microbiome's activity also significantly impact metabolism affecting different organs and tissues, and thereby drugs' toxicity/efficacy effects. Other physiological interfaces (i.e., gut, lungs, and skin) also represent complex media with features about microbiota's composition. In addition, there have been described key regulatory effects of microbes on immunotherapy, because of its potential harnessing the host immune system, mental disorders by modulating neuroendocrine systems and cancer. These alterations are responsible of physiological variations in the response(s) between individuals and populations. However, the study of population-based differences in intestinal microbial-related drug metabolism has been largely inferential. This review outlines major reciprocal implications between drugs and microbes regulatory capacities in pharmacotherapy.
Collapse
Affiliation(s)
- Juan Antonio Gimenez-Bastida
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Lucia Martinez Carreras
- Nutrition Precision in Cancer Unit, Madrid Institute for Advanced Studies in Food (IMDEA Food), Madrid 28049, Spain
| | - Angela Moya-Pérez
- Department of Developmental and Cell Biology, University of California, Irvine, California 92617
| | - José Moisés Laparra Llopis
- Nutrition Precision in Cancer Unit, Madrid Institute for Advanced Studies in Food (IMDEA Food), Madrid 28049, Spain.
| |
Collapse
|
28
|
de Matos AM, de Macedo MP, Rauter AP. Bridging Type 2 Diabetes and Alzheimer's Disease: Assembling the Puzzle Pieces in the Quest for the Molecules With Therapeutic and Preventive Potential. Med Res Rev 2017; 38:261-324. [PMID: 28422298 DOI: 10.1002/med.21440] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/18/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes (T2D) and Alzheimer's disease (AD) are two age-related amyloid diseases that affect millions of people worldwide. Broadly supported by epidemiological data, the higher incidence of AD among type 2 diabetic patients led to the recognition of T2D as a tangible risk factor for the development of AD. Indeed, there is now growing evidence on brain structural and functional abnormalities arising from brain insulin resistance and deficiency, ultimately highlighting the need for new approaches capable of preventing the development of AD in type 2 diabetic patients. This review provides an update on overlapping pathophysiological mechanisms and pathways in T2D and AD, such as amyloidogenic events, oxidative stress, endothelial dysfunction, aberrant enzymatic activity, and even shared genetic background. These events will be presented as puzzle pieces put together, thus establishing potential therapeutic targets for drug discovery and development against T2D and diabetes-induced cognitive decline-a heavyweight contributor to the increasing incidence of dementia in developed countries. Hoping to pave the way in this direction, we will present some of the most promising and well-studied drug leads with potential against both pathologies, including their respective bioactivity reports, mechanisms of action, and structure-activity relationships.
Collapse
Affiliation(s)
- Ana Marta de Matos
- Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016, Lisbon, Portugal.,CEDOC Chronic Diseases, Nova Medical School, Rua Câmara Pestana n 6, 6-A, Ed. CEDOC II, 1150-082, Lisbon, Portugal
| | - Maria Paula de Macedo
- CEDOC Chronic Diseases, Nova Medical School, Rua Câmara Pestana n 6, 6-A, Ed. CEDOC II, 1150-082, Lisbon, Portugal
| | - Amélia Pilar Rauter
- Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016, Lisbon, Portugal
| |
Collapse
|
29
|
Xu J, Chen HB, Li SL. Understanding the Molecular Mechanisms of the Interplay Between Herbal Medicines and Gut Microbiota. Med Res Rev 2017; 37:1140-1185. [PMID: 28052344 DOI: 10.1002/med.21431] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/21/2016] [Accepted: 11/16/2016] [Indexed: 02/06/2023]
Abstract
Herbal medicines (HMs) are much appreciated for their significant contribution to human survival and reproduction by remedial and prophylactic management of diseases. Defining the scientific basis of HMs will substantiate their value and promote their modernization. Ever-increasing evidence suggests that gut microbiota plays a crucial role in HM therapy by complicated interplay with HM components. This interplay includes such activities as: gut microbiota biotransforming HM chemicals into metabolites that harbor different bioavailability and bioactivity/toxicity from their precursors; HM chemicals improving the composition of gut microbiota, consequently ameliorating its dysfunction as well as associated pathological conditions; and gut microbiota mediating the interactions (synergistic and antagonistic) between the multiple chemicals in HMs. More advanced experimental designs are recommended for future study, such as overall chemical characterization of gut microbiota-metabolized HMs, direct microbial analysis of HM-targeted gut microbiota, and precise gut microbiota research model development. The outcomes of such research can further elucidate the interactions between HMs and gut microbiota, thereby opening a new window for defining the scientific basis of HMs and for guiding HM-based drug discovery.
Collapse
Affiliation(s)
- Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, P.R. China.,Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, P.R. China
| |
Collapse
|
30
|
Xiao J, Chen H, Kang D, Shao Y, Shen B, Li X, Yin X, Zhu Z, Li H, Rao T, Xie L, Wang G, Liang Y. Qualitatively and quantitatively investigating the regulation of intestinal microbiota on the metabolism of panax notoginseng saponins. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:324-336. [PMID: 27637802 DOI: 10.1016/j.jep.2016.09.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/21/2016] [Accepted: 09/13/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intestinal microflora plays crucial roles in modulating pharmacokinetic characteristics and pharmacological actions of active ingredients in traditional Chinese medicines (TCMs). However, the exact impact of altered intestinal microflora affecting the biotransformation of TCMs remains poorly understood. AIMS OF THE STUDY This study aimed to reveal the specific enterobacteria which dominate the metabolism of panax notoginseng saponins (PNSs) via exploring the relationship between bacterial community structures and the metabolic profiles of PNSs. MATERIALS AND METHODS 2, 4, 6-Trinitrobenzenesulphonic acid (TNBS)-challenged and pseudo germ-free (pseudo GF) rats, which prepared by treating TNBS and antibiotic cocktail, respectively, were employed to investigate the influence of intestinal microflora on the PNS metabolic profiles. Firstly, the bacterial community structures of the conventional, TNBS-challenged and pseudo GF rat intestinal microflora were compared via 16S rDNA amplicon sequencing technique. Then, the biotransformation of protopanaxadiol-type PNSs (ginsenoside Rb1, Rb2 and Rd), protopanaxatriol-type PNSs (ginsenoside Re, Rf, Rg1 and notoginsenoside R1) and Panax notoginseng extract (PNE) in conventional, TNBS-challenged and pseudo GF rat intestinal microbiota was systematically studied from qualitative and quantitative angles based on LC-triple-TOF/MS system. Besides, glycosidases (β-glucosidase and β-xylosidase), predominant enzymes responsible for the deglycosylation of PNSs, were measured by the glycosidases assay kits. RESULTS Significant differences in the bacterial community structure on phylum, class, order, family, and genera levels were observed among the conventional, TNBS-challenged and pseudo GF rats. Most of the metabolites in TNBS-challenged rat intestinal microflora were identified as the deglycosylation products, and had slightly lower exposure levels than those in the conventional rats. In the pseudo GF group, the peak area of metabolites formed by loss of glucose, xylose and rhamnose was significantly lower than that in the conventional group. Importantly, the exposure levels of the deglycosylated metabolites were found have a high correlation with the alteration of glycosidase activities and proteobacteria population. Several other metabolites, which formed by oxidation, dehydrogenation, demethylation, etc, had higher relative exposure in pseudo GF group, which implicated that the up-regulation of Bacteroidetes could enhance the activities of some redox enzymes in intestinal microbiota. CONCLUSION The metabolism of PNSs was greatly influenced by intestinal microflora. Proteobacteria may affect the deglycosylated metabolism of PNSs via regulating the activities of glycosidases. Besides, up-regulation of Bacteroidetes was likely to promote the redox metabolism of PNSs via improving the activities of redox metabolic enzymes in intestinal microflora.
Collapse
Affiliation(s)
- Jingcheng Xiao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Huimin Chen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Dian Kang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Yuhao Shao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Boyu Shen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Xinuo Li
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Xiaoxi Yin
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Zhangpei Zhu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Haofeng Li
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Tai Rao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China.
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China.
| |
Collapse
|
31
|
Kang A, Zhang S, Zhu D, Dong Y, Shan J, Xie T, Wen H, Di L. Gut microbiota in the pharmacokinetics and colonic deglycosylation metabolism of ginsenoside Rb1 in rats: Contrary effects of antimicrobials treatment and restraint stress. Chem Biol Interact 2016; 258:187-96. [DOI: 10.1016/j.cbi.2016.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/09/2016] [Accepted: 09/06/2016] [Indexed: 02/02/2023]
|