1
|
Zhang L, Zhao XL, Cao ZJ, Li KD, Xu LY, Tang F, Zhang JN, Peng C, Ao H. Ginsenoside CK inhibits EMT and overcomes oxaliplatin resistance in gastric cancer by targeting the PI3K/Akt pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156516. [PMID: 40054176 DOI: 10.1016/j.phymed.2025.156516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Gastric cancer remains a leading cause of cancer mortality, with oxaliplatin (L-OHP) resistance posing a major therapeutic challenge. Ginsenosides have shown potential in addressing chemoresistance. PURPOSE This study aimed to investigate whether ginsenoside Compound K (CK), a derivative of protopanaxadiol ginsenosides, could overcome L-OHP resistance in gastric cancer cells. METHODS The anti-cancer effects of CK were investigated using L-OHP-resistant HGC27/L cells through comprehensive in vitro experiments. Cell viability, migration, invasion, apoptosis, and colony formation were evaluated under CK treatment alone or combined with L-OHP. Drug efflux was specifically assessed using Rhodamine 123 staining. To understand the molecular mechanism, network pharmacology and molecular docking analyses were employed, which identified the PI3K/Akt pathway as a crucial target of CK. This finding was further validated through Western blotting and RT-qPCR analyses, focusing on PI3K/Akt signaling components and EMT markers. Finally, drug-resistant gastric cancer xenograft models were established to evaluate the therapeutic efficacy of CK alone and in combination with L-OHP in vivo. RESULTS CK effectively suppressed cell viability, migration, invasion, drug efflux, and colony formation while enhancing apoptosis in resistant cells. Mechanistically, CK inhibited the PI3K/Akt pathway, leading to reduced P-glycoprotein (P-gp) expression and EMT reversal. These effects were confirmed using PI3K pathway modulators. In xenograft models, CK significantly inhibited tumor growth and reduced PI3K/Akt activity, P-gp expression, and EMT markers. CONCLUSION This study demonstrates that CK overcomes L-OHP resistance through PI3K/Akt pathway inhibition and EMT prevention, suggesting that combining CK with L-OHP may improve outcomes in chemoresistant gastric cancer patients.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhang-Jing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ke-Di Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Li K, Wang YJ, Chen C, Wang XJ, Li W. Targeting pyroptosis: A novel strategy of ginseng for the treatment of diabetes and its chronic complications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156430. [PMID: 39892311 DOI: 10.1016/j.phymed.2025.156430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/28/2024] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Pyroptosis is a recently identified form of programmed cell death that plays a crucial role in the pathogenesis and progression of diabetes and associated chronic complications, while the occurrence mechanism remains unclear. Ginseng (Panax Ginseng C. A. Mey.) is a valuable traditional medicinal material with proved therapeutic effects on prevention and treatment of diabetes and diabetic complications. Targeting pyroptosis pathway has become a focus of study for ginseng in improvement of diabetes and related chronic complications. PURPOSE The review aims to elucidate the happening mechanism of pyroptosis in diabetes and diabetic chronic complications, evaluate the effects of ginseng and its active components on diabetes and its chronic complications via pyroptosis-related pathways, and provide a new perspective for the management of diabetes. METHODS We conducted the literature retrieval with PubMed, Web of Science, and ScienceDirect databases in a systematic manner (up to August 2024). The keywords included pyroptosis, diabetes, diabetic nephropathy, diabetic retinopathy, diabetic cardiomyopathy, diabetic neuropathy, ginseng, ginseng extract, and ginsenoside. The obtained literatures were comprehensively sorted out. RESULTS Oxidative stress, endoplasmic reticulum stress (ERS), and inflammatory responses were primary contributors to pyroptosis in diabetes and associated chronic complications. In addition, some RNA molecules (miRNAs, circRNAs, and lncRNAs) also contributed to pyroptosis under hyperglycemia. The signaling pathways mainly included Nrf2/HO-1, IκB/NF-κB/NLRP3, NOX1/NOX4/TXNIP, and P2X7R/TXNIP/NLRP3. Ginseng extracts, some ginsenosides and flavonoid (Quercetin) could exert anti-diabetic effect by regulating pyroptosis-related pathways. We also discussed the toxicity, side effects and clinical applications of ginseng. CONCLUSION In summary, this review elucidates the happening mechanisms of pyroptosis in diabetes and associated chronic complications, and summarizes published studies about ginseng and its active ingredients in improving diabetes by regulating pyroptosis-related pathways. However, almost all researches are limited to animal and cell experiments, and more clinical trials are required to prove the therapeutic effect of ginseng on diabetes by targeting pyroptosis.
Collapse
Affiliation(s)
- Ke Li
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China; College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China
| | - Ya-Jun Wang
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China; College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Chen Chen
- Endocrinology and Metabolism, School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
| | - Xiao-Jie Wang
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China; School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Wei Li
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China; College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China.
| |
Collapse
|
3
|
Shang S, Yang H, Qu L, Fan D, Deng J. Ginsenoside, a potential natural product against liver diseases: a comprehensive review from molecular mechanisms to application. Crit Rev Food Sci Nutr 2025:1-25. [PMID: 39810734 DOI: 10.1080/10408398.2025.2451761] [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: 01/16/2025]
Abstract
Liver disease constitutes a significant cause of global mortality, with its pathogenesis being multifaceted. Identifying effective pharmacological and preventive strategies is imperative for liver protection. Ginsenosides, the major bioactive compounds found in ginseng, exhibit multiple pharmacological activities including protection against liver-related diseases by mitigating liver fat accumulation and inflammation, preventing hepatic fibrosis, and exerting anti-hepatocarcinogenic effects. However, a comprehensive overview elucidating the regulatory pathways associated with ginsenosides in liver disease remains elusive. This review aims to consolidate the molecular mechanisms through which different ginsenosides ameliorate distinct liver diseases, alongside the pathogenic factors underlying liver ailments. Notably, ginsenosides Rb1 and Rg1 demonstrate significantly effective in treating fatty liver, hepatitis, and liver fibrosis, and ginsenosides CK and Rh2 exhibit potent anti-hepatocellular carcinogenic effects. Their molecular mechanisms underlying these effects primarily involve the modulation of AMPK, NF-κB, TGF-β, NFR2, JNK, and other pathways, thereby attenuating hepatic fat accumulation, inflammation, inhibition of hepatic stellate cell activation, and promoting apoptosis in hepatocellular carcinoma cells. Furthermore, it provides insights into the safety profile and current applications of ginsenosides, thereby facilitating their clinical development. Consequently, ginsenosides present promising prospects for liver disease management, underscoring their potential as valuable therapeutic agents in this context.
Collapse
Affiliation(s)
- Shiyan Shang
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
4
|
Niu Z, Liu Y, Shen R, Jiang X, Wang Y, He Z, Li J, Hu Y, Zhang J, Jiang Y, Hu W, Si C, Wei S, Shen T. Ginsenosides from Panax ginseng as potential therapeutic candidates for the treatment of inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155474. [PMID: 38471369 DOI: 10.1016/j.phymed.2024.155474] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by a chronic inflammation of the intestine, which significantly affects patients' quality of life. As a perennial plant with the homology of medicine and food, Panax ginseng is known for its substantial anti-inflammatory effects in various inflammatory disorders. Ginsenosides, the main bioactive compounds of P. ginseng, are recognized for their efficacy in ameliorating inflammation. PURPOSE Over the past decade, approximately 150 studies have investigated the effects of P. ginseng and ginsenosides on IBD treatment and new issues have arisen. However, there has yet to be a comprehensive review assessing the potential roles of ginsenosides in IBD therapy. METHOD This manuscript strictly adheres to the PRISMA guidelines, thereby guaranteeing systematic synthesis of data. The research articles referenced were sourced from major scientific databases, including Google Scholar, PubMed, and Web of Science. The search strategy employed keywords such as "ginsenoside", "IBD", "colitis", "UC", "inflammation", "gut microbiota", and "intestinal barrier". For image creation, Figdraw 2.0 was methodically employed. RESULTS Treatment with various ginsenosides markedly alleviated clinical IBD symptoms. These compounds have been observed to restore intestinal epithelia, modulate cellular immunity, regulate gut microbiota, and suppress inflammatory signaling pathways. CONCLUSION An increasing body of research supports the potential of ginsenosides in treating IBD. Ginsenosides have emerged as promising therapeutic agents for IBD, attributed to their remarkable efficacy, safety, and absence of side effects. Nevertheless, their limited bioavailability presents a substantial challenge. Thus, efforts to enhance the bioavailability of ginsenosides represent a crucial and promising direction for future IBD research.
Collapse
Affiliation(s)
- Zhiqiang Niu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yanan Liu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ruyi Shen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaojian Jiang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Yanting Wang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ziliang He
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Junyao Li
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yeye Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ji Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Yunyao Jiang
- Institute for Chinese Materia Medica, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chuanling Si
- Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Shuai Wei
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Ting Shen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
5
|
Morshed MN, Akter R, Karim MR, Iqbal S, Kang SC, Yang DC. Bioconversion, Pharmacokinetics, and Therapeutic Mechanisms of Ginsenoside Compound K and Its Analogues for Treating Metabolic Diseases. Curr Issues Mol Biol 2024; 46:2320-2342. [PMID: 38534764 DOI: 10.3390/cimb46030148] [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: 02/14/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Rare ginsenoside compound K (CK) is an intestinal microbial metabolite with a low natural abundance that is primarily produced by physicochemical processing, side chain modification, or metabolic transformation in the gut. Moreover, CK exhibits potent biological activity compared to primary ginsenosides, which has raised concerns in the field of ginseng research and development, as well as ginsenoside-related dietary supplements and natural products. Ginsenosides Rb1, Rb2, and Rc are generally used as a substrate to generate CK via several bioconversion processes. Current research shows that CK has a wide range of pharmacological actions, including boosting osteogenesis, lipid and glucose metabolism, lipid oxidation, insulin resistance, and anti-inflammatory and anti-apoptosis properties. Further research on the bioavailability and toxicology of CK can advance its medicinal application. The purpose of this review is to lay the groundwork for future clinical studies and the development of CK as a therapy for metabolic disorders. Furthermore, the toxicology and pharmacology of CK are investigated as well in this review. The findings indicate that CK primarily modulates signaling pathways associated with AMPK, SIRT1, PPARs, WNTs, and NF-kB. It also demonstrates a positive therapeutic effect of CK on non-alcoholic fatty liver disease (NAFLD), obesity, hyperlipidemia, diabetes, and its complications, as well as osteoporosis. Additionally, the analogues of CK showed more bioavailability, less toxicity, and more efficacy against disease states. Enhancing bioavailability and regulating hazardous variables are crucial for its use in clinical trials.
Collapse
Affiliation(s)
- Md Niaj Morshed
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Reshmi Akter
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Md Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Se Chan Kang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Republic of Korea
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea
| |
Collapse
|
6
|
Cao Y, Tao F, Yu Y, Song L, Zhang R, Feng J, Zhai Q, Xue P. Safety evaluation of rare ginsenosides of stems and leaves from American ginseng: 90-day exposure toxicity study combined with intestinal flora analysis and metabonomics in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115429. [PMID: 37660532 DOI: 10.1016/j.ecoenv.2023.115429] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/24/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Rare ginsenosides have already been widely applied in many fields, including health food and bio-medicine. The human being can expose to rare ginsenosides directly or indirectly increasingly. However, there are few studies on the safety assessment of rare ginsenoside mixtures. In the present study, the sub-chronic toxicity of rare ginsenosides for 90 days on SD rats was performed by combining the intestinal flora analysis and urine metabonomics aiming to illustrate the safety of long-term consumption of rare ginsenosides and the potential damage for liver and intestinal. 48 adult rats were divided into four groups: control (0 mg/kg), low-dose (60 mg/kg), medium-dose (200 mg/kg), and high-dose (600 mg/kg). Rats in the high-dose group showed inflammatory changes in their livers and intestines. The strong bactericidal effect of rare ginsenosides caused intestinal flora disorder and changed the structure of intestinal flora in rats, thus inducing intestinal damage in rats. In the high-dose group, levels of alanine aminotransferase (ALT), lactate dehydrogenase (LDH), and alkaline phosphatase (AKP) increased significantly. As a result of the high-dose treatment, certain metabolic pathways were altered, such as vitamin B6 metabolism, methionine metabolism, glutathione metabolism, and others. These results indicated that high doses of rare ginsenosides induced liver injury by affecting the above metabolic pathways. Rare ginsenosides with no observed adverse effect level (NOAEL) were below 200 mg/kg/day in vivo. Thus, this present study provides insight into the rational use of rare ginsenosides.
Collapse
Affiliation(s)
- Yuqing Cao
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Feiyan Tao
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Yuan Yu
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Linmeng Song
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Ruoyu Zhang
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jing Feng
- School of Rehabilitation, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Qingfeng Zhai
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China.
| | - Peng Xue
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|
7
|
Xu J, Wu G, Yu X, Dong Z, Yan J, Wu L, Bao L, Liu Q. Exploring the mechanism of MP gel against skin photoaging based on network pharmacology, molecular docking, and experimental validation. J Cosmet Dermatol 2023; 22:1108-1123. [PMID: 36465034 DOI: 10.1111/jocd.15542] [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: 04/07/2022] [Revised: 11/02/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Long-term and high exposure to UV radiation can lead to the development of skin photoaging diseases. Therefore, there is an ongoing need for more natural and safe drugs to prevent or treat skin photoaging diseases. METHODS The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database were used to collect the active compounds and corresponding targets of Cnidii Fructus, Arnebiae Radix, Angelicae Sinensis Radix, Poria, and Borneolum. The GeneCards database and the NCBI Gene database were used to collect the targets of skin photoaging diseases. The STRING database was used to construct a protein-protein interaction network formed by the intersecting targets of drugs and diseases. The Metascape database was applied for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the targets. Molecular docking between active compounds and targets was verified by Autodock. After that, the skin photoaging model of mice was established and treated with MP gel. The skin characterization on the back of mice was observed, and the ameliorative effect of MP gel on skin photoaging was evaluated by histological and epidermal thickness assays. The MDA content and SOD activity were measured. Caspase-3 expression in mouse skin tissues was detected by immunohistochemistry, quantitative real-time polymerase chain reaction assay, and Western blot. RESULTS The results of network pharmacology experiments showed that the natural drugs have multi-component, multi-target therapeutic disease characteristics. The results of animal studies showed that MP gel improved the health of photoaged skin, promoted skin structural integrity, had antioxidant properties and significantly inhibited caspase-3 expression. CONCLUSION The experimental validation of the results of the preliminary network pharmacology analysis was carried out in animal experiments, which confirmed part of the mechanism of action of MP gel in the prevention and treatment of skin photoaging.
Collapse
Affiliation(s)
- Jinfan Xu
- Department of Pharmacology, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Guodong Wu
- Department of Pharmacology, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Xianglin Yu
- Department of Pharmacology, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Zhiheng Dong
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jibiao Yan
- Inner Mongolia Puze Biological Products Co., Ltd., Hohhot, Inner Mongolia, China
| | - Lan Wu
- Mongolia Medical School, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lidao Bao
- Hohhot Mongolian Hospital of Traditional Chinese Medicine, Hohhot, Inner Mongolia, China
| | - Quanli Liu
- Department of Pharmacology, Baotou Medical College, Baotou, Inner Mongolia, China.,Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, Inner Mongolia, China
| |
Collapse
|
8
|
He Z, Xu X, Wang C, Li Y, Dong B, Li S, Zeng J. Effect of Panax quinquefolius extract on Mycobacterium abscessus biofilm formation. BIOFOULING 2023; 39:24-35. [PMID: 36644897 DOI: 10.1080/08927014.2023.2166405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Mycobacterium abscessus (M. abscessus) can exist either as planktonic bacteria or as a biofilm. Biofilm formation is one of the important causes of conversion to resistance to antibiotics of bacteria that were previously sensitive when in their planktonic form, resulting in infections difficult to manage. Panax quinquefolius and its active ingredient ginsenosides have the potential ability in fighting pathogenic infections. In this study, the P. quinquefolius extract (PQE) showed good antibacterial/bactericidal activity against the M. abscessus planktonic cells. The extract reduced the biomass, thickness, and number of M. abscessus in the biofilm and altered its morphological characteristics as well as the spatial distribution of dead/alive bacteria. Moreover, the ginsenoside CK monomer had a similar inhibitory effect on M. abscessus planktonic bacteria and biofilm formation. Therefore, PQE and its monomer CK might be potential novel antimicrobial agents for the clinical prevention and treatment of M. abscessus, including biofilms in chronic infections.
Collapse
Affiliation(s)
- Zhiqun He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xinyue Xu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chuan Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Baoyu Dong
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Shuai Li
- Pharmaceutical Research Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Malik M, Bhatt P, Singh J, Kaushik RD, Sharma G, Kumar V. Preclinical Safety Assessment of Chemically Cross-Linked Modified Mandua Starch: Acute and Sub-Acute Oral Toxicity Studies in Swiss Albino Mice. ACS OMEGA 2022; 7:35506-35514. [PMID: 36249409 PMCID: PMC9558613 DOI: 10.1021/acsomega.2c01309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
In the present era, 28 days of repeated-dose-toxicity study following the Organization for Economic Cooperation and Development (OECD) guidelines 407 is compulsory for every drug to go through phase 1 clinical trials. The increasing demand for high-resistant starch containing nutraceuticals and the applicability of modified starch in development of targeted drug delivery inspired us to investigate the toxic profile of mandua starch chemically cross-linked by epichlorohydrin and compare it with alkali-isolated starch in healthy adult Swiss albino mice, which can be the first step for exploring the use of epichlorohydrin-cross-linked mandua starch (ECC-MS) as a pharmaceutical excipient. Histopathological examinations of the kidney and liver did not expose noteworthy abnormalities in the treated mice. There were no clinical and mortality symptoms of toxicity observed during the repeated-dose-toxicity study. The oral consumption of ECC-MS did not pose any harm as it was neither lethal nor had any harmful hematological, biochemical, psychological, anatomical, and behavioral effects. The use of ECC-MS and alkali-isolated mandua starch (AMS) was found safe at a dose of 2000 mg/kg body weight in the acute toxicity study and at doses of 2000, 1500, and 1000 mg/kg body weight in the sub-acute toxicity study as no detrimental effects were observed after oral administration in mice for 14 and 28 days, respectively.
Collapse
Affiliation(s)
- Mayank
Kumar Malik
- Department
of Pharmaceutical Sciences, Faculty of Medical Science & Health, Gurukula Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
- Department
of Chemistry, Faculty of Sciences, Gurukula
Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
| | - Pankaj Bhatt
- Department
of Pharmaceutical Sciences, Faculty of Medical Science & Health, Gurukula Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
| | - Jaspal Singh
- Department
of Chemistry, Faculty of Sciences, Gurukula
Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
| | - Rajneesh Dutt Kaushik
- Department
of Chemistry, Faculty of Sciences, Gurukula
Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
| | - Gaurav Sharma
- Bilwal
Medchem and Research Laboratory Pvt. Ltd., Jaipur 302023, Rajasthan, India
| | - Vipin Kumar
- Department
of Pharmaceutical Sciences, Faculty of Medical Science & Health, Gurukula Kangri (Deemed to be University), Haridwar 249407, Uttrakhand, India
| |
Collapse
|
10
|
Zhao JQ, Zhao Z, Zhang C, Sun JX, Liu FJ, Yu T, Jiang Y, Li HJ. Long-term oral administration of Epimedii Folium induced cholestasis in mice by interfering with bile acid transport. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115254. [PMID: 35381309 DOI: 10.1016/j.jep.2022.115254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/18/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedii Folium (EF) is a common traditional Chinese medicine that functions as a tonifying kidney yang to strengthen bones and muscles and dispel wind dampness (limb pain, lethargy, nausea, anorexia, and loose stools). Several studies have reported the potential risk of cholestatic liver damage from EF use; however, there have been few investigations of EF-induced cholestasis, particularly the underlying mechanisms. AIMS OF THE STUDY The purpose of this study was to evaluate the risk of EF-induced cholestasis in vivo and to explore the mechanisms of action. MATERIALS AND METHODS ICR mice were orally administered a water extract of EF (WEF) in doses of 6.5 and 19.5 g/kg/day for 14 weeks. Liver-to-body weight ratios, body weight, histopathological examination, and biochemical analyses were performed to assess WEF-induced cholestasis in the mice. Genes associated with bile acid (BA) metabolism and transport, including sodium taurocholate cotransporting polypeptide (NTCP), cytochrome P450 8B1 (CYP8B1), bile-salt export pump (BSEP), multidrug resistance P-glycoproteins 1 (MDR1), and farnesoid X receptor (FXR), were measured at the transcript and protein levels to investigate the potential mechanisms through which cholestasis is aroused by EF. RESULTS After administration of WEF for 14 weeks, mice in the high-dose WEF group showed poor health with an increased liver-to-body weight ratio as well as higher serum aminotransferase, alkaline phosphatase, direct bilirubin, and total BA levels. Compared with the control group, mRNA expression of NTCP and cholesterol 7a-hydroxylase (CYP7A1) increased, and levels of BSEP, MDR1, multidrug resistance-associated protein 2, and multidrug resistance-associated protein 3 decreased in the WEF-treated group. NTCP, BSEP, MDR1, and CYP8B1 showed similar mRNA and protein expression trends. CONCLUSION We demonstrated that the long-term oral administration of WEF causes cholestatic liver injury in mice, which is consistent with reported clinical cases. Furthermore, we found that the destruction of BA metabolism and transport is involved in WEF-induced cholestasis. The fine-scale molecular mechanisms of WEF-induced cholestasis and the active compounds of EF need further study.
Collapse
Affiliation(s)
- Jin-Quan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Cai Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Jia-Xing Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Feng-Jie Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Ting Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Yan Jiang
- Nanjing Forestry University, Nanjing, 210037, China.
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
11
|
Ye R, Yan C, Zhou H, Zhang C, Huang Y, Dong M, Zhang H, Lin J, Jiang X, Yuan S, Chen L, Jiang R, Cheng Z, Zheng K, Yu A, Zhang Q, Quan LH, Jin W. Brown adipose tissue activation by ginsenoside compound K treatment ameliorates polycystic ovary syndrome. Br J Pharmacol 2022; 179:4563-4574. [PMID: 35751868 DOI: 10.1111/bph.15909] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Polycystic ovary syndrome (PCOS) is a common metabolic and endocrine disease affecting women of reproductive age. Due to its complex etiology, there is no effective cure for PCOS currently. Brown adipose tissue (BAT) activity is significantly decreased in PCOS patients and BAT activation has beneficial effects on PCOS animal models. Here, we investigated the therapeutic effect of ginsenoside compound K (CK) on an animal model of PCOS and its mechanism of BAT activation EXPERIMENTAL APPROACH: Primary brown adipocyte, Db/Db mice and dehydroepiandrosterone (DHEA)-induced PCOS rats were used. The core body temperature, oxygen consumption, energy metabolism related gene and protein expression were assessed to identify the function of CK on energy metabolism. Estrous cycle, serum sex hormone, ovarian steroidogenic enzyme gene expression and ovarian morphology were evaluated following CK treatment. KEY RESULTS Our results indicated that CK treatment could significantly protect against body weight gain in Db/Db mice via BAT activation. Furthermore, we found that CK treatment could normalize hyperandrogenism, estrous cyclicity, normalize steroidogenic enzyme expression and decrease the number of cystic follicles in PCOS rats. Interestingly, as a potential endocrine intermediate, C-X-C motif chemokine ligand-14 protein (CXCL14) was significantly upregulated following CK administration. In addition, exogenous CXC14 supplementation was found to reverse DHEA-induced PCOS in a phenotypically similar manner to CK treatment. CONCLUSION AND IMPLICATIONS In summary, CK treatment significantly activates BAT, increases CXCL14 expression and ameliorates PCOS. These findings suggest that CK might be a potential drug candidate for PCOS treatment.
Collapse
Affiliation(s)
- Rongcai Ye
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Chunlong Yan
- College of Agriculture, Yanbian University, Yanji, China
| | - Huiqiao Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Chuanhai Zhang
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Yuanyuan Huang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Jun Lin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaoxiao Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Shouli Yuan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Li Chen
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Rui Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Ziyu Cheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Kexin Zheng
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Anni Yu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Qiaoli Zhang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lin-Hu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Tao Q, Peng D, Li P, Lai L, Li W, Du B. Genotoxicity, acute and subchronic toxicity evaluation of fermented Morinda officinalis. Food Chem Toxicol 2022; 163:113003. [PMID: 35413384 DOI: 10.1016/j.fct.2022.113003] [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: 01/04/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/15/2022]
Abstract
Morinda officinalis has diverse pharmacological effects and has the potential to be used as functional food and medicine. Fermentation is traditionally used to process Morinda officinalis. However, the toxicological profile of fermented Morinda officinalis (FMO) is not reported. In the present study, the toxicological characteristics of FMO were assessed for the first time. FMO did not show any genotoxicity based on the Ames test, mammalian erythrocyte micronucleus test, and mouse primary spermatocyte chromosome aberration test. FMO administered by gavage in mice and rats at a dose of 20 g/kg BW did not induce death or toxicity based on acute study, indicating that FMO could be regarded as non-toxic at the tested dose. In the 90-day subchronic toxicity study, rats fed with FMO at the maximum dose of 8 g/kg BW did not affect mortalities, BW, food consumption, organ weights, hematology, serum biochemistry, or urinalysis. The no observed adverse effect level of FMO in both sexes was not less than 8 g/kg BW/day based on subchronic toxicity. The obtained results support the safe use of FMO as functional food and medicine.
Collapse
Affiliation(s)
- Qian Tao
- Infinitus (China) Co., Ltd., Guangzhou, 510623, China
| | - Dong Peng
- South China Agricultural University, College of Food Science, Guangzhou, 510642, China
| | - Pan Li
- South China Agricultural University, College of Food Science, Guangzhou, 510642, China
| | - Lanyu Lai
- South China Agricultural University, College of Food Science, Guangzhou, 510642, China
| | - Wenzhi Li
- Infinitus (China) Co., Ltd., Guangzhou, 510623, China.
| | - Bing Du
- South China Agricultural University, College of Food Science, Guangzhou, 510642, China.
| |
Collapse
|
13
|
Jia H, Zeng X, Cai R, Wang Z, Yuan Y, Yue T. Fabrication of Epsilon-Polylysine-Based Magnetic Nanoflowers with Effective Antibacterial Activity against Alicyclobacillus acidoterrestris. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:857-868. [PMID: 35040323 DOI: 10.1021/acs.jafc.1c06885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The risk of fruit juice contamination caused by microorganisms, especially Alicyclobacillus acidoterrestris, has been reported worldwide. To develop cost-effective control methods, in this work, flower-like magnetic molybdenum disulfide (Fe3O4@MoS2) nanoparticles (NPs) were fabricated by a facile two-step hydrothermal method. After further modifying polyacrylic acid (PAA) on the surface of the NPs, epsilon-polylysine (EPL) was immobilized via N-(3-dimethylaminopropyl)-N-carbodiimide hydrochloride/N-hydroxysuccinimide coupling reaction to obtain the Fe3O4@MoS2@PAA-EPL nanocomposites. Antibacterial results exhibited that the synthesized nanocomposites showed effective antibacterial activity against A. acidoterrestris with a minimum inhibitory concentration of 0.31 mg mL-1. Investigation on the antibacterial mechanism revealed that the presence of nanocomposites caused damage and disruption of the bacterial membrane through dent formation, resulting in the leakage of intracellular protein. Moreover, the activity of dehydrogenase enzymes was inhibited with the treatment of Fe3O4@MoS2@PAA-EPL, causing the reduction of metabolic activity and adenosine triphosphate levels in bacteria. Simultaneously, the presence of nanocomposites improved intracellular reactive oxygen species levels, and this disrupted the antioxidant defense system and caused oxidative damage to bacteria. Furthermore, Fe3O4@MoS2@PAA-EPL nanocomposites were confirmed to possess satisfactory biocompatibility by performing in vitro cytotoxicity and in vivo acute toxicity experiments. The aim of this research was to develop a new pathway for the inhibition of A. acidoterrestris in the juice industry.
Collapse
Affiliation(s)
- Hang Jia
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Xuejun Zeng
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Rui Cai
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Zhouli Wang
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Yahong Yuan
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Tianli Yue
- College of Food Science and Engineering, Northwest A & F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-Products (Yangling), Ministry of Agriculture, Yangling 712100, China
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
| |
Collapse
|
14
|
Liu J, Wang Y, Yu Z, Lv G, Huang X, Lin H, Ma C, Lin Z, Qu P. Functional Mechanism of Ginsenoside Compound K on Tumor Growth and Metastasis. Integr Cancer Ther 2022; 21:15347354221101203. [PMID: 35615883 PMCID: PMC9152193 DOI: 10.1177/15347354221101203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ginsenosides, as the most important constituents of ginseng, have been extensively investigated in cancer chemoprevention and therapeutics. Among the ginsenosides, Compound K (CK), a rare protopanaxadiol type of ginsenoside, has been most broadly used for cancer treatment due to its high anticancer bioactivity. However, the functional mechanism of CK in cancer is not well known. This review describes the structure, transformation and pharmacological activity of CK and discusses the functional mechanisms of CK and its metabolites, which regulate signaling pathways related to tumor growth and metastasis. CK inhibits tumor growth by inducing tumor apoptosis and tumor cell differentiation, regulates the tumor microenvironment by suppressing tumor angiogenesis-related proteins, and downregulates the roles of immunosuppressive cells, such as myeloid-derived suppressor cells (MDSCs). There is currently much research on the potential development of CK as a new strategy when administered alone or in combination with other compounds.
Collapse
Affiliation(s)
- Jinlong Liu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuchen Wang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhun Yu
- Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guangfu Lv
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaowei Huang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - He Lin
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Chao Ma
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhe Lin
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Peng Qu
- National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
15
|
Sng KS, Li G, Zhou LY, Song YJ, Chen XQ, Wang YJ, Yao M, Cui XJ. Ginseng extract and ginsenosides improve neurological function and promote antioxidant effects in rats with spinal cord injury: A meta-analysis and systematic review. J Ginseng Res 2022; 46:11-22. [PMID: 35058723 PMCID: PMC8753526 DOI: 10.1016/j.jgr.2021.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/26/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is defined as damage to the spinal cord that temporarily or permanently changes its function. There is no definite treatment established for neurological complete injury patients. This study investigated the effect of ginseng extract and ginsenosides on neurological recovery and antioxidant efficacies in rat models following SCI and explore the appropriate dosage. Searches were done on PubMed, Embase, and Chinese databases, and animal studies matches the inclusion criteria were selected. Pair-wise meta-analysis and subgroup analysis were performed. Ten studies were included, and the overall methodological qualities were low quality. The result showed ginseng extract and ginsenosides significantly improve neurological function, through the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale (pooled MD = 4.40; 95% CI = 3.92 to 4.88; p < 0.00001), significantly decrease malondialdehyde (MDA) (n = 290; pooled MD = −2.19; 95% CI = −3.16 to −1.22; p < 0.0001) and increase superoxide dismutase (SOD) levels (n = 290; pooled MD = 2.14; 95% CI = 1.45 to 2.83; p < 0.00001). Both low (<25 mg/kg) and high dosage (≥25 mg/kg) showed significant improvement in the motor function recovery in SCI rats. Collectively, this review suggests ginseng extract and ginsenosides has a protective effect on SCI, with good safety and a clear mechanism of action and may be suitable for future clinical trials and applications.
Collapse
|
16
|
Xu C, Li L, Wang C, Jiang J, Li L, Zhu L, Jin S, Jin Z, Lee JJ, Li G, Yan G. Effects of G-Rh2 on mast cell-mediated anaphylaxis via AKT-Nrf2/NF-κB and MAPK-Nrf2/NF-κB pathways. J Ginseng Res 2021; 46:550-560. [PMID: 35818417 PMCID: PMC9270651 DOI: 10.1016/j.jgr.2021.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/04/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022] Open
Abstract
Background The effect of ginsenoside Rh2 (G-Rh2) on mast cell-mediated anaphylaxis remains unclear. Herein, we investigated the effects of G-Rh2 on OVA-induced asthmatic mice and on mast cell-mediated anaphylaxis. Methods Asthma model was established for evaluating airway changes and ear allergy. RPMCs and RBL-2H3 were used for in vitro experiments. Calcium uptake, histamine release and degranulation were detected. ELISA and Western blot measured cytokine and protein levels, respectively. Results G-Rh2 inhibited OVA-induced airway remodeling, the production of TNF-α, IL-4, IL-8, IL-1β and the degranulation of mast cells of asthmatic mice. G-Rh2 inhibited the activation of Syk and Lyn in lung tissue of OVA-induced asthmatic mice. G-Rh2 inhibited serum IgE production in OVA induced asthmatic mice. Furthermore, G-Rh2 reduced the ear allergy in IgE-sensitized mice. G-Rh2 decreased the ear thickness. In vitro experiments G-Rh2 significantly reduced calcium uptake and inhibited histamine release and degranulation in RPMCs. In addition, G-Rh2 reduced the production of IL-1β, TNF-α, IL-8, and IL-4 in IgE-sensitized RBL-2H3 cells. Interestingly, G-Rh2 was involved in the FcεRI pathway activation of mast cells and the transduction of the Lyn/Syk signaling pathway. G-Rh2 inhibited PI3K activity in a dose-dependent manner. By blocking the antigen-induced phosphorylation of Lyn, Syk, LAT, PLCγ2, PI3K ERK1/2 and Raf-1 expression, G-Rh2 inhibited the NF-κB, AKT-Nrf2, and p38MAPK-Nrf2 pathways. However, G-Rh2 up-regulated Keap-1 expression. Meanwhile, G-Rh2 reduced the levels of p-AKT, p38MAPK and Nrf2 in RBL-2H3 sensitized IgE cells and inhibited NF-κB signaling pathway activation by activating the AKT-Nrf2 and p38MAPK-Nrf2 pathways. Conclusion G-Rh2 inhibits mast cell-induced allergic inflammation, which might be mediated by the AKT-Nrf2/NF-κB and p38MAPK-Nrf2/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Lianhua Zhu
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Shan Jin
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Zhehu Jin
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Jung Joon Lee
- College of Pharmacy, Yanbian University, Yanji, China
| | - Guanhao Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Food Research Center of Yanbian University, Yanji, China
- Corresponding author. Food Research Center of Yanbian University, No. 977 Gongyuan Road, Yanji, 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Corresponding author. Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, No. 977 Gongyuan Road, Yanji, 133002, PR China.
| |
Collapse
|
17
|
Xu J, Pan Y, Liu Y, Na S, Zhou H, Li L, Chen F, Song H. A review of anti-tumour effects of ginsenoside in gastrointestinal cancer. J Pharm Pharmacol 2021; 73:1292-1301. [PMID: 33836068 DOI: 10.1093/jpp/rgab048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/23/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Gastrointestinal cancer, one of the major causes of cancer-related deaths in the world, refers to malignant conditions of the gastrointestinal (GI) tract and other organs. Although conventional therapy has been successful to some extent in cancer treatment, drug resistance and cancer recurrence still limit the therapeutic efficacy. There is increasing evidence indicating that ginsenoside, as a kind of high nutritional value and widely used traditional Chinese medicine, could contribute to the promotion of treatment in GI cancer, which deserves further investigation. KEY FINDINGS Based on previous studies, the possible mechanisms mainly include regulation of autophagy, apoptosis, proliferation, migration and angiogenesis. However, no studies recently have conducted a more in-depth review of the anti-cancer effects of ginsenoside in GI cancer. SUMMARY Therefore, this review will summarise and analyse the latest developments in the anti-tumour effects of ginsenosides in GI cancer, thus may promote further research of the anti-tumour efficacy of ginsenoside.
Collapse
Affiliation(s)
- Jing Xu
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yunxia Pan
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yanyan Liu
- Department of Biochemistry and Molecular Biology, School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| | - Sha Na
- Department of Biochemistry and Molecular Biology, School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| | - Hui Zhou
- Department of Biochemistry and Molecular Biology, School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| | - Lu Li
- Department of Biochemistry and Molecular Biology, School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| | - Fengyuan Chen
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| | - Hang Song
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Department of Biochemistry and Molecular Biology, School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrative Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Medicinal Formula of Anhui Province, Hefei, China
| |
Collapse
|
18
|
Han L, Chen K, Liu P, Yang L, Kang Y, Gao Y, Li C, Sun C, Li Y, Fan W, Hou H. Toxicological evaluation of S. involucrata culture: Acute, 90-day subchronic and genotoxicity studies. Regul Toxicol Pharmacol 2021; 124:104980. [PMID: 34186141 DOI: 10.1016/j.yrtph.2021.104980] [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: 03/27/2021] [Revised: 06/04/2021] [Accepted: 06/24/2021] [Indexed: 12/01/2022]
Abstract
Saussurea involucrata is an endangered plant that is used in traditional Chinese medicine. Through the use of plant cell culture techniques, preparations of Saussurea involucrata (S. involucrata) cell cultures have been developed and used to generate medicinal preparations. There have been few evidence-based analyses of the toxicological effects of S. involucrata culture conducted to date. Here, we conducted the experiments designed to assess the acute, subchronic, and genotoxic toxicological effects of S. involucrata culture. The genotoxic study was assessed through Ames, marrow micronucleus, and sperm malformation assays. The acute toxicity was assessed by orally administering in rats and mice at dose of 7500 mg/kg. Subchronic toxicity studies were then conducted by administering rats at doses of 500, 1000, or 1500 mg/kg for 90 days. No genotoxicity was observed at any tested dose levels, nor was any evidence of acute toxicity detected in treated mice or rats. Similarly, subchronic study of S. involucrata culture administration was not associated with any changes in rat food intake, weight, hematological parameters, organ weight, or organ histology. Then, we determined that the no observed adverse effect level of S. involucrata culture was greater than 1500 mg/kg in our 90-day toxicity study.
Collapse
Affiliation(s)
- Lei Han
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Kaili Chen
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Ping Liu
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Lintong Yang
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Yating Kang
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Yonglin Gao
- School of Life Science,Yantai University, Yantai, 264005, PR China.
| | - Chunmei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Chengfeng Sun
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Yanshen Li
- School of Life Science,Yantai University, Yantai, 264005, PR China
| | - Wenxia Fan
- Dalian Practical Biotechnology Co., Ltd., Dalian, 116001, China
| | - Hu Hou
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
19
|
Ginsenoside CK Inhibits Hypoxia-Induced Epithelial-Mesenchymal Transformation through the HIF-1α/NF-κB Feedback Pathway in Hepatocellular Carcinoma. Foods 2021; 10:foods10061195. [PMID: 34073155 PMCID: PMC8227303 DOI: 10.3390/foods10061195] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a kind of malignant tumor with high morbidity and mortality rates worldwide. Epithelial-mesenchymal transformation (EMT) is crucial for HCC progression and prognosis. Characteristics of the tumor microenvironment, such as hypoxia, and excessive activation of the NF-κB signaling pathway have been identified as the key inducers of EMT in HCC. In our study, we verified the crosstalk between HIF-1α signaling and NF-κB pathway and their effects on EMT in HCC cells. The results show that CoCl2-induced hypoxia could promote IκB phosphorylation to activate NF-κB signaling and vice versa. Moreover, we found that ginsenoside CK, a metabolite of protopanaxadiol saponins, could inhibit the proliferation and colony formation of different HCC cell lines. Furthermore, ginsenoside CK could impair the metastatic potential of HCC cell lines under hypoxic conditions. Mechanistically, ginsenoside CK suppressed HIF-1α/NF-κB signaling and expression level of EMT-related proteins and cytokines in hypoxia-induced or TNFα-stimulated HCC cell lines. An in vivo study revealed that the oral delivery of ginsenoside CK could inhibit the growth of xenograft tumors and block HIF-1α and NF-κB signaling as well as EMT marker expression. Our study suggests that ginsenoside CK is a potential therapy for HCC patients that functions by targeting the HIF-1α/NF-κB crosstalk.
Collapse
|
20
|
Tang M, Xie X, Yang Y, Li F. Ginsenoside compound K- a potential drug for rheumatoid arthritis. Pharmacol Res 2021; 166:105498. [PMID: 33609698 DOI: 10.1016/j.phrs.2021.105498] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/28/2021] [Accepted: 02/14/2021] [Indexed: 12/26/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory and autoimmune disease, if prescription of effective delayed, the articular disturbances may lead to disability. Ginsenoside compound K (GCK) is the main degradation product of oral ginsenosides in the human intestine. Numerous researches in vitro and in vivo have recorded the anti-arthritic effect of GCK, we discuss the mechanisms from the following three aspects, including anti-inflammatory, immune-regulatory, and bone-protective, respectively, in this review, and the anti-arthritic mechanism of GCK may be related to the effect on TNF-α-TNFR2, glucocorticoid receptor (GR) and β-arrestin1/2. We also describe the anti-anemia effect of GCK to open the possibility that GCK can be used as an effective disease-modifying anti-rheumatic drug (DMARD).
Collapse
Affiliation(s)
- Mengshi Tang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, No.139, Renmin Middle Road, Changsha, Hunan 410011, China.
| | - Xi Xie
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, No.139, Renmin Middle Road, Changsha, Hunan 410011, China.
| | - Yongyu Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Fen Li
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, No.139, Renmin Middle Road, Changsha, Hunan 410011, China.
| |
Collapse
|
21
|
Assessment of the toxicity and toxicokinetics of the novel potent tropomyosin receptor kinase (Trk) inhibitor LPM4870108 in rhesus monkeys. Regul Toxicol Pharmacol 2021; 122:104886. [PMID: 33556418 DOI: 10.1016/j.yrtph.2021.104886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 01/28/2023]
Abstract
LPM4870108 is a tropomyosin receptor kinase (Trk) inhibitor that is currently under consideration for human clinical trials. We characterized the toxicity and toxicokinetic properties of LPM4870108 following its oral administration to rhesus monkeys (5, 10, or 20 mg/kg/day for 4 weeks with a 4-week recovery period). No evidence of LPM4870108 toxicity was observed over this study as reflected by an absence of difference in body weight, ophthalmoscopy, urinalysis, gross, or histopathology findings. No significant differences in toxicity-related outcomes were detected when comparing LPM4870108 and control groups, and no significant treatment-related changes in food consumption, electrocardiogram results, blood pressure, hematological parameters, biochemical values, organ weight, or bone marrow parameters were observed. Treatment caused dose-dependent effects of gait disturbance, impaired balance, poor coordination, and decreased grip strength in all LPM4870108-treated animals, with these effects being attributable to excessive on-target Trk receptor inhibition. After the 4-week recovery period, all these abnormal treatment-related findings had fully or partially resolved. The toxicokinetic study of monkeys revealed that the LPM4870108 exposure increased with dose. Overall, LPM4870108 exhibited a safety profile in treated monkeys, indicating that the Highest Non-Severely Toxic Dose (HNSTD) for LPM4870108 in monkeys was 20 mg/kg/day.
Collapse
|
22
|
Lee YC, Wong WT, Li LH, Chu LJ, Menon MP, Ho CL, Chernikov OV, Lee SL, Hua KF. Ginsenoside M1 Induces Apoptosis and Inhibits the Migration of Human Oral Cancer Cells. Int J Mol Sci 2020; 21:ijms21249704. [PMID: 33352689 PMCID: PMC7766606 DOI: 10.3390/ijms21249704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) accounts for 5.8% of all malignancies in Taiwan, and the incidence of OSCC is on the rise. OSCC is also a common malignancy worldwide, and the five-year survival rate remains poor. Therefore, new and effective treatments are needed to control OSCC. In the present study, we prepared ginsenoside M1 (20-O-beta-d-glucopyranosyl-20(S)-protopanaxadiol), a major deglycosylated metabolite of ginsenoside, through the biotransformation of Panax notoginseng leaves by the fungus SP-LSL-002. We investigated the anti-OSCC activity and associated mechanisms of ginsenoside M1 in vitro and in vivo. We demonstrated that ginsenoside M1 dose-dependently inhibited the viability of human OSCC SAS and OEC-M1 cells. To gain further insight into the mode of action of ginsenoside M1, we demonstrated that ginsenoside M1 increased the expression levels of Bak, Bad, and p53 and induced apoptotic DNA breaks, G1 phase arrest, PI/Annexin V double-positive staining, and caspase-3/9 activation. In addition, we demonstrated that ginsenoside M1 dose-dependently inhibited the colony formation and migration ability of SAS and OEC-M1 cells and reduced the expression of metastasis-related protein vimentin. Furthermore, oral administration or subcutaneous injection of ginsenoside M1 significantly reduced tumor growth in SAS xenograft mice. These results indicate that ginsenoside M1 can be translated into a potential therapeutic against OSCC.
Collapse
Affiliation(s)
- Yu-Chieh Lee
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan; (Y.-C.L.); (W.-T.W.); (M.P.M.)
| | - Wei-Ting Wong
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan; (Y.-C.L.); (W.-T.W.); (M.P.M.)
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan;
- National Defense Medical Center, Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan;
- Liver Research Center, Chang Gung Memorial Hospital at Linkou, Gueishan, Taoyuan 33302, Taiwan
| | - Mridula P. Menon
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan; (Y.-C.L.); (W.-T.W.); (M.P.M.)
| | - Chen-Lung Ho
- Division of Wood Cellulose, Taiwan Forestry Research Institute, Taipei 100051, Taiwan;
| | - Oleg V. Chernikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022 Vladivostok, Russia;
| | - Sheau-Long Lee
- Wellhead Biological Technology Corporation, Taoyuan 325, Taiwan;
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan; (Y.-C.L.); (W.-T.W.); (M.P.M.)
- National Defense Medical Center, Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 406040, Taiwan
- Correspondence: ; Tel.: +886-3931-7626
| |
Collapse
|
23
|
Li C, Su C, Wang Y, Gao Y, Wang H, Tian J, Fu F. A 26-week toxicological study of Xuezhikang (XZK), red yeast rice extract, in Beagle dogs with a 4-week recovery period. Regul Toxicol Pharmacol 2020; 117:104781. [DOI: 10.1016/j.yrtph.2020.104781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022]
|
24
|
Kim KT, Cho DW, Yang MJ, Kim DH, Shin SH, Hwang JH, Woo DH, Shin DG, Seo JH, Yang YS, Han SC. Thirteen-week oral toxicity study of fermented ginseng, GBCK25, in Sprague-Dawley rats. Regul Toxicol Pharmacol 2020; 118:104812. [PMID: 33122046 DOI: 10.1016/j.yrtph.2020.104812] [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: 03/16/2020] [Revised: 09/12/2020] [Accepted: 10/23/2020] [Indexed: 10/23/2022]
Abstract
Ginseng (Panax ginseng) is commonly used in Asia as a medicinal herb. In particular, fermented ginseng, GBCK25, has been recently developed to increase ginsenoside absorption. It also has other beneficial biological effects such as hemodynamic and anti-inflammation functions. Here, we investigated the potential toxicity of GBCK25 in Sprague-Dawley rats following 13 weeks of GBCK25 treatment by oral gavage at doses of 250, 500, or 1000 mg/kg/day and reversible toxic effects over a 4-week recovery phase. Ten male and female rats per group were randomly allocated to the main toxicology groups and five male and female rats per group were allocated to the 0 and 1000 mg/kg/day recovery groups, respectively. There was no mortality; significant clinical toxicity or microscopic findings; and changes in body weight, food consumption, hematological parameters, serum biochemistry, or absolute and relative organ weights in any of the groups. In conclusion, there were no toxicological changes upon repeated oral gavage of GBCK25 at doses of 250, 500, or 1000 mg/kg/day in Sprague-Dawley rats over 13 weeks. The no-observed-adverse-effect level of GBCK25 was 1000 mg/kg/day in both sexes of Sprague-Dawley rat.
Collapse
Affiliation(s)
- Kyung-Tai Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Doo-Wan Cho
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Da-Hee Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Seung-Hyuk Shin
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Jeong Ho Hwang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea
| | - Dong Ho Woo
- Research Center for Convergence Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Dong Gyu Shin
- Research & Development Center of GENERAL BIO Co., Ltd, 254, Yongtusan-ro, Songdong-myeon, Namwon City, Jeollabuk-Do, 55793, Republic of Korea
| | - Jeong Hun Seo
- Research & Development Center of GENERAL BIO Co., Ltd, 254, Yongtusan-ro, Songdong-myeon, Namwon City, Jeollabuk-Do, 55793, Republic of Korea
| | - Young-Su Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea.
| | - Su-Cheol Han
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabuk-Do, 56212, Republic of Korea.
| |
Collapse
|
25
|
Fan S, Zhang Z, Su H, Xu P, Qi H, Zhao D, Li X. Panax ginseng clinical trials: Current status and future perspectives. Biomed Pharmacother 2020; 132:110832. [PMID: 33059260 DOI: 10.1016/j.biopha.2020.110832] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/31/2022] Open
Abstract
Panax Ginseng has been widely used in Asian for thousand years. In order to evaluate the efficacy and safety of ginseng, more and more ginseng clinical trials (GCTs) have been conducted recently. However, there is a lack of an extensive review summarizing the current status for the quality and quantity of ginseng clinical researches until now. Therefore, clinical trials for ginseng were retrieved from International Clinical Trials Registration Platform and collected through the system retrieval method of Preferred Reporting Items for Systematic Reviews and Meta-Analyses in PubMed, the Web of Science, the Korean Studies Information Service System, and SCOPUS database. We summarized the clinical characteristics of 152 registered ginseng clinical trials (R-GCTs) and119 published ginseng clinical trials (P-GCTs), such as source register, recruitment status, primary purpose, duration, sample size, conditions, and outcomes. Among them, ginseng has mainly been studied in clinical trials in the single-center and less than 200 subjects. In the most GCTs, healthy subjects and patients with various conditions, such as cardiovascular and metabolic diseases are administrated with ginseng, ginsenosides or the prescriptions containing ginseng for less than 3 months to investigate the protective and therapeutic functions of ginseng. 95 (79.8 %) published articles showed that ginseng has plenty of positive effects. This review could assist the basic researchers and clinical doctors to understand current status and problem of ginseng clinical research, and perhaps could benefit for the reasonable and accurate design of future clinical studies.
Collapse
Affiliation(s)
- Shasha Fan
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zepeng Zhang
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China; Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hang Su
- Practice Innovations Center, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Peng Xu
- Neurology Department, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hongyu Qi
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China.
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China.
| |
Collapse
|
26
|
Sharma A, Lee HJ. Ginsenoside Compound K: Insights into Recent Studies on Pharmacokinetics and Health-Promoting Activities. Biomolecules 2020; 10:E1028. [PMID: 32664389 PMCID: PMC7407392 DOI: 10.3390/biom10071028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Ginseng (Panax ginseng) is an herb popular for its medicinal and health properties. Compound K (CK) is a secondary ginsenoside biotransformed from major ginsenosides. Compound K is more bioavailable and soluble than its parent ginsenosides and hence of immense importance. The review summarizes health-promoting in vitro and in vivo studies of CK between 2015 and 2020, including hepatoprotective, anti-inflammatory, anti-atherosclerosis, anti-diabetic, anti-cancer, neuroprotective, anti-aging/skin protective, and others. Clinical trial data are minimal and are primarily based on CK-rich fermented ginseng. Besides, numerous preclinical and clinical studies indicating the pharmacokinetic behavior of CK, its parent compound (Rb1), and processed ginseng extracts are also summarized. With the limited evidence available from animal and clinical studies, it can be stated that CK is safe and well-tolerated. However, lower water solubility, membrane permeability, and efflux significantly diminish the efficacy of CK and restrict its clinical application. We found that the use of nanocarriers and cyclodextrin for CK delivery could overcome these limitations as well as improve the health benefits associated with them. However, these derivatives have not been clinically evaluated, thus requiring a safety assessment for human therapy application. Future studies should be aimed at investigating clinical evidence of CK.
Collapse
Affiliation(s)
- Anshul Sharma
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
| |
Collapse
|
27
|
Gao Y, Chen X, Li C, Wang H, Tian J, Fu F. Toxicological evaluation of, red rice yeast extract, Xuezhikang: Acute, 26-week chronic and genotoxicity studies. Regul Toxicol Pharmacol 2020; 114:104654. [DOI: 10.1016/j.yrtph.2020.104654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/27/2020] [Accepted: 03/31/2020] [Indexed: 10/24/2022]
|
28
|
Li CW, Deng MZ, Gao ZJ, Dang YY, Zheng GD, Yang XJ, Chao YX, Cai YF, Wu XL. Effects of compound K, a metabolite of ginsenosides, on memory and cognitive dysfunction in db/db mice involve the inhibition of ER stress and the NLRP3 inflammasome pathway. Food Funct 2020; 11:4416-4427. [PMID: 32374299 DOI: 10.1039/c9fo02602a] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Accumulating clinical and epidemiological evidence indicates a close relationship between diabetes mellitus and dementia. The ginsenoside compound K (CK) has been reported to ameliorate diabetes mellitus and confer protection to the central nervous system. In this study, we investigated whether CK could improve memory impairment and cognitive dysfunction in diabetic db/db mice. Firstly, we found that CK treatments significantly improved behavioral impairment and cognitive dysfunction based on Morris water maze, Y-maze, and fear conditioning tests. Besides, CK decreased the fasting glucose level, increased lipid metabolism, and ameliorated glucose tolerance, insulin sensitivity, and dyslipidemia in diabetic db/db mice. In addition, CK treatments alleviated oxidative stress and inhibited the inflammatory response in hippocampal tissue. Further investigations showed that CK treatments inhibited the NLRP3 inflammasome pathway, as evidenced by the declined expression of TXNIP, NLRP3 inflammasomes, ASC, cleaved caspase-1, and mature IL-1β in hippocampal tissues. Moreover, CK treatments alleviated ER stress via down-regulating the level of BiP, CHOP, p-PERK, p-IRE1α and ATF6 in the hippocampus of db/db mice. These results suggest that CK improves memory and cognitive dysfunction, possibly by ameliorating glucose tolerance, insulin sensitivity, and dyslipidemia, suppressing oxidative stress and inflammatory response and modulating the NLRP3 inflammasome pathway and ER stress.
Collapse
Affiliation(s)
- Chu-Wen Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Li C, Wang Z, Wang T, Wang G, Li G, Sun C, Lin J, Sun L, Sun X, Cho S, Wang H, Gao Y, Tian J. Repeated-dose 26-week oral toxicity study of ginsenoside compound K in Beagle dogs. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112323. [PMID: 31639487 DOI: 10.1016/j.jep.2019.112323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/06/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginsenoside compound K (CK), a product produced by the intestinal bacteria-mediated breakdown of ginsenoside, exhibits a wide array of pharmacological activities against diverse targets. However, few of preclinical safety evaluation of CK is reported. AIMS OF THE STUDY The present study therefore sought to assess the toxicity of oral CK in Beagle dogs over a 26-week period. MATERIAL AND METHODS All dogs received 4, 12, or 36 mg/kg oral CK doses for 26 weeks with regular monitoring, followed by a 4-week recovery period. Animals were monitored through measurements of temperature, weight, food intake, blood chemistry and hematological findings, electrocardiogram (ECG) measurements, urinalysis, gross necropsy and organ weight and tissue histopathology. RESULTS Animals in the 36 mg/kg group exhibited an apparent reduction in body weight over the study period, in addition to the presence of focal liver necrosis and increased plasma enzyme levels (alanine aminotransferase, ALT; alkaline phosphatase, ALP) consistent with hepatotoxicity, although there was some evidence suggesting this toxicity was reversible. Animals in the 4 and 12 mg/kg groups did not exhibit any apparent toxicity for any measured parameters. CONCLUSION These results thus indicate that the no observed adverse effect level (NOAEL) in dogs is 12 mg/kg.
Collapse
Affiliation(s)
- Chunmei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Zhezhe Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Tong Wang
- School of Life Science, Yantai University, Yantai, 264005, PR China
| | - Guangfei Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Guisheng Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Chengfeng Sun
- School of Life Science, Yantai University, Yantai, 264005, PR China
| | - Jian Lin
- School of Life Science, Yantai University, Yantai, 264005, PR China
| | - Liqin Sun
- School of Life Science, Yantai University, Yantai, 264005, PR China
| | - Xilin Sun
- Yantai Laishan Changen Hospital, Yantai, 264005, PR China
| | - Susan Cho
- NutraSource, Inc., Clarksville, MD, 21029, USA
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Yonglin Gao
- School of Life Science, Yantai University, Yantai, 264005, PR China.
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| |
Collapse
|