1
|
Li P, Zhang M, Chen M, Liu G, Meng L, Zhang D. Systematic studies on the kinetic process of 20(S)-protopanaxadiol in rats and dogs: absorption, distribution, metabolism and excretion. Front Pharmacol 2024; 15:1430780. [PMID: 38966555 PMCID: PMC11222998 DOI: 10.3389/fphar.2024.1430780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Background and Objective Ginseng has been regarded as a precious medicinal herb with miraculous effects in Eastern culture. The primary chemical constituents of ginseng are saponins, and the physiological activities of ginsenosides determine their edible and medicinal value. The aim of this study is to comprehensively and systematically investigate the kinetic processes of 20(S)-protopanaxadiol (PPD) in rats and dogs, in order to promote the rational combination of ginseng as a drug and dietary ingredient. Methods PPD was administered, and drug concentration in different biological samples were detected by liquid chromatography tandem mass spectrometry (LC/MS/MS) and radioactive tracer methods. Pharmacokinetic parameters such as absorption, bioavailability, tissue distribution, plasma protein binding rate, excretion rate, and cumulative excretion were calculated, along with inference of major metabolites. Results This study systematically investigated the absorption, distribution, metabolism, excretion (ADME) of PPD in rats and dogs for the first time. The bioavailabilities of PPD were relatively low, with oral absorption nearly complete, and the majority underwent first-pass metabolism. PPD had a high plasma protein binding rate and was relatively evenly distributed in the body. Following oral administration, PPD underwent extensive metabolism, potentially involving one structural transformation and three hydroxylation reactions. The metabolites were primarily excreted through feces and urine, indicating the presence of enterohepatic circulation. The pharmacokinetic processes of PPD following intravenous administration aligned well with a three-compartment model. In contrast, after gastric administration, it fitted better with a two-compartment model, conforming to linear pharmacokinetics and proportional elimination. There were evident interspecies differences between rats and dogs regarding PPD, but individual variations of this drug were minimal within the same species. Conclusion This study systematically studied the kinetic process of PPD in rats and also investigated the kinetic characteristics of PPD in dogs for the first time. These findings lay the foundation for further research on the dietary nutrition and pharmacological effects of PPD.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Pharmacy, Beijing Anding Hospital, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Min Zhang
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Meng Chen
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangxu Liu
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Linghui Meng
- Department of Pharmacy, Beijing Anding Hospital, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Dan Zhang
- Department of Clinical Pharmacology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
2
|
Nam OH, Kim JH, Kang SW, Chae YK, Jih MK, You HH, Koh JT, Kim Y. Ginsenoside Rb1 alleviates lipopolysaccharide-induced inflammation in human dental pulp cells via the PI3K/Akt, NF-κB, and MAPK signalling pathways. Int Endod J 2024; 57:759-768. [PMID: 38436525 DOI: 10.1111/iej.14058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
AIM Among numerous constituents of Panax ginseng, a constituent named Ginsenoside Rb1 (G-Rb1) has been studied to diminish inflammation associated with diseases. This study investigated the anti-inflammatory properties of G-Rb1 on human dental pulp cells (hDPCs) exposed to lipopolysaccharide (LPS) and aimed to determine the underlying molecular mechanisms. METHODOLOGY The KEGG pathway analysis was performed after RNA sequencing in G-Rb1- and LPS-treated hDPCs. Reverse-transcription polymerase chain reaction (RT-PCR) and western blot analysis were used for the assessment of cell adhesion molecules and inflammatory cytokines. Statistical analysis was performed with one-way ANOVA and the Student-Newman-Keuls test. RESULTS G-Rb1 did not exhibit any cytotoxicity within the range of concentrations tested. However, it affected the levels of TNF-α, IL-6 and IL-8, as these showed reduced levels with exposure to LPS. Additionally, less mRNA and protein expressions of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) were shown. With the presence of G-Rb1, decreased levels of PI3K/Akt, phosphorylated IκBα and p65 were also observed. Furthermore, phosphorylated ERK and JNK by LPS were diminished within 15, 30 and 60 min of G-Rb1 exposure; however, the expression of non-phosphorylated ERK and JNK remained unchanged. CONCLUSIONS G-Rb1 suppressed the LPS-induced increase of cell adhesion molecules and inflammatory cytokines, while also inhibiting PI3K/Akt, phosphorylation of NF-κB transcription factors, ERK and JNK of MAPK signalling in hDPCs.
Collapse
Affiliation(s)
- Ok Hyung Nam
- Department of Pediatric Dentistry, School of Dentistry, Kyung Hee University, Seoul, Korea
- Department of Pediatric Dentistry, Kyung Hee University College of Dentistry, Kyung Hee Universtiy Medical Center, Seoul, Korea
| | - Jae-Hwan Kim
- Department of Pediatric Dentistry, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Si Won Kang
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Yong Kwon Chae
- Department of Pediatric Dentistry, Kyung Hee University College of Dentistry, Kyung Hee Universtiy Medical Center, Seoul, Korea
| | - Myeong-Kwan Jih
- Department of Pediatric Dentistry, School of Dentistry, Chosun University, Gwangju, Korea
| | - Hyekyoung Hannah You
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Jeong-Tae Koh
- Department of Pharmacology and Dental Therapeutics, Hard-tissue Biointerface Research Center, School of Dentistry, Dental Science Research Institute, Chonnam National University, Gwangju, Korea
| | - Young Kim
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, Korea
| |
Collapse
|
3
|
Dong WR, Gao X, Li CX, Song Y, Chai JH, Liang J. Detection and Characterization of the Metabolites of Ciwujianoside B in Rats Based on UPLC-Fusion Lumos Orbitrap Mass Spectrometry. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2024; 2024:3187511. [PMID: 38813478 PMCID: PMC11136543 DOI: 10.1155/2024/3187511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/21/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
We previously conducted a systematic study on the metabolic process and products of hederasaponin B in rats. We hypothesized that the sugar chain structures play a key role in the metabolism of triterpenoid saponins. To verify this hypothesis, we conducted metabolic research on ciwujianoside B ascribed to the same sugar chains and a distinct aglycone and compared it with hederasaponin B. Specifically, we collected feces, urine, and plasma of rats after gavage with ciwujianoside B and identified 42 metabolites by UPLC-Fusion Lumos Orbitrap mass spectrometry. Finally, ciwujianoside B metabolism and hederasaponin B metabolism were compared, reaching the following conclusions: (i) more than 40 metabolites were identified in both, with the majority of metabolites identified in feces; (ii) the corresponding metabolic pathways in vivo were basically similar, including deglycosylation, acetylation, hydroxylation, glucuronidation, oxidation, and glycosylation; and (iii) deglycosylation was considered the main metabolic reaction, and its metabolites accounted for approximately 50% of all metabolites. Overall, this study provides a foundation for further research on the metabolism of triterpenoid saponins.
Collapse
Affiliation(s)
- Wan-Ru Dong
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Xue Gao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Chen-Xue Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Yan Song
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Jun-Hong Chai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| | - Jun Liang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Harbin 150040, China
| |
Collapse
|
4
|
Ma Y, Zhao Y, Luo M, Jiang Q, Liu S, Jia Q, Bai Z, Wu F, Xie J. Advancements and challenges in pharmacokinetic and pharmacodynamic research on the traditional Chinese medicine saponins: a comprehensive review. Front Pharmacol 2024; 15:1393409. [PMID: 38774213 PMCID: PMC11106373 DOI: 10.3389/fphar.2024.1393409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
Recent research on traditional Chinese medicine (TCM) saponin pharmacokinetics has revealed transformative breakthroughs and challenges. The multicomponent nature of TCM makes it difficult to select representative indicators for pharmacokinetic studies. The clinical application of saponins is limited by their low bioavailability and short half-life, resulting in fluctuating plasma concentrations. Future directions should focus on novel saponin compounds utilizing colon-specific delivery and osmotic pump systems to enhance oral bioavailability. Optimizing drug combinations, such as ginsenosides with aspirin, shows therapeutic potential. Rigorous clinical validation is essential for practical applications. This review emphasizes a transformative era in saponin research, highlighting the need for clinical validation. TCM saponin pharmacokinetics, guided by traditional principles, are in development, utilizing multidisciplinary approaches for a comprehensive understanding. This research provides a theoretical basis for new clinical drugs and supports rational clinical medication.
Collapse
Affiliation(s)
- Yuhan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yongxia Zhao
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mingxia Luo
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qin Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Sha Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qi Jia
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Zhixun Bai
- Organ Transplant Center, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Faming Wu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jian Xie
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
Park JD. Metabolism and drug interactions of Korean ginseng based on the pharmacokinetic properties of ginsenosides: Current status and future perspectives. J Ginseng Res 2024; 48:253-265. [PMID: 38707645 PMCID: PMC11068998 DOI: 10.1016/j.jgr.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 05/07/2024] Open
Abstract
Orally administered ginsenosides, the major active components of ginseng, have been shown to be biotransformed into a number of metabolites by gastric juice, digestive and bacterial enzymes in the gastrointestinal tract and also in the liver. Attention is brought to pharmacokinetic studies of ginseng that need further clarification to better understand the safety and possible active mechanism for clinical application. Experimental results demonstrated that ginsenoside metabolites play an important role in the pharmacokinetic properties such as drug metabolizing enzymes and drug transporters, thereby can be applied as a metabolic modulator. Very few are known on the possibility of the consistency of detected ginsenosides with real active metabolites if taken the recommended dose of ginseng, but they have been found to act on the pharmacokinetic key factors in any clinical trial, affecting oral bioavailability. Since ginseng is increasingly being taken in a manner more often associated with prescription medicines, ginseng and drug interactions have been also reviewed. Considering the extensive oral administration of ginseng, the aim of this review is to provide a comprehensive overview and perspectives of recent studies on the pharmacokinetic properties of ginsenosides such as deglycosylation, absorption, metabolizing enzymes and transporters, together with ginsenoside and drug interactions.
Collapse
Affiliation(s)
- Jong Dae Park
- R&D Center, REBIO Co., Ltd., Seoul, Republic of Korea
| |
Collapse
|
6
|
Wang Y, Li C, Chen J, Cui X, Wang B, Wang Y, Wang D, Liu J, Li J. Pyxinol Fatty Acid Ester Derivatives J16 against AKI by Selectively Promoting M1 Transition to M2c Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7074-7088. [PMID: 38525502 DOI: 10.1021/acs.jafc.3c06979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Acute kidney injury (AKI) is a common, multicause clinical condition that, if ignored, often progresses to chronic kidney disease (CKD) and end-stage kidney disease, with a mortality rate of 40-50%. However, there is a lack of universal treatment for AKI. Inflammation is the basic pathological change of early kidney injury, and inflammation can exacerbate AKI. Macrophages are the primary immune cells involved in the inflammatory microenvironment of kidney disease. Therefore, regulating the function of macrophages is a crucial breakthrough for the AKI intervention. Our team chemically modified pyxinol, an ocotillol-type ginsenoside, to prepare PJ16 with higher solubility and bioavailability. In vitro, using a model of macrophages stimulated by LPS, it was found that PJ16 could regulate macrophage function, including inhibiting the secretion of inflammatory factors, promoting phagocytosis, inhibiting M1 macrophages, and promoting M1 transition to the M2c macrophage. Further investigation revealed that PJ16 may shield renal tubular epithelial cells (HK-2) damaged by LPS in vitro. Based on this, PJ16 was validated in the animal model of unilateral ureteral obstruction, which showed that it improves renal function and inhibits renal tissue fibrosis by decreasing inflammatory responses, reducing macrophage inflammatory infiltration, and preferentially upregulating M2c macrophages. In conclusion, our study is the first to show that PJ16 resists AKI and fibrosis by mechanistically regulating macrophage function by modulating the phenotypic transition from M1 to M2 macrophages, mainly M2c macrophages.
Collapse
Affiliation(s)
- Yaru Wang
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Changcheng Li
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Jingyi Chen
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Xiaoli Cui
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Binghuan Wang
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Yuezeng Wang
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Dayu Wang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Jinping Liu
- Research Center of Natural Drug, School of Pharmaceutical Sciences of Jilin University, Changchun, Jilin 130012, China
| | - Jing Li
- Department of Pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
7
|
Wang Y, Adekolurejo OO, Wang B, McDermott K, Do T, Marshall LJ, Boesch C. Bioavailability and excretion profile of betacyanins - Variability and correlations between different excretion routes. Food Chem 2024; 437:137663. [PMID: 37879158 DOI: 10.1016/j.foodchem.2023.137663] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/02/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023]
Abstract
The present study addresses the knowledge gap in betalain bioavailability, transformation and excretion. Analysis of renal and fecal excretion profiles in humans after consumption of beetroot revealed very low bioavailability (renal recovery of 0.13 %) and fast elimination of pigments (renal elimination rate constant of 0.16 h-1), while the majority of betalains underwent severe depletion during GI transit, evidenced by decarboxylation, deglucosidation and dehydrogenation. Betacyanin metabolite levels in human urine were positively associated with those in stools (p < 0.05), indicating significant impact of pigment metabolism in the gut on their bioavailability. In addition, the current study revealed large inter-individual and compositional variabilities of pigment after colonic fermentation compared with systemic metabolism, likely attributed to the increasing complexity of intestinal environment with diverse gut microbiota. To conclude, intestinal uptake and systemic metabolism of betacyanins are intimately associated with their intestinal biotransformation, with gut microbiota serving as a crucial factor.
Collapse
Affiliation(s)
- Yunqing Wang
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, LS2 9JT, United Kingdom
| | - Opeyemi O Adekolurejo
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, LS2 9JT, United Kingdom; School of Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT, United Kingdom
| | - Binying Wang
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, LS2 9JT, United Kingdom
| | - Katie McDermott
- School of Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT, United Kingdom
| | - Thuy Do
- School of Dentistry, Faculty of Medicine and Health, University of Leeds, LS2 9LU, United Kingdom
| | - Lisa J Marshall
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, LS2 9JT, United Kingdom
| | - Christine Boesch
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
8
|
Chu J, Zhao L, Xu X, Li Y, Wu B, Qin S, He B. Evolving the 3-O/6-O regiospecificity of a microbial glycosyltransferase for efficient production of ginsenoside Rh1 and unnatural ginsenoside. Int J Biol Macromol 2024; 261:129678. [PMID: 38280704 DOI: 10.1016/j.ijbiomac.2024.129678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
Glycosyltransferase is a popular and promising enzyme to produce high-value-added natural products. Rare ginsenoside Rh1 and unnatural ginsenoside 3β-O-Glc-PPT are promising candidates for drugs. Herein, the microbial glycosyltransferase UGTBL1 was able to catalyze the 20(S)-protopanaxatriol (PPT) 3-O/6-O-glycosylation with poor 6-O-regiospecificity. A structure-guided strategy of mutations involving loop engineering, PSPG motif evolution, and access tunnel engineering was proposed to engineer the enzyme UGTBL1. The variant I62R/M320H/P321Y/N170A from protein engineering achieved a great improvement in 6-O regioselectivity which increased from 10.98 % (WT) to 96.26 % and a booming conversion of 95.57 % for ginsenoside Rh1. A single mutant M320W showed an improved 3-O regioselectivity of 84.83 % and an increased conversion of 98.13 % for the 3β-O-glc-PPT product. Molecular docking and molecular dynamics (MD) simulations were performed to elucidate the possible molecular basis of the regiospecificity and catalytic activity. The unprecedented high titer of ginsenoside Rh1 (20.48 g/L) and 3β-O-Glc-PPT (18.04 g/L) was attained with high regioselectivity and yields using fed-batch cascade reactions from UDPG recycle, which was the highest yield reported to date. This work could provide an efficient and cost-effective approach to the valuable ginsenosides.
Collapse
Affiliation(s)
- Jianlin Chu
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Lu Zhao
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Xiaoli Xu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Yuting Li
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Bin Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Song Qin
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China.
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China; College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China.
| |
Collapse
|
9
|
Xu L, Xiao S, Chai Z, Li T, Joon Lee J, Su G, Zhao Y. Study of novel ginsenoside metabolites targeting HSP70 as anti-prostate cancer drugs. Bioorg Chem 2024; 144:107131. [PMID: 38271824 DOI: 10.1016/j.bioorg.2024.107131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024]
Abstract
Ginsenoside 20 (R)-25-methoxy-dammarane-3 β, twelve β, 20 triol (AD-1) is a promising new drug for the treatment of prostate cancer, but its bioavailability is low. This study investigated the effects of the main metabolites PD and M6 of AD-1 on prostate cancer cell PC3. The in vitro experimental results showed that the IC50 values of PC3 cells treated with PD and M6 were 65.61 and 11.72, respectively. Both PD and M6 inhibited the migration of PC3 cells, and the cell cycle was blocked in the G1 phase. The apoptosis rates of cells following M6 treatment at concentrations of 7.5, 15, and 30 μM were 13.4 %, 17.5 %, and 41.4 %, respectively, which stimulated the expression of apoptosis protein and significantly increased intracellular ROS levels. In xenograft models, PD and M6 have been reported to significantly inhibit tumor growth. We used a genome-wide mRNA expression profile to study the effects of PD and M6 on gene expression in PC3 cancer cells. PD and M6 induced downregulation of HSP70 subtypes HSPA1A and HSPA1B. RT-PCR confirmed that the significant down-regulation of HSP70 subtype expressions was consistent with the results of Transcriptome analysis. Moreover, M6 significantly downregulated the expression of AR, which was further proved by Western blot analysis. In summary, our research findings provide a scientific basis for interpreting the significant activity of AD-1 in prostate cancer, and for the research and development of PD and M6 as novel HSP70 inhibitors.
Collapse
Affiliation(s)
- Lei Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, China; Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Shengnan Xiao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, China; Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Zhi Chai
- Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Tao Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, China
| | - Jung Joon Lee
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, China
| | - Guangyue Su
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, China.
| |
Collapse
|
10
|
Wang Y, Bao X, Xian H, Wei F, Song Y, Zhao S, Zhang Y, Wang Y, Wang Y. Glucocorticoid receptors involved in ginsenoside compound K ameliorate adjuvant arthritis by inhibiting the glycolysis of fibroblast-like synoviocytes via the NF-κB/HIF-1α pathway. PHARMACEUTICAL BIOLOGY 2023; 61:1162-1174. [PMID: 37559380 PMCID: PMC10416744 DOI: 10.1080/13880209.2023.2241512] [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: 12/04/2022] [Revised: 05/12/2023] [Accepted: 07/22/2023] [Indexed: 08/11/2023]
Abstract
CONTEXT Ginsenoside metabolite compound K (CK) is an active metabolite produced by ginsenosides in vivo that has an anti-arthritic effect related to the glucocorticoid receptor (GR). However, the potential mechanisms of CK remain unclear. OBJECTIVE This study explores the role and potential mechanisms of CK in vivo and in vitro. MATERIALS AND METHODS Adjuvant arthritis (AA) model was induced in Sprague-Dawley (SD) rats; the rats were randomly divided into four groups (n = 10): normal, AA, CK (80 mg/kg), and dexamethasone (Dex) group (1 mg/kg). From day 15, rats were treated with CK (once a day, i.g.) and Dex (once every 3 days, i.p.) for 18 days. To further verify the mechanism of CK, fibroblast-like synoviocytes (FLS) were stimulated by tumour necrosis factor α (TNF-α) to establish an inflammatory model in vitro. RESULTS CK (80 mg/kg) reduced paw swelling (52%) and arthritis global assessment (31%) compared to that in AA rats. In addition, CK (80 mg/kg) suppressed GLUT1 (38%), HK2 (50%), and PKM2 (56%) levels compared with those in AA FLS. However, the effects of CK (30 μM) on these events were weakened or enhanced after GR knockdown or overexpression in FLS stimulated by TNF-α (30 ng/mL). CK (80 mg/kg) also downregulated the expression of P65 (61%), p-IκB (92%), and HIF-1α (59%). DISCUSSION AND CONCLUSIONS The inhibition of CK on glycolysis and the NF-κB/HIF-1α pathway is potentially mediated through activating GR. These findings provide experimental evidence for elucidating the molecular mechanism of CK in treating rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- Yating Wang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| | - Xiurong Bao
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| | - Hao Xian
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| | - Fang Wei
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| | - Yining Song
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| | - Siyu Zhao
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Yujie Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Yumeng Wang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
| | - Ying Wang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, P.R. China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, P.R. China
| |
Collapse
|
11
|
Mao A, Zhao W, Zhu Y, Kong F, Chen D, Si H, Xu C. Gut Bacterial Community Determines the Therapeutic Effect of Ginsenoside on Canine Inflammatory Bowel Disease by Modulating the Colonic Mucosal Barrier. Microorganisms 2023; 11:2616. [PMID: 38004628 PMCID: PMC10672857 DOI: 10.3390/microorganisms11112616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammatory bowel disease (IBD) comprises systemic inflammatory conditions primarily affecting the gastrointestinal tract, including Crohn's disease and ulcerative colitis. This research aims to analyze the clinical symptoms and pathogenesis of a Dextran sodium sulfate (DSS)-induced canine IBD model and evaluate the restorative effect of ginsenoside from a pathogenesis perspective. We established the DSS-induced canine IBD model and studied the pathological mechanisms. Additionally, we examined the therapeutic effect of ginsenosides by assessing the Canine Inflammatory Bowel Disease Activity Index (CIBDAI), C-reactive protein (CRP) levels, colonic tissue morphology, protein expression, and mucosal bacterial community analysis. Our findings revealed a total ginsenoside content of 22.7% in the ginsenoside extract. Animal experiments demonstrated that dogs with IBD exhibited decreased mental state, significantly increased CIBDAI and CRP levels, disrupted colonic epithelial tissue structure, decreased expression of mucin, tight junctions, and adherens junctions, as well as reduced diversity of the colonic mucosal bacterial community. Furthermore, correlation analysis highlighted a total of 38 bacterial strains correlated with physiological indices. Significantly, ginsenoside treatment could improve these symptoms and reverse the relative abundance of some bacterial communities. In conclusion, alterations in the properties of the colonic mucus layer or the reduction in MUC2, its core component, in dogs with IBD can lead to bacterial penetration of the mucus layer and subsequent contact with intestinal epithelial cells, resulting in inflammation. Remarkably, ginsenoside intervention showcased the capacity to positively influence the relative abundance of bacteria and impact the colonic mucus layer properties, thereby offering promising prospects for IBD management and recovery.
Collapse
Affiliation(s)
- Aipeng Mao
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Research Center for Microbial Feed Engineering of Special Animals in Jilin Province, Innovation Center for Feeding and Utilization of Special Animals in Jilin Province, Changchun 130112, China; (A.M.); (W.Z.); (F.K.); (D.C.)
| | - Weigang Zhao
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Research Center for Microbial Feed Engineering of Special Animals in Jilin Province, Innovation Center for Feeding and Utilization of Special Animals in Jilin Province, Changchun 130112, China; (A.M.); (W.Z.); (F.K.); (D.C.)
| | - Yuhang Zhu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China;
| | - Fantao Kong
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Research Center for Microbial Feed Engineering of Special Animals in Jilin Province, Innovation Center for Feeding and Utilization of Special Animals in Jilin Province, Changchun 130112, China; (A.M.); (W.Z.); (F.K.); (D.C.)
| | - Danyang Chen
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Research Center for Microbial Feed Engineering of Special Animals in Jilin Province, Innovation Center for Feeding and Utilization of Special Animals in Jilin Province, Changchun 130112, China; (A.M.); (W.Z.); (F.K.); (D.C.)
| | - Huazhe Si
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China;
| | - Chao Xu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Research Center for Microbial Feed Engineering of Special Animals in Jilin Province, Innovation Center for Feeding and Utilization of Special Animals in Jilin Province, Changchun 130112, China; (A.M.); (W.Z.); (F.K.); (D.C.)
| |
Collapse
|
12
|
Wang Y, Han Q, Zhang S, Xing X, Sun X. New perspective on the immunomodulatory activity of ginsenosides: Focus on effective therapies for post-COVID-19. Biomed Pharmacother 2023; 165:115154. [PMID: 37454595 DOI: 10.1016/j.biopha.2023.115154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
More than 700 million confirmed cases of Coronavirus Disease-2019 (COVID-19) have been reported globally, and 10-60% of patients are expected to exhibit "post-COVID-19 symptoms," which will continue to affect human life and health. In the absence of safer, more specific drugs, current multiple immunotherapies have failed to achieve satisfactory efficacy. Ginseng, a traditional Chinese medicine, is often used as an immunomodulator and has been used in COVID-19 treatment as a tonic to increase blood oxygen saturation. Ginsenosides are the main active components of ginseng. In this review, we summarize the multiple ways in which ginsenosides affect post-COVID-19 symptoms, including inhibition of lipopolysaccharide, tumor necrosis factor signaling, modulation of chemokine receptors and inflammasome activation, induction of macrophage polarization, effects on Toll-like receptors, nuclear factor kappa-B, the mitogen-activated protein kinase pathway, lymphocytes, intestinal flora, and epigenetic regulation. Ginsenosides affect virus-mediated tissue damage, local or systemic inflammation, immune modulation, and other links, thus alleviating respiratory and pulmonary symptoms, reducing the cardiac burden, protecting the nervous system, and providing new ideas for the rehabilitation of patients with post-COVID-19 symptoms. Furthermore, we analyzed its role in strengthening body resistance to eliminate pathogenic factors from the perspective of ginseng-epidemic disease and highlighted the challenges in clinical applications. However, the benefit of ginsenosides in modulating organismal imbalance post-COVID-19 needs to be further evaluated to better validate the pharmacological mechanisms associated with their traditional efficacy and to determine their role in individualized therapy.
Collapse
Affiliation(s)
- Yixin Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Qin Han
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Shuxia Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| |
Collapse
|
13
|
Ren Q, Lin J, Wang H, Huang M, Tan X, Huang W, Xu Y. Effects of ginseng consumption on the biomarkers of oxidative stress: A systematic review and meta-analysis. Phytother Res 2023; 37:3262-3274. [PMID: 37216939 DOI: 10.1002/ptr.7893] [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: 02/18/2023] [Revised: 04/07/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
Oxidative stress (OS) is a key factor involved in the initiation and development of chronic diseases. Despite its widespread acceptance as an antioxidant, the effects of ginseng on OS in human clinical trials have not been comprehensively analyzed. Therefore, this study aimed to synthesize the results of previous randomized clinical trials (RCTs) examining the impact of ginseng consumption on OS indicators. PubMed, Web of Science, Scopus, and Cochrane databases were searched for articles on the effects of ginseng consumption on oxidative stress markers up to March 20, 2023. Standardized mean difference (SMD) and 95% confidence intervals (CIs) were used to assess effect sizes. Twelve RCTs with 15 effect sizes revealed that the effects of ginseng lowered serum malondialdehyde (MDA) levels (SMD = 0.45, 95% CI: -0.87, -0.08; p = 0.03) and significantly increased the serum total antioxidant capacity (TAC) (SMD = 0.23, 95% CI: 0.01, 0.45; p = 0.04), oxidative dismutase (SOD) (SMD = 0.39, 95% CI: 0.21, 0.57; p < 0.0001), glutathione (GSH) (SMD = 0.36; 95% CI: 0.11, 0.61; p = 0.005), and glutathione reductase (GR) (SMD = 0.56; 95% CI: 0.31, 0.81; p < 0.0001) levels compared to the effects of placebo. However, the effects on serum glutathione peroxidase (GPx) and catalase (CAT) were not significant. Moreover, subgroup analysis based on intervention duration showed that ginseng consumption increased GPx (SMD = 0.91, 95% CI: 0.05, 1.78; p = 0.039) and CAT (SMD = 0.74, 95% CI: 0.27, 1.21; p = 0.002) levels after more than 4 weeks of intervention. According to the results of this meta-analysis, ginseng supplementation dramatically reduced MDA levels and increased TAC, SOD, GSH, and GR levels. Our results open up a new line of defense against oxidative stress-induced diseases.
Collapse
Affiliation(s)
- Qian Ren
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| | - Jie Lin
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| | - Hongya Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| | - Mengting Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Niu Y, Zhang J, Shi D, Zang W, Niu J. Glycosides as Potential Medicinal Components for Ulcerative Colitis: A Review. Molecules 2023; 28:5210. [PMID: 37446872 DOI: 10.3390/molecules28135210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic, non-specific disease of unknown etiology. The disease develops mainly in the rectum or colon, and the main clinical symptoms include abdominal pain, diarrhea, and purulent bloody stools, with a wide variation in severity. The specific causative factors and pathogenesis of the disease are not yet clear, but most scholars believe that the disease is caused by the interaction of genetic, environmental, infectious, immune, and intestinal flora factors. As for the treatment of UC, medications are commonly used in clinical practice, mainly including aminosalicylates, glucocorticoids, and immunosuppressive drugs. However, due to the many complications associated with conventional drug therapy and the tendency for UC to recur, there is an urgent need to discover new, safer, and more effective drugs. Natural compounds with biodiversity and chemical structure diversity from medicinal plants are the most reliable source for the development of new drug precursors. Evidence suggests that glycosides may reduce the development and progression of UC by modulating anti-inflammatory responses, inhibiting oxidative stress, suppressing abnormal immune responses, and regulating signal transduction. In this manuscript, we provide a review of the epidemiology of UC and the available drugs for disease prevention and treatment. In addition, we demonstrate the protective or therapeutic role of glycosides in UC and describe the possible mechanisms of action to provide a theoretical basis for preclinical studies in drug development.
Collapse
Affiliation(s)
- Yating Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Jun Zhang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Dianhua Shi
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Weibiao Zang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
15
|
Kim WH, Kim JE, Kim S, Na Y, Hong YD, Choi J, Park WS, Shim SM. Bioconversion of BIOGF1K, a compound-K-rich fraction from ginseng root and its effect on epidermal barrier function. Heliyon 2023; 9:e14803. [PMID: 37025761 PMCID: PMC10070716 DOI: 10.1016/j.heliyon.2023.e14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
BIOGF1K, the ginseng root-based and hydrolyzed ginsenoside-rich fraction, is known to improve skin damage, but there are rare studies on the kinetic of ginsenosides in the epidermis and their effects on epidermal barrier function. The current study investigated the effect of BIOGF1K on epidermal barrier function and its kinetics on epidermal transport. HPLC and LC/MS were used to verify the ginsenosides and the metabolites of BIOGF1K. Human immortalized keratinocytes (HaCaT) and epidermis-dermis artificial skin were treated with BIOGF1K and their metabolites were analyzed by HPLC and LC/MS. The epidermal barrier function was evaluated by transepithelial electrical resistance (TEER). In BIOGF1K, ginsenoside Rg1, Rd, F1, F2, compound Mc, compound Y (CY), and compound K (CK) were detected and CK and CY were the most and second abundant ginsenosides. TEER of HaCaT with 100 and 200 μg/mL BIOGF1K treatment was significantly higher than the control during 600 min of incubation. CK was permeated to the epidermis in a time-dependent manner and its maximum transported rate was observed at 600 min. In the case of artificial skin, CY and CK were permeated to the epidermis-dermis skin as time-dependent. Also, 24 h after treatment of CY, CK was detected as 19.59% of CY. It was proposed that CY was hydrolyzed into CK while permeating the epidermis. Results from the current study suggest that bioconversion of BIOGF1K rich in CK effectively enhances epidermal barrier function and it could be a useful cosmeceutical to exhibit its functionality to the skin.
Collapse
Affiliation(s)
- Woo-Hyun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Jeong-Eun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Sehyun Kim
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yongjoo Na
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yong-Deok Hong
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Joonho Choi
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Won-Seok Park
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
- Corresponding author.
| | - Soon-Mi Shim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
- Corresponding author.
| |
Collapse
|
16
|
Eun S, Seo H, Suh HJ, Jeong S, Lee S. Modulation of Gut Microbiota and Intestinal Barrier Integrity and Inflammation Profile in High Fat-fed Rats. BIOTECHNOL BIOPROC E 2023. [DOI: 10.1007/s12257-022-0379-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
17
|
Wu F, Lai S, Fu D, Liu J, Wang C, Feng H, Liu J, Li Z, Li P. Neuroprotective Effects and Metabolomics Study of Protopanaxatriol (PPT) on Cerebral Ischemia/Reperfusion Injury In Vitro and In Vivo. Int J Mol Sci 2023; 24:ijms24021789. [PMID: 36675303 PMCID: PMC9861888 DOI: 10.3390/ijms24021789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Stroke, one of the leading causes of disability and death worldwide, is a severe neurological disease that threatens human life. Protopanaxatriol (PPT), panaxatriol-type saponin aglycone, is a rare saponin that exists in Panax ginseng and Panax Noto-ginseng. In this study, we established an oxygen-glucose deprivation (OGD)-PC12 cell model and middle cerebral artery occlusion/reperfusion (MCAO/R) model to evaluate the neuroprotective effects of PPT in vitro and in vivo. In addition, metabolomics analysis was performed on rat plasma and brain tissue samples to find relevant biomarkers and metabolic pathways. The results showed that PPT could significantly regulate the levels of LDH, MDA, SOD, TNF-α and IL-6 factors in OGD-PC12 cells in vitro. PPT can reduce the neurological deficit score and infarct volume of brain tissue in rats, restore the integrity of the blood-brain barrier, reduce pathological damage, and regulate TNF-α, IL-1β, IL-6, MDA, and SOD factors. In addition, the results of metabolomics found that PPT can regulate 19 biomarkers involving five metabolic pathways, including amino acid metabolism, arachidonic acid metabolism, sphingolipid metabolism, and glycerophospholipid metabolism. Thus, it could be inferred that PPT might serve as a novel natural agent for MCAO/R treatment.
Collapse
Affiliation(s)
- Fulin Wu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Sihan Lai
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Dongxing Fu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Juntong Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hao Feng
- College of Basic Medicine Sciences, Jilin University, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuo Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
- Correspondence: (Z.L.); (P.L.); Tel.: +86-0431-8561-9803 (P.L.)
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
- Correspondence: (Z.L.); (P.L.); Tel.: +86-0431-8561-9803 (P.L.)
| |
Collapse
|
18
|
Ye XW, Li CS, Zhang HX, Li Q, Cheng SQ, Wen J, Wang X, Ren HM, Xia LJ, Wang XX, Xu XF, Li XR. Saponins of ginseng products: a review of their transformation in processing. Front Pharmacol 2023; 14:1177819. [PMID: 37188270 PMCID: PMC10175582 DOI: 10.3389/fphar.2023.1177819] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The primary processed product of Panax ginseng C.A. Meyer (P. ginseng) is red ginseng. As technology advances, new products of red ginseng have arisen. Red ginseng products, e.g., traditional red ginseng, sun ginseng, black ginseng, fermented red ginseng, and puffed red ginseng, are commonly used in herbal medicine. Ginsenosides are the major secondary metabolites of P. ginseng. The constituents of P. ginseng are significantly changed during processing, and several pharmacological activities of red ginseng products are dramatically increased compared to white ginseng. In this paper, we aimed to review the ginsenosides and pharmacological activities of various red ginseng products, the transformation law of ginsenosides in processing, and some clinical trials of red ginseng products. This article will help to highlight the diverse pharmacological properties of red ginseng products and aid in the future development of red ginseng industrialization.
Collapse
Affiliation(s)
- Xian-Wen Ye
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun-Shuai Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Hai-Xia Zhang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Shui-Qing Cheng
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Wen
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xuan Wang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Hong-Min Ren
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Liang-Jing Xia
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Xing Wang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Fang Xu
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xin-Fang Xu, ; Xiang-Ri Li,
| | - Xiang-Ri Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xin-Fang Xu, ; Xiang-Ri Li,
| |
Collapse
|
19
|
Anticancer natural products targeting immune checkpoint protein network. Semin Cancer Biol 2022; 86:1008-1032. [PMID: 34838956 DOI: 10.1016/j.semcancer.2021.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/13/2021] [Accepted: 11/23/2021] [Indexed: 01/27/2023]
Abstract
Normal cells express surface proteins that bind to immune checkpoint proteins on immune cells to turn them off, whereby the immune system does not attack normal healthy cells. Cancer cells can also utilize this same protective mechanism by expressing surface proteins that can interact with checkpoint proteins on immune cells to overcome the immune surveillance. Immunotherapy is making the best use of the body's own immune system to reinforce anti-tumor responses. The most generally used immunotherapy is the control of immune checkpoints including the cytotoxic T lymphocyte-associated molecule 4 (CTLA-4), programmed cell deathreceptor 1 (PD-1), or programmed cell death ligand-1 (PD-L1). In spite of the clinical effectiveness of immune checkpoint inhibitors, the overall response rate still remains low. Therefore, there have been considerable efforts in searching for alternative immune checkpoint proteins that may work as new therapeutic targets for treatment of cancer. Recent studies have identified several additional novel immune checkpoint targets, including lymphocyte activation gene-3, T cell immunoglobulin and mucin-domain containing-3, T cell immunoglobulin and immunoreceptor tyrosine-based inhibition motif domain, V-domain Ig suppressor of T cell activation, B7 homolog 3 protein, B and T cell lymphocyte attenuator, and inducible T cell COStimulator. Natural compounds, especially those present in medicinal or dietary plants, have been investigated for their anti-tumor effects in various in vitro and in vivo models. Some phytochemicals exert anti-tumor activities based on immunoregulatioby blocking interaction between proteins involved in immune checkpoint signal transduction or regulating their expression/activity. Recently, synergistic anti-cancer effects of diverse phytochemicals with anti-PD-1/PD-L1 or anti-CTLA-4 monoclonal antibody drugs have been continuously reported. Considering an increasing attention to noteworthy therapeutic effects of immune checkpoint inhibitors in the cancer therapy, this review focuses on regulatory effects of selected phytochemicals on immune checkpoint protein network and their combinational effectiveness with immune checkpoint inhibitors targeting tumor cells.
Collapse
|
20
|
Wan Y, Liu D, Xia J, Xu JF, Zhang L, Yang Y, Wu JJ, Ao H. Ginsenoside CK, rather than Rb1, possesses potential chemopreventive activities in human gastric cancer via regulating PI3K/AKT/NF-κB signal pathway. Front Pharmacol 2022; 13:977539. [PMID: 36249752 PMCID: PMC9556731 DOI: 10.3389/fphar.2022.977539] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Ginsenoside Rb1, a main component of ginseng, is often transformed into ginsenoside CK by intestinal flora to exert various pharmacological activity. However, it remains unclear whether ginsenoside CK is responsible for the anti-gastric cancer effect of ginsenoside Rb1 in vivo. In this study, network pharmacology was applied to predict the key signal pathways of ginsenoside Rb1 and ginsenoside CK when treating gastric cancer. The anti-proliferative effects of ginsenoside Rb1 and ginsenoside CK and the underlying mechanism in gastric cancer cells were explored by MTT, Hoechst3328 staining, ELISA, RT-qPCR and Western blotting. The results showed that PI3K-AKT/NF-κB signal pathway was the common important pathway of ginsenoside Rb1 and CK in the treatment of gastric cancer. The results of MTT assay showed that ginsenoside Rb1 could hardly inhibit the proliferation of HGC-27 cells, whereas ginsenoside CK could inhibit the proliferation of HGC-27 cells. Hoechst3328 staining showed that cells in the ginsenoside CK group were densely stained bright blue and nuclear fragmented, indicating that apoptosis occurred. ELISA results showed that ginsenoside CK could effectively downregulate the levels of cyclin CyclinB1 and CyclinD1, but ginsenoside Rb1 had no significant effect. Also, the results of Western blot and RT-qPCR showed that ginsenoside CK inhibited the expressions of anti-apoptosis-related protein Bcl-2 and apoptosis-related pathway PI3K/AKT/NF-κB, and promoted the expression of pro-apoptosis proteins Bax and Caspase 3, whereas ginsenoside Rb1 exerted no effect. In short, ginsenoside Rb1 had no anti-gastric cancer cell activity in vitro, but ginsenoside CK could effectively inhibit cell proliferation and induce cell apoptosis in HGC-27 cells. The mechanism might relate to the inhibitory effect of ginsenoside CK on the PI3K/AKT/NF-κB pathway. These results suggest that ginsenoside CK might be the in vivo material basis for the anti-gastric cancer activity of ginsenosides.
Collapse
Affiliation(s)
- Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Xia
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-Feng Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiao-Jiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Hui Ao,
| |
Collapse
|
21
|
Feng H, Xue M, Deng H, Cheng S, Hu Y, Zhou C. Ginsenoside and Its Therapeutic Potential for Cognitive Impairment. Biomolecules 2022; 12:1310. [PMID: 36139149 PMCID: PMC9496100 DOI: 10.3390/biom12091310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment (CI) is one of the major clinical features of many neurodegenerative diseases. It can be aging-related or even appear in non-central nerve system (CNS) diseases. CI has a wide spectrum that ranges from the cognitive complaint with normal screening tests to mild CI and, at its end, dementia. Ginsenosides, agents extracted from a key Chinese herbal medicine (ginseng), show great promise as a new therapeutic option for treating CI. This review covered both clinical trials and preclinical studies to summarize the possible mechanisms of how ginsenosides affect CI in different diseases. It shows that ginsenosides can modulate signaling pathways associated with oxidative stress, apoptosis, inflammation, synaptic plasticity, and neurogenesis. The involved signaling pathways mainly include the PI3K/Akt, CREB/BDNF, Keap1/Nrf2 signaling, and NF-κB/NLRP3 inflammasome pathways. We hope to provide a theoretical basis for the treatment of CI for related diseases by ginsenosides.
Collapse
Affiliation(s)
- Hui Feng
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Mei Xue
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Hao Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300073, China
| | - Shiqi Cheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330008, China
| | - Yue Hu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| |
Collapse
|
22
|
Yamamoto M. KAMPOmics: A framework for multidisciplinary and comprehensive research on Japanese traditional medicine. Gene X 2022; 831:146555. [PMID: 35569769 DOI: 10.1016/j.gene.2022.146555] [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: 11/24/2021] [Revised: 03/18/2022] [Accepted: 05/06/2022] [Indexed: 11/04/2022] Open
Abstract
Traditional Japanese medicines, known as "Kampo medicines", are pharmaceutical-grade multi-herbal treatments that are integrated within the modern medical system in Japan. Although basic and clinical research including placebo-controlled double-blind trials is attempting to clarify their effectiveness and mechanisms of action, such studies are seriously limited due to the multi-targeted, multi-component "long-tail" properties of Kampo medicines, which are fundamentally different from modern western therapeutics. However, recent progress in high-throughput analytical technology, coupled with an exponential increase in biomedical information on various levels from molecular biology to clinical "big" data, is enabling us to commence a multidisciplinary and comprehensive investigation of Kampo medicines based on multi-omics, bio-informatics, and systems biology. In addition to deriving an inclusive understanding of the benefits and mechanisms of Kampo medicines, "KAMPOmics" may lead to the development of new principles to control and treat diseases in a systems-oriented manner. Furthermore, elucidation of "sho" and "mibyo" - classical concepts of Kampo, which loosely approximate to the notions of "precise medicine" and "pre-symptomatic aberration", respectively - may contribute to the development of patient-oriented medicine, an area attracting enormous growth and interest in contemporary medicine.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Yoshiwara 3586, Ami, Inashiki, Ibaraki 300-1192, Japan.
| |
Collapse
|
23
|
Gupta M, Chandan K, Sarwat M. Natural Products and their Derivatives as Immune Check Point Inhibitors: Targeting Cytokine/Chemokine Signalling in Cancer. Semin Cancer Biol 2022; 86:214-232. [PMID: 35772610 DOI: 10.1016/j.semcancer.2022.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022]
Abstract
Cancer immunotherapy is the new generation and widely accepted form of tumour treatment. It is, however, associated with exclusive challenges which include organ-specific inflammation, and single-target strategies. Therefore, approaches that can enhance the efficiency of existing immunotherapies and expand their indications are required for the further development of immunotherapy. Natural products and medicines are stated to have this desired effect on cancer immunotherapy (adoptive immune-cells therapy, cancer vaccines, and immune-check point inhibitors). They refurbish the immunosuppressed tumour microenvironment, which is the primary location of interaction of tumour cells with the host immune system. Various immune cell subsets, via interaction with cytokine/chemokine receptors, are recruited into this microenvironment, and these subsets have roles in tumour progression and treatment responsiveness. This review summarises cytokine/chemokine signalling, types of cancer immunotherapy and the herbal medicine-derived natural products targeting cytokine/chemokines and immune checkpoints. These natural compounds possess immunomodulatory activities and exert their anti-tumour effect by either blocking the interaction or modulating the expression of the proteins linked with immune checkpoint signaling pathways. Some compounds also show a synergistic effect in combination with existing monoclonal antibody drugs to reverse the tumour microenvironment. Additionally, we have also reported some studies about the derivatives and formulations used to overcome the limitations of natural forms. This review can provide important insights for directing future research.
Collapse
Affiliation(s)
- Meenakshi Gupta
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Kumari Chandan
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India.
| |
Collapse
|
24
|
Liu J, Xin Y, Qiu Z, Zhang Q, He T, Qiu Y, Wang W. Cordyceps sinensis-mediated biotransformation of notoginsenoside R1 into 25-OH-20( S/ R)-R2 with elevated cardioprotective effect against DOX-induced cell injury. RSC Adv 2022; 12:12938-12946. [PMID: 35497008 PMCID: PMC9049007 DOI: 10.1039/d2ra01470j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023] Open
Abstract
Notoginsenoside R1 is a dammarane saponin in Panax notoginseng with promising cardioprotective effects. The bioactivity–structure relationship of such saponins suggested that the presence of a hydroxyl group at C25 could elevate its performance. To fulfill that goal, bioconversion of notoginsenoside R1 was mediated by a biocatalytic system of Cordyceps sinensis that had successfully produced multiple 25-OH derivatives from ginsenoside Re and Rg1. The major metabolic products of notoginsenoside R1 were identified as 25-OH-20(S/R)-R2 via the techniques of HRMS, 13C-NMR, 1H-NMR, HSQC and HMBC. Time-course experiments were designed to monitor the reaction process, establishing a biocatalytic pathway of “R1→20(S/R)-R2→25-OH-20(S/R)-R2”. The bioconversion rate of these 25-OH derivatives added up to 69.87% which greatly precedes the previous report. Afterwards, the effect of these biocatalytic products against doxorubicin-induced cardiotoxicity was evaluated, indicating a significant increase in efficacy after the hydration of the C24–C25 double bond on the dammarane skeleton. In conclusion, the biocatalytic system employed in this paper is able to harvest 25-OH-20(S/R)-R2 in high yield from notoginsenoside R1, which will provide lead compounds or drug candidates to alleviate myocardial injury caused by doxorubicin. The biocatalytic system in this paper preferably yielded 25-OH notoginsenoside R2 from R1 in a regioselective manner. Such a process significantly elevated the effects of these 25-OH derivatives against DOX-induced cardiomyocyte injury.![]()
Collapse
Affiliation(s)
- Jishuang Liu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Yu Xin
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Zhidong Qiu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Qi Zhang
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Tianzhu He
- School of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Ye Qiu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Weinan Wang
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| |
Collapse
|
25
|
Zhou L, Li ZK, Li CY, Liang YQ, Yang F. Anticancer properties and pharmaceutical applications of ginsenoside compound K: A review. Chem Biol Drug Des 2021; 99:286-300. [PMID: 34793617 PMCID: PMC9541358 DOI: 10.1111/cbdd.13983] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/31/2021] [Accepted: 11/06/2021] [Indexed: 12/16/2022]
Abstract
Ginsenoside compound K (CK) is the major intestinal bacterial metabolite of ginsenosides that exhibits anticancer potential in various cancer cells both in vitro and in vivo. The anticancer types, mechanisms, and effects of CK in the past decade have been summarized in this review. Briefly, CK exerts anticancer effects via multiple molecular mechanisms, including the inhibition of proliferation, invasion, and migration, the induction of apoptosis and autophagy, and anti‐angiogenesis. Some signaling pathways play a significant role in related processes, such as PI3K/Akt/mTOR, JNK/MAPK pathway, and reactive oxygen species (ROS). Moreover, the effects of CK combined with nanocarriers for anticancer efficiency are discussed in this review. Furthermore, we aimed to review the research progress of CK against cancer in the past decade, which might provide theoretical support and effective reference for further research on the medicinal value of small molecules, such as CK.
Collapse
Affiliation(s)
- Li Zhou
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Zhong-Kun Li
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Cong-Yuan Li
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yue-Qin Liang
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Fan Yang
- Joint Surgery, General Hospital of Tibetan Military Command Lhasa, Lhasa, China
| |
Collapse
|
26
|
Xu L, Zhang X, Xiao S, Li X, Jiang H, Wang Z, Sun B, Zhao Y. Panaxadiol as a major metabolite of AD-1 can significantly inhibit the proliferation and migration of breast cancer cells: In vitro and in vivo study. Bioorg Chem 2021; 116:105392. [PMID: 34619469 DOI: 10.1016/j.bioorg.2021.105392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Previous studies have shown that 20 (R)-25-methoxyl-dammarane-3β, 12β, 20 triol (AD-1) can inhibit various cancer cell lines. This study aimed to explore the effect and mechanism of AD-1 metabolite M2 (Panaxadiol; PD) on breast cancer cells of nude mice. Five AD-1 metabolites were isolated and identified using various chromatographic techniques. PD was the main component. In vitro results showed that PD could inhibit the proliferation and migration of MDA-MB-231 cells by inducing G1-phase arrest. In addition, PD down-regulated the expression of Cyclin D1, cdk2, cdk4, cdk6, P-p38, and MMP9, and up-regulated p21 and p27. In vivo results showed that PD could effectively reduce the volume, weight, and migration of breast cancer Transcriptomics analyzed 491 differentially expressed genes by GO and KEGG enrichment. RT-PCR verification confirmed that the significant down-regulation of MMP9 was consistent with transcriptomics results. In further research showed that PD regulated the protein expression of P-p38 and MMP9 in MAPK pathway. In summary, in vivo and in vitro studies showed that PD significantly inhibit the occurrence and development of breast cancer, possibly through the MAPK pathway.
Collapse
Affiliation(s)
- Lei Xu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaoshu Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shengnan Xiao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaofei Li
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Jiang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ziyi Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Baoshan Sun
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yuqing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China; Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
27
|
Qingre Yiqi Method along with Oral Hypoglycemic Drugs in Treating Adults with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4395228. [PMID: 34552649 PMCID: PMC8452389 DOI: 10.1155/2021/4395228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/09/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023]
Abstract
Objective To evaluate the efficacy of the Qingre Yiqi method in the treatment of type 2 diabetes mellitus (T2DM) with meta-analysis. Method The randomized controlled trials (RCTs) of the Qingre Yiqi method in the treatment of T2DM in the PubMed, Medline, EMBase, Cochrane Library, Web of Science, Weipu Journal, China Knowledge Network (CNKI), and Wanfang database were conducted. Three reviewers independently conducted the screening, extracted the data, and assessed methodological quality. Data analysis was performed using Rev Man 5.3 software for statistical analysis. Results A total of 15 RCTs, including 1440 patients, were included. The results showed that compared with oral hypoglycemic drugs alone, the add-on treatment of the Qingre Yiqi method could significantly improve Chinese medicine syndrome (OR (95%CI) = 3.66 [2.47,5.42], P < 0.00001) and lower the level of HbA1c (MD (95%CI) = −0.68 [0.91, −0.45], P < 0.00001), triglyceride (TG) (MD (95%CI) = −0.38 [−0.58,-0.17], P=0.0004), low-density lipoprotein cholesterol (LDL-C) (MD (95%CI) = −0.25 [−0.37, −0.13], P < 0.0001), and total cholesterol(TC) (MD(95%CI) = −0.40[−0.67, −0.13], P=0.003). In terms of fasting blood glucose (FBG) and postprandial blood sugar (PBG), subgroup analysis showed that the baseline of FBG and the number of combined oral hypoglycemic drugs of PBG were the major sources of heterogeneity. Conclusion Compared with the standard treatment, the Qingre Yiqi method along with oral hypoglycemic drugs showed the more beneficial effects for T2DM on improving TCM syndromes and reducing the blood glucose and partial lipid parameter.
Collapse
|
28
|
Cui M, Sun T, Li S, Pan H, Liu J, Zhang X, Li L, Li S, Wei C, Yu C, Yang C, Ma N, Ma B, Lu S, Chang J, Zhang W, Wang H. NIR light-responsive bacteria with live bio-glue coatings for precise colonization in the gut. Cell Rep 2021; 36:109690. [PMID: 34525358 DOI: 10.1016/j.celrep.2021.109690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 04/09/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Recombinant bacterial colonization plays an indispensable role in disease prevention, alleviation, and treatment. Successful application mainly depends on whether bacteria can efficiently spatiotemporally colonize the host gut. However, a primary limitation of existing methods is the lack of precise spatiotemporal regulation, resulting in uncontrolled methods that are less effective. Herein, we design upconversion microgels (UCMs) to convert near-infrared light (NIR) into blue light to activate recombinant light-responsive bacteria (Lresb) in vivo, where autocrine "functional cellular glues" made of adhesive proteins assist Lresb inefficiently colonizing the gut. The programmable engineering platform is further developed for the controlled and effective colonization of Escherichia coli Nissle 1917 (EcN) in the gut. The colonizing bacteria effectively alleviate DSS-induced colitis in mice. We anticipate that this approach could facilitate the clinical application of engineered microbial therapeutics to accurately and effectively regulate host health.
Collapse
Affiliation(s)
- Meihui Cui
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Tao Sun
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin 300072, China; Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Shubin Li
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Huizhuo Pan
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Jing Liu
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Xinyu Zhang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Lianyue Li
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Shanshan Li
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Chunyang Wei
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Chengzhuang Yu
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Chun Yang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Ning Ma
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Binglin Ma
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Shenjunjie Lu
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Jin Chang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Weiwen Zhang
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin 300072, China; Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Hanjie Wang
- School of Life Sciences, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
29
|
Bell L, Whyte A, Duysburgh C, Marzorati M, Van den Abbeele P, Le Cozannet R, Fança-Berthon P, Fromentin E, Williams C. A randomized, placebo-controlled trial investigating the acute and chronic benefits of American Ginseng (Cereboost®) on mood and cognition in healthy young adults, including in vitro investigation of gut microbiota changes as a possible mechanism of action. Eur J Nutr 2021; 61:413-428. [PMID: 34396468 PMCID: PMC8783888 DOI: 10.1007/s00394-021-02654-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 08/04/2021] [Indexed: 12/01/2022]
Abstract
Purpose Cereboost®, an American ginseng extract, has shown improved short-term memory and attention/alertness in healthy young and middle-aged individuals, potentially via modulation of the gut microbiome and upregulation of neurotransmitters such as acetylcholine. Here, we explored the effects of Cereboost® on cognition and mood in the first 6 h post intervention (acute), after 2 weeks daily supplementation (chronic), and whether 2 weeks daily supplementation altered the response to a single acute dose (acute-on-chronic). A concurrent in vitro study evaluated effects of repeated Cereboost® administration on human gut microbiota. Methods Cognitive effects of Cereboost® were assessed using a double-blind, randomized, placebo-controlled clinical trial, with 61 healthy young adults. Modulation of the gut microbiome was concurrently modelled using the Simulator of the Human Microbial Ecosystem (SHIME®), using a young adult donor. Results Consistent with previous findings, Cereboost® improved working memory and attention during the immediate postprandial period; effects that were amplified following two weeks’ treatment (acute-on-chronic) compared to acute testing alone. Chronic supplementation improved cognition on an acetylcholine-sensitive attention task and improved mental fatigue and self-assurance aspects of mood. The parallel in vitro study revealed significantly increased acetate, propionate, and butyrate levels in simulated proximal and distal colon regions, linked with observed increases in Akkermansia muciniphila and Lactobacillus. Conclusion This study confirmed the promising effects of Cereboost® on cognitive function and mood, while suggesting a possible link to alterations of the gut microbiome and modulation of acetylcholine. Further studies will be required to unravel the underlying mechanisms that are involved. Registration The study was pre-registered at ClinicalTrials.gov on 6th July 2018 (Identifier: NCT03579095). Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02654-5.
Collapse
Affiliation(s)
- Lynne Bell
- School of Psychology & Clinical Language Sciences, University of Reading, Earley Gate, Whiteknights Road, Reading, RG6 6ES, UK
| | - Adrian Whyte
- School of Psychology & Clinical Language Sciences, University of Reading, Earley Gate, Whiteknights Road, Reading, RG6 6ES, UK
| | | | | | | | | | | | | | - Claire Williams
- School of Psychology & Clinical Language Sciences, University of Reading, Earley Gate, Whiteknights Road, Reading, RG6 6ES, UK.
| |
Collapse
|
30
|
Production of Minor Ginsenosides C-K and C-Y from Naturally Occurring Major Ginsenosides Using Crude β-Glucosidase Preparation from Submerged Culture of Fomitella fraxinea. Molecules 2021; 26:molecules26164820. [PMID: 34443407 PMCID: PMC8401847 DOI: 10.3390/molecules26164820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
Minor ginsenosides, such as compounds (C)-K and C-Y, possess relatively better bioactivity than those of naturally occurring major ginsenosides. Therefore, this study focused on the biotransformation of major ginsenosides into minor ginsenosides using crude β-glucosidase preparation isolated from submerged liquid culture of Fomitella fraxinea (FFEP). FFEP was prepared by ammonium sulfate (30–80%) precipitation from submerged culture of F. fraxinea. FFEP was used to prepare minor ginsenosides from protopanaxadiol (PPD)-type ginsenoside (PPDG-F) or total ginsenoside fraction (TG-F). In addition, biotransformation of major ginsenosides into minor ginsenosides as affected by reaction time and pH were investigated by TLC and HPLC analyses, and the metabolites were also identified by UPLC/negative-ESI-Q-TOF-MS analysis. FFEP biotransformed ginsenosides Rb1 and Rc into C-K via the following pathways: Rd → F2 → C-K for Rb1 and both Rd → F2→ C-K and C-Mc1 → C-Mc → C-K for Rc, respectively, while C-Y is formed from Rb2 via C-O. FFEP can be applied to produce minor ginsenosides C-K and C-Y from PPDG-F or TG-F. To the best of our knowledge, this study is the first to report the production of C-K and C-Y from major ginsenosides by basidiomycete F. fraxinea.
Collapse
|
31
|
Anti-Cancer Effect of Panax Ginseng and Its Metabolites: From Traditional Medicine to Modern Drug Discovery. Processes (Basel) 2021. [DOI: 10.3390/pr9081344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer incidence and mortality rate are growing worldwide. The effectiveness of cancer therapy depends on the degree of cancer development. Anticancer prevention, screening tests, detection of precancerous conditions or cancers at an early stage of development help to prevent the development of cancer, and in the event of cancer development, they provide the best chance for a full recovery. However, in most cases of advanced cancer, there is no method that can fully cure this disease. Recently, natural products have gained more attention in cancer therapy. Panax ginseng (PG), one of the most popular natural products, is reported to have a wide range of pharmacological activities in cancer. Therefore, the anti-cancer effects and mechanisms of PG and its metabolites (compound K, Ginsenoside Rh1, Rh2, Rh3 and F1) in five major cancers (lung cancer, breast cancer, colon cancer, prostate cancer and stomach cancer) are reviewed in this study. It is confirmed that PG and its metabolites regulated apoptosis, epithelial mesenchymal transition (EMT), angiogenesis, cell cycle arrest and multidrug resistance (MDR) in vitro and in vivo cancer models. In particular, ginsenoside Rh2 showed anticancer effects in all five major cancers. This review could improve the understanding of anticancer mechanisms of PG and its metabolites against major five cancers. Further clinical studies are needed for development anti-cancer drugs using PG and its metabolites.
Collapse
|
32
|
Chopra P, Chhillar H, Kim YJ, Jo IH, Kim ST, Gupta R. Phytochemistry of ginsenosides: Recent advancements and emerging roles. Crit Rev Food Sci Nutr 2021; 63:613-640. [PMID: 34278879 DOI: 10.1080/10408398.2021.1952159] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Ginsenosides, a group of tetracyclic saponins, accounts for the nutraceutical and pharmaceutical relevance of the ginseng (Panax sp.) herb. Owing to the associated therapeutic potential of ginsenosides, their demand has been increased significantly in the last two decades. However, a slow growth cycle, low seed production, and long generation time of ginseng have created a gap between the demand and supply of ginsenosides. The biosynthesis of ginsenosides involves an intricate network of pathways with multiple oxidation and glycosylation reactions. However, the exact functions of some of the associated genes/proteins are still not completely deciphered. Moreover, ginsenoside estimation and extraction using analytical techniques are not feasible with high efficiency. The present review is a step forward in recapitulating the comprehensive aspects of ginsenosides including their distribution, structural diversity, biotransformation, and functional attributes in both plants and animals including humans. Moreover, ginsenoside biosynthesis in the potential plant sources and their metabolism in the human body along with major regulators and stimulators affecting ginsenoside biosynthesis have also been discussed. Furthermore, this review consolidates biotechnological interventions to enhance the biosynthesis of ginsenosides in their potential sources and advancements in the development of synthetic biosystems for efficient ginsenoside biosynthesis to meet their rising industrial demands.
Collapse
Affiliation(s)
- Priyanka Chopra
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Himanshu Chhillar
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Yu-Jin Kim
- Department of Life Science and Environmental Biochemistry, College of Natural Resources and Life Sciences, Pusan National University, Miryang, South Korea
| | - Ick Hyun Jo
- Department of Herbal Crop Research, Rural Development Administration, Eumseong, South Korea
| | - Sun Tae Kim
- Department of Plant Bioscience, College of Natural Resources and Life Sciences, Pusan National University, Miryang, South Korea
| | - Ravi Gupta
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.,Department of Forestry, Environment, and Systems, College of Science and Technology, Kookmin University, Seoul, South Korea
| |
Collapse
|
33
|
Anti-Tumor Effects of Ginsenoside 20(S)-Protopanaxadiol and 1,25-Dihydroxyvitamin D3 Combination in Castration Resistant Prostate Cancer. MEDICINES 2021; 8:medicines8060028. [PMID: 34199743 PMCID: PMC8227560 DOI: 10.3390/medicines8060028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/16/2022]
Abstract
In spite of possessing desirable anticancer properties, currently, limited clinical success has been achieved with 20(S)-protopanaxadiol (aPPD) and 1,25-dihydroxyvitamin D3 (calcitriol). This study is designed to evaluate if the combination of aPPD with calcitriol can inhibit human prostate cancer xenograft growth by using nuclear receptor signaling. Athymic male nude mice were utilized to establish an androgen-independent human prostate cancer C4-2 cell castration-resistant prostate cancer (CRPC) xenograft model. Mice were treated orally for six weeks with 70 mg/kg aPPD administered once daily or three times per week with 4 µg/kg calcitriol or in combination or only with vehicle control. Contrary to our expectations, calcitriol treatment alone increased C4-2 tumor growth. However, the addition of calcitriol substantially increased aPPD-mediated tumor growth suppression (76% vs. 53%, combination vs. aPPD alone). The combination treatment significantly increased levels of cleaved caspase-3 apoptotic marker compared to vehicle-treated or aPPD-treated C4-2 tumors. The mechanistic elucidations indicate that tumor inhibition by the aPPD and calcitriol combination was accompanied by elevated vitamin D receptor (VDR) protein expression. In silico data suggest that aPPD weakly binds to the native LBD pocket of VDR. Interestingly, the combination of aPPD and calcitriol activated VDR at a significantly higher level than calcitriol alone and this indicates that aPPD may be an allosteric activator of VDR. Overall, aPPD and calcitriol combination significantly inhibited tumor growth in vivo with no acute or chronic toxic effects in the C4-2 xenograft CRPC nude mice. The involvement of VDR and downstream apoptotic pathways are potential mechanistic routes of antitumor effects of this combination.
Collapse
|
34
|
A comprehensive review of natural products against atopic dermatitis: Flavonoids, alkaloids, terpenes, glycosides and other compounds. Biomed Pharmacother 2021; 140:111741. [PMID: 34087696 DOI: 10.1016/j.biopha.2021.111741] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/24/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Atopic dermatitis (AD) is considered a great challenge for human communities and imposes both physiological and mental burdens on patients. Natural products have widely been used to treat a wide range of diseases, including cancer, gastrointestinal diseases, asthma, neurological disorders, and infections. To seek potential natural products against AD, in the current review, we searched the terms "atopic dermatitis" and "natural product" in Pubmed, Medline, Web of Science,Science Direct, Embase, EBSCO, CINAHL, ACS. The results show that many natural products, especially puerarin, ferulic acid and ginsenosides, cound protect against AD. Meanwhile, we discussed the therapeutic mechanisms and showed that the natural products exert their anti-inflammatory effects by suppressing the quantity and activity of many inflammatory cell types and cytokines, including neutrophils, monocytes, lymphocytes, Langerhans cells, interleukins (ILs, including IL-1α, IL-1β, IL-4), TNF-α, and TSLP, IgE. via inhibition of JAK/STAT, MAPKs and NF-κB signaling pathways, thereby, halting the inflammatory cascade. Future investigations should focus on studies with more reflective of the clinical characteristics and demographics, so as to develop natural products that will be hopefully available for the treatment of human AD disease.
Collapse
|
35
|
You Y, Liu YL, Ai ZY, Wang YS, Liu JM, Piao CH, Wang YH. Lactobacillus fermentum KP-3-fermented ginseng ameliorates alcohol-induced liver disease in C57BL/6N mice through the AMPK and MAPK pathways. Food Funct 2021; 11:9801-9809. [PMID: 33079125 DOI: 10.1039/d0fo02396e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Panax ginseng was fermented using Lactobacillus fermentum KP-3, and the levels of the minor ginsenosides were measured. Then, the effect of fermented ginseng on alcohol-induced liver injury was investigated. C57BL/6N mice were randomly assigned to 4 groups: pair fed (PF), alcohol fed (AF), alcohol with non-fermented ginseng (AF + NFG) and alcohol with fermented ginseng (AF + FG) groups. After treatment for 8 weeks, fermented ginseng intervention significantly reduced the levels of serum ALT, AST, LPS, TG and TC compared with the AF group. The western-blotting results showed that fermented ginseng activated the adenosine-monophosphate-activated protein kinase (AMPK) pathway to inhibit de novo lipogenesis in the liver and inhibited phosphorylation of p38 through the mitogen-activated protein kinase (MAPK) pathway to alleviate hepatic inflammation, and these effects were superior than those of non-fermented ginseng. Furthermore, fermented ginseng reduced alcohol-induced liver oxidative damage by upregulating the levels of antioxidant enzymes. These findings suggested that the L. fermentum KP-3-fermented ginseng product may be used as a potential dietary nutraceutical for alleviating alcoholic liver injury.
Collapse
Affiliation(s)
- Ying You
- College of Food science and Engineering, Jilin Agricultural University, Changchun, China.
| | | | | | | | | | | | | |
Collapse
|
36
|
Yue H, Liu C, Han Y, Zhuang Z, Yu H, Wang Z, Sun C, Im WT, Jin F. Preparation of minor ginsenosides C-K and C-Mx from protopanaxadiol ginsenosides of American ginseng leaves by a enzyme from Aspergillus sp.agl-84 strain. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Chen Y, Wang S, Yang S, Li R, Yang Y, Chen Y, Zhang W. Inhibitory role of ginsenoside Rb2 in endothelial senescence and inflammation mediated by microRNA‑216a. Mol Med Rep 2021; 23:415. [PMID: 33786633 PMCID: PMC8025470 DOI: 10.3892/mmr.2021.12054] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/09/2021] [Indexed: 01/02/2023] Open
Abstract
Targeting microRNAs (miRs) using small chemical molecules has become a promising strategy for disease treatment. miR‑216a has been reported to be a potential therapeutic target in endothelial senescence and atherosclerosis via the Smad3/NF‑κB signaling pathway. Ginsenoside Rb2 (Rb2) is the main bioactive component extracted from the plant Panax ginseng, and is a widely used traditional Chinese medicine. In the present study, Rb2 was identified to have a high score for miR‑216a via bioinformatics analysis based on its sequence and structural features. The microscale thermophoresis experiment further demonstrated that Rb2 had a specific binding affinity for miR‑216a and the dissociation constant was 17.6 µM. In both young and senescent human umbilical vein endothelial cells (HUVECs), as well as human aortic endothelial cells, Rb2 decreased the expression of endogenous miR‑216a. Next, a replicative endothelial senescence model of HUVECs was established by infection with pre‑miR‑216a recombinant lentiviruses (Lv‑miR‑216a) and the number of population‑doubling level (PDL) was calculated. Stable overexpression of miR‑216a induced a premature senescent‑like phenotype, whereas the senescent features and increased activity of senescence‑associated β‑galactosidase (SA‑β‑gal) were reversed after Rb2 treatment. The percentage of SA‑β‑gal‑positive cells in senescent PDL25 cells transfected with Lv‑miR‑216a was decreased 76% by Rb2 treatment compared with the Lv‑miR‑216a group without Rb2 treatment (P=0.01). Mechanistically, miR‑216a inhibited Smad3 protein expression, promoted IκBα degradation and activated NF‑κB‑responsive genes, such as vascular cell adhesion molecule 1 (VCAM1), which promoted the adhesiveness of endothelial cells to monocytes. These pro‑inflammatory effects of miR‑216a were significantly suppressed by Rb2 treatment. When Smad3 was suppressed by small interfering RNA, the elevated expression levels of intercellular adhesion molecule 1 and VCAM1 induced by miR‑216a were significantly reversed. Collectively, to the best of our knowledge, the present study demonstrated for the first time that Rb2 exerted an anti‑inflammation effect on the process of endothelial cell senescence and could be a potential therapeutic drug by targeting miR‑216a.
Collapse
Affiliation(s)
- Yutong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, P.R. China
| | - Shuting Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Shujun Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Rongxia Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Yunyun Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Yu Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Weili Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| |
Collapse
|
38
|
Systematic optimization of the yeast cell factory for sustainable and high efficiency production of bioactive ginsenoside compound K. Synth Syst Biotechnol 2021; 6:69-76. [PMID: 33869813 PMCID: PMC8040117 DOI: 10.1016/j.synbio.2021.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/23/2021] [Accepted: 03/16/2021] [Indexed: 02/08/2023] Open
Abstract
Ginsenoside Compound K (CK) has been recognized as a major functional component that is absorbed into the systemic circulation after oral administration of ginseng. CK demonstrates diverse bioactivities. A phase I clinical study indicated that CK was a potential candidate for arthritis therapy. However, a phase II clinical study was suspended because of the high cost associated with the present CK manufacturing approach, which is based on the traditional planting-extracting-biotransforming process. We previously elucidated the complete CK biosynthetic pathway and realized for the first time de novo biosynthesis of CK from glucose by engineered yeast. However, CK production was not sufficient for industrial application. Here, we systematically engineered Saccharomyces cerevisiae to achieve high titer production of CK from glucose using a previously constructed protopanaxadiol (PPD)-producing chassis, optimizing UGTPg1 expression, improving UDP-glucose biosynthesis, and tuning down UDP-glucose consumption. Our final engineered yeast strain produced CK with a titer of 5.74 g/L in fed-batch fermentation, which represents the highest CK production in microbes reported to date. Once scaled-up, this high titer de novo microbial biosynthesis platform will enable a robust and stable supply of CK, thus facilitating study and medical application of CK.
Collapse
|
39
|
DURHAN A, KOŞMAZ K, ŞENLİKCİ A, ERGÜDER E, SÜLEYMAN M, DUYMUŞ ME, BAĞ YM, PEKCİCİ MR, ŞENEŞ M, ALKAN KUŞABBİ İ, ESER EP, HÜCÜMENOĞLU S. Does red ginseng ameliorate liver damage caused by obstructive jaundice? : an experimental study. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2021. [DOI: 10.32322/jhsm.900023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
40
|
Wada Y, Tokuda K, Morine Y, Okikawa S, Yamashita S, Ikemoto T, Imura S, Saito Y, Yamada S, Shimada M. The inhibitory effect of TU-100 on hepatic stellate cell activation in the tumor microenvironment. Oncotarget 2020; 11:4593-4604. [PMID: 33346211 PMCID: PMC7733620 DOI: 10.18632/oncotarget.27835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION The tumor microenvironment is involved in acquiring tumor malignancies of colorectal liver metastasis (CRLM). We have reported that TU-100 (Daikenchuto) suppresses hepatic stellate cell (HSC) activation in obstructive jaundice. In this study, we report new findings as the direct and indirect inhibitory effects of TU-100 on cancer cell growth through the suppression of HSC activation. MATERIALS AND METHODS The HSCs (LX2) were cultured in colon cancer cells (HCT116 and HT29)-conditioned medium (CM) with or without TU-100 treatment (90, 270, 900 μg/ml). Activated HSCs (aHSCs) were detected by α-SMA and IL-6 mRNA expressions and cytokine arrays of HSC's culture supernatants. Cancer cell growth was analyzed for proliferation and migration ability, compared with TU-100 treatment. To investigate the direct anti-tumor effect of TU-100, cancer cells were cultured in the presence of aHSC-CM and TU-100 (90, 270, 900) or aHSC-CM alone, and assessed autophagosomes, conversion to LC3-II protein, and Beclin-1 mRNA expression. RESULTS Colon cancer-CM significantly increased α-SMA and IL-6 mRNA expressions of aHSC. α-SMA and IL-6 mRNA expressions of aHSC, and IL-6 secretions from aHSCs were significantly decreased with TU-100 (270, 900) treatment, compared to colon cancer-CM alone. Compared with normal culture medium, aHSC-CM led to a significantly increased cell number and modified HSC-CM (TU-100; 270, 900) significantly suppressed cancer cell growth and migration. TU-100 (900) treatment induced autophagy and significantly promoted the autophagic cell death. CONCLUSIONS TU-100 inhibited colon cancer cell malignant potential by both suppressing HSC activation and inducing directly autophagy of cancer cells.
Collapse
Affiliation(s)
- Yuma Wada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,These authors contributed equally to this work
| | - Kazunori Tokuda
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,These authors contributed equally to this work
| | - Yuji Morine
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shohei Okikawa
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shoko Yamashita
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Satoru Imura
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yu Saito
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shinichiro Yamada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
41
|
Liu J, Liu Y, Yu H, Zhang Y, Hsu ACY, Zhang M, Gou Y, Sun W, Wang F, Li P, Liu J. Design, synthesis and biological evaluation of novel pyxinol derivatives with anti-heart failure activity. Biomed Pharmacother 2020; 133:111050. [PMID: 33378957 DOI: 10.1016/j.biopha.2020.111050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 01/20/2023] Open
Abstract
Heart failure (HF) is an important and leading cause of substantial morbidity and mortality globally. The angiotensin-converting enzymatic (ACE) is the causative source for congestive heart failure. Natural products and its derivatives play a vital role in drug discovery and development owing to their efficacy and low toxicity. Pyxinol is a potent natural agent for cardiovascular disease. Thus we investigated the effect on ACE and HF of pyxinol derivatives. We designed and synthesized 32 novel fatty acid ester derivatives of pyxinol via esterification. Among them, compounds 2e (IC50=105 nM) and 3b (IC50=114 nM) displayed excellent ACE inhibitory activity in vitro, and exhibited non-toxic to H9c2 cells. The interactions between ACE and compounds were predicted by molecular docking respectively. In verapamil-induced zebrafish HF model, the activity assay showed that these two derivatives could improve cardiovascular physiological indexes including heart beats, venous congestion, heart dilation, cardiac output, ejection fraction and fractional shortening in a dose-dependent manner. A UPLC-QTOF-MS-based serum metabolomics approach was applied to explore the latent mechanism. A total of 25 differentiated metabolites and 8 perturbed metabolic pathways were identified. These results indicated that pyxinol fatty acid ester derivatives 2e and 3b might be considered as potent drug candidates against heart failure and deserved further research and development.
Collapse
Affiliation(s)
- Junli Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Yunhe Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Hui Yu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China; The First Hospital of Jilin University, Changchun 130021, China
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Mingming Zhang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yawei Gou
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Wei Sun
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China.
| |
Collapse
|
42
|
Bastola T, Pariyar R, Jeon BM, Baek JI, Chang BY, Kim SC, Kim SY, Seo J. Protective effects of SGB121, ginsenoside F1-enriched ginseng extract, on scopolamine-induced cytotoxicity and memory impairments. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
43
|
Sui X, Liu J, Xin Y, Qu M, Qiu Y, He T, Luo H, Wang W, Qiu Z. Highly regioselective biotransformation of ginsenoside Rg1 to 25-OH derivatives of 20(S/R)-Rh1 by Cordyceps Sinensis. Bioorg Med Chem Lett 2020; 30:127504. [PMID: 32827631 DOI: 10.1016/j.bmcl.2020.127504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/08/2020] [Accepted: 08/17/2020] [Indexed: 11/20/2022]
Abstract
25-OH ginsenosides are potent and rare prodrugs in natural sources. However current strategies for such modification always end up in undesirable side products and unsatisfied yield that hinders them from further applications. Herein, ginsenoside Rg1 was thoroughly converted into 20(S/R)-Rh1 and 25-OH-20(S/R)-Rh1 by Cordyceps Sinensis in an optimum medium. The chemical correctness of either 25-OH-20(S/R)-Rh1 epimers was validated by LC-IT-TOF-MSn and 13C NMR spectrometry. The biocatalytic pathway was established as Rg1 → 20(S/R)-Rh1 → 25-OH-20(S/R)-Rh1. The molar bioconversion rate for total 25-OH-20(S/R)-Rh1 was calculated to be 82.5%, of which S-configuration accounted for 43.2% while R-configuration 39.3%. These two 25-OH derivatives are direct hydration products from 20(S/R)-Rh1 without other side metabolites, suggesting this is a highly regioselective process. In conclusion, this biocatalytic system could be harnessed to facilitate the preparation of diversified 25-OH ginsenosides with high yields of the target compound and simple chemical background in the reaction mixture.
Collapse
Affiliation(s)
- Xin Sui
- Changchun University of Chinese Medicine, Changchun 130117, China; The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jishuang Liu
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yu Xin
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Mo Qu
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Ye Qiu
- Changchun University of Chinese Medicine, Changchun 130117, China; National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Tianzhu He
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Weinan Wang
- Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Zhidong Qiu
- Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
44
|
Prebiotics enhance the biotransformation and bioavailability of ginsenosides in rats by modulating gut microbiota. J Ginseng Res 2020; 45:334-343. [PMID: 33841014 PMCID: PMC8020290 DOI: 10.1016/j.jgr.2020.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/13/2020] [Accepted: 08/02/2020] [Indexed: 12/02/2022] Open
Abstract
Background Gut microbiota mainly function in the biotransformation of primary ginsenosides into bioactive metabolites. Herein, we investigated the effects of three prebiotic fibers by targeting gut microbiota on the metabolism of ginsenoside Rb1 in vivo. Methods Sprague Dawley rats were administered with ginsenoside Rb1 after a two-week prebiotic intervention of fructooligosaccharide, galactooligosaccharide, and fibersol-2, respectively. Pharmacokinetic analysis of ginsenoside Rb1 and its metabolites was performed, whilst the microbial composition and metabolic function of gut microbiota were examined by 16S rRNA gene amplicon and metagenomic shotgun sequencing. Results The results showed that peak plasma concentration and area under concentration time curve of ginsenoside Rb1 and its intermediate metabolites, ginsenoside Rd, F2, and compound K (CK), in the prebiotic intervention groups were increased at various degrees compared with those in the control group. Gut microbiota dramatically responded to the prebiotic treatment at both taxonomical and functional levels. The abundance of Prevotella, which possesses potential function to hydrolyze ginsenoside Rb1 into CK, was significantly elevated in the three prebiotic groups (P < 0.05). The gut metagenomic analysis also revealed the functional gene enrichment for terpenoid/polyketide metabolism, glycolysis, gluconeogenesis, propanoate metabolism, etc. Conclusion These findings imply that prebiotics may selectively promote the proliferation of certain bacterial stains with glycoside hydrolysis capacity, thereby, subsequently improving the biotransformation and bioavailability of primary ginsenosides in vivo.
Collapse
Key Words
- ANOVA, analysis of variance
- AUC, area under the concentration-time curve
- Bioavailability
- Biotransformation
- CAT, CAZymes Analysis Toolkit
- CAZymes, carbohydrate active enzymes
- CK, compound K
- Cmax, peak plasma concentration
- FDR, false discovery rate
- FOS, fructooligosaccharide
- GOS, galactooligosaccharide
- Ginsenoside
- Gut microbiota
- IS, internal standard
- KEGG, the Kyoto Encyclopaedia of Genes and Genomes
- LCA, lowest common ancestor
- LDA, linear discriminant analysis
- LEfSe, LDA effect size
- LLOQs, lower limits of quantifications
- MANOVA, multivariate ANOVA
- MRM, multiple reaction monitoring
- NMDS, non-metric multidimensional scaling
- PCA, principal component analysis
- PCoA, principal coordinates analysis
- Prebiotic
- SD, Sprague Dawley
- SRA, Sequence Read Archive
- Tmax, time of maximum plasma concentration
- UPLC-ESI-QqQ-MS/MS, ultra-high pressure liquid chromatography coupled to an electrospray ionization source and a triple-quadrupole mass spectrometer
Collapse
|
45
|
Yao YY, Ling EA, Lu D. Microglia mediated neuroinflammation - signaling regulation and therapeutic considerations with special reference to some natural compounds. Histol Histopathol 2020; 35:1229-1250. [PMID: 32662061 DOI: 10.14670/hh-18-239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroinflammation plays a central role in multiple neurodegenerative diseases and neurological disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemic injury etc. In this connection, microglia, the key players in the central nervous system, mediate the inflammatory response process. In brain injuries, activated microglia can clear the cellular debris and invading pathogens and release neurotrophic factors; however, prolonged microglia activation may cause neuronal death through excessive release of inflammatory mediators. Therefore, it is of paramount importance to understand the underlying molecular mechanisms of microglia activation to design an effective therapeutic strategy to alleviate neuronal injury. Recent studies have shown that some natural compounds and herbal extracts possess anti-inflammatory properties that may suppress microglial activation and ameliorate neuroinflammation and hence are neuroprotective. In this review, we will update some of the common signaling pathways that regulate microglia activation. Among the various signaling pathways, the Notch-1, mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) have been reported to exacerbate microglia mediated neuroinflammation that is implicated in different neuropathological diseases. The search for natural compounds or agents, specifically those derived from natural herbal extracts such as Gastrodin, scutellarin, RG1 etc. has been the focus of many of our recent studies because they have been found to regulate microglia activation. The pharmacological effects of these agents and their potential mechanisms for regulating microglia activation are systematically reviewed here for a fuller understanding of their biochemical action and therapeutic potential for treatment of microglia mediated neuropathological diseases.
Collapse
Affiliation(s)
- Yue-Yi Yao
- Technology Transfer Center, Kunming Medical University, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Young Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Di Lu
- Technology Transfer Center, Kunming Medical University, Kunming, China.
| |
Collapse
|
46
|
Gao Y, Zhang S, Li J, Zhao J, Xiao Q, Zhu Y, Zhang J, Huang W. Effect and mechanism of ginsenoside Rg1-regulating hepatic steatosis in HepG2 cells induced by free fatty acid. Biosci Biotechnol Biochem 2020; 84:2228-2240. [PMID: 32654591 DOI: 10.1080/09168451.2020.1793293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ginsenoside Rg1 (G-Rg1) is a bioactive phytochemical that has been found to be beneficial for the treatment of several diseases including nonalcoholic fatty liver disease (NAFLD). But there is a lack of literature reporting the effect of G-Rg1 on lipid metabolism balance in NAFLD. We investigated the effect and mechanism of G-Rg1 on lipid metabolism in vitro. We found that G-Rg1 decreased the levels of TG, TC, and MDA, and increased activity of SOD. Results of RT-PCR and western blotting showed that supplementation with G-Rg1 downregulated the expression of PPAR γ, FABP1, FATP2/5, CD36, SREBP1 c, and FASN, while the expression of PPAR ɑ, CPT1, ACOX1, MTTP, and ApoB100 was upregulated, after induction by a free fatty acid. Taken together, we conclude that G-Rg1 inhibits lipid synthesis and lipid uptake, and enhances lipid oxidation and lipid export to reduce hepatic steatosis of HepG2 cells by regulating PPAR ɑ and PPAR γ expression.
Collapse
Affiliation(s)
- Yue Gao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jiajun Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jinqiu Zhao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Qing Xiao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Yali Zhu
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jia Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Wenxiang Huang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| |
Collapse
|
47
|
Han X, Li W, Ma X, Fan D. Enzymatic hydrolysis and extraction of ginsenoside recovered from deep eutectic solvent-salt aqueous two-phase system. J Biosci Bioeng 2020; 130:390-396. [PMID: 32591224 DOI: 10.1016/j.jbiosc.2020.05.008] [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: 01/08/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/29/2022]
Abstract
Rare ginsenoside CK was recognized as a popular functional food because of superior pharmacological activity, but it is restricted by further applications by the difficulty in preparation. In this study, deep eutectic solvent (DES)-based aqueous two-phase system (ATPS) was established to transform and extract ginsenoside CK in situ for the first time. The phase formation conditions for preparing ATPS using choline chloride-based DES were studied, and the optimal conditions for extractive bioconversion were explored using conventional single-factor experiments. The conditions for ATPS establishment were as follows: 31.9% (w/w) DES (ChCl-ethylene glycol)/24.5% (w/w) K2HPO4, 55 °C, pH 5.0. Under the optimal conditions, 75.79% product and 61.14% β-glucosidase were recovered from the top and bottom phase, respectively. In addition, DES and β-glucosidase can be recovered and recycled again for the next extractive bioconversion of CK. These results indicated that this green and efficient method exhibited considerable value in integrated production and extraction processes, and demonstrated the potential for obtaining highly recycled functional foods and similar products.
Collapse
Affiliation(s)
- Xin Han
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. &Biomed. Research Institute, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Weina Li
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. &Biomed. Research Institute, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Xiaoxuan Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. &Biomed. Research Institute, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. &Biomed. Research Institute, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
48
|
Yang N, Liu D, Zhang X, Li J, Wang M, Xu T, Liu Z. Effects of ginsenosides on bone remodelling for novel drug applications: a review. Chin Med 2020; 15:42. [PMID: 32391072 PMCID: PMC7201946 DOI: 10.1186/s13020-020-00323-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ginsenosides are pharmacologically active compounds that are often extracted from the Panax plant for their medicinal properties. Ginsenosides have multiple effects, including antitumor effects which have been widely studied. In recent years, studies have found that ginsenosides promote proliferation and osteogenesis of osteoblast-related cells, as well as inhibit the activity of osteoclasts. MAIN BODY We briefly introduces the molecules and BMP, WNT, and RANKL signalling pathways involved in bone formation and bone resorption. Next, recent studies on the mechanism of action of ginsenosides in bone remodelling are reviewed from three perspectives: the effects on proliferation of osteoblast-related cells, effects on osteogenesis and effects on osteoclasts. To expedite the development of drugs containing ginsenosides, we summarize the multiple beneficial roles of various types of ginsenosides in bone remodelling; including the promotion of bone formation, inhibition of bone resorption, and anti-inflammatory and antioxidant effects. CONCLUSION Many ginsenosides can promote bone formation and inhibit bone resorption, such as Rb1, Rb2 and Re. Ginsenosides have the potential to be new drugs for the treatment of osteoporosis, promote fracture healing and are strong candidates for cytokines in the tissue-engineered bone. This review provides a theoretical basis for clinical drug applications and proposes several future directions for exploring the beneficial role of ginseng compounds in bone remodelling.
Collapse
Affiliation(s)
- Nan Yang
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Dingkun Liu
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Xiao Zhang
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Jianing Li
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Mi Wang
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Tongtong Xu
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Zhihui Liu
- Department of Prosthodontics, Stomatology Hospital of Jilin University, Changchun, 130021 Jilin China
| |
Collapse
|
49
|
Kim D, Park M, Haleem I, Lee Y, Koo J, Na YC, Song G, Lee J. Natural Product Ginsenoside 20(S)-25-Methoxyl-Dammarane-3β, 12β, 20-Triol in Cancer Treatment: A Review of the Pharmacological Mechanisms and Pharmacokinetics. Front Pharmacol 2020; 11:521. [PMID: 32425780 PMCID: PMC7212460 DOI: 10.3389/fphar.2020.00521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Panax ginseng has been used as an herbal medicine for thousands of years. Most of its pharmacological effects are attributed to its constituent ginsenosides, including 20(S)-25-methoxyl-dammarane-3β, 12β, 20-triol (20(S)-25-OCH3-PPD), which is one of the protopanaxadiol type ginsenosides. It has been found to exhibit anticancer effects by interacting with multiple pharmacological pathways, such as the Wnt/β-catenin, MDM2, ERK/MAPK, and STAT3 signaling pathways. However, its therapeutic potential could be limited by its low bioavailability mainly due to its low aqueous solubility. Thus, several studies have been conducted on its pharmacokinetics and its delivery systems, so as to increase its oral bioavailability. In this review, comprehensive information on its varying pharmacological pathways in cancer, as well as its pharmacokinetic behavior and pharmaceutical strategies, is provided. This information would be useful in the understanding of its diverse mechanisms and pharmacokinetics as an anticancer drug, leading to the design of superior 20(S)-25-OCH3-PPD-containing formulations that maximize its therapeutic potential.
Collapse
Affiliation(s)
- Dohyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Minwoo Park
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Iqra Haleem
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Younghong Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jain Koo
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Young Chae Na
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Gidong Song
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| |
Collapse
|
50
|
Ahn HJ, You HJ, Park MS, Li Z, Choe D, Johnston TV, Ku S, Ji GE. Microbial biocatalysis of quercetin-3-glucoside and isorhamnetin-3-glucoside in Salicornia herbacea and their contribution to improved anti-inflammatory activity. RSC Adv 2020; 10:5339-5350. [PMID: 35498283 PMCID: PMC9049170 DOI: 10.1039/c9ra08059g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Salicornia herbacea (glasswort) is a traditional Asian medicinal plant which exhibits multiple nutraceutical and pharmaceutical properties. Quercetin-3-glucoside and isorhamnetin-3-glucoside are the major flavonoid glycosides found in S. herbacea. Multiple researchers have shown that flavonoid glycosides can be structurally transformed into minor aglycone molecules, which play a significant role in exerting physiological responses in vivo. However, minor aglycone molecule levels in S. herbacea are very low. In this study, Bifidobacterium animalis subsp. lactis AD011, isolated from infant feces, catalyzed >85% of quercetin-3-glucoside and isorhamnetin-3-glucoside into quercetin and isorhamnetin, respectively, in 2 h, without breaking down flavonoid backbones. Functionality analysis demonstrated that the quercetin and isorhamnetin produced showed improved anti-inflammatory activity vs. the original source molecules against lipopolysaccharide induced RAW 264.7 macrophages. Our report highlights a novel protocol for rapid quercetin and isorhamnetin production from S. herbacea flavonoids and the applicability of quercetin and isorhamnetin as nutraceutical molecules with enhanced anti-inflammatory properties.
Collapse
Affiliation(s)
- Hyung Jin Ahn
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University Seoul 08826 Republic of Korea
| | - Hyun Ju You
- Center for Human and Environmental Microbiome, Institute of Health and Environment Seoul 08826 Republic of Korea
| | - Myeong Soo Park
- Research Center, BIFIDO Co., Ltd. Hongcheon 25117 Republic of Korea
| | - Zhipeng Li
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University Seoul 08826 Republic of Korea
| | - Deokyeong Choe
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University Murfreesboro TN 37132 USA
| | - Tony Vaughn Johnston
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University Murfreesboro TN 37132 USA
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University Murfreesboro TN 37132 USA
| | - Geun Eog Ji
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University Seoul 08826 Republic of Korea
- Research Center, BIFIDO Co., Ltd. Hongcheon 25117 Republic of Korea
| |
Collapse
|