1
|
Hernández-Guzmán C, Hernández-Montiel LG, Velázquez-Lizarraga AE, Ríos-González LJ, Huerta-Ochoa S, Cervantes-Güicho VDJ, Morales-Martínez TK, Mejía-Ruíz CH, Reyes AG. Enhanced Antioxidant, Antifungal, and Herbicidal Activities through Bioconversion of Diosgenin by Yarrowia lipolytica P01a. PLANTS (BASEL, SWITZERLAND) 2024; 13:2629. [PMID: 39339605 PMCID: PMC11434977 DOI: 10.3390/plants13182629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
This study explores the bioconversion of diosgenin by Yarrowia lipolytica P01a, focusing on enhancing the antioxidant, antifungal, and herbicidal activities of the resulting extracts. The bioconversion process, involving glycosylation and hydroxylation, produced significant amounts of protodioscin and soyasaponin I. The extracts showed superior antioxidant activity, with up to 97.02% inhibition of ABTS· radicals and 33.30% inhibition of DPPH· radicals at 1000 mg L-1 of diosgenin. Antifungal assays revealed strong inhibitory effects against Botrytis cinerea, Alternaria sp., and Aspergillus niger, with maximum inhibition rates of 67.34%, 35.63%, and 65.53%, respectively. Additionally, the herbicidal activity of the bioconverted extracts was comparable to commercial herbicides, achieving 100% inhibition of seed germination in both monocotyledonous and dicotyledonous plants. These findings suggest that the Y. lipolytica P01a-mediated bioconversion of diosgenin could provide a sustainable and eco-friendly alternative for developing natural biofungicides and bioherbicides.
Collapse
Affiliation(s)
- Christian Hernández-Guzmán
- Unidad Iztapalapa, Departamento de Biotecnología, Universidad Autónoma Metropolitana, México City 02128, Mexico
| | - Luis G Hernández-Montiel
- Centro de Investigaciones Biológicas del Noroeste, Programa de Agricultura en Zonas Áridas, Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, Mexico
| | | | - Leopoldo J Ríos-González
- Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Saltillo, Coahuila 25280, Mexico
| | - Sergio Huerta-Ochoa
- Unidad Iztapalapa, Departamento de Biotecnología, Universidad Autónoma Metropolitana, México City 02128, Mexico
| | | | | | - Claudio H Mejía-Ruíz
- Centro de Investigaciones Biológicas del Noroeste, Programa de Agricultura en Zonas Áridas, Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, Mexico
| | - Ana G Reyes
- Centro de Investigaciones Biológicas del Noroeste, Programa de Agricultura en Zonas Áridas, Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, Mexico
| |
Collapse
|
2
|
Ben-Eltriki M, Shankar G, Tomlinson Guns ES, Deb S. Pharmacokinetics and pharmacodynamics of Rh2 and aPPD ginsenosides in prostate cancer: a drug interaction perspective. Cancer Chemother Pharmacol 2023; 92:419-437. [PMID: 37709921 DOI: 10.1007/s00280-023-04583-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Ginsenoside Rh2 and its aglycon (aPPD) are one of the major metabolites from Panax ginseng. Preclinical studies suggest that Rh2 and aPPD have antitumor effects in prostate cancer (PCa). Our aims in this review are (1) to describe the pharmacokinetic (PK) properties of Rh2 and aPPD ginsenosides; 2) to provide an overview of the preclinical findings on the use of Rh2 and aPPD in the treatment of PCa; and (3) to highlight the mechanisms of its PK and pharmacodynamic (PD) drug interactions. Increasing evidence points to the potential efficacy of Rh2 or aPPD for PCa treatment. Based on the laboratory studies, Rh2 or aPPD combinations revealed an additive or synergistic interaction or enhanced sensitivity of anticancer drugs toward PCa. This review reveals that enhanced anticancer activities were demonstrated in preclinical studies through interactions of Rh2 and/or aPPD with the proteins related to PK (e.g., cytochrome P450 enzymes, transporters) or PD of the other anticancer drugs or PCa signaling pathways. In conclusion, combining Rh2 or aPPD with anti-prostate cancer drugs leads to PK or PD interactions which could facilitate either therapeutically beneficial or toxic effects.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC, Canada.
- Community Pharmacist, Vancouver Area, BC, Canada.
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Gehana Shankar
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
3
|
Abstract
As a steroid skeleton-based saponin, ginsenoside Rh2 (G-Rh2) is one of the major bioactive ginsenosides from the plants of genus Panax L. Many studies have reported the notable pharmacological activities of G-Rh2 such as anticancer, antiinflammatory, antiviral, antiallergic, antidiabetic, and anti-Alzheimer's activities. Numerous preclinical studies have demonstrated the great potential of G-Rh2 in the treatment of a wide range of carcinomatous diseases in vitro and in vivo. G-Rh2 is able to inhibit proliferation, induce apoptosis and cell cycle arrest, retard metastasis, promote differentiation, enhance chemotherapy and reverse multi-drug resistance against multiple tumor cells. The present review mainly summarizes the anticancer effects and related mechanisms of G-Rh2 in various models as well as the recent advances in G-Rh2 delivery systems and structural modification to ameliorate its anticancer activity and pharmacokinetics characteristics.
Collapse
|
4
|
Hao Q, Wu Y, Vadgama JV, Wang P. Phytochemicals in Inhibition of Prostate Cancer: Evidence from Molecular Mechanisms Studies. Biomolecules 2022; 12:1306. [PMID: 36139145 PMCID: PMC9496067 DOI: 10.3390/biom12091306] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Prostate cancer is one of the leading causes of death for men worldwide. The development of resistance, toxicity, and side effects of conventional therapies have made prostate cancer treatment become more intensive and aggressive. Many phytochemicals isolated from plants have shown to be tumor cytotoxic. In vitro laboratory studies have revealed that natural compounds can affect cancer cell proliferation by modulating many crucial cellular signaling pathways frequently dysregulated in prostate cancer. A multitude of natural compounds have been found to induce cell cycle arrest, promote apoptosis, inhibit cancer cell growth, and suppress angiogenesis. In addition, combinatorial use of natural compounds with hormone and/or chemotherapeutic drugs seems to be a promising strategy to enhance the therapeutic effect in a less toxic manner, as suggested by pre-clinical studies. In this context, we systematically reviewed the currently available literature of naturally occurring compounds isolated from vegetables, fruits, teas, and herbs, with their relevant mechanisms of action in prostate cancer. As there is increasing data on how phytochemicals interfere with diverse molecular pathways in prostate cancer, this review discusses and emphasizes the implicated molecular pathways of cell proliferation, cell cycle control, apoptosis, and autophagy as important processes that control tumor angiogenesis, invasion, and metastasis. In conclusion, the elucidation of the natural compounds' chemical structure-based anti-cancer mechanisms will facilitate drug development and the optimization of drug combinations. Phytochemicals, as anti-cancer agents in the treatment of prostate cancer, can have significant health benefits for humans.
Collapse
Affiliation(s)
- Qiongyu Hao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Yanyuan Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Piwen Wang
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| |
Collapse
|
5
|
Liu Y, Yu S, Xing X, Qiao J, Yin Y, Wang J, Liu M, Zhang W. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex. Mol Med Rep 2021; 24:873. [PMID: 34713297 PMCID: PMC8569524 DOI: 10.3892/mmr.2021.12513] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside Rh2 (G-Rh2) is a monomeric compound that extracted from ginseng and possesses anti-cancer activities both in vitro and in vivo. Previously, we reported that G-Rh2 induces apoptosis in HeLa cervical cancer cells and that the process was related to reactive oxygen species (ROS) accumulation and mitochondrial dysfunction. However, the upstream mechanisms of G-Rh2, along with its cellular targets, remain to be elucidated. In the present study, the Cell Counting Kit-8 assay, flow cytometry and Hoechst staining revealed that G-Rh2 significantly inhibited cell viability and induced apoptosis of cervical cancer cells. However, G-Rh2 was demonstrated to be non-toxic to End1/e6e7 cells. JC-1, rhodamine 123 staining, oxidative phosphorylation and glycolysis capacity assays demonstrated that G-Rh2 exposure caused an immediate decrease in mitochondrial transmembrane potential due to its inhibition of mitochondrial oxidative phosphorylation, as well as glycolysis, both of which reduced cellular ATP production. Western blotting and electron transport chain (ETC) activity assays revealed that G-Rh2 significantly inhibited the activity of ETC complexes I, III and V. Overexpression of ETC complex III partially significantly restored mitochondrial ROS and inhibited the apoptosis of cervical cancer cells induced by G-Rh2. The predicted results of binding energy in molecular docking, confirmed that G-Rh2 was highly likely to induce mitochondrial ROS production and promote cell apoptosis by targeting the ETC complex, especially for ETC complex III. Taken together, the present results revealed the potential anti-cervical cancer activity of G-Rh2 and provide direct evidence for the contribution of impaired ETC complex activity to cervical cancer cell death.
Collapse
Affiliation(s)
- Ying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Shiting Yu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xin Xing
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Juhui Qiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yiqiu Yin
- GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin 130012, P.R. China
| | - Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Wei Zhang
- Scientific Research Department, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
6
|
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: 0.8] [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
|
7
|
Li X, Chu S, Lin M, Gao Y, Liu Y, Yang S, Zhou X, Zhang Y, Hu Y, Wang H, Chen N. Anticancer property of ginsenoside Rh2 from ginseng. Eur J Med Chem 2020; 203:112627. [PMID: 32702586 DOI: 10.1016/j.ejmech.2020.112627] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
Ginseng has been used as a well-known traditional Chinese medicine since ancient times. Ginsenosides as its main active constituents possess a broad scope of pharmacological properties including stimulating immune function, enhancing cardiovascular health, increasing resistance to stress, improving memory and learning, developing social functioning and mental health in normal persons, and chemotherapy. Ginsenoside Rh2 (Rh2) is one of the major bioactive ginsenosides from Panax ginseng. When applied to cancer treatment, Rh2 not only exhibits the anti-proliferation, anti-invasion, anti-metastasis, induction of cell cycle arrest, promotion of differentiation, and reversal of multi-drug resistance activities against multiple tumor cells, but also alleviates the side effects after chemotherapy or radiotherapy. In the past decades, nearly 200 studies on Rh2 in the treatment of cancer have been published, however no specific reviews have been conducted by now. So the purpose of this review is to provide a systematic summary and analysis of the anticancer effects and the potential mechanisms of Rh2 extracted from Ginseng then give a future prospects about it. In the end of this paper the metabolism and derivatives of Rh2 also have been documented.
Collapse
Affiliation(s)
- Xun Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Shifeng Chu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Meiyu Lin
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Yan Gao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yingjiao Liu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Xin Zhou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yani Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Yaomei Hu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Huiqin Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Naihong Chen
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
| |
Collapse
|
8
|
Mathiyalagan R, Wang C, Kim YJ, Castro-Aceituno V, Ahn S, Subramaniyam S, Simu SY, Jiménez-Pérez ZE, Yang DC, Jung SK. Preparation of Polyethylene Glycol-Ginsenoside Rh1 and Rh2 Conjugates and Their Efficacy against Lung Cancer and Inflammation. Molecules 2019; 24:E4367. [PMID: 31795352 PMCID: PMC6930446 DOI: 10.3390/molecules24234367] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Low solubility and tumor-targeted delivery of ginsenosides to avoid off-target cytotoxicity are challenges for clinical trials. In the present study, we report on a methodology for the synthesis of polyethylene glycol (PEG)-ginsenoside conjugates through a hydrolysable ester bond using the hydrophilic polymer polyethylene glycol with the hydrophobic ginsenosides Rh1 and Rh2 to enhance water solubility and passive targeted delivery. The resulting conjugates were characterized by 1H nuclear magnetic resonance (1H NMR) and Fourier-transform infrared spectroscopy (FT-IR). 1H NMR revealed that the C-6 and C-3 sugar hydroxyl groups of Rh1 and Rh2 were esterified. The conjugates showed spherical shapes that were monitored by field-emission transmission electron microscopy (FE-TEM), and the average sizes of the particles were 62 ± 5.72 nm and 134 ± 8.75 nm for PEG-Rh1and PEG-Rh2, respectively (measured using a particle size analyzer). Owing to the hydrophilic enhancing properties of PEG, PEG-Rh1 and PEG-Rh2 solubility was greatly enhanced compared to Rh1 and Rh2 alone. The release rates of Rh1 and Rh2 were increased in lower pH conditions (pH 5.0), that for pathophysiological sites as well as for intracellular endosomes and lysosomes, compared to normal-cell pH conditions (pH 7.4). In vitro cytotoxicity assays showed that the PEG-Rh1conjugates had greater anticancer activity in a human non-small cell lung cancer cell line (A549) compared to Rh1 alone, whereas PEG-Rh2 showed lower cytotoxicity in lung cancer cells. On the other hand, both PEG-Rh1 and PEG-Rh2 showed non-cytotoxicity in a nondiseased murine macrophage cell line (RAW 264.7) compared to free Rh1 and Rh2, but PEG-Rh2 exhibited increased efficacy against inflammation by greatly inhibiting nitric oxide production. Thus, the overall conclusion of our study is that PEG conjugation promotes the properties of Rh1 for anticancer and Rh2 for inflammation treatments. Depends on the disease models, they could be potential drug candidates for further studies.
Collapse
Affiliation(s)
- Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (R.M.); (S.Y.S.); (Z.E.J.-P.)
| | - Chao Wang
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo 255000, Shandong, China
| | - Yeon Ju Kim
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
| | - Verónica Castro-Aceituno
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
| | - Sungeun Ahn
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
| | - Sathiyamoorthy Subramaniyam
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
- Department of Biotechnology, Dr.N.G.P., Arts and Science College, Coimbatore 641048, Tamil Nadu, India
| | - Shakina Yesmin Simu
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (R.M.); (S.Y.S.); (Z.E.J.-P.)
| | - Zuly Elizabeth Jiménez-Pérez
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (R.M.); (S.Y.S.); (Z.E.J.-P.)
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (R.M.); (S.Y.S.); (Z.E.J.-P.)
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
| | - Seok-Kyu Jung
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea; (C.W.); (Y.J.K.); (V.C.-A.); (S.A.); (S.S.)
| |
Collapse
|
9
|
Zhao N, Cheng M, Huang S, Liu D, Zhao Q, Bai Y, Zhang X. Various Multicharged Anions of Ginsenosides in Negative Electrospray Ionization with QTOF High-Resolution Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:403-418. [PMID: 30644055 DOI: 10.1007/s13361-018-2089-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/06/2018] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
When characterizing components from ginseng, we found a vast number of multicharged anions presented in the liquid chromatography-mass spectrometry (LC-MS) chromatograms. The source of these anions is unclear yet, while ginsenosides, the major components of ginseng, are the main suspected type of molecules because of their sugar moiety. Our investigation using 14 pure ginsenosides affirmed that the multicharged anions were formed by ginsenosides rather than other types of ingredients in ginseng. Various anions could be observed for each ginsenoside. These anions contain ions ([M-2H]2-, [M+Adduct]2-), as well as those formed by polymerization of at least two ginsenosides, such as [nM-2H]2-, [nM-H+Adduct]2-, and [nM-3H]3-. The presence of so different types of ions from a ginsenoside explains the reason for the large number of anions in the LC-MS analysis of ginseng. We further found that formation of [nM-2H]2- ions was influenced by the number of sugar chains: ginsenosides containing two sugar chains produced all [nM-2H]2- ion types, whereas ginsenosides containing one sugar chain did not produce [2M-2H]2-. Thus, [2M-2H]2- and [3M-2H]2- can be utilized to rapidly identify monodesmosidic and/or bidesmosidic ginsenosides as joint diagnostic anions. The position of the glycosyl radical might be the key factor affecting the formation of multicharged multimer ions from monodesmosidic ginsenosides. Consequently, three groups of ginsenoside isomers were differentiated by characteristic [nM-2H]2- anions. Using concentration-dependent characteristics and collision-induced dissociation (CID), we confirmed that [nM-2H]2- ions are non-covalently bound multimers whose aggregation has marked distinction between monodesmosidic and bidesmosidic ginsenosides, accounting for the differentiated formation of [nM-2H]2- between them. Graphical Abstract.
Collapse
Affiliation(s)
- Nan Zhao
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
- University of Chinese Academy of Sciences, Yuquan Road 19, 100049, Beijing, People's Republic of China
| | - Mengchun Cheng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
| | - Shuai Huang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
| | - Dan Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
| | - Qiang Zhao
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Yunpeng Bai
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China
| | - Xiaozhe Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, 116023, Dalian, People's Republic of China.
| |
Collapse
|
10
|
Pharmacokinetic interaction of calcitriol with 20(S)-protopanaxadiol in mice: Determined by LC/MS analysis. Eur J Pharm Sci 2019; 130:173-180. [PMID: 30654110 DOI: 10.1016/j.ejps.2019.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/23/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
Abstract
The physiological and anti-cancer functions of vitamin D3 are accomplished primarily via 1α,25-dihydroxyvitamin D3 (calcitriol), whereas 20(S)-protopanaxadiol (aPPD) is a ginsenoside, which is isolated from Panax ginseng, with potential anti-cancer benefits. In the present study, we report a pharmacokinetic (PK) herb-nutrient interaction between calcitriol and aPPD in mice. A liquid chromatography mass spectrometry (LC/MS) method was developed using 4-phenyl-1,2,4-triazoline-3,5-dione derivatizing agent and we subsequently used the method to quantitate calcitriol in mouse serum. The limit of quantitation was 0.01 ng/ml which is approximately 100 fold lower than the previously reported assay from our laboratory. Calcitriol PK parameters were determined in non-tumor-bearing or C4-2 human prostate tumor-bearing nude mice following oral co-administration of calcitriol either alone or in combination with aPPD. Mice were pretreated with oral aPPD (70 mg/kg) or vehicle control twice daily for seven consecutive days, followed by a single oral dose of 4 μg/kg calcitriol alone or in combination with aPPD. Our PK results demonstrated that co-administration of calcitriol with aPPD (following pre-treatment with vehicle for seven days) resulted in a 35% increase in the area under the curve (AUC0-24 h) and a 41% increase in the maximum serum concentration (Cmax) compared to the calcitriol only group. aPPD therefore significantly increased calcitriol serum exposure. We also saw a reduction in the time required to reach Cmax. In contrast, calcitriol PK in mice co-administered with calcitriol and aPPD as well as those pretreated seven consecutive days with aPPD was no different than that determined for the mice that received vehicle for seven days as pre-treatment. Co-administration of calcitriol with aPPD therefore could increase health benefits of vitamin D3, however any increased risk of hypercalcemia, resulting from this combination approach, requires further investigation. Lastly, we surmise that a cytochrome P450 inhibition-based mechanism may contribute to the observed PK interaction.
Collapse
|
11
|
20(S)-protopanaxadiol regio-selectively targets androgen receptor: anticancer effects in castration-resistant prostate tumors. Oncotarget 2018; 9:20965-20978. [PMID: 29765513 PMCID: PMC5940378 DOI: 10.18632/oncotarget.24695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/24/2018] [Indexed: 01/21/2023] Open
Abstract
We have explored the effects of 20(S)-protopanaxadiol (aPPD), a naturally derived ginsenoside, against androgen receptor (AR) positive castration resistant prostate cancer (CRPC) xenograft tumors and have examined its interactions with AR. In silico docking studies for aPPD binding to AR, alongside transactivation bioassays and in vivo efficacy studies were carried out in the castration-resistant C4-2 xenograft model. Immunohistochemical (IHC) and Western blot analyses followed by evaluation of AR, apoptotic, cell cycle and proliferative markers in excised tumors was performed. The growth of established CRPC tumors was inhibited by 53% with aPPD and a corresponding decrease in serum PSA was seen compared to controls. The IHC data revealed that Ki-67 was significantly lower for aPPD treated tumors and was associated with elevated p21 and cleaved caspase-3 expression, compared to vehicle treatment. Furthermore, aPPD decreased AR protein expression in xenograft tumors, while significantly upregulating p27 and Bax protein levels. In vitro data supporting this suggests that aPPD binds to and significantly inhibits the N-terminal or the DNA binding domains of AR. The AR androgen binding site docking score for androgen (dihydrotestosterone) was −11.1, while that of aPPD was −7.1. The novel findings described herein indicate aPPD potently inhibits PCa in vivo partly via inhibition of a site on the AR N-terminal domain. This manifested as cell cycle arrest and concurrent induction of apoptosis via an increase in Bax, cleaved-caspase-3, p27 and p21 expression.
Collapse
|
12
|
Lo SH, Hsu CT, Niu HS, Niu CS, Cheng JT, Chen ZC. Ginsenoside Rh2 Improves Cardiac Fibrosis via PPARδ-STAT3 Signaling in Type 1-Like Diabetic Rats. Int J Mol Sci 2017; 18:ijms18071364. [PMID: 28672855 PMCID: PMC5535857 DOI: 10.3390/ijms18071364] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Accepted: 06/22/2017] [Indexed: 02/08/2023] Open
Abstract
Ginsenoside Rh2 (Rh2) is an active principal ingredient contained in ginseng (Panax ginseng Meyer), a medicinal herb used to enhance health worldwide. The present study is designed to investigate the effect of Rh2 on myocardial fibrosis in diabetic rats. In a streptozotocin-induced model of type-1 diabetic rats (STZ-diabetic rats), the increased fasting blood glucose levels and heart weight/body weight (HW/BW) ratio were substantially alleviated by Rh2. Moreover, Rh2 improved cardiac performance in STZ-diabetic rats. Histological results from Masson staining showed that Rh2 attenuated cardiac fibrosis in STZ-diabetic rats. The effects of Rh2 were reversed by GSK0660 at a dose sufficient to inhibit peroxisome proliferator-activated receptor δ (PPARδ) in STZ-diabetic rats. The role of PPARδ was subsequently investigated in vitro. Rh2 restored the decreased PPARδ expression level in high glucose-cultured cardiomyocytes. Moreover, increased protein levels of fibrotic signals, including signal transducer and activator of transcription 3 (STAT3), connective tissue growth factor (CCN2) and fibronectin, were reduced by Rh2 in high glucose-cultured cardiomyocytes. These effects of Rh2 were reversed by GSK0660 or siRNA specific for PPARδ Taken together, PPARδ activation may inhibit STAT3 activation to reduce CCN2 and fibronectin expression in diabetic rats with cardiac fibrosis. Moreover, Rh2 improves cardiac function and fibrosis by increasing PPARδ signaling. Therefore, Rh2 is suitable to develop as an alternative remedy for cardiac fibrosis.
Collapse
Affiliation(s)
- Shih-Hsiang Lo
- Division of Cardiology, Department of Internal Medicine, Zhongxing Branch of Taipei City Hospital, Taipei 10341, Taiwan.
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien 97041, Taiwan.
| | - Chao-Tien Hsu
- Department of Pathology, E-DA Hospital, I-Shou University, Yanchao, Kaohsiung 82401, Taiwan.
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien 97041, Taiwan.
| | - Chiang-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien 97041, Taiwan.
| | - Juei-Tang Cheng
- Department of Cardiology and Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan 71003, Taiwan.
- Institute of Medical Sciences, Chang Jung Christian University, Guiren, Tainan 71101, Taiwan.
| | - Zhih-Cherng Chen
- Department of Cardiology and Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan 71003, Taiwan.
- Department of Pharmacy, Chia Nan University of Pharmacy & Science, Jean-Tae 71701, Taiwan.
| |
Collapse
|
13
|
Chen F, Sun Y, Zheng SL, Qin Y, Julian McClements D, Hu JN, Deng ZY. Antitumor and immunomodulatory effects of ginsenoside Rh2 and its octyl ester derivative in H22 tumor-bearing mice. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.03.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
14
|
Siddiqi MZ, Cui CH, Park SK, Han NS, Kim SC, Im WT. Comparative analysis of the expression level of recombinant ginsenoside-transforming β-glucosidase in GRAS hosts and mass production of the ginsenoside Rh2-Mix. PLoS One 2017; 12:e0176098. [PMID: 28423055 PMCID: PMC5396970 DOI: 10.1371/journal.pone.0176098] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/05/2017] [Indexed: 01/29/2023] Open
Abstract
The ginsenoside Rh2, a pharmaceutically active component of ginseng, is known to have anticancer and antitumor effects. However, white ginseng and red ginseng have extremely low concentrations of Rh2 or Rh2-Mix [20(S)-Rh2, 20(R)-Rh2, Rk2, and Rh3]. To enhance the production of food-grade ginsenoside Rh2, an edible enzymatic bioconversion technique was developed adopting GRAS host strains. A β-glucosidase (BglPm), which has ginsenoside conversion ability, was expressed in three GRAS host strains (Corynebacterium glutamicum, Saccharomyces cerevisiae and Lactococus lactis) by using a different vector system. Enzyme activity in these three GRAS hosts were 75.4%, 11.5%, and 9.3%, respectively, compared to that in the E. coli pGEX 4T-1 expression system. The highly expressed BglPm_C in C. glutamicum can effectively transform the ginsenoside Rg3-Mix [20(S)-Rg3, 20(R)-Rg3, Rk1, Rg5] to Rh2-Mix [20(S)-Rh2, 20(R)-Rh2, Rk2, Rh3] using a scaled-up biotransformation reaction, which was performed in a 10-L jar fermenter at pH 6.5/7.0 and 37°C for 24 h. To our knowledge, this is the first report in which 50 g of PPD-Mix (Rb1, Rb2, Rb3, Rc, and Rd) as a starting substrate was converted to ginsenoside Rg3-Mix by acid heat treatment and then 24.5-g Rh2-Mix was obtained by enzymatic transformation of Rg3-Mix through by BglPm_C. Utilization of this enzymatic method adopting a GRAS host could be usefully exploited in the preparation of ginsenoside Rh2-Mix in cosmetics, functional food, and pharmaceutical industries, thereby replacing the E. coli expression system.
Collapse
Affiliation(s)
- Muhammad Zubair Siddiqi
- Department of Biotechnology, Hankyoung National University, Kyonggi-do, Republic of Korea
- Center for Genetic Information, Graduate School of Bio and Information Technology, Hankyoung National University, Kyonggi-do, Republic of Korea
| | - Chang-Hao Cui
- Intelligent Synthetic Biology Center, Yuseong-gu, Daejeon, Republic of Korea
| | - Seul-Ki Park
- Intelligent Synthetic Biology Center, Yuseong-gu, Daejeon, Republic of Korea
| | - Nam Soo Han
- Brain Korea 21 Center for Bio-Resource Development, Division of Animal, Horticultural and Food Sciences, Chungbuk National University, Cheongju, Korea
| | - Sun-Chang Kim
- Intelligent Synthetic Biology Center, Yuseong-gu, Daejeon, Republic of Korea
| | - Wan-Taek Im
- Department of Biotechnology, Hankyoung National University, Kyonggi-do, Republic of Korea
- Center for Genetic Information, Graduate School of Bio and Information Technology, Hankyoung National University, Kyonggi-do, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Xu XH, Li T, Fong CMV, Chen X, Chen XJ, Wang YT, Huang MQ, Lu JJ. Saponins from Chinese Medicines as Anticancer Agents. Molecules 2016; 21:molecules21101326. [PMID: 27782048 PMCID: PMC6272920 DOI: 10.3390/molecules21101326] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022] Open
Abstract
Saponins are glycosides with triterpenoid or spirostane aglycones that demonstrate various pharmacological effects against mammalian diseases. To promote the research and development of anticancer agents from saponins, this review focuses on the anticancer properties of several typical naturally derived triterpenoid saponins (ginsenosides and saikosaponins) and steroid saponins (dioscin, polyphyllin, and timosaponin) isolated from Chinese medicines. These saponins exhibit in vitro and in vivo anticancer effects, such as anti-proliferation, anti-metastasis, anti-angiogenesis, anti-multidrug resistance, and autophagy regulation actions. In addition, related signaling pathways and target proteins involved in the anticancer effects of saponins are also summarized in this work.
Collapse
Affiliation(s)
- Xiao-Huang Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Chi Man Vivienne Fong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Xiao-Jia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Ming-Qing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
16
|
Ben-Eltriki M, Deb S, Adomat H, Tomlinson Guns ES. Calcitriol and 20(S)-protopanaxadiol synergistically inhibit growth and induce apoptosis in human prostate cancer cells. J Steroid Biochem Mol Biol 2016; 158:207-219. [PMID: 26709138 DOI: 10.1016/j.jsbmb.2015.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
The potential cancer preventive roles of calcitriol, the dihydroxylated metabolite of Vitamin D3, as well as 20(S)-protopanaxadiol (aPPD), the aglycone of the protopanaxadiol family of ginsenosides, have gained much attention in recent years for the prevention/treatment of prostate cancer (PCa). In the present study, we evaluated the anticancer and chemosensitization effects of calcitriol at clinically relevant concentrations and aPPD, either alone or in combination, in two well-characterized human PCa cell lines: androgen-sensitive non-metastatic LNCaP cells and androgen-independent metastatic C4-2 cells. The effects of the treatments on PCa cell viability and proliferation rates were evaluated by MTS and Brdu assays, respectively. Combination Indices (CI) and Dose Reduction Indices (DRI) were estimated to assess synergistic anticancer activity using Calcusyn software (Biosoft, Cambridge, UK). Then, we determined the potential Pharmacodynamic interaction mechanisms as follows: The protein expression levels of the genes those are known to control cell cycle (cyclin D1 and cdk2); apoptosis (Bcl-2, Bax, and Capspases 3), androgen receptor and Vitamin D receptors were examined upon combinational treatment. The cell viability assay data show that addition of 10nM calcitriol to aPPD significantly lowered its IC50 values from the range of 41-53μM to 13-23μM, in LNCaP and C4-2 prostate cancer cells. The cell proliferation rate was significantly lower for combination treatments compared to the cells treated with aPPD alone. Similarly, Western blot results indicate that aPPD significantly upregulated Vitamin D receptor (VDR) expression, while calcitriol further enhanced the ability of aPPD to induce pro-apoptotic BAX, increased cleaved caspase-3 and downregulate cdk2 protein levels. Thus, the pharmacodynamic interaction between aPPD and calcitriol in impacting growth inhibition and apoptosis appears to be synergistic in nature. In conclusion, calcitriol sensitizes PCa cells to aPPD-mediated anticancer effects by enhancing its ability to induce apoptosis and reduce cell proliferation, and this synergism may limit calcitriol toxicity by facilitating the use of lower calcitriol doses. The associated increase in VDR expression and calcitriol half-life may be mechanistically associated with this sensitization effect.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; Department of Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Subrata Deb
- Department of Biopharmaceutical Sciences, College of Pharmacy at Roosevelt University, Schaumburg, IL, USA
| | - Hans Adomat
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
17
|
Wong AST, Che CM, Leung KW. Recent advances in ginseng as cancer therapeutics: a functional and mechanistic overview. Nat Prod Rep 2015; 32:256-72. [PMID: 25347695 DOI: 10.1039/c4np00080c] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Ginseng, a key ingredient in traditional Chinese medicine, shows great promise as a new treatment option. As listed by the U.S. National Institutes of Health as a complementary and alternative medicine, its anti-cancer functions are being increasingly recognized. This review covers the mechanisms of action of ginsenosides and their metabolites, which can modulate signaling pathways associated with inflammation, oxidative stress, angiogenesis, metastasis, and stem/progenitor-like properties of cancer cells. The emerging use of structurally modified ginsenosides and recent clinical studies on the use of ginseng either alone or in combination with other herbs or Western medicines which are exploited as novel therapeutic strategies will also be explored.
Collapse
Affiliation(s)
- Alice S T Wong
- State Key Laboratory of Oncogenes and Related Genes, and School of Biological Sciences, The University of Hong Kong, Hong Kong.
| | | | | |
Collapse
|
18
|
Deb S, Chin MY, Adomat H, Guns EST. Ginsenoside-mediated blockade of 1α,25-dihydroxyvitamin D3 inactivation in human liver and intestine in vitro. J Steroid Biochem Mol Biol 2014; 141:94-103. [PMID: 24486455 DOI: 10.1016/j.jsbmb.2014.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/21/2013] [Accepted: 01/22/2014] [Indexed: 12/19/2022]
Abstract
The beneficial effects of vitamin D3 are exerted through 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3], the dihydroxy metabolite of vitamin D3. Hepatic and intestinal biotransformation of 1α,25(OH)2D3 and modifiers of metabolic capacity could be important determinants of bioavailability in serum and tissues. Ginsenosides and their aglycones, mainly 20(S)-protopanaxadiol (aPPD) and 20(S)-protopanaxatriol (aPPT), are routinely ingested as health supplements. The purpose of the present study was to investigate the potential of ginsenosides and their aglycones to block hepatic and intestinal inactivation of 1α,25(OH)2D3, which is the most potent ligand of vitamin D receptor. In vitro biotransformation reactions were initiated with NADPH regenerating solutions following initial preincubation of pooled human hepatic or intestinal microsomal protein or human recombinant CYP3A4 supersomes with 1α,25(OH)2D3 or midazolam. Formation of hydroxylated metabolites of 1α,25(OH)2D3 or midazolam was analyzed using liquid chromatography-mass spectrometry. Co-incubation of 1α,25(OH)2D3 with various ginsenosides (Rg1, Rh2, aPPD, aPPT and total ginsenosides) led to differential inhibition (30-100%) of its hydroxylation. Results suggest that aPPD, aPPT and Rh2 strongly attenuated the hydroxylation of 1α,25(OH)2D3. Follow up inhibition studies with aPPD and aPPT at varying concentrations (0.5-100μM) led to up to 91-100% inhibition of formation of hydroxylated metabolites of 1α,25(OH)2D3 thus preventing inactivation of active vitamin D3. The IC50 values of aPPD or aPPT for the most abundant hydroxylated metabolites of 1α,25(OH)2D3 ranged from 3.3 to 9.0μM in human microsomes. The inhibitory mechanism of aPPD or aPPT for CYP3A4-mediated biotransformation of 1α,25(OH)2D3 was competitive in nature (apparent Ki: 1.7-2.9μM). Similar inhibitory effects were also observed upon addition of aPPD or aPPT into midazolam hydroxylation assay. In summary, our results suggest that ginsenosides, specifically aPPD and aPPT, inhibit the CYP3A4-mediated catabolism of active vitamin D3 in human liver and intestine, potentially providing additional vitamin D-related benefits to patients with cancer, neurodegenerative and metabolic diseases.
Collapse
Affiliation(s)
- Subrata Deb
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, Canada V6H 3Z6
| | - Mei Yieng Chin
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, Canada V6H 3Z6
| | - Hans Adomat
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, Canada V6H 3Z6
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, Canada V6H 3Z6.
| |
Collapse
|
19
|
TAIRA S, UEMATSU K, KANEKO D, KATANO H. Mass Spectrometry Imaging: Applications to Food Science. ANAL SCI 2014; 30:197-203. [DOI: 10.2116/analsci.30.197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Shu TAIRA
- Department of Bioscience, Fukui Prefectural University
| | - Kohei UEMATSU
- Department of Bioscience, Fukui Prefectural University
| | - Daisaku KANEKO
- Frontier Research Academy for Young Researchers, Department of Applied Chemistry, Faculty of Engineering, Kyushu Institute of Technology
| | - Hajime KATANO
- Department of Bioscience, Fukui Prefectural University
| |
Collapse
|
20
|
Chiu NTC, Guns EST, Adomat H, Jia W, Deb S. Identification of human cytochrome P450 enzymes involved in the hepatic and intestinal biotransformation of 20(S)-protopanaxadiol. Biopharm Drug Dispos 2013; 35:104-18. [DOI: 10.1002/bdd.1873] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/10/2013] [Accepted: 10/09/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Nga Ting Colette Chiu
- Department of Pathology and Laboratory Science; University of British Columbia; Vancouver BC Canada
| | - Emma S. Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital; 2660 Oak Street Vancouver BC Canada V6H 3Z6
| | - Hans Adomat
- The Vancouver Prostate Centre at Vancouver General Hospital; 2660 Oak Street Vancouver BC Canada V6H 3Z6
| | - William Jia
- Department of Surgery; University of British Columbia; Vancouver BC Canada
| | - Subrata Deb
- The Vancouver Prostate Centre at Vancouver General Hospital; 2660 Oak Street Vancouver BC Canada V6H 3Z6
| |
Collapse
|
21
|
Choi JS, Chun KS, Kundu J, Kundu JK. Biochemical basis of cancer chemoprevention and/or chemotherapy with ginsenosides (Review). Int J Mol Med 2013; 32:1227-38. [PMID: 24126942 DOI: 10.3892/ijmm.2013.1519] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/26/2013] [Indexed: 11/06/2022] Open
Abstract
Cancer still imposes a global threat to public health. After decades of research on cancer biology and enormous efforts in developing anticancer therapies, we now understand that the majority of cancers can be prevented. Bioactive phytochemicals present in edible plants have been shown to reduce the risk of various types of cancer. Ginseng (Panax ginseng C.A. Meyer), which contains a wide variety of saponins, known as ginsenosides, is an age-old remedy for human ailments, including cancer. Numerous laboratory-based studies have revealed the anticancer properties of ginsenosides, which compel tumor cells to commit suicide, arrest the proliferation of cancer cells in culture and inhibit experimentally-induced tumor formation in laboratory animals. Ginsenosides have been reported to inhibit tumor angiogenesis, as well as the invasion and metastasis of various types of cancer cells. Moreover, ginsenosides as combination therapy enhance the sensitivity of chemoresistant tumors to clinically used chemotherapeutic agents. This review sheds light on the molecular mechanisms underlying the cancer chemopreventive and/or chemotherapeutic activity of ginsenosides and their intestinal metabolites with particular focus on the modulation of cell signaling pathways associated with oxidative stress, inflammation, cell proliferation, apoptosis, angiogenesis and the metastasis of cancer cells.
Collapse
Affiliation(s)
- Joon-Seok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | |
Collapse
|
22
|
Koczurkiewicz P, Podolak I, Skrzeczyńska-Moncznik J, Sarna M, Wójcik KA, Ryszawy D, Galanty A, Lasota S, Madeja Z, Czyż J, Michalik M. Triterpene saponosides from Lysimachia ciliata differentially attenuate invasive potential of prostate cancer cells. Chem Biol Interact 2013; 206:6-17. [PMID: 23954719 DOI: 10.1016/j.cbi.2013.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 07/25/2013] [Accepted: 08/05/2013] [Indexed: 01/06/2023]
Abstract
Neither androgen ablation nor chemotherapeutic agents are effective in reducing the risk of prostate cancer progression. On the other hand, multifaceted effects of phytochemicals, such as triterpene saponins, on cancer cells have been suggested. A promising safety and tolerability profile indicate their possible application in the treatment of advanced prostate cancers. We analyzed the specificity, selectivity and versatility of desglucoanagalloside B effects on human prostate cancer cells derived from prostate cancer metastases to brain (DU-145 cells) and bone (PC-3 cells). Prominent growth arrest and apoptotic response of both cell types was observed in the presence of sub-micromolar desglucoanagalloside B concentrations. This was accompanied by cytochrome c release and caspase 3/7 activation. A relatively low cytostatic and pro-apoptotic response of cancer cells to a desglucoanagalloside B analog, anagallosaponin IV, illustrated the specificity of the effects of desglucoanagalloside B, whereas the low sensitivity of normal prostate PNT2 cells to desglucoanagalloside B showed the selectivity of its action. Inhibition of cancer cell motility was observed in the presence of both saponins, however only desglucoanagalloside B attenuated cancer cell invasive potential, predominantly through an effect on cell elastic properties. These data demonstrate the versatility of its effects on prostate cancer cells. In contrast to PNT2 cells, cancer cells tested in this study were relatively resistant to mitoxantrone. The multifaceted action of desglucoanagalloside B on basic cellular traits, crucial for prostate cancer progression, opens perspectives for elaboration of combined palliative therapies and new prostate cancer prophylaxis regimens.
Collapse
Affiliation(s)
- Paulina Koczurkiewicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; Department of Pharmacognosy, Pharmaceutical Faculty, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kornienko A, Mathieu V, Rastogi SK, Lefranc F, Kiss R. Therapeutic Agents Triggering Nonapoptotic Cancer Cell Death. J Med Chem 2013; 56:4823-39. [DOI: 10.1021/jm400136m] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University—San Marcos, San Marcos, Texas 78666,
United States
| | - Véronique Mathieu
- Laboratoire
de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, CP205/1,
Boulevard du Triomphe, Brussels, Belgium
| | - Shiva K. Rastogi
- Department of Chemistry and Biochemistry, Texas State University—San Marcos, San Marcos, Texas 78666,
United States
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, ULB, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Robert Kiss
- Laboratoire
de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, CP205/1,
Boulevard du Triomphe, Brussels, Belgium
| |
Collapse
|
24
|
Augustin JM, Drok S, Shinoda T, Sanmiya K, Nielsen JK, Khakimov B, Olsen CE, Hansen EH, Kuzina V, Ekstrøm CT, Hauser T, Bak S. UDP-glycosyltransferases from the UGT73C subfamily in Barbarea vulgaris catalyze sapogenin 3-O-glucosylation in saponin-mediated insect resistance. PLANT PHYSIOLOGY 2012; 160:1881-95. [PMID: 23027665 PMCID: PMC3510118 DOI: 10.1104/pp.112.202747] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/30/2012] [Indexed: 05/18/2023]
Abstract
Triterpenoid saponins are bioactive metabolites that have evolved recurrently in plants, presumably for defense. Their biosynthesis is poorly understood, as is the relationship between bioactivity and structure. Barbarea vulgaris is the only crucifer known to produce saponins. Hederagenin and oleanolic acid cellobioside make some B. vulgaris plants resistant to important insect pests, while other, susceptible plants produce different saponins. Resistance could be caused by glucosylation of the sapogenins. We identified four family 1 glycosyltransferases (UGTs) that catalyze 3-O-glucosylation of the sapogenins oleanolic acid and hederagenin. Among these, UGT73C10 and UGT73C11 show highest activity, substrate specificity and regiospecificity, and are under positive selection, while UGT73C12 and UGT73C13 show lower substrate specificity and regiospecificity and are under purifying selection. The expression of UGT73C10 and UGT73C11 in different B. vulgaris organs correlates with saponin abundance. Monoglucosylated hederagenin and oleanolic acid were produced in vitro and tested for effects on P. nemorum. 3-O-β-d-Glc hederagenin strongly deterred feeding, while 3-O-β-d-Glc oleanolic acid only had a minor effect, showing that hydroxylation of C23 is important for resistance to this herbivore. The closest homolog in Arabidopsis thaliana, UGT73C5, only showed weak activity toward sapogenins. This indicates that UGT73C10 and UGT73C11 have neofunctionalized to specifically glucosylate sapogenins at the C3 position and demonstrates that C3 monoglucosylation activates resistance. As the UGTs from both the resistant and susceptible types of B. vulgaris glucosylate sapogenins and are not located in the known quantitative trait loci for resistance, the difference between the susceptible and resistant plant types is determined at an earlier stage in saponin biosynthesis.
Collapse
|
25
|
A novel oral dosage formulation of the ginsenoside aglycone protopanaxadiol exhibits therapeutic activity against a hormone-insensitive model of prostate cancer. Anticancer Drugs 2012; 23:543-52. [PMID: 22481061 DOI: 10.1097/cad.0b013e32835006f5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study focuses on determining the pharmacokinetics, biodistribution, and efficacy of the ginsenoside aglycone protopanaxadiol (aPPD) administered as a single agent in a novel oral dosage formulation. To obtain these data and to characterize the stability of aPPD, appropriate analytical assay development was carried out. The solubility and stability of aPPD were determined, and the compound was formulated for oral gavage. aPPD levels in blood and tissues following oral administration to nu/nu nude mice were determined using liquid chromatography-mass spectrometry/mass spectrometry. The efficacy of aPPD was determined upon oral administration to nu/nu nude mice bearing PC-3 human prostate cancer xenograft tumors. Immunohistochemical analysis of tumor tissues was performed to establish apoptotic indices and Ki-67 expression as markers of proliferation. The maximum solubility of aPPD in ethanol was 68.4 mg/ml. aPPD administered at a dose of 70 mg/kg yielded a T(max) of approximately 40 min and a C(max) value of 3.9 ± 1.4 μg/ml, and no toxicity was observed. aPPD accumulated largely in the stomach and small intestine and was also present in the brain. This dose engendered a significant delay in PC-3 tumor growth, an increase in apoptotic index, and a decrease in Ki-67 levels. We have shown that aPPD is a stable compound that can be formulated for oral gavage. Pharmacokinetic studies demonstrate the ability of this compound to be absorbed after oral administration. Future studies will assess the activity and pharmacokinetics of aPPD when administered in combination with standard chemotherapy.
Collapse
|
26
|
Yang Z, Wang JR, Niu T, Gao S, Yin T, You M, Jiang ZH, Hu M. Inhibition of P-glycoprotein leads to improved oral bioavailability of compound K, an anticancer metabolite of red ginseng extract produced by gut microflora. Drug Metab Dispos 2012; 40:1538-44. [PMID: 22584255 PMCID: PMC3400789 DOI: 10.1124/dmd.111.044008] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/10/2012] [Indexed: 11/22/2022] Open
Abstract
Ginsenosides are hydrolyzed extensively by gut microflora after oral administration, and their metabolites are pharmacologically active against lung cancer cells. In this study, we measured the metabolism of various ginsenosides by gut microflora and determined the mechanisms responsible for the observed pharmacokinetic behaviors of its active metabolite, Compound K (C-K). The results showed that biotransformation into C-K is the major metabolic pathway of ginsenosides after the oral administration of the red ginseng extract containing both protopanaxadiol and protopanaxatriol ginsenosides. Pharmacokinetic studies in normal mice showed that C-K exhibited low oral bioavailability. To define the mechanisms responsible for this low bioavailability, two P-glycoprotein (P-gp) inhibitors, verapamil and cyclosporine A, were used, and their presence substantially decreased C-K's efflux ratio in Caco-2 cells (from 26.6 to <3) and significantly increased intracellular concentrations (by as much as 40-fold). Similar results were obtained when transcellular transport of C-K was determined using multidrug resistance 1 (MDR1)-overexpressing Madin-Darby canine kidney II cells. In MDR1a/b(-/-) FVB mice, its plasma C(max) and AUC(0-24h) were increased substantially by 4.0- and 11.7-fold, respectively. These increases appear to be due to slower elimination and faster absorption of C-K in MDR1a/b(-/-) mice. In conclusion, C-K is the major active metabolite of ginsenosides after microflora hydrolysis of primary ginsenosides in the red ginseng extract, and inhibition/deficiency of P-gp can lead to large enhancement of its absorption and bioavailability.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 1441 Moursund St., Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
QUISPE-FUENTES ISSIS, VEGA-GÁLVEZ ANTONIO, MIRANDA MARGARITA, LEMUS-MONDACA ROBERTO, LOZANO MARIBEL, AH-HEN KONG. A KINETIC APPROACH TO SAPONIN EXTRACTION DURING WASHING OF QUINOA (CHENOPODIUM QUINOAWILLD.) SEEDS. J FOOD PROCESS ENG 2012. [DOI: 10.1111/j.1745-4530.2012.00673.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Stereoisomers ginsenosides-20(S)-Rg₃ and -20(R)-Rg₃ differentially induce angiogenesis through peroxisome proliferator-activated receptor-gamma. Biochem Pharmacol 2012; 83:893-902. [PMID: 22234331 DOI: 10.1016/j.bcp.2011.12.039] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 12/23/2011] [Accepted: 12/27/2011] [Indexed: 01/22/2023]
Abstract
Ginsenosides are considered the major constituents that are responsible for most of the pharmacological actions of ginseng. However, some ginsenosides exist as stereoisomeric pairs, detailed and molecular exposition based on the structural differences of ginsenoside stereoisomers has not been emphasized in most studies. Here we explore the functional differences of ginsenoside Rg₃ stereoisomers on angiogenesis. In this study, we demonstrated the distinctive differential angiogenic activities of 20(S)-Rg₃ and 20(R)-Rg₃ stereoisomers. 20(S)-Rg₃ at micromolar concentration promotes human endothelial cells proliferation, migration and tube formation in vitro, as well as ex vivo endothelial sprouting. The effects induced by 20(S)-Rg₃ are significantly more potent than 20(R)-Rg₃. These effects are partially mediated through the activation of AKT/ERK-eNOS signaling pathways. Moreover, knockdown of peroxisome proliferator-activated receptor-gamma (PPARγ) by specific small interference RNA abolished the 20(S)-Rg₃-induced angiogenesis, indicating that PPARγ is responsible for mediating the angiogenic activity of Rg₃. Using reporter gene assay, the PPARγ agonist activity of 20(S)-Rg₃ has been found 10-fold higher than that of 20(R)-Rg₃. Computer modeling also revealed the differential binding is due to the chiral center of 20(S)-Rg₃ can form a critical hydrogen bond with Tyr473 of PPARγ ligand binding domain. The present study elucidated the differential angiogenic effects of Rg₃ stereoisomers by acting as agonist of PPARγ. The results shed light on the structural difference between two ginsenoside stereoisomers that can lead to significant differential physiological outcomes which should be carefully considered in the future development of ginsenoside-based therapeutics.
Collapse
|
29
|
Choi S, Oh JY, Kim SJ. Ginsenoside Rh2 induces Bcl-2 family proteins-mediated apoptosis in vitro and in xenografts in vivo models. J Cell Biochem 2011; 112:330-40. [PMID: 21080338 DOI: 10.1002/jcb.22932] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The cancer chemoprevention effects of ginseng saponins have been demonstrated against a variety of experimental tumors; however, their molecular mechanisms in vitro and in in vivo models are not well studied. This study was undertaken to gain insights into the molecular mechanisms of ginsenoside Rh2 (Rh2)-induced cell death in human breast cancer cell lines as well as in in vivo xenografts. Rh2 treatment significantly inhibited viability of both MCF-7 and MDA-MB-231 human breast cells in a concentration-dependent manner, which correlated with mitochondria-mediated apoptosis. Rh2-induced apoptosis was accompanied by the down-regulation of antiapoptotic proteins Bcl-2, Bcl-xL, and Mcl-1. It also caused induction of the proapoptotic members Bak, Bax, and Bim leading to mitochondrial translocation of Bax and activation of caspases. Moreover, Rh2-induced apoptosis was partially, yet significantly protected by transient transfection of MCF-7 cells with Bax- and Bak-targeted siRNAs. Oral gavage of 5 mg Rh2/kg of mouse (three times a week) significantly caused apoptosis of MDA-MB-231 xenografts. An increase in Bax and Bak and a decrease in Bcl-2 and Bcl-xL transcript levels, in accordance with their protein expression, were observed in tumor tissue. Tumors from Rh2-treated mice exhibited a markedly higher count of apoptotic bodies and reduced proliferation index compared with control tumors. Our data suggest that Rh2 used in traditional oriental medicine for the treatment of various ailments, may be an attractive agent for the treatment and/or prevention of human breast cancers.
Collapse
Affiliation(s)
- Sunga Choi
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 301747, Korea.
| | | | | |
Collapse
|
30
|
Qi LW, Wang CZ, Yuan CS. Ginsenosides from American ginseng: chemical and pharmacological diversity. PHYTOCHEMISTRY 2011; 72:689-99. [PMID: 21396670 PMCID: PMC3103855 DOI: 10.1016/j.phytochem.2011.02.012] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 01/11/2011] [Accepted: 02/14/2011] [Indexed: 05/19/2023]
Abstract
Ginseng occupies a prominent position in the list of best-selling natural products in the world. Compared to the long history of use and widespread research on Asian ginseng, the study of American ginseng is relatively limited. In the past decade, some promising advances have been achieved in understanding the chemistry, pharmacology and structure-function relationship of American ginseng. To date, there is no systematic review of American ginseng. In this review, the different structures of the ginsenosides in American ginseng are described, including naturally occurring compounds and those resulting from steaming or biotransformation. Preclinical and clinical studies published in the past decade are also discussed. Highlighted are the chemical and pharmacological diversity and potential structural-activity relationship of ginsenosides. The goal is that this article is a useful reference to chemists and biologists researching American ginseng, and will open the door to agents in drug discovery.
Collapse
Affiliation(s)
- Lian-Wen Qi
- Tang Center for Herbal Medicine Research and Department of Anesthesia and Critical Care, The Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
31
|
Augustin JM, Kuzina V, Andersen SB, Bak S. Molecular activities, biosynthesis and evolution of triterpenoid saponins. PHYTOCHEMISTRY 2011; 72:435-57. [PMID: 21333312 DOI: 10.1016/j.phytochem.2011.01.015] [Citation(s) in RCA: 434] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/07/2011] [Accepted: 01/11/2011] [Indexed: 05/19/2023]
Abstract
Saponins are bioactive compounds generally considered to be produced by plants to counteract pathogens and herbivores. Besides their role in plant defense, saponins are of growing interest for drug research as they are active constituents of several folk medicines and provide valuable pharmacological properties. Accordingly, much effort has been put into unraveling the modes of action of saponins, as well as in exploration of their potential for industrial processes and pharmacology. However, the exploitation of saponins for bioengineering crop plants with improved resistances against pests as well as circumvention of laborious and uneconomical extraction procedures for industrial production from plants is hampered by the lack of knowledge and availability of genes in saponin biosynthesis. Although the ability to produce saponins is rather widespread among plants, a complete synthetic pathway has not been elucidated in any single species. Current conceptions consider saponins to be derived from intermediates of the phytosterol pathway, and predominantly enzymes belonging to the multigene families of oxidosqualene cyclases (OSCs), cytochromes P450 (P450s) and family 1 UDP-glycosyltransferases (UGTs) are thought to be involved in their biosynthesis. Formation of unique structural features involves additional biosynthetical enzymes of diverse phylogenetic background. As an example of this, a serine carboxypeptidase-like acyltransferase (SCPL) was recently found to be involved in synthesis of triterpenoid saponins in oats. However, the total number of identified genes in saponin biosynthesis remains low as the complexity and diversity of these multigene families impede gene discovery based on sequence analysis and phylogeny. This review summarizes current knowledge of triterpenoid saponin biosynthesis in plants, molecular activities, evolutionary aspects and perspectives for further gene discovery.
Collapse
Affiliation(s)
- Jörg M Augustin
- Plant Biochemistry Laboratory, Department of Plant Biology and Biotechnology, Center for Synthetic Biology, VKR Research Centre Pro-Active Plants, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
32
|
Park EK, Lee EJ, Lee SH, Koo KH, Sung JY, Hwang EH, Park JH, Kim CW, Jeong KC, Park BK, Kim YN. Induction of apoptosis by the ginsenoside Rh2 by internalization of lipid rafts and caveolae and inactivation of Akt. Br J Pharmacol 2010; 160:1212-23. [PMID: 20590613 DOI: 10.1111/j.1476-5381.2010.00768.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Lipid rafts and caveolae are membrane microdomains with important roles in cell survival signalling involving the Akt pathway. Cholesterol is important for the structure and function of these microdomains. The ginsenoside Rh2 exhibits anti-tumour activity. Because Rh2 is structurally similar to cholesterol, we investigated the possibility that Rh2 exerted its anti-tumour effect by modulating rafts and caveolae. EXPERIMENTAL APPROACH A431 cells (human epidermoid carcinoma cell line) were treated with Rh2 and the effects on cell apoptosis, raft localization and Akt activation measured. We also examined the effects of over-expression of Akt and active-Akt on Rh2-induced cell death. KEY RESULTS Rh2 induced apoptosis concentration- and time-dependently. Rh2 reduced the levels of rafts and caveolae in the plasma membrane and increased their internalization. Furthermore, Akt activity was decreased and consequently, Akt-dependent phosphorylation of Bad, a pro-survival protein, was decreased whereas the pro-apoptotic proteins, Bim and Bax, were increased upon Rh2 treatment. Unlike microdomain internalization induce by cholesterol depletion, Rh2-mediated internalization of rafts and caveolae was not reversed by cholesterol addition. Also, cholesterol addition did not restore Akt activation or rescue cells from Rh2-induced cell death. Rh2-induced cell death was attenuated in MDA-MB-231 cells over-expressing either wild-type or dominant-active Akt. CONCLUSIONS AND IMPLICATIONS Rh2 induced internalization of rafts and caveolae, leading to Akt inactivation, and ultimately apoptosis. Because elevated levels of membrane rafts and caveolae, and Akt activation have been correlated with cancer development, internalization of these microdomains by Rh2 could potentially be used as an anti-cancer therapy.
Collapse
Affiliation(s)
- E-K Park
- Division of Specific Organs Cancer, Pediatric Oncology Division, National Cancer Center, Ilsan-gu, Goyang-si, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Musende AG, Eberding A, Jia W, Ramsay E, Bally MB, Guns ET. Rh2 or its aglycone aPPD in combination with docetaxel for treatment of prostate cancer. Prostate 2010; 70:1437-47. [PMID: 20687217 DOI: 10.1002/pros.21179] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Docetaxel is one of the few chemotherapeutic drugs that are considered highly effective when used to treat prostate cancer patients that have relapsed and/or metastatic disease, it is therefore reasonable to expect further improvements in treatment outcomes when it is combined with other therapeutic agents active in prostate cancer. This study assesses the combination of well tolerated and orally bioavailable formulations of ginsenoside Rh2 or its aglycone aPPD with docetaxel. METHODS The in vitro activity of Rh2, aPPD, and docetaxel was determined in four prostate cancer cell lines: PC-3, LNCaP, DU145, and C4-2. Combinations of Rh2 or aPPD with docetaxel were assessed using the constant ratio combination design. Combination Indices (CI) and Dose Reduction Indices (DRI) were subsequently estimated using Calcusyn. In vivo efficacy studies and Immunohistochemical analyses (PC-3 model) were also evaluated. RESULTS In PC-3, DU145 and C4-2 prostate cancer cells combinations of Rh2 or aPPD with docetaxel were predominantly additive or synergistic. Combinations of Rh2 + docetaxel and aPPD + docetaxel caused established PC-3 tumors to regress from their initial size by 15% and 27%, respectively. Tumor cell proliferation rate (measured by Ki-67 positive cells) was significantly lower for combinations of Rh2 + docetaxel and aPPD + docetaxel, compared to animals treated with docetaxel alone. CONCLUSIONS Rh2 and aPPD can be combined with docetaxel to yield additive or synergistic activity in vitro and in vivo. Pending further assessment of toxicity and pharmacodynamic behavior, this study supports testing of combinations of ginsenoside Rh2 or its aglycone aPPD with docetaxel in a clinical setting.
Collapse
Affiliation(s)
- Alain G Musende
- The Prostate Centre at Vancouver General Hospital, Vancouver, BC, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Taira S, Ikeda R, Yokota N, Osaka I, Sakamoto M, Kato M, Sahashi Y. Mass spectrometric imaging of ginsenosides localization in Panax ginseng root. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2010; 38:485-93. [PMID: 20503467 DOI: 10.1142/s0192415x10008007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We performed mass spectrometric imaging (MSI) to localize ginsenosides (Rb(1), Rb(2) or Rc, and Rf) in cross-sections of the Panax ginseng root at a resolution of 100 microm using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS). Tandem mass spectrometry (MS/MS) of alkali metal-adducted ginsenoside ions revealed structural information of the corresponding saccharides and aglycone. MALDI-MSI confirmed that ginsenosides were located more in the cortex and the periderm than that in the medulla of a lateral root. In addition, it revealed that localization of ginsenosides in a root tip (diameter, 2.7 mm) is higher than that in the center of the root (diameter, 7.3 mm). A quantitative difference was detected between localizations of protopanaxadiol-type ginsenoside (Rb(1), Rb(2), or Rc) and protopanaxatriol-type ginsenoside (Rf) in the root. This imaging approach is a promising technique for rapid evaluation and identification of medicinal saponins in plant tissues.
Collapse
Affiliation(s)
- Shu Taira
- Japan Advanced Institute of Science and Technology, Nomi, Ishikawa, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Qi LW, Wang CZ, Yuan CS. American ginseng: potential structure-function relationship in cancer chemoprevention. Biochem Pharmacol 2010; 80:947-54. [PMID: 20599804 DOI: 10.1016/j.bcp.2010.06.023] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 02/06/2023]
Abstract
Ginseng has a prominent position on the list of best-selling herbal products in the world, and its main active constituents are thought to be ginsenosides. Compared with the long history of use and widespread research on Asian ginseng, studies of American ginseng are relatively limited, especially regarding cancer chemoprevention. In recent studies of American ginseng, steaming or heating altered the ginsenoside profile and thereby increased anticancer effects. Yet the ginsenoside structures and their activities have not been systematically elucidated. In this commentary, we introduce the different ginsenosides in American ginseng, both the naturally occurring compounds and those resulting from steaming or biotransformation. We briefly review American ginseng's reported anticancer effects and their mechanisms of action, and explore the possible structural-function relationship with a focus on sugar molecules, hydroxyl groups and stereoselectivity in ginsenosides. Understanding these relationships may produce insights into chemical and pharmacological approaches for enhancing the chemopreventive effects of ginsenoside and for developing novel anticancer agents.
Collapse
Affiliation(s)
- Lian-Wen Qi
- Tang Center for Herbal Medicine Research, Department of Anesthesia & Critical Care, The Pritzker School of Medicine, University of Chicago, 5841 South Maryland Avenue, MC 4028, Chicago, IL 60637, United States
| | | | | |
Collapse
|
36
|
Liu J, Shimizu K, Yu H, Zhang C, Jin F, Kondo R. Stereospecificity of hydroxyl group at C-20 in antiproliferative action of ginsenoside Rh2 on prostate cancer cells. Fitoterapia 2010; 81:902-5. [PMID: 20554003 DOI: 10.1016/j.fitote.2010.05.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 05/27/2010] [Accepted: 05/30/2010] [Indexed: 10/19/2022]
Abstract
Prostate cancer is the fifth most common neoplasm worldwide, and the second most common cancer among men. Ginsenosides, the main component of ginseng, have been known for their medicinal effects such as anti-inflammatory and anti-proliferative activities. In this study, we investigated the inhibitory effects of ginsenosides (ginsenoside 20(R)-Rh2 and ginsenoside 20(S)-Rh2) on prostate cancer cells in vitro. Only ginsenoside 20(S)-Rh2 showed proliferation inhibition on androgen-dependent and -independent prostate cancer cells. These results implied that the stereochemistry of the hydroxyl group at C-20 may play an important role in antitumor activities.
Collapse
Affiliation(s)
- Jie Liu
- Department of Forest and Forest Products Science, Faculty of Agriculture, Kyushu University, Fukuoka, 812-8581, Japan
| | | | | | | | | | | |
Collapse
|
37
|
N(3)-o-toluyl-fluorouracil inhibits human hepatocellular carcinoma cell growth via sustained release of 5-FU. Cancer Chemother Pharmacol 2009; 66:11-9. [PMID: 19756602 PMCID: PMC2854362 DOI: 10.1007/s00280-009-1128-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 09/02/2009] [Indexed: 01/01/2023]
Abstract
Purpose N3-o-toluyl-fluorouracil (TFU), the prodrug of 5-fluorouracil (5-FU), is the metabolite of N1-acetyl-N3-o-toluyl-fluorouracil (atofluding). In the present study, we aimed to evaluate the efficacy of TFU on the inhibition of human hepatocellular carcinoma cells via sustained release of 5-FU. The metabolism of TFU underlying the inhibitory effect was also analyzed. Methods In vitro assays, inhibition of cell growth by TFU was evaluated by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide method. The levels of TFU and 5-FU in the cell culture supernatant fluid were measured by high-performance liquid chromatography (HPLC). In vivo assays, the efficacy of TFU was evaluated in a human hepatocellular carcinoma xenograft mice model after 3 weeks of oral administration. The distributions of TFU and 5-FU in plasma and homogenate tissues including liver, lung and tumor were determined by HPLC. Results N3-o-toluyl-fluorouracil weakly inhibited the proliferation of SMMC-7721 and PLC/PRF/5 cells in the absence of liver microsomal enzymes. In contrast, the inhibition rates were significantly increased in the presence of these enzymes. HPLC results revealed that TFU was metabolized slowly by liver microsomal enzymes and therefore the concentration of 5-FU was gradually increased with a longer retention time in cell culture supernatant fluid. The efficacy of TFU was confirmed in SMMC-7721 xenografts in Balb/c athymic (nu+/nu+) mice model. TFU treatment induced inhibition of SMMC-7721 growth with few side effects. HPLC results showed that high levels of TFU were still in liver 48 h after the end of oral administration, implying that TFU preferentially accumulated in liver with slow conversion to 5-FU by enzymes. This led to a long-lasting concentration of 5-FU in plasma. Further, a high level of 5-FU was found in tumors with a relatively low level in lungs. These results suggest that the metabolite of TFU was preferentially converted or taken up by tumor cells. The distributions of 5-FU may contribute to its high anti-tumor activity and low adverse reactions in vivo. Conclusion These results demonstrate that TFU is a promising prodrug of 5-FU for cancer treatment via sustained release of 5-FU in liver.
Collapse
|