1
|
Li F, Cai C, Wang F, Zhang N, Zhao Q, Chen Y, Cui X, Wang S, Zhang W, Liu D, Cai Y, Jin J. 20(S)-ginsenoside Rg3 suppresses gastric cancer cell proliferation by inhibiting E2F-DP dimerization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156740. [PMID: 40252583 DOI: 10.1016/j.phymed.2025.156740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Gastric cancer (GC) is a common and aggressive malignancy, with treatment options often limited by drug resistance and the adverse effects of targeted therapies and immunotherapy. Ginsenoside Rg3, a bioactive compound derived from ginseng, has shown promise in inhibiting the growth of various tumor types, including GC. However, the molecular mechanisms underlying its therapeutic effects against GC remain insufficiently understood. OBJECTIVE This study aimed to elucidate the molecular mechanisms underlying the anti-cancer effects of ginsenoside Rg3 against GC. METHODS To explore the molecular mechanisms underlying Rg3's anti-GC effects, RNA sequencing (RNA-Seq) was conducted to identify potential Rg3-regulated targets. The interaction between Rg3 and E2F was analyzed using several approaches, including the cellular thermal shift assay (CETSA), Rg3-PEGA pull-down, Rg3 pull-down protein mass spectrometry, and 3D molecular docking. Additionally, quantitative reverse transcription PCR (qRT-PCR), co-transfection followed by immunoprecipitation, Western blotting, flow cytometry, Annexin V-FITC staining, Hoechst staining, and luciferase reporter assays were employed to elucidate the molecular effects of Rg3. The inhibitory effect of Rg3 on GC proliferation was assessed through colony formation assays in vitro and tumor xenograft experiments in C57BL/6 mice in vivo. RESULTS Rg3-mediated gene expression profiling in GC cells revealed several transcription factors, including E2F, and biological processes potentially influenced by Rg3. Consistent with these findings, Rg3 suppressed E2F expression and impeded GC cell proliferation by inducing G1/S cell cycle arrest, reducing cell growth both in vitro and in vivo, enhancing apoptosis, and inhibiting CDC6 transactivation. CETSA and Rg3 pull-down assays confirmed an interaction between Rg3 and E2F. Additionally, 3D molecular docking analysis demonstrated that Rg3 binds with high affinity to E2F at the heterodimeric domain via hydrogen bonding, potentially disrupting E2F-DP heterodimer formation and subsequently inhibiting cell cycle gene expression. In agreement with this, Rg3-treated GC cells exhibited reduced expression of cyclin D1, CDK4, cyclin A, CDK1, and CDK2. Moreover, Rg3 activated the tumor suppressors p53 and p21, further inhibiting RB phosphorylation by suppressing cyclin/CDK activity, thereby blocking transcription of G1/S transition-related genes. CONCLUSION This study provides the first evidence that Rg3 directly binds to E2F proteins, disrupting E2F-DP heterodimer formation and inhibiting the transcription of E2F-DP-regulated target genes. Furthermore, Rg3 activates the p53-p21 pathway while suppressing the cyclin/CDK-RB signaling pathway, effectively inhibiting cancer cell proliferation. These findings highlight a potential therapeutic strategy for developing small molecules structurally similar to Rg3 to target tumors with high E2F expression.
Collapse
Affiliation(s)
- Fuqiang Li
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China; School of Pharmacy, Changchun University of Chinese Medicine, Boshuo Road, Jingyue Development Zone, Changchun, Jilin 130117, China
| | - Chengyu Cai
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Fei Wang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Na Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Qingzhi Zhao
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Yuyang Chen
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Xueli Cui
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Siyang Wang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Wenjie Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Boshuo Road, Jingyue Development Zone, Changchun, Jilin 130117, China.
| | - Yong Cai
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China.
| | - Jingji Jin
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China.
| |
Collapse
|
2
|
Yang Y, Nan Y, Du Y, Liu W, Ning N, Chen G, Gu Q, Yuan L. Ginsenosides in cancer: Proliferation, metastasis, and drug resistance. Biomed Pharmacother 2024; 177:117049. [PMID: 38945081 DOI: 10.1016/j.biopha.2024.117049] [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: 03/25/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024] Open
Abstract
Ginseng, the dried root of Panax ginseng C.A. Mey., is widely used in Chinese herbal medicine. Ginsenosides, the primary active components of ginseng, exhibit diverse anticancer functions through various mechanisms, such as inhibiting tumor cell proliferation, promoting apoptosis, and suppressing cell invasion and migration. In this article, the mechanism of action of 20 ginsenoside subtypes in tumor therapy and the research progress of multifunctional nanosystems are reviewed, in order to provide reference for clinical prevention and treatment of cancer.
Collapse
Affiliation(s)
- Yi Yang
- School of Basic Medical, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Yuhua Du
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Na Ning
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Guoqing Chen
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Qian Gu
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Ling Yuan
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China.
| |
Collapse
|
3
|
Wang L, Zhang Y, Song Z, Liu Q, Fan D, Song X. Ginsenosides: a potential natural medicine to protect the lungs from lung cancer and inflammatory lung disease. Food Funct 2023; 14:9137-9166. [PMID: 37801293 DOI: 10.1039/d3fo02482b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Lung cancer is the malignancy with the highest morbidity and mortality. Additionally, pulmonary inflammatory diseases, such as pneumonia, acute lung injury, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis (PF), also have high mortality rates and can promote the development and progression of lung cancer. Unfortunately, available treatments for them are limited, so it is critical to search for effective drugs and treatment strategies to protect the lungs. Ginsenosides, the main active components of ginseng, have been shown to have anti-cancer and anti-inflammatory activities. In this paper, we focus on the beneficial effects of ginsenosides on lung diseases and their molecular mechanisms. Firstly, the molecular mechanism of ginsenosides against lung cancer was summarized in detail, mainly from the points of view of proliferation, apoptosis, autophagy, angiogenesis, metastasis, drug resistance and immunity. In in vivo and in vitro lung cancer models, ginsenosides Rg3, Rh2 and CK were reported to have strong anti-lung cancer effects. Then, in the models of pneumonia and acute lung injury, the protective effect of Rb1 was particularly remarkable, followed by Rg3 and Rg1, and its molecular mechanism was mainly associated with targeting NF-κB, Nrf2, MAPK and PI3K/Akt pathways to alleviate inflammation, oxidative stress and apoptosis. Additionally, ginsenosides may also have a potential health-promoting effect in the improvement of COPD, asthma and PF. Furthermore, to overcome the low bioavailability of CK and Rh2, the development of nanoparticles, micelles, liposomes and other nanomedicine delivery systems can significantly improve the efficacy of targeted lung cancer treatment. To conclude, ginsenosides can be used as both anti-lung cancer and lung protective agents or adjuvants and have great potential for future clinical applications.
Collapse
Affiliation(s)
- Lina Wang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Yanxin Zhang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Zhimin Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Qingchao Liu
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Biotechnology & Biomedicine Research Institute, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaoping Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
4
|
How ginseng regulates autophagy: Insights from multistep process. Biomed Pharmacother 2023; 158:114139. [PMID: 36580724 DOI: 10.1016/j.biopha.2022.114139] [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: 10/20/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Although autophagy is a recognized contributor to the pathogenesis of human diseases, chloroquine and hydroxychloroquine are the only two FDA-approved autophagy inhibitors to date. Emerging evidence has revealed the potential therapeutic benefits of various extracts and active compounds isolated from ginseng, especially ginsenosides and their derivatives, by mediating autophagy. Mechanistically, active components from ginseng mediate key regulators in the multistep processes of autophagy, namely, initiation, autophagosome biogenesis and cargo degradation. AIM OF REVIEW To date, a review that systematically described the relationship between ginseng and autophagy is still lacking. Breakthroughs in finding the key players in ginseng-autophagy regulation will be a promising research area, and will provide positive insights into the development of new drugs based on ginseng and autophagy. KEY SCIENTIFIC CONCEPTS OF REVIEW Here, we comprehensively summarized the critical roles of ginseng-regulated autophagy in treating diseases, including cancers, neurological disorders, cardiovascular diseases, inflammation, and neurotoxicity. The dual effects of the autophagy response in certain diseases are worthy of note; thus, we highlight the complex impacts of both ginseng-induced and ginseng-inhibited autophagy. Moreover, autophagy and apoptosis are controlled by multiple common upstream signals, cross-regulate each other and affect certain diseases, especially cancers. Therefore, this review also discusses the cross-signal transduction pathways underlying the molecular mechanisms and interaction between ginseng-regulated autophagy and apoptosis.
Collapse
|
5
|
Recent advances in ginsenosides against respiratory diseases: Therapeutic targets and potential mechanisms. Biomed Pharmacother 2023; 158:114096. [PMID: 36502752 DOI: 10.1016/j.biopha.2022.114096] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Respiratory diseases mainly include asthma, influenza, pneumonia, chronic obstructive pulmonary disease, pulmonary hypertension, lung fibrosis, and lung cancer. Given their high prevalence and poor prognosis, the prevention and treatment of respiratory diseases are increasingly essential. In particular, the development for the novel strategies of drug treatment has been a hot topic in the research field. Ginsenosides are the major component of Panax ginseng C. A. Meyer (ginseng), a food homology and well-known medicinal herb. In this review, we summarize the current therapeutic effects and molecular mechanisms of ginsenosides in respiratory diseases. METHODS The reviewed studies were retrieved via a thorough analysis of numerous articles using electronic search tools including Sci-Finder, ScienceDirect, PubMed, and Web of Science. The following keywords were used for the online search: ginsenosides, asthma, influenza, pneumonia, chronic obstructive pulmonary disease (COPD), pulmonary hypertension (PH), lung fibrosis, lung cancer, and clinical trials. We summarized the findings and the conclusions from 176 manuscripts on ginsenosides, including research articles and reviews. RESULTS Ginsenosides Rb1, Rg1, Rg3, Rh2, and CK, which are the most commonly reported ginsenosides for treating of respiratory diseases, and other ginsenosides such as Rh1, Rk1, Rg5, Rd and Re, all primarily reduce pneumonia, fibrosis, and inhibit tumor progression by targeting NF-κB, TGF-β/Smad, PI3K/AKT/mTOR, and JNK pathways, thereby ameliorating respiratory diseases. CONCLUSION This review provides novel ideas and important aspects for the future research of ginsenosides for treating respiratory diseases.
Collapse
|
6
|
Comparison of the Saponins in Three Processed American Ginseng Products by Ultra-High Performance Liquid Chromatography-Quadrupole Orbitrap Tandem Mass Spectrometry and Multivariate Statistical Analysis. Int J Anal Chem 2022; 2022:6721937. [PMID: 35521625 PMCID: PMC9064508 DOI: 10.1155/2022/6721937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 11/20/2022] Open
Abstract
A method with ultrahigh performance liquid chromatography Quadrupole-Orbitrap tandem mass spectrometry (UHPLC-Q-Orbitrap-MS/MS) was applied for the quality evaluation of different processing and drying of American ginseng, including natural drying (ND), steam drying (SD), and vacuum freeze-drying (VFD). A total of 51 saponins were successfully identified in three processed products. Three processed American ginseng products were well-differentiated in orthogonal partial least-squares discriminant analysis (OPLS-DA). The S-plot also identified the marker compounds in each product, while the major ginsenosides of ND (malonyl (M)-Rd, M-Rb1, Rg1), SD (20 (S)-Rg3, 20 (S)-Rg2), and VFD (M-Rd, M-Rb1) were found. The results indicate that the method by vacuum freeze-drying can retain the content of rare ginsenosides and malonyl-ginsenosides. The marker compounds selected will benefit the holistic evaluation of related American ginseng products.
Collapse
|
7
|
Xu JF, Wan Y, Tang F, Chen L, Yang Y, Xia J, Wu JJ, Ao H, Peng C. Emerging Significance of Ginsenosides as Potentially Reversal Agents of Chemoresistance in Cancer Therapy. Front Pharmacol 2022; 12:720474. [PMID: 34975466 PMCID: PMC8719627 DOI: 10.3389/fphar.2021.720474] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022] Open
Abstract
Chemoresistance has become a prevalent phenomenon in cancer therapy, which alleviates the effect of chemotherapy and makes it difficult to break the bottleneck of the survival rate of tumor patients. Current approaches for reversing chemoresistance are poorly effective and may cause numerous new problems. Therefore, it is urgent to develop novel and efficient drugs derived from natural non-toxic compounds for the reversal of chemoresistance. Researches in vivo and in vitro suggest that ginsenosides are undoubtedly low-toxic and effective options for the reversal of chemoresistance. The underlying mechanism of reversal of chemoresistance is correlated with inhibition of drug transporters, induction of apoptosis, and modulation of the tumor microenvironment(TME), as well as the modulation of signaling pathways, such as nuclear factor erythroid-2 related factor 2 (NRF2)/AKT, lncRNA cancer susceptibility candidate 2(CASC2)/ protein tyrosine phosphatase gene (PTEN), AKT/ sirtuin1(SIRT1), epidermal growth factor receptor (EGFR)/ phosphatidylinositol 3-kinase (PI3K)/AKT, PI3K/AKT/ mammalian target of rapamycin(mTOR) and nuclear factor-κB (NF-κB). Since the effects and the mechanisms of ginsenosides on chemoresistance reversal have not yet been reviewed, this review summarized comprehensively experimental data in vivo and in vitro to elucidate the functional roles of ginsenosides in chemoresistance reversal and shed light on the future research of ginsenosides.
Collapse
Affiliation(s)
- Jin-Feng Xu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Wan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Xia
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiao-Jiao Wu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Huang J, Li JX, Ma LR, Xu DH, Wang P, Li LQ, Yu LL, Li Y, Li RZ, Zhang H, Zheng YH, Tang L, Yan PY. Traditional Herbal Medicine: A Potential Therapeutic Approach for Adjuvant Treatment of Non-small Cell Lung Cancer in the Future. Integr Cancer Ther 2022; 21:15347354221144312. [PMID: 36567455 PMCID: PMC9806388 DOI: 10.1177/15347354221144312] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/27/2022] Open
Abstract
Lung carcinoma is the primary reason for cancer-associated mortality, and it exhibits the highest mortality and incidence in developed and developing countries. Non-small cell lung cancer (NSCLC) and SCLC are the 2 main types of lung cancer, with NSCLC contributing to 85% of all lung carcinoma cases. Conventional treatment mainly involves surgery, chemoradiotherapy, and immunotherapy, but has a dismal prognosis for many patients. Therefore, identifying an effective adjuvant therapy is urgent. Historically, traditional herbal medicine has been an essential part of complementary and alternative medicine, due to its numerous targets, few side effects and substantial therapeutic benefits. In China and other East Asian countries, traditional herbal medicine is increasingly popular, and is highly accepted by patients as a clinical adjuvant therapy. Numerous studies have reported that herbal extracts and prescription medications are effective at combating tumors. It emphasizes that, by mainly regulating the P13K/AKT signaling pathway, the Wnt signaling pathway, and the NF-κB signaling pathway, herbal medicine induces apoptosis and inhibits the proliferation and migration of tumor cells. The present review discusses the anti-NSCLC mechanisms of herbal medicines and provides options for future adjuvant therapy in patients with NSCLC.
Collapse
Affiliation(s)
- Jie Huang
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Jia-Xin Li
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Lin-Rui Ma
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Dong-Han Xu
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Peng Wang
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Li-Qi Li
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Li-Li Yu
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Yu Li
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Run-Ze Li
- Second Affiliated Hospital of Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hao Zhang
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Yu-Hong Zheng
- Macau University of Science and
Technology, Taipa, Macau, China
| | - Ling Tang
- Southern Medical University, Guangzhou,
Guangdong, China
- Guangdong Provincial Key Laboratory of
Chinese Medicine Pharmaceutics, Guangzhou, Guangdong, China
- Guangdong Provincial Engineering
Laboratory of Chinese Medicine Preparation Technology, Guangzhou, Guangdong,
China
| | - Pei-Yu Yan
- Macau University of Science and
Technology, Taipa, Macau, China
| |
Collapse
|
9
|
Chen P, Dai CH, Shi ZH, Wang Y, Wu JN, Chen K, Su JY, Li J. Synergistic inhibitory effect of berberine and icotinib on non-small cell lung cancer cells via inducing autophagic cell death and apoptosis. Apoptosis 2021; 26:639-656. [PMID: 34743246 DOI: 10.1007/s10495-021-01694-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 01/12/2023]
Abstract
Resistance to epidermal growth factor receptor-tyrosin kinase inhibitors (TKIs, e.g. icotinib) remains a major clinical challenge. Non-small cell lung cancer patients with wild-type EGFR and/or K-RAS mutation are primary resistance to EGFR-TKIs. Berberine has been found to have potent anticancer activities via distinct molecular mechanism. In this study, we sought to investigate the therapeutic utility of BBR in combination with icotinib to overcome icotinib resistance in NSCLC cells, and explore the molecular mechanism of synergism of icotinib and BBR to EGFR-resistant NSCLC cells. We used the two EGFR-resistant NSCLC cell lines H460 and H1299 for testing the inhibitory effect of icotinib and/or BBR on them. Moreover, xenograft mouse model was applied for assessing the anti-tumor activities of BBR and icotinib in combination. Results showed that BBR and icotinib have a synergistic inhibitory effect on H460 and H1299 cells through induction of autophagic cell death and apoptosis. Accordingly, the anti-cancer effect of BBR plus icotinib was further confirmed in the NSCLC xenograft mouse models. Combination of BBR and icotinib significantly inhibited the protein expression and the activity of EGFR by inducing autophagic EGFR degradation. BBR plus icotinib resulted in intracellular ROS accumulation, which could mediated autophagy and apoptosis and involved in the suppression of cell migration and invasion. In conclusions, combination application of BBR and icotinib could be an effective strategy to overcome icotinib resistance in the treatment of NSCLC.
Collapse
Affiliation(s)
- Ping Chen
- Department of Pulmonary Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chun-Hua Dai
- Department of Radiation Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhi-Hao Shi
- Department of Pulmonary Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yi Wang
- Center of Medical Experiment, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jian-Nong Wu
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kang Chen
- Department of Pulmonary Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jin-Yu Su
- Department of Pulmonary Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jian Li
- Department of Pulmonary Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
10
|
Liu W, Zhang SX, Ai B, Pan HF, Zhang D, Jiang Y, Hu LH, Sun LL, Chen ZS, Lin LZ. Ginsenoside Rg3 Promotes Cell Growth Through Activation of mTORC1. Front Cell Dev Biol 2021; 9:730309. [PMID: 34589493 PMCID: PMC8473834 DOI: 10.3389/fcell.2021.730309] [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: 06/24/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Ginsenoside Rg3 is a steroidal saponin isolated from Panax ginseng. Previous studies have shown that Rg3 treatment downregulates the activity of rapamycin complex 1 (mTORC1) activity and inhibits the growth of cancer cells. However, the inhibitory effect of Rg3 on cancer cells is associated with high concentrations of Rg3 that are difficult to achieve in vivo. The human cervix adenocarcinoma HeLa cells were treated with Rg3. The protein levels of AMP-activated protein kinase alpha (AMPKα), protein kinase B(Akt), ribosomal S6 protein(S6), and Erk were determined by immunoblotting analyses. We used a fluorescent probe to detect reactive oxygen species (ROS) production in living cells. The oxygen consumption rate (OCR) was examined by the Seahorse Extracellular Flux Analyzer. The content of adenosine triphosphate (ATP) was measured by ATPlite kit and Mitotracker was applied to detect the mitochondria. We showed that at lower concentrations, Rg3 activates mTORC1 independent of AKT and AMP-activated protein kinase (AMPK). Rg3 promotes mitochondrial biogenesis and function, increases the oxygen consumption of mitochondria and the content of ATP. This effect is in contrast to that of high concentrations of Rg3, which inhibits cell growth. These findings demonstrate a pro-growth activity of Rg3 that acts through mTORC1 and mitochondrial biogenesis and suggest a dose-dependent effect of Rg3 on tumor cell growth.
Collapse
Affiliation(s)
- Wei Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Integrative Cancer Centre, The First Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | | | - Bo Ai
- Department of Thoracic Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Feng Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lei-Hao Hu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Integrative Cancer Centre, The First Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Ling-Ling Sun
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Integrative Cancer Centre, The First Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John's University, Queens, NY, United States
| | - Li-Zhu Lin
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Integrative Cancer Centre, The First Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
11
|
Zhang JJ, Zhou YD, Liu YB, Wang JQ, Li KK, Gong XJ, Lin XH, Wang YP, Wang Z, Li W. Protective Effect of 20(R)-Ginsenoside Rg3 Against Cisplatin-Induced Renal Toxicity via PI3K/AKT and NF-[Formula: see text]B Signaling Pathways Based on the Premise of Ensuring Anticancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1739-1756. [PMID: 34461812 DOI: 10.1142/s0192415x21500828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although the protective effect of ginsenoside on cisplatin-induced renal injury has been extensively studied, whether ginsenoside interferes with the antitumor effect of cisplatin has not been confirmed. In this paper, we verified the main molecular mechanism of 20(R)-ginsenoside Rg3 (R-Rg3) antagonizing cisplatin-induced acute kidney injury (AKI) through the combination of in vivo and in vitro models. It is worth mentioning that the two cell models of HK-2 and HepG2 were used simultaneously for the first time to explore the effect of the activation site of tumor-associated protein p53 on apoptosis and tumor suppression. The results showed that a single injection of cisplatin (20 mg/kg) led to weight loss, the kidney index of the mice increased, and creatinine (CRE) and blood urea nitrogen (BUN) levels in mice sharply increased. Continuous administration of R-Rg3 at doses of 10 and 20 mg/kg for 10 days could significantly alleviate this symptom. Similarly, R-Rg3 treatment reduced oxidative stress damage caused by cisplatin. Moreover, R-Rg3 could observably reduce the apoptosis and inflammatory infiltration of renal tubular cells induced by cisplatin. We used western blotting analysis to demonstrate that R-Rg3 restored cisplatin-induced AKI might be related to PI3K/AKT and NF-[Formula: see text]B mediated apoptosis and inflammation pathways. In the meantime, we also verified that R-Rg3 could activate different sites of p53 to control renal cell apoptosis induced by cisplatin without affecting its antitumor effect.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yan-Dan Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yong-Bo Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jian-Qiang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Ke-Ke Li
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiao-Jie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiang-Hui Lin
- Liaoning XIFENG Pharmaceutical Group Co., Ltd., Huanren 117200, P. R. China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China.,Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| |
Collapse
|
12
|
Cai Z, Teng Y, Chen Y. The Effect of Shenyi Capsule on Non-Small-Cell Lung Cancer Combined with Chemotherapy from the Yin-Yang Perspective. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1653750. [PMID: 34512772 PMCID: PMC8426066 DOI: 10.1155/2021/1653750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/26/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
As an example of Shenyi capsule on non-small-cell lung cancer combined with chemotherapy, this review discusses the synergistic effect and mechanism of natural drugs in oncotherapy from the yin-yang perspective in ancient Chinese philosophy, so as to reflect the therapeutic principle of natural drugs for tumor more comprehensively. The major focuses of this review are on the philosophical thinking of yin-yang as a tool which can not only explain the effect of Shenyi capsule in NSCLC combined with chemotherapy but also explore the mechanism of Shenyi capsule at the cellular and molecular level. Learning from the "yin-yang" thinking of ancient Chinese philosophy will bring more enlightenment to the research and development of traditional Chinese drugs in the future.
Collapse
Affiliation(s)
- Zhixing Cai
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Yue Teng
- Outpatient Department of Clinic Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai 200437, China
| | - Yue Chen
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| |
Collapse
|
13
|
He GH, Xing DJ, Jin D, Lu Y, Guo L, Li YL, Li D. Scutellarin improves the radiosensitivity of non-small cell lung cancer cells to iodine-125 seeds via downregulating the AKT/mTOR pathway. Thorac Cancer 2021; 12:2352-2359. [PMID: 34255431 PMCID: PMC8410549 DOI: 10.1111/1759-7714.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 01/12/2023] Open
Abstract
Background In our previous study, we indicated that scutellarin (SCU) induced an anticancer effect in A549 cells. However, whether SCU regulates the radiosensitivity of non‐small cell lung cancer (NSCLC) and its related mechanism is still unclear. Methods In this study, we explored the anticancer effect induced by iodine‐125 (125I) and SCU at a sensitizing concentration in A549 and H1975 cells. Cellular apoptosis and proliferation were detected by flow cytometry, Bcl‐2/Bax expression level, cell cycle, CCK‐8, and EdU staining. A tumor model using nude mice was also carried out to investigate the combined effect of 125I and SCU in vivo. In addition, the expression level of AKT/mTOR pathway was detected to investigate whether it is linked to the anticancer effect of 125I and SCU. Results SCU at a sensitizing concentration promoted the 125I‐induced apoptosis and antiproliferative effect in A549 and H1975 cells. Moreover, the same results were obtained in vivo. Based on our findings, the AKT/mTOR pathway was significantly downregulated after combined treatment with 125I and SCU. Conclusions The results of our study suggested that SCU promotes the anticancer effects induced by 125I in NSCLC cells by downregulating the AKT/mTOR pathway and lays a foundation for future application of this combined treatment.
Collapse
Affiliation(s)
- Guang-Hui He
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Interventional Therapy, The Second People's Hospital of Weifang, Weifang, China
| | - Dian-Jin Xing
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Die Jin
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Yue Lu
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Lei Guo
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Vascular Anomalies and Interventional Radiology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Yu-Liang Li
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Dong Li
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
14
|
Zhou X, Liu H, Zhang M, Li C, Li G. Spectrum-effect relationship between UPLC fingerprints and anti-lung cancer effect of Panax ginseng. PHYTOCHEMICAL ANALYSIS : PCA 2021; 32:339-346. [PMID: 32808367 PMCID: PMC8048684 DOI: 10.1002/pca.2980] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/21/2020] [Accepted: 07/27/2020] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Lung cancer has the highest mortality rate among the various types of cancer. Panax ginseng (C. A. Mey). is a popular anti-cancer herbal supplement. The quality control of ginseng is crucial to ensure its clinical efficacy. This study aimed to establish new quality control methods for ginseng and to identify its main active components responsible for lung cancer treatment. METHODS Ultra-high-performance liquid chromatography (UPLC) was used to establish fingerprints of 18 batches of ginseng. CCK-8 test was performed to evaluate the inhibitory activity of ginseng on Lewis lung cancer (LLC) cells. The spectrum-effect relationship analysis of ginseng was assessed by canonical correlation analysis (CCA) and bioactivity validation. KEY FINDINGS Six common peaks were identified and the variation coefficients were determined. The 18 batches of ginseng inhibited the proliferation of LLC cells to different degrees, showing different half maximal inhibitory concentration (IC50 ) values. Spectrum-effect relationship analysis showed that ginsenoside Ro is the main anti-proliferative constituent of LLC cell. CONCLUSIONS Spectrum-effect relationship is suitable for quality control of ginseng used for lung cancer. It is also effective in discovering the active ingredients related to the clinical efficacy of traditional Chinese medicine.
Collapse
Affiliation(s)
- Xiaowei Zhou
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haiyang Liu
- Tonghua Institute for Food and Drug ControlTonghuaChina
| | - Mingyu Zhang
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chunyu Li
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Guohui Li
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
15
|
Paving the Road Toward Exploiting the Therapeutic Effects of Ginsenosides: An Emphasis on Autophagy and Endoplasmic Reticulum Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:137-160. [PMID: 33861443 DOI: 10.1007/978-3-030-64872-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Programmed cell death processes such as apoptosis and autophagy strongly contribute to the onset and progression of cancer. Along with these lines, modulation of cell death mechanisms to combat cancer cells and elimination of resistance to apoptosis is of great interest. It appears that modulation of autophagy and endoplasmic reticulum (ER) stress with specific agents would be beneficial in the treatment of several disorders. Interestingly, it has been suggested that herbal natural products may be suitable candidates for the modulation of these processes due to few side effects and significant therapeutic potential. Ginsenosides are derivatives of ginseng and exert modulatory effects on the molecular mechanisms associated with autophagy and ER stress. Ginsenosides act as smart phytochemicals that confer their effects by up-regulating ATG proteins and converting LC3-I to -II, which results in maturation of autophagosomes. Not only do ginsenosides promote autophagy but they also possess protective and therapeutic properties due to their capacity to modulate ER stress and up- and down-regulate and/or dephosphorylate UPR transducers such as IRE1, PERK, and ATF6. Thus, it would appear that ginsenosides are promising agents to potentially restore tissue malfunction and possibly eliminate cancer.
Collapse
|
16
|
Wen T, Song L, Hua S. Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Med 2021; 10:2396-2422. [PMID: 33650320 PMCID: PMC7982634 DOI: 10.1002/cam4.3660] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer‐related mortality both in men and women and accounts for 18.4% of all cancer‐related deaths. Although advanced therapy methods have been developed, the prognosis of lung cancer patients remains extremely poor. Over the past few decades, clinicians and researchers have found that chemical compounds extracted from natural products may be useful for treating lung cancer. Drug formulations derived from natural compounds, such as paclitaxel, doxorubicin, and camptothecin, have been successfully used as chemotherapeutics for lung cancer. In recent years, hundreds of new natural compounds that can be used to treat lung cancer have been found through basic and sub‐clinical research. However, there has not been a corresponding increase in the number of drugs that have been used in a clinical setting. The probable reasons may include low solubility, limited absorption, unfavorable metabolism, and severe side effects. In this review, we present a summary of the natural compounds that have been proven to be effective for the treatment of lung cancer, as well as an understanding of the mechanisms underlying their pharmacological effects. We have also highlighted current controversies and have attempted to provide solutions for the clinical translation of these compounds.
Collapse
Affiliation(s)
- Tingting Wen
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Lei Song
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Shucheng Hua
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
17
|
Liang Y, Zhang T, Jing S, Zuo P, Li T, Wang Y, Xing S, Zhang J, Wei Z. 20(S)-Ginsenoside Rg3 Inhibits Lung Cancer Cell Proliferation by Targeting EGFR-Mediated Ras/Raf/MEK/ERK Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:753-765. [DOI: 10.1142/s0192415x2150035x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lung cancer is the leading cause of cancer death in the world and classified into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). As tyrosine kinase inhibitors (TKIs), several triterpenoid saponins can target to epidermal growth factor receptor (EGFR), a widely used molecular therapeutic target, to exhibit remarkable anti-proliferative activities in cancer cells. As one of triterpenoid saponins, 20([Formula: see text])-ginsenoside Rg3 [20([Formula: see text])-Rg3] was confirmed to be an EGFR-TKI in this work. According to the quantitative real-time reverse transcription-PCR (qRT-PCR) and immunoblotting analysis, 20([Formula: see text])-Rg3 was certified to play a key role on EGFR/Ras/Raf/MEK/ERK signal pathway regulation. Our data demonstrated that 20([Formula: see text])-Rg3 might block the cell cycle at the G0/G1 phase by downregulating CDK2, Cyclin A2, and Cyclin E1. Molecular docking suggested that the combination of both hydrophobic and hydrogen-bonding interactions may help stabilizing the 20([Formula: see text])-Rg3-EGFR binding. Furthermore, their binding stability was assessed by molecular dynamics simulation. Taken together, these data provide the evidence that 20([Formula: see text])-Rg3 could prohibit A549 cell proliferation, probably by arresting the cell cycle at the G0/G1 phase via the EGFR/Ras/Raf/MEK/ERK pathway.
Collapse
Affiliation(s)
- Yuan Liang
- College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Siyuan Jing
- College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Peng Zuo
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun 130033, P. R. China
| | - Tiezhu Li
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun 130033, P. R. China
| | - Yongjun Wang
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun 130033, P. R. China
| | - Shaochen Xing
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun 130033, P. R. China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, P. R. China
| | - Zhengyi Wei
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun 130033, P. R. China
| |
Collapse
|
18
|
Hong H, Baatar D, Hwang SG. Anticancer Activities of Ginsenosides, the Main Active Components of Ginseng. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8858006. [PMID: 33623532 PMCID: PMC7875636 DOI: 10.1155/2021/8858006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
Cancer incidence rate has been increasing drastically in recent years. One of the many cancer treatment methods is chemotherapy. Traditional medicine, in the form of complementary and alternative therapy, is actively used to treat cancer, and many herbs and active ingredients of such therapies are being intensely studied to integrate them into modern medicine. Ginseng is traditionally used as a nourishing tonic and for treating various diseases in Asian countries. The therapeutic potential of ginseng in modern medicine has been studied extensively; the main bioactive component of ginseng is ginsenosides, which have gathered attention, particularly for their prospects in the treatment of fatal diseases such as cancer. Ginsenosides displayed their anticancer and antimetastatic properties not only via restricting cancer cell proliferation, viability, invasion, and migration but also by promoting apoptosis, cell cycle arrest, and autophagy in several cancers, such as breast, brain, liver, gastric, and lung cancer. Additionally, ginsenosides can work synergistically with already existing cancer therapies. Thus, ginsenosides may be used alone or in combination with other pharmaceutical agents in new therapeutic strategies for cancer. To date however, there is little systematic summary available for the anticancer effects and therapeutic potential of ginsenosides. Therefore, we have reviewed and discussed all available literature in order to facilitate further research of ginsenosides in this manuscript.
Collapse
Affiliation(s)
- Heeok Hong
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Delgerzul Baatar
- Laboratory of Genetics, Institute of Biology, Mongolian Academy of Sciences, Peace Avenue 13330, Ulaanbaatar, Mongolia
| | - Seong Gu Hwang
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong City 17579, Republic of Korea
| |
Collapse
|
19
|
Wang L, Han X, Zheng X, Zhou Y, Hou H, Chen W, Li X, Zhao L. [Ginsenoside 20(S)-Rg3 upregulates tumor suppressor VHL gene expression by suppressing DNMT3A-mediated promoter methylation in ovarian cancer cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:100-106. [PMID: 33509760 DOI: 10.12122/j.issn.1673-4254.2021.01.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To explore the mechanism by which ginsenoside 20(S)-Rg3 upregulates the expression of tumor suppressor von Hippel-Lindau (VHL) gene in ovarian cancer cells. METHODS Ovarian cancer cell line SKOV3 treated with 20(S)-Rg3 were examined for mRNA and protein levels of VHL, DNMT1, DNMT3A and DNMT3B by real-time PCR and Western blotting, respectively. The changes in VHL mRNA expression in SKOV3 cells in response to treatment with 5-Aza-CdR, a DNA methyltransferase inhibitor, were detected using real-time PCR. VHL gene promoter methylation was examined with methylation-specific PCR and VHL expression levels were determined with real-time PCR and Western blotting in non-treated or 20(S)-Rg3-treated SKOV3 cells and in 20(S)-Rg3-treated DNMT3A-overexpressing SKOV3 cells. VHL and DNMT3A protein levels were detected by immunohistochemistry in subcutaneous SKOV3 cell xenografts in nude mice. RESULTS Treatment of SKOV3 cells with 20(S)-Rg3 significantly upregulated VHL and downregulated DNMT3A expressions at both the mRNA and protein levels (P < 0.05) and upregulated DNMT3B expression only at the mRNA level, but did not cause significant changes in either the mRNA or protein level of DNMT1. Treatment of the cells with 2 and 5 μmol/L 5-Aza-CdR obviously increased VHL mRNA expression by by over 3 folds (P < 0.05). 20(S)-Rg3 significantly decreased the methylation level in the promoter region of VHL gene, and this effect was abrogated by DNMT3A overexpression in the cells (P < 0.05). Immunohistochemisty showed a significantly increased VHL expression but a lowered DNMT3A expression in subcutaneous SKOV3 cell xenografts in 20 (S)-Rg3-treated nude mice. CONCLUSIONS Ginsenoside 20(S)-Rg3 upregulates VHL expression in ovarian cancer cells by suppressing DNMT3A-mediated DNA methylation.
Collapse
Affiliation(s)
- Lijie Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Department of Gynecology, Lanzhou University Second Hospital, Lan Zhou 730030, China
| | - Xi Han
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xia Zheng
- the Second Affiliated Hospital of Zhejiang University School of medicine, Hangzhou 310009, China
| | - Yuanyuan Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Huilian Hou
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wei Chen
- Center for Laboratory Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xu Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Le Zhao
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
20
|
Xiao N, He H, Wang J, Zhang L, Chow B, Feng F, Xu Y, Huang J, Zhou X, Dong R. Meta-Analysis of Aidi Injection and First-Generation Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor Therapy in Treating Advanced Non-Small Cell Lung Cancer. J Evid Based Integr Med 2021; 26:2515690X211010733. [PMID: 33926244 PMCID: PMC8114743 DOI: 10.1177/2515690x211010733] [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: 12/22/2020] [Revised: 02/24/2021] [Accepted: 03/27/2021] [Indexed: 12/01/2022] Open
Abstract
The combination of Aidi injection (ADI) and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) in treating non-small cell lung cancer (NSCLC) has been reported, but the effects of this therapy have not been systematically assessed. Randomized controlled trials (RCTs) published before June 2020 were searched from 6 databases. Two reviewers independently assessed the methodological quality of 8 RCTs involving 667 patients diagnosed with stage III-IV NSCLC. We found that ADI combined with EGFR-TKI increased the objective response rate (ORR) significantly (relative risk [RR]: 1.60; 95% confidence interval [CI]: 1.28-1.99, P < 0.0001). There was also improvement in the disease control rate (DCR) (RR: 1.25; 95% CI: 1.11-1.40, P = 0.0002) as compared with EGFR-TKI alone. This therapy also increased the percentage of CD3+ cells (weighted mean difference [WMD]: 9.86; 95% CI: 4.62-15.10), CD4+ cells (WMD: 6.10; 95% CI: 1.67-10.53), and the CD4+/CD8+ (WMD: 0.35; 95% CI: 0.28-0.43). With regard to drug toxicity, the occurrence of rash was significantly reduced by ADI combined with EGFR-TKI (RR: 0.78, 95% CI: 0.63-0.97, P = 0.03); however, we did not find a significant reduction in the occurrence of dry skin, nausea and vomiting, as well as diarrhea between the 2 therapies. ADI combined with first-generation EGFR-TKIs may be more effective in improving tumor response, reducing the occurrence of rash, and enhancing immune function in NSCLC than EGFR-TKI alone.
Collapse
Affiliation(s)
- Na Xiao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Hailang He
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jing Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Brandon Chow
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, AZ, USA
| | - Fanchao Feng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingyi Huang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Rui Dong
- Nanjing University of Chinese Medicine, Nanjing, China
- Beijing Kangyide Pulmonary Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| |
Collapse
|
21
|
Liu L, Xu FR, Wang YZ. Traditional uses, chemical diversity and biological activities of Panax L. (Araliaceae): A review. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:112792. [PMID: 32311488 DOI: 10.1016/j.jep.2020.112792] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/22/2020] [Accepted: 03/22/2020] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax L. (Araliaceae) is globally-recognized plant resource suitable for the globalization of traditional Chinese medicines. It has traditionally been used as tonic agents in various ethnomedicinal systems of East Asia, especially in China. It is often used to regulate bodily functions and considered as adjuvant therapy for tumor, resuscitation of traumatic hemorrhagic shock, etc. AIM OF THIS REVIEW: This review systematically summarized the information on distributions, botanical characteristics, traditional uses, chemical components and biological activities of the genus Panax, in order to explore and exploit the therapeutic potential of this plant. MATERIALS AND METHODS The available information about genus Panax was collected via the online search on Web of Science, Google Scholar, PubMed, Baidu Scholar, Science Direct, China National Knowledge Infrastructure and Springer search. The keywords used include Panax, saponin, secondary metabolites, chemical components, biological activity, pharmacology, traditional medicinal uses, safety and other related words. The Plant List (www.theplantlist.org) and Catalogue of Life: 2019 Annual Checklist (www.catalogueoflife.org/col/) databases were used to provide the scientific names, subspecies classification and distribution information of Panax. RESULTS Panax is widely assessed concerning its phytochemistry and biological activities. To date, at least 748 chemical compounds from genus Panax were isolated, including saponins, flavonoids, polysaccharides, steroids and phenols. Among them, triterpenoid saponins and polysaccharides were the representative active ingredients of Panax plants, which have been widely investigated. Modern pharmacological studies showed that these compounds exhibited a wide range of biological activities in vitro and in vivo including antineoplastic, anti-inflammatory, hepatorenal protective, neuroprotective, immunoregulatory, cardioprotective and antidiabetic activities. Many studies also confirmed that the mechanisms of organ-protective were closely related to molecular signaling pathways, the expression of related proteins and antioxidant reactions. To sum up, genus Panax has high medicinal and social value, deserving further investigation. CONCLUSIONS The genus Panax is very promising to be fully utilized in the development of nutraceutical and pharmaceutical products. However, there is a lack of in-depth studies on ethnomedicinal uses of Panax plants. In addition, further studies of single chemical component should be performed based on the diversity of chemical structure, significant biological activities and clinical application. If the bioactive molecules and multicomponent interactions are discovered, it will be of great significance to the clinical application of Panax plants. It is an urgent requirement to carry out detailed phytochemical, pharmacology and clinical research on Panax classical prescriptions for the establishment of modern medication guidelines. Exploring the molecular basis of herbal synergistic actions may provide a new understanding of the complex disease mechanisms and accelerate the process of pharmaceutical development.
Collapse
Affiliation(s)
- Lu Liu
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China; College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Fu-Rong Xu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China.
| | - Yuan-Zhong Wang
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China.
| |
Collapse
|
22
|
20(S)-Rg3 upregulates FDFT1 via reducing miR-4425 to inhibit ovarian cancer progression. Arch Biochem Biophys 2020; 693:108569. [PMID: 32877662 DOI: 10.1016/j.abb.2020.108569] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/11/2020] [Accepted: 08/27/2020] [Indexed: 01/15/2023]
Abstract
We previously found that ginsenoside 20(S)-Rg3 diminishes the proliferative and invasive capacities of ovarian cancer cells by decreasing miR-4425 level. Yet the mechanism of action of miR-4425 in ovarian cancer remains unclear. Here we report that miR-4425 is upregulated in ovarian cancer tissues relative to normal ovarian tissues, and transfection of miR-4425 inhibitor impairs the proliferation, migration and invasion of SKOV3 and 3AO ovarian cancer cells. Further, miR-4425 antagomiR reduces cell proliferation in a subcutaneous SKOV3 xenograft model using BALB/c nude mice. We identifies farnesyl-diphosphate farnesyltransferase 1 (FDFT1) as a direct target of miR-4425 by Western blotting and a luciferase reporter assay. Forced expression of FDFT1 via transfection of an FDFT1-expressing plasmid into ovarian cancer cells not only retards cell proliferation, motility and invasiveness, but also negates the tumorigenic properties of a miR-4425 mimic. By contrast, silencing of FDFT1 by siRNAs abrogates suppression of the proliferation, migration and invasion of ovarian cancer cells treated with a miR-4425 inhibitor. Finally, transfection of either a miR-4425 mimic or FDFT1 siRNAs into 20(S)-Rg3-treated ovarian cancer cells counteracts the tumor-inhibitory activity of the ginsenoside. In conclusion, 20(S)-Rg3 exerts anti-ovarian cancer activity by downregulating oncogenic miR-4425 that inhibits the expression of the tumor suppressor gene FDFT1. These results expand our current understanding of the molecular pathways leading to ovarian cancer progression, and unveil the mechanism of action of 20(S)-Rg3 in ovarian cancer inhibition.
Collapse
|
23
|
Liang Y, Zhang T, Zhang J. Natural tyrosine kinase inhibitors acting on the epidermal growth factor receptor: Their relevance for cancer therapy. Pharmacol Res 2020; 161:105164. [PMID: 32846211 DOI: 10.1016/j.phrs.2020.105164] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Epidermal growth factor receptor (EGFR), also known as ErbB-1/HER-1, plays a key role in the regulation of the cell proliferation, migration, differentiation, and survival. Since the constitutive activation or overexpression of EGFR is nearly found in various cancers, the applications focused on EGFR are the most widely used in the clinical level, including the therapeutic drugs of targeting EGFR, monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs).Over the past decades, the compounds from natural sources have been a productive source of novel drugs, especially in both discovery and development of anti-tumor drugs by targeting the EGFR pathways as the TKIs. This work presents a review of the compounds from natural sources as potential EGFR-TKIs involved in the regulation of cancer. Moreover, high-throughput drug screening of EGFR-TKIs from the natural compounds has also been summarized.
Collapse
Affiliation(s)
- Yuan Liang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
24
|
Wang P, Song D, Wan D, Li L, Mei W, Li X, Han L, Zhu X, Yang L, Cai Y, Zhang R. Ginsenoside panaxatriol reverses TNBC paclitaxel resistance by inhibiting the IRAK1/NF-κB and ERK pathways. PeerJ 2020; 8:e9281. [PMID: 32547883 PMCID: PMC7275687 DOI: 10.7717/peerj.9281] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/12/2020] [Indexed: 12/17/2022] Open
Abstract
Background Paclitaxel (PTX) resistance is a major obstacle in the treatment of triple-negative breast cancer (TNBC). Previously, we have reported that interleukin-1 receptor-associated kinase 1 (IRAK1) and its downstream pathways are associated with PTX resistance in TNBC cells. In this study, we sought to investigate the combination treatment of ginsenoside panaxatriol (GPT), one of the main active components in Panax ginseng, with PTX on viability and apoptosis of TNBC PTX resistant cells, and explore the role of IRAK1 mediated signaling pathways in the therapeutic effects. Methods CellTiter-Glo and colony formation assays were used to assess cell viability. Flow cytometry was used to analyze subG1 and apoptosis. Western blot was used to detect expressions of proteins involved in apoptosis and the IRAK1/NF-κB and ERK pathways. The mRNA expression of inflammatory cytokines, S100A7/8/9 and cancer stem cell (CSC)-related genes were examined by qPCR. Stem cells were identified by tumor sphere assay. Cell invasion ability was examined by transwell assay. Results We show that GPT inhibits MDA-MB-231 PTX resistant (MB231-PR) cell viability in a dose-dependent manner. When combined with PTX, GPT synergistically causes more cell death, induces subG1 accumulation and cell apoptosis. Besides, up-regulation of BAX/BCL-2 ratio, and down-regulation of MCL-1 are also observed. Moreover, this combination inhibits IRAK1, NF-κB and ERK1/2 activation, and leads to down-regulation of inflammatory cytokines (IL6, IL8, CXCL1, CCL2), S100A7/9 and CSC-related genes (OCT4, SOX2, NANOG, ALDH1, CD44) expression. In addition, the combination treatment suppresses MB231-PR cell invasion ability, and impairs tumor sphere growth both in MB231-PR and SUM159 PTX resistant (SUM159-PR) cells. Conclusion Our study demonstrates that GPT can resensitize TNBC PTX resistant cells to PTX by inhibiting the IRAK1/NF-κB and ERK pathways and reducing stem cell characteristics.
Collapse
Affiliation(s)
- Panpan Wang
- College of Pharmacy, Jinan University, Guangzhou, China.,First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dan Song
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Danhong Wan
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lingyu Li
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenhui Mei
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaoyun Li
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Li Han
- First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaofeng Zhu
- First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Yang
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou, China.,Cancer Research Institute, Jinan University, Guangzhou, China
| | - Ronghua Zhang
- College of Pharmacy, Jinan University, Guangzhou, China.,Cancer Research Institute, Jinan University, Guangzhou, China
| |
Collapse
|
25
|
Wang Z, Wu W, Guan X, Guo S, Li C, Niu R, Gao J, Jiang M, Bai L, Leung EL, Hou Y, Jiang Z, Bai G. 20( S)-Protopanaxatriol promotes the binding of P53 and DNA to regulate the antitumor network via multiomic analysis. Acta Pharm Sin B 2020; 10:1020-1035. [PMID: 32642409 PMCID: PMC7332671 DOI: 10.1016/j.apsb.2020.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
Abstract
Although the tumor suppressor P53 is known to regulate a broad network of signaling pathways, it is still unclear how certain drugs influence these P53 signaling networks. Here, we used a comprehensive single-cell multiomics view of the effects of ginsenosides on cancer cells. Transcriptome and proteome profiling revealed that the antitumor activity of ginsenosides is closely associated with P53 protein. A miRNA–proteome interaction network revealed that P53 controlled the transcription of at least 38 proteins, and proteome-metabolome profiling analysis revealed that P53 regulated proteins involved in nucleotide metabolism, amino acid metabolism and “Warburg effect”. The results of integrative multiomics analysis revealed P53 protein as a potential key target that influences the anti-tumor activity of ginsenosides. Furthermore, by applying affinity mass spectrometry (MS) screening and surface plasmon resonance fragment library screening, we confirmed that 20(S)-protopanaxatriol directly targeted adjacent regions of the P53 DNA-binding pocket and promoted the stability of P53–DNA interactions, which further induced a series of omics changes.
Collapse
|
26
|
Effects of Ginsenoside Biopolymer Nanoparticles on the Malignant Behavior of Non-Small-Cell Lung Cancer. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/1796701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. To explore the effects and mechanism of ginsenoside Rg3 nanoparticles on the malignant behavior of non-small-cell lung cancer. Methods. The nanoparticle carriers were prepared by using an electrostatic system, and the coverage of ginsenoside Rg3 was determined by HPLC after coating the nanoparticle carriers with the ginsenoside Rg3 monomer. The proliferation of H125 cells was measured using MTT assay, and the Transwell assay was used to detect the invasiveness of H125 cells. Cell scratch test was used to determine the migration ability of H125 cells, and Western blotting was used to measure the expression level of PTEN in H125 cells; the expression level of miR-192 in H125 cells was measured via RT-qPCR, and the apoptosis level of H125 cells was detected by Tunel assay. Results. Firstly, gelatin nanoparticles and hyaluronic acid nanoparticles were uniformly distributed, uniform in size and spherical in shape, and after coating ginsenoside Rg3, the sizes of the nanoparticles were significantly increased. Secondly, the expression level of miR-192 was upregulated in H125 cells, which could be effectively inhibited by the treatment of Rg3 monomer and HA-Rg3 nanoparticles. Thirdly, the knockdown of miR-192 significantly inhibited H125 cell proliferation, invasion, and migration and also enhanced H125 cell apoptosis. In addition, PTEN was demonstrated as a target gene of miR-192. Finally, by inhibiting the expression level of miR-192 in H125 cells, the Rg3 monomer and HA-Rg3 nanoparticles upregulated the expression of PTEN and thus exerted its antitumor effect; the effects of HA-Rg3 were comparatively more significant than those of the Rg3 monomer. Conclusions. The Rg3 monomer and HA-Rg3 nanoparticles mitigated the malignant behavior of human non-small-cell lung cancer H125 cells through the miR-192/PTEN molecular axis, and HA-Rg3 nanoparticles showed better antitumor effects.
Collapse
|
27
|
Zhang T, Liang Y, Zuo P, Jing S, Li T, Wang Y, Lv C, Li D, Zhang J, Wei Z. 20(S)-Protopanaxadiol blocks cell cycle progression by targeting epidermal growth factor receptor. Food Chem Toxicol 2020; 135:111017. [DOI: 10.1016/j.fct.2019.111017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022]
|
28
|
Zhang S, Luo J, Xie J, Wang Z, Xiao W, Zhao L. Cooperated biotransformation of ginsenoside extracts into ginsenoside 20(
S
)‐Rg3 by three thermostable glycosidases. J Appl Microbiol 2019; 128:721-734. [DOI: 10.1111/jam.14513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022]
Affiliation(s)
- S. Zhang
- Co‐Innovation Center for Sustainable Forestry in Southern China Nanjing Forestry University Nanjing China
- College of Chemical Engineering Nanjing Forestry University Nanjing China
| | - J. Luo
- Co‐Innovation Center for Sustainable Forestry in Southern China Nanjing Forestry University Nanjing China
- College of Chemical Engineering Nanjing Forestry University Nanjing China
| | - J. Xie
- Co‐Innovation Center for Sustainable Forestry in Southern China Nanjing Forestry University Nanjing China
- College of Chemical Engineering Nanjing Forestry University Nanjing China
| | - Z. Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd Lianyungang China
| | - W. Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd Lianyungang China
| | - L. Zhao
- Co‐Innovation Center for Sustainable Forestry in Southern China Nanjing Forestry University Nanjing China
- College of Chemical Engineering Nanjing Forestry University Nanjing China
| |
Collapse
|
29
|
Shi Y, Wang H, Zheng M, Xu W, Yang Y, Shi F. Ginsenoside Rg3 suppresses the NLRP3 inflammasome activation through inhibition of its assembly. FASEB J 2019; 34:208-221. [PMID: 31914640 DOI: 10.1096/fj.201901537r] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 12/23/2022]
Abstract
Ginsenoside Rg3 is one of the main constituents of Panax ginseng. Compelling evidence has demonstrated that ginsenoside Rg3 is capable of inhibiting inflammation. However, the mechanism mediating its anti-inflammatory effects remain unclear. Here we show that ginsenoside Rg3 blocks IL-1β secretion and caspase-1 activation through inhibiting LPS priming and the NLRP3 inflammasome activation in human and mouse macrophages. Rg3 specifically inhibits activation of NLRP3 but not the NLRC4 or AIM2 inflammasomes. In addition, Rg3 has no effect on upstream regulation of NLRP3 inflammasome, such as K+ efflux, ROS production, or mitochondrial membrane potential. Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. More importantly, Rg3 can reduce IL-1β secretion induced by LPS in mice and protect mice from lethal endotoxic shock. Thus, our findings reveal an anti-inflammatory mechanism for Rg3 and suggest its potential use in NLRP3-driven diseases.
Collapse
Affiliation(s)
- Yuhua Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Huanan Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Mengjie Zheng
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yang Yang
- College of Animal Science and Technology, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Zhejiang A&F University, Hangzhou, China
| | - Fushan Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
30
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
31
|
Liu T, Zuo L, Guo D, Chai X, Xu J, Cui Z, Wang Z, Hou C. Ginsenoside Rg3 regulates DNA damage in non-small cell lung cancer cells by activating VRK1/P53BP1 pathway. Biomed Pharmacother 2019; 120:109483. [PMID: 31629252 DOI: 10.1016/j.biopha.2019.109483] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. Ginsenoside Rg3 is the main ingredient of Ginseng which is used to treat non-small cell lung cancer (NSCLC). It has been found to enhance the efficiency of chemotherapy thereby reducing its side effects. Previous studies found that ginsenoside Rg3 can reduce the occurrence of NSCLC by inducing DNA damage. Yet, its anti-DNA damaging effects and mechanisms in tumor cells are still not fully understood. This study explored the effect of ginsenoside Rg3 on DNA repair and VRK1/P53BP1 signaling pathway. Ginsenoside Rg3 treatment significantly decreased the incidence and invasionin a mouse model of lung cancer induced by urethane. The results of cell survival assay and single cell gel electrophoresis showed that ginsenoside Rg3 protected lung adenocarcinoma cells from DNA damage as well as inhibited the proliferation of tumor cells. Ginsenoside Rg3 increased the mRNA and protein expression of VRK1 in NSCLC cells as measured by RT-qPCR and western blot, respectively. These findings suggests that ginsenoside Rg3 regulates VRK1 signaling. Immunofluorescence assays showed that P53BP1 and VRK1 protein level increased, and the VRK1 protein translocated between the nuclei and cytoplasm. Finally, this conclusion was confirmed by the reverse validation in VRK1-knockdown cells. Taken together, these results show that ginsenoside Rg3 upregulate VRK1 expression and P53BP1 foci formation in response to DNA damage thereby inhibiting the tumorigenesis and viability of cancer cells. These findings reveal the role of Rg3 in lung cancer and provides therapeutic targets for developing new drugs in the prevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Tianhua Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ling Zuo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongqing Guo
- School of Life science, Beijing University of Chinese Medicine, Beijing, China
| | - Xinlou Chai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaorui Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiyi Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunying Hou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|