1
|
Peng L, Li S, Cai H, Chen X, Tang Y. Ginsenoside Rg1 treats chronic heart failure by downregulating ERK1/2 protein phosphorylation. In Vitro Cell Dev Biol Anim 2024; 60:1085-1098. [PMID: 39251466 DOI: 10.1007/s11626-024-00960-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
In this study, we investigated the potential therapeutic mechanism of ginsenoside Rg1 (GRg1) in chronic heart failure (CHF), focusing on its regulation of ERK1/2 protein phosphorylation. H9c2 cardiomyocytes and SD rats were divided into the control group, CHF (ADR) group, and CHF+ginsenoside Rg1 group using an isolated cardiomyocyte model and an in vivo CHF rat model induced by adriamycin (ADR). Cell viability, proliferation, apoptosis, and the expression of relevant proteins were measured to assess the effects of GRg1. The results showed that treatment with GRg1 increased cell activity and proliferation, while significantly reducing levels of inflammatory and apoptotic factors compared to the CHF (ADR) group. Moreover, the CHF+ginsenoside Rg1 group exhibited higher levels of Bcl-2 mRNA and protein expression, as well as lower levels of Caspase3 and Bax mRNA and protein expression, compared to the CHF (ADR) group. Notably, the CHF+ginsenoside Rg1 group displayed decreased serum NT-proBNP levels and heart weight/body weight (HW/BW) index. Furthermore, the electrocardiogram of rats in the CHF+ginsenoside Rg1 group resembled that of rats in the control group. Overall, our findings suggested that GRg1 alleviated CHF by inhibiting ERK1/2 protein phosphorylation, thereby inhibiting apoptosis, enhancing cell activity and proliferation, and reducing cardiac inflammatory responses.
Collapse
Affiliation(s)
- Liqi Peng
- First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Shaodong Li
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Huzhi Cai
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Xueliang Chen
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yanping Tang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
2
|
Lin L, Chen D, Li S, Wang T. Ginsenoside Rg1 inhibits multiple myeloma and overcomes bortezomib resistance through AMPK-mTOR pathway. Heliyon 2024; 10:e33935. [PMID: 39071579 PMCID: PMC11283037 DOI: 10.1016/j.heliyon.2024.e33935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Background The resistance of multiple myeloma (MM) to bortezomib (BTZ) has brought multiple challenges to its clinical use. Numerous ginsenosides have potential anti-tumor effects, however, the research on the role of Rg1 in MM has not been reported. Objective To examine the inhibitory impact of Rg1 on the growth of MM and reduce the drug resistance of MM to BTZ through in vivo and in vitro experiments, and to explore their potential mechanism. Methods BTZ drug-resistant cell line RPMI8226R was constructed. Mouse tumor-bearing model was developed by abdominal subcutaneous injection of MM cells. MM cells were treated with AMPK inhibitor Compound C or autophagy inhibitor Chloroquine together with Rg1. RPMI8226R cells were treated with BTZ and Rg1. Cell multiplication was detected using Methylthiazolyldiphenyl-tetrazolium bromide assay. Apoptosis was assessed using flow cytometry. Immunofluorescence assay was employed to assess the autophagy markers LC3. Western blot was utilized to assess the protein expression. Immunohistochemistry was used to detect cell proliferation and apoptosis in tumor tissues. Results In vitro experiments demonstrated that Rg1 could hinder the proliferation of MM cells, promote apoptosis and enhance autophagy. Rg1 could also increase the sensitivity of RPMI8226R to BTZ. In vivo experiments illustrated that Rg1 could hinder the development of MM cells in mice, weaken the proliferation of tumor cells and enhance their apoptosis. Further study found that the anti-MM impact of Rg1 was linked to AMPK-mTOR pathway, the autophagy degree of RPMI8226R was higher than that of RPMI8226, and that Rg1 could inhibit MM and overcome drug resistance through autophagy induced by AMPK-mTOR pathway. Conclusion Rg1 has significant anti-MM effect and can overcome BTZ resistance, and its potential mechanism is related to the regulation of autophagy induced by AMPK-mTOR pathway. Rg1 is a promising adjuvant drug for the treatment of MM.
Collapse
Affiliation(s)
- Li Lin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Dong Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tiantian Wang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
3
|
Szponar J, Niziński P, Dudka J, Kasprzak-Drozd K, Oniszczuk A. Natural Products for Preventing and Managing Anthracycline-Induced Cardiotoxicity: A Comprehensive Review. Cells 2024; 13:1151. [PMID: 38995002 PMCID: PMC11240786 DOI: 10.3390/cells13131151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.
Collapse
Affiliation(s)
- Jarosław Szponar
- Clinical Department of Toxicology and Cardiology, Toxicology Clinic, Stefan Wyszyński Regional Specialist Hospital, Medical University of Lublin, 20-718 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11 Street, 20-080 Lublin, Poland;
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
4
|
Mancuso C. Panax notoginseng: Pharmacological Aspects and Toxicological Issues. Nutrients 2024; 16:2120. [PMID: 38999868 PMCID: PMC11242943 DOI: 10.3390/nu16132120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/30/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Current evidence suggests a beneficial role of herbal products in free radical-induced diseases. Panax notoginseng (Burk.) F. H. Chen has long occupied a leading position in traditional Chinese medicine because of the ergogenic, nootropic, and antistress activities, although these properties are also acknowledged in the Western world. The goal of this paper is to review the pharmacological and toxicological properties of P. notoginseng and discuss its potential therapeutic effect. A literature search was carried out on Pubmed, Scopus, and the Cochrane Central Register of Controlled Trials databases. The following search terms were used: "notoginseng", "gut microbiota", "immune system", "inflammation", "cardiovascular system", "central nervous system", "metabolism", "cancer", and "toxicology". Only peer-reviewed articles written in English, with the full text available, have been included. Preclinical evidence has unraveled the P. notoginseng pharmacological effects in immune-inflammatory, cardiovascular, central nervous system, metabolic, and neoplastic diseases by acting on several molecular targets. However, few clinical studies have confirmed the therapeutic properties of P. notoginseng, mainly as an adjuvant in the conventional treatment of cardiovascular disorders. Further clinical studies, which both confirm the efficacy of P. notoginseng in free radical-related diseases and delve into its toxicological aspects, are mandatory to broaden its therapeutic potential.
Collapse
Affiliation(s)
- Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168 Rome, Italy; ; Tel.: +39-06-30154367; Fax: +39-06-3050159
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
5
|
Ni B, Xue K, Wang J, Zhou J, Wang L, Wang X, Liu T, Ye N, Jiang J. Integrating Chinese medicine into mainstream cancer therapies: a promising future. Front Oncol 2024; 14:1412370. [PMID: 38957318 PMCID: PMC11217489 DOI: 10.3389/fonc.2024.1412370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
Malignant tumors are complex systemic chronic diseases and one of the major causes of human mortality. Targeted therapy, chemotherapy, immunotherapy, and radiotherapy are examples of mainstream allopathic medicine treatments that effective for intermediate and advanced malignant tumors. The ongoing use of conventional allopathic medicine has resulted in adverse responses and drug resistance, which have hampered its efficacy. As an important component of complementary and alternative medicine, Chinese medicine has been found to have antitumor effects and has played an important role in enhancing the therapeutic sensitivity of mainstream allopathic medicine, reducing the incidence of adverse events and improving immune-related functions. The combined application of adjuvant Chinese medicine and mainstream allopathic medicine has begun to gain acceptance and is gradually used in the field of antitumor therapy. Traditional natural medicines and their active ingredients, as well as Chinese patent medicines, have been proven to have excellent therapeutic efficacy and good safety in the treatment of various malignant tumors. This paper focuses on the mechanism of action and research progress of combining the above drugs with mainstream allopathic medicine to increase therapeutic sensitivity, alleviate drug resistance, reduce adverse reactions, and improve the body's immune function. To encourage the clinical development and use of Chinese herb adjuvant therapy as well as to provide ideas and information for creating safer and more effective anticancer medication combinations, the significant functions of Chinese herb therapies as adjuvant therapies for cancer treatment are described in detail.
Collapse
Affiliation(s)
- Baoyi Ni
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Kaiyuan Xue
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Jilai Zhou
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lankang Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinmiao Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ting Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Naijing Ye
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiakang Jiang
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Yu X, Wang Q, Dai Z. Ginsenosides Inhibit the Proliferation of Lung Cancer Cells and Suppress the Rate of Metastasis by Modulating EGFR/VEGF Signaling Pathways. J Oleo Sci 2024; 73:219-230. [PMID: 38311411 DOI: 10.5650/jos.ess23120] [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] [Indexed: 02/10/2024] Open
Abstract
Ginsenosides Rg3 and Rg5 obtained from Panax (ginseng) have shown significant anticancer activity via the PI3K-Akt signaling pathway. This study evaluated the anticancer and antimetastatic effects of a combination of Rg3 and Rg5 on lung cancer cells. A combination of Rg3 and Rg5 was treated for lung cancer cell line A549 and human lung tumor xenograft mouse model, and anti-metastatic effects on Matrigel plug implantation in mice. The combination of Rg3 and Rg5 showed potent antiproliferative effects on A549 cells with IC50 values of 44.6 and 36.0 μM for Rg3 and Rg5 respectively. The combination of Rg3 and Rg5 (30 µM each) showed 48% cell viability as compared to Rg3 (72% viability) and Rg5 (64% viability) at 30 µM concentrations. The combination of Rg3 and Rg5 induced apoptosis in A549 cells characterized by activation of caspase-9 and caspase-3 and cleavage of PARP, as well as suppression of the autophagic marker LC3A/B. The antitumoral potentials of the combination of Rg3 and Rg5 were ascertained in a lung tumor xenograft mouse model with high efficacy as compared to individual ginsenosides. The metastasislimiting properties of the combination of Rg3 and Rg5 were assessed in Matrigel plug implantation in mice which showed the potent efficacy of the combination as compared to individual ginsenoside. Mechanistically, the combination of Rg3 and Rg5 inhibited the expression of PI3K/Akt/mTOR and EGFR/VEGF signaling pathways in lung cancer cells. Results suggest that the combination of Rg3 and Rg5 suppressed the tumor cell proliferation in lung cancer cells and limited the rate of metastasis which further suggest that the combination has a significant effect as compared to the administration of single ginsenoside.
Collapse
Affiliation(s)
- Xuelian Yu
- Department of Pulmonary, Muping Chinese Traditional Medical Hospital
| | - Qihu Wang
- Department of Pulmonary, Muping Chinese Traditional Medical Hospital
| | - Zhaoxin Dai
- Department of Pulmonary, Muping Chinese Traditional Medical Hospital
| |
Collapse
|
7
|
Jiang RY, Fang ZR, Zhang HP, Xu JY, Zhu JY, Chen KY, Wang W, Jiang X, Wang XJ. Ginsenosides: changing the basic hallmarks of cancer cells to achieve the purpose of treating breast cancer. Chin Med 2023; 18:125. [PMID: 37749560 PMCID: PMC10518937 DOI: 10.1186/s13020-023-00822-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/16/2023] [Indexed: 09/27/2023] Open
Abstract
In 2021, breast cancer accounted for a substantial proportion of cancer cases and represented the second leading cause of cancer deaths among women worldwide. Although tumor cells originate from normal cells in the human body, they possess distinct biological characteristics resulting from changes in gene structure and function of cancer cells in contrast with normal cells. These distinguishing features, known as hallmarks of cancer cells, differ from those of normal cells. The hallmarks primarily include high metabolic activity, mitochondrial dysfunction, and resistance to cell death. Current evidence suggests that the fundamental hallmarks of tumor cells affect the tissue structure, function, and metabolism of tumor cells and their internal and external environment. Therefore, these fundamental hallmarks of tumor cells enable tumor cells to proliferate, invade and avoid apoptosis. Modifying these hallmarks of tumor cells represents a new and potentially promising approach to tumor treatment. The key to breast cancer treatment lies in identifying the optimal therapeutic agent with minimal toxicity to normal cells, considering the specific types of tumor cells in patients. Some herbal medicines contain active ingredients which can precisely achieve this purpose. In this review, we introduce Ginsenoside's mechanism and research significance in achieving the therapeutic effect of breast cancer by changing the functional hallmarks of tumor cells, providing a new perspective for the potential application of Ginsenoside as a therapeutic drug for breast cancer.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Zi-Ru Fang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Jun-Yao Xu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Ke-Yu Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Wei Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Xiao Jiang
- Department of Basic Medical Sciences, Guangxi University of Chinese Medicine, NO. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China.
| | - Xiao-Jia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
8
|
Li X, Cao D, Sun S, Wang Y. Anticancer therapeutic effect of ginsenosides through mediating reactive oxygen species. Front Pharmacol 2023; 14:1215020. [PMID: 37564184 PMCID: PMC10411515 DOI: 10.3389/fphar.2023.1215020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Dysregulation of reactive oxygen species (ROS) production and ROS-regulated pathways in cancer cells leads to abnormal accumulation of reactive oxygen species, displaying a double-edged role in cancer progression, either supporting transformation/proliferation and stimulating tumorigenesis or inducing cell death. Cancer cells can accommodate reactive oxygen species by regulating them at levels that allow the activation of pro-cancer signaling pathways without inducing cell death via modulation of the antioxidant defense system. Therefore, targeting reactive oxygen species is a promising approach for cancer treatment. Ginsenosides, their derivatives, and related drug carriers are well-positioned to modulate multiple signaling pathways by regulating oxidative stress-mediated cellular and molecular targets to induce apoptosis; regulate cell cycle arrest and autophagy, invasion, and metastasis; and enhance the sensitivity of drug-resistant cells to chemotherapeutic agents of different cancers depending on the type, level, and source of reactive oxygen species, and the type and stage of the cancer. Our review focuses on the pro- and anticancer effects of reactive oxygen species, and summarizes the mechanisms and recent advances in different ginsenosides that bring about anticancer effects by targeting reactive oxygen species, providing new ideas for designing further anticancer studies or conducting more preclinical and clinical studies.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Siming Sun
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Xie Y, Wang C. Herb-drug interactions between Panax notoginseng or its biologically active compounds and therapeutic drugs: A comprehensive pharmacodynamic and pharmacokinetic review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116156. [PMID: 36754189 DOI: 10.1016/j.jep.2023.116156] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herbs, along with the use of herb-drug interactions (HDIs) to combat diseases, are increasing in popularity worldwide. HDIs have two effects: favorable interactions that tend to improve therapeutic outcomes and/or minimize the toxic effects of drugs, and unfavorable interactions aggravating the condition of patients. Panax notoginseng (Burk.) F.H. Chen is a medicinal plant that has long been commonly used in traditional Chinese medicine to reduce swelling, relieve pain, clear blood stasis, and stop bleeding. Numerous studies have demonstrated the existence of intricate pharmacodynamic (PD) and pharmacokinetic (PK) interactions between P. notoginseng and conventional drugs. However, these HDIs have not been systematically summarized. AIM OF THE REVIEW To collect the available literature on the combined applications of P. notoginseng and drugs published from 2005 to 2022 and summarize the molecular mechanisms of interactions to circumvent the potential risks of combination therapy. MATERIALS AND METHODS This work was conducted by searching PubMed, Scopus, Web of Science, and CNKI databases. The search terms included "notoginseng", "Sanqi", "drug interaction," "synergy/synergistic", "combination/combine", "enzyme", "CYP", and "transporter". RESULTS P. notoginseng and its bioactive ingredients interact synergistically with numerous drugs, including anticancer, antiplatelet, and antimicrobial agents, to surmount drug resistance and side effects. This review elaborates on the molecular mechanisms of the PD processed involved. P. notoginseng shapes the PK processes of the absorption, distribution, metabolism, and excretion of other drugs by regulating metabolic enzymes and transporters, mainly cytochrome P450 enzymes and P-glycoprotein. This effect is a red flag for drugs with a narrow therapeutic window. Notably, amphipathic saponins in P. notoginseng act as auxiliary materials in drug delivery systems to enhance drug solubility and absorption and represent a new entry point for studying interactions. CONCLUSION This article provides a comprehensive overview of HDIs by analyzing the results of the in vivo and in vitro studies on P. notoginseng and its bioactive components. The knowledge presented here offers a scientific guideline for investigating the clinical importance of combination therapies. Physicians and patients need information on possible interactions between P. notoginseng and other drugs, and this review can help them make scientific predictions regarding the consequences of combination treatments.
Collapse
Affiliation(s)
- Yujuan Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
10
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
|
11
|
Telang NT, Nair HB, Wong GYC. Growth Inhibitory Efficacy of Chinese Herbs in a Cellular Model for Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:ph14121318. [PMID: 34959717 PMCID: PMC8708199 DOI: 10.3390/ph14121318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor-α progesterone receptor and human epidermal growth factor receptor-2. Treatment for this breast cancer subtype is restricted to multidrug chemotherapy and survival pathway-based molecularly targeted therapy. The long-term treatment options are associated with systemic toxicity, spontaneous and/or acquired tumor resistance and the emergence a of drug-resistant stem cell population. These limitations lead to advanced stage metastatic cancer. Current emphasis is on research directions that identify efficacious, naturally occurring agents representing an unmet need for testable therapeutic alternatives for therapy resistant breast cancer. Chinese herbs are widely used in traditional Chinese medicine in women for estrogen related health issues and also for integrative support for cancer treatment. This review discusses published evidence on a TNBC model for growth inhibitory effects of several mechanistically distinct nontoxic Chinese herbs, most of them nutritional in nature, and identifies susceptible pathways and potential molecular targets for their efficacy. Documented anti-proliferative and pro-apoptotic effects of these herbs are associated with downregulation of RB, RAS, PI3K, and AKT signaling, modulation of Bcl-2/BAX protein expressions and increased caspase activity. This review provides a proof of concept for Chinese herbs as testable alternatives for prevention/therapy of TNBC.
Collapse
Affiliation(s)
- Nitin T. Telang
- Cancer Prevention Research Program, Palindrome Liaisons Consultants, Montvale, NJ 07645, USA
- Correspondence: ; Tel.: +1-646-512-3277
| | - Hareesh B. Nair
- Department of Obstetrics and Gynecology, University of Texas Health Sciences Center, San Antonio, TX 78229, USA;
| | - George Y. C. Wong
- Breast Center, Maimonides Medical Center, Brooklyn, NY 11219, USA;
- American Foundation for Chinese Medicine, New York, NY 11103, USA
| |
Collapse
|