1
|
Chen C, Bu X, Deng L, Xia J, Wang X, Chen L, Li W, Huang J, Chen Q, Wang C. Astragaloside IV as a promising therapeutic agent for liver diseases: current landscape and future perspectives. Front Pharmacol 2025; 16:1574154. [PMID: 40337517 PMCID: PMC12055773 DOI: 10.3389/fphar.2025.1574154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Astragaloside IV (C41H68O14, AS-IV) is a naturally occurring saponin isolated from the root of Astragalus membranaceus, a widely used traditional Chinese botanical drug in medicine. In recent years, AS-IV has attracted considerable attention for its hepatoprotective properties, which are attributed to its low toxicity as well as its anti-inflammatory, antioxidant and antitumour effects. Numerous preclinical studies have demonstrated its potential in the prevention and treatment of various liver diseases, including multifactorial liver injury, metabolic-associated fatty liver disease, liver fibrosis and liver cancer. Given the promising hepatoprotective potential of AS-IV and the growing interest in its research, this review provides a comprehensive summary of the current state of research on the hepatoprotective effects of AS-IV, based on literature available in databases such as CNKI, PubMed, ScienceDirect, Google Scholar and Web of Science. The hepatoprotective mechanisms of AS-IV are multifaceted, encompassing the inhibition of inflammatory responses, reduction of oxidative stress, improvement of insulin and leptin resistance, modulation of the gut microbiota, suppression of hepatocellular carcinoma cell proliferation and induction of tumour cell apoptosis. Notably, key molecular pathways involved in these effects include Nrf2/HO-1, NF-κB, NLRP3/Caspase-1, JNK/c-Jun/AP-1, PPARα/FSP1 and Akt/GSK-3β/β-catenin. Toxicity studies indicate that AS-IV has a high level of safety. In addition, this review discusses the sources, physicochemical properties, and current challenges in the development and clinical application of AS-IV, providing valuable insights into its potential as a hepatoprotective agent in the pharmaceutical and nutraceutical industries.
Collapse
Affiliation(s)
- Chunyan Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaolan Bu
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Liping Deng
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jiayan Xia
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xinming Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Li Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wen Li
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jie Huang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qixiang Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Cheng Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
2
|
Wang T, Chen X, Gao Q, Huang C, Wang K, Qiu F. Herb-drug interaction potential of Astragali Radix: a metabolic perspective. Drug Metab Rev 2025; 57:9-25. [PMID: 39692050 DOI: 10.1080/03602532.2024.2441235] [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: 08/02/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
Astragali Radix (AR) is one of the most widely used herbs in Asia and has a wide range of biological activities. These activities are attributed to its various compounds like flavonoids, saponins, and polysaccharides. AR and its major components are often used in combination with other drugs for the treatment of diseases such as cancer and cerebral ischemia. With the expanding range of AR combinations, the potential for herb-drug interaction (HDI) has been raised. Key targets in HDI studies include drug-metabolizing enzymes (DMEs) and transporters. Existing studies have shown that AR and its major components have various regulatory effects on these targets, notably CYP2C9, CYP3A4, UGT1A6, and P-gp. AR may contribute to HDI when it is taken with substrates of these biomolecules, such as tolbutamide, midazolam, and digoxin. However, there are also different views in the current study, such as the effect of AR on CYP3A4. To better understand the interactions of AR with drugs, we review the metabolic pathways and pharmacokinetic parameters of the main components of AR. Meanwhile, the regulatory effects and mechanisms of AR on DMEs and transporters are summarized to provide a theoretical and technical basis for the rational use of AR in clinical practice.
Collapse
Affiliation(s)
- Tianwang Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- State Key Laboratory of Component based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Xiaofei Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- State Key Laboratory of Component based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Qing Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Chonggang Huang
- Chongqing Academy of Chinese Materia Medica, Chongqing, P.R. China
| | - Kai Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Tianjin Key Laboratory of Therapeutic substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- State Key Laboratory of Component based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
3
|
Koide E, Pietz HL, Beltran J, Chen J. Structural basis for the transport and regulation mechanism of the multidrug resistance-associated protein 2. Nat Commun 2025; 16:484. [PMID: 39779684 PMCID: PMC11711199 DOI: 10.1038/s41467-024-55810-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: 06/21/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Multidrug resistance-associated protein 2 (MRP2) is an ATP-powered exporter important for maintaining liver homeostasis and a potential contributor to chemotherapeutic resistance. Using cryogenic electron microscopy (cryo-EM), we determine the structures of human MRP2 in three conformational states: an autoinhibited state, a substrate-bound pre-translocation state, and an ATP-bound post-translocation state. In the autoinhibited state, the cytosolic regulatory (R) domain plugs into the transmembrane substrate-binding site and extends into the cytosol to form a composite ATP-binding site at the surface of nucleotide-binding domain 2. Substrate displaces the R domain, permitting conformational changes necessary for transport. These observations suggest that the R domain functions as a selectivity gauge, where only at sufficiently high concentrations can the substrate effectively initiate transport. Comparative structural analyzes of MRP2 bound to various substrates, as determined in this study and others, reveal how MRP2 recognizes a diverse array of compounds, supporting its role in multidrug resistance.
Collapse
Affiliation(s)
- Eriko Koide
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Harlan L Pietz
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Jean Beltran
- Department of Biology, Davidson College, Davidson, NC, USA
| | - Jue Chen
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, The Rockefeller University, 1230 York Ave, New York, NY, USA.
| |
Collapse
|
4
|
Li J, Niu Y, Yuan L, Jiang W, Jiao T, Dou H, Nan Y. Research Progress in the Medicine-Food Dual Use of Astragalus for Gastrointestinal Tumors. J Med Food 2024; 27:1145-1157. [PMID: 39431943 DOI: 10.1089/jmf.2024.k.0057] [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: 10/22/2024] Open
Abstract
Gastrointestinal tumors have a major impact on human life expectancy and quality of life and are a major cause of personal and social hygiene stress. Gastrointestinal tumors are the main cause of cancer-related death, and the main treatment methods are surgery, radiotherapy, and chemotherapy. However, they also cause great damage to the body and have a poor prognosis after surgery. Therefore, we urgently need safe and effective drugs to intervene in gastrointestinal tumors. In recent years, Traditional Chinese Medicine has been widely used in tumor treatment as a complementary and alternative therapy. Astragalus membranaceus is one of the main herbal medicines with tonic effect and one of the important components of many antitumor herbal compounds. Astragalus polysaccharides, saponins, and flavonoids are the main active components of Astragalus, all of which have antitumor effects. In this article, we studied the mechanism of action of Astragalus and its active ingredients in the intervention of gastrointestinal tumors in recent years and suggested a new approach for the study of Astragalus intervention in gastrointestinal tumors from the perspective of the homology of medicine and food.
Collapse
Affiliation(s)
- Jiaqing Li
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, China
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, China
| | - Yang Niu
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, China
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, China
| | - Ling Yuan
- Pharmacy College of Ningxia Medical University, Yinchuan, China
| | - Wenjie Jiang
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, China
| | - Taiqiang Jiao
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, China
| | - Hongli Dou
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, China
- Marxist College of Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yi Nan
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Yinchuan, China
| |
Collapse
|
5
|
Xiu W, Zhang Y, Tang D, Lee SH, Zeng R, Ye T, Li H, Lu Y, Qin C, Yang Y, Yan X, Wang X, Hu X, Chu M, Sun Z, Xu W. Inhibition of EREG/ErbB/ERK by Astragaloside IV reversed taxol-resistance of non-small cell lung cancer through attenuation of stemness via TGFβ and Hedgehog signal pathway. Cell Oncol (Dordr) 2024; 47:2201-2215. [PMID: 39373858 DOI: 10.1007/s13402-024-00999-7] [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] [Accepted: 09/25/2024] [Indexed: 10/08/2024] Open
Abstract
PURPOSE Taxol is the first-line chemo-drug for advanced non-small cell lung cancer (NSCLC), but it frequently causes acquired resistance, which leads to the failure of treatment. Therefore, it is critical to screen and characterize the mechanism of the taxol-resistance reversal agent that could re-sensitize the resistant cancer cells to chemo-drug. METHOD The cell viability, sphere-forming and xenografts assay were used to evaluate the ability of ASIV to reverse taxol-resistance. Immunohistochemistry, cytokine application, small-interfering RNA, small molecule inhibitors, and RNA-seq approaches were applied to characterize the molecular mechanism of inhibition of epiregulin (EREG) and downstream signaling by ASIV to reverse taxol-resistance. RESULTS ASIV reversed taxol resistance through suppression of the stemness-associated genes of spheres in NSCLC. The mechanism exploration revealed that ASIV promoted the K48-linked polyubiquitination of EREG along with degradation. Moreover, EREG could be triggered by chemo-drug treatment. Consequently, EREG bound to the ErbB receptor and activated the ERK signal to regulate the expression of the stemness-associated genes. Inhibition of EREG/ErbB/ERK could reverse the taxol-resistance by inhibiting the stemness-associated genes. Finally, it was observed that TGFβ and Hedgehog signaling were downstream of EREG/ErbB/ERK, which could be targeted using inhibitors to reverse the taxol resistance of NSCLC. CONCLUSIONS These findings revealed that inhibition of EREG by ASIV reversed taxol-resistance through suppression of the stemness of NSCLC via EREG/ErbB/ERK-TGFβ, Hedgehog axis.
Collapse
Affiliation(s)
- Wenhao Xiu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yujia Zhang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Clinical Medicine, Suzhou Vocational Health College, Suzhou, Jiangsu, China
| | - Dongfang Tang
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Sau Har Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Rui Zeng
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tingjie Ye
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yanlin Lu
- Department of Oncology and Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Changtai Qin
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuxi Yang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaofeng Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoling Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xudong Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Maoquan Chu
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Zhumei Sun
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wei Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
6
|
Yang C, Deng X, Tang Y, Tang H, Xia C. Natural products reverse cisplatin resistance in the hypoxic tumor microenvironment. Cancer Lett 2024; 598:217116. [PMID: 39002694 DOI: 10.1016/j.canlet.2024.217116] [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: 03/21/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Cisplatin is one of the most commonly used drugs for cancer treatment. Despite much progress in improving patient outcomes, many patients are resistant to cisplatin-based treatments, leading to limited treatment efficacy and increased treatment failure. The fact that solid tumors suffer from hypoxia and an inadequate blood supply in the tumor microenvironment has been widely accepted for decades. Numerous studies have shown that a hypoxic microenvironment significantly reduces the sensitivity of tumor cells to cisplatin. Therefore, understanding how hypoxia empowers tumor cells with cisplatin resistance is essential. In the fight against tumors, developing innovative strategies for overcoming drug resistance has attracted widespread interest. Natural products have historically made major contributions to anticancer drug research due to their obvious efficacy and abundant candidate resources. Intriguingly, natural products show the potential to reverse chemoresistance, which provides new insights into cisplatin resistance in the hypoxic tumor microenvironment. In this review, we describe the role of cisplatin in tumor therapy and the mechanisms by which tumor cells generate cisplatin resistance. Subsequently, we call attention to the linkage between the hypoxic microenvironment and cisplatin resistance. Furthermore, we summarize known and potential natural products that target the hypoxic tumor microenvironment to overcome cisplatin resistance. Finally, we discuss the current challenges that limit the clinical application of natural products. Understanding the link between hypoxia and cisplatin resistance is the key to unlocking the full potential of natural products, which will serve as new therapeutic strategies capable of overcoming resistance.
Collapse
Affiliation(s)
- Chuansheng Yang
- Department of Breast, Thyroid and Head-Neck Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, 512099, China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yunyun Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Chenglai Xia
- Foshan Maternity and Child Health Care Hospital, Foshan, 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China.
| |
Collapse
|
7
|
Li Z, Liu J, Cui H, Qi W, Tong Y, Wang T. Astragalus membranaceus: A Review of Its Antitumor Effects on Non-Small Cell Lung Cancer. Cancer Manag Res 2024; 16:909-919. [PMID: 39081698 PMCID: PMC11287463 DOI: 10.2147/cmar.s466633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/08/2024] [Indexed: 08/02/2024] Open
Abstract
The rising global morbidity and mortality rates of non-small cell lung cancer (NSCLC) underscore the urgent need for more effective treatments. Current therapeutic modalities-including surgery, radiotherapy, chemotherapy, and targeted therapy-face several limitations. Recently, Astragalus membranaceus, a traditional Chinese medicine (TCM), has captured significant attention due to its broad pharmacological properties, such as immune regulation, anti-inflammatory effects, and the modulation of reactive oxygen species (ROS) and enzyme activities. This review delivers a comprehensive summary of the most recent advancements and ongoing applications of Astragalus membranaceus in NSCLC treatment, underlining its potential for integration into existing treatment protocols. It also highlights essential areas for future research, including the elucidation of its molecular mechanisms, optimization of dosage and administration, and evaluation of its efficacy and safety alongside standard therapies, all of which could potentially improve therapeutic outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Zhenyu Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, People’s Republic of China
| | - Jimin Liu
- Department of Respiratory, The Third Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130117, People’s Republic of China
| | - Haishan Cui
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130117, People’s Republic of China
| | - Wenlong Qi
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, People’s Republic of China
| | - Yangyang Tong
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, People’s Republic of China
| | - Tan Wang
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, People’s Republic of China
| |
Collapse
|
8
|
Sheng F, Yang S, Li M, Wang J, Liu L, Zhang L. Research Progress on the Anti-Cancer Effects of Astragalus membranaceus Saponins and Their Mechanisms of Action. Molecules 2024; 29:3388. [PMID: 39064966 PMCID: PMC11280308 DOI: 10.3390/molecules29143388] [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: 06/12/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Astragalus membranaceus saponins are the main components of A. membranaceus, a plant widely used in traditional Chinese medicine. Recently, research on the anti-cancer effects of A. membranaceus saponins has received increasing attention. Numerous in vitro and in vivo experimental data indicate that A. membranaceus saponins exhibit significant anti-cancer effects through multiple mechanisms, especially in inhibiting tumor cell proliferation, migration, invasion, and induction of apoptosis, etc. This review compiles relevant studies on the anti-cancer properties of A. membranaceus saponins from various databases over the past two decades. It introduces the mechanism of action of astragalosides, highlighting their therapeutic benefits in the management of cancer. Finally, the urgent problems in the research process are highlighted to promote A. membranaceus saponins as an effective drug against cancer.
Collapse
Affiliation(s)
- Feiya Sheng
- College of Pharmacy, Chengdu University, Chengdu 610106, China; (F.S.); (S.Y.); (M.L.); (J.W.)
| | - Siyu Yang
- College of Pharmacy, Chengdu University, Chengdu 610106, China; (F.S.); (S.Y.); (M.L.); (J.W.)
| | - Mi Li
- College of Pharmacy, Chengdu University, Chengdu 610106, China; (F.S.); (S.Y.); (M.L.); (J.W.)
| | - Jiaojiao Wang
- College of Pharmacy, Chengdu University, Chengdu 610106, China; (F.S.); (S.Y.); (M.L.); (J.W.)
| | - Lianghong Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Lele Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| |
Collapse
|
9
|
Guo J, Le Y, Yuan A, Liu J, Chen H, Qiu J, Wang C, Dou X, Yuan X, Lu D. Astragaloside IV ameliorates cisplatin-induced liver injury by modulating ferroptosis-dependent pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118080. [PMID: 38521426 DOI: 10.1016/j.jep.2024.118080] [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: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The use of antineoplastic drugs, such as cisplatin, in clinical practice can cause adverse effects in patients, such as liver injury, which limits their long-term use. Therefore, there is an urgent need to develop alternative therapeutic strategies or drugs to minimize cisplatin-induced liver injury. Huangqi, the root of Astragalus membranaceus, is extensively used in traditional Chinese medicine (TCM) and has been employed in treating diverse liver injuries. Astragalus membranaceus contains several bioactive constituents, including triterpenoid saponins, one of which, astragaloside IV (ASIV), has been reported to have anti-inflammatory and antioxidant stress properties. However, its potential in ameliorating cisplatin-induced liver injury has not been explored. AIM OF THE STUDY The objective of this study was to examine the mechanism by which ASIV protects against cisplatin-induced liver injury. MATERIALS AND METHODS This study established a model of cisplatin-induced liver injury in mice, followed by treatment with various doses of astragaloside IV (40 mg/kg, 80 mg/kg). In addition, a model of hepatocyte ferroptosis in AML-12 cells was established using RSL3. The mechanism of action of astragaloside IV was investigated using a range of methods, including Western blot assay, qPCR, immunofluorescence, histochemistry, molecular docking, and high-content imaging system. RESULTS The findings suggested a significant improvement in hepatic injury, inflammation and oxidative stress phenotypes with the administration of ASIV. Furthermore, network pharmacological analyses provided evidence that a major pathway for ASIV to attenuate cisplatin-induced hepatic injury entailed the cell death cascade pathway. It was observed that ASIV effectively inhibited ferroptosis both in vivo and in vitro. Subsequent experimental outcomes provided further validation of ASIV's ability to hinder ferroptosis through the inhibition of PPARα/FSP1 signaling pathway. The current findings suggest that ASIV could function as a promising phytotherapy composition to alleviate cisplatin-induced liver injury. CONCLUSIONS The current findings suggest that astragaloside IV could function as a promising phytotherapy composition to alleviate cisplatin-induced liver injury.
Collapse
Affiliation(s)
- Jianan Guo
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Aini Yuan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Jing Liu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Hang Chen
- Department of Medical Research Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China.
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Cui Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Xingyu Yuan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Dezhao Lu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
10
|
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
|
11
|
Mazza T, Roumeliotis TI, Garitta E, Drew D, Rashid ST, Indiveri C, Choudhary JS, Linton KJ, Beis K. Structural basis for the modulation of MRP2 activity by phosphorylation and drugs. Nat Commun 2024; 15:1983. [PMID: 38438394 PMCID: PMC10912322 DOI: 10.1038/s41467-024-46392-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
Multidrug resistance-associated protein 2 (MRP2/ABCC2) is a polyspecific efflux transporter of organic anions expressed in hepatocyte canalicular membranes. MRP2 dysfunction, in Dubin-Johnson syndrome or by off-target inhibition, for example by the uricosuric drug probenecid, elevates circulating bilirubin glucuronide and is a cause of jaundice. Here, we determine the cryo-EM structure of rat Mrp2 (rMrp2) in an autoinhibited state and in complex with probenecid. The autoinhibited state exhibits an unusual conformation for this class of transporter in which the regulatory domain is folded within the transmembrane domain cavity. In vitro phosphorylation, mass spectrometry and transport assays show that phosphorylation of the regulatory domain relieves this autoinhibition and enhances rMrp2 transport activity. The in vitro data is confirmed in human hepatocyte-like cells, in which inhibition of endogenous kinases also reduces human MRP2 transport activity. The drug-bound state reveals two probenecid binding sites that suggest a dynamic interplay with autoinhibition. Mapping of the Dubin-Johnson mutations onto the rodent structure indicates that many may interfere with the transition between conformational states.
Collapse
Affiliation(s)
- Tiziano Mazza
- Department of Life Sciences, Imperial College London, SW7 2AZ, London, UK
- Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, Oxfordshire, OX11 0FA, UK
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Theodoros I Roumeliotis
- Functional Proteomics group, Chester Beatty Laboratories, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Elena Garitta
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, E1 2A, London, UK
| | - David Drew
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - S Tamir Rashid
- Department of Metabolism, Digestion & Reproduction, Imperial College London, W12 0NN, London, UK
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, 87036, Arcavacata di Rende, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), 70126, Bari, Italy
| | - Jyoti S Choudhary
- Functional Proteomics group, Chester Beatty Laboratories, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Kenneth J Linton
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, E1 2A, London, UK
| | - Konstantinos Beis
- Department of Life Sciences, Imperial College London, SW7 2AZ, London, UK.
- Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, Oxfordshire, OX11 0FA, UK.
| |
Collapse
|
12
|
Ziyang T, Xirong H, Chongming A, Tingxin L. The potential molecular pathways of Astragaloside-IV in colorectal cancer: A systematic review. Biomed Pharmacother 2023; 167:115625. [PMID: 37793276 DOI: 10.1016/j.biopha.2023.115625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023] Open
Abstract
Astragaloside IV (AS-IV), a traditional Chinese medicine, is often used to treat cancer. Colorectal cancer imposes a heavy burden on patients and society. It is essential to update the clinical evidence supporting AS-IV in the treatment of colorectal cancer. The purpose of this review is to systematically evaluate the molecular pathway and safety of AS-IV in colorectal cancer. 7 databases were queried for Jan 2012-Dec 2022. A total of 37 related articles were retrieved. 8 papers were included to evaluate the role of AS-IV in colorectal cancer and make a review. AS-IV plays vital roles in colorectal cancer, especially in the suppression of proliferation, inducing tumor cell apoptosis, increasing immune function and reducing drug resistance. Furthermore, AS-IV has been proved to regulate many signaling pathways, which are usually affected by most cancers. However, a large-scale and well-designed multicenter randomized controlled study ensures that the safety and optimal dose of AS-IV will be determined in the future.
Collapse
Affiliation(s)
- Tang Ziyang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, Chengdu, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Hu Xirong
- Faculty of Nursing, Xi'an Jiaotong University, Xi'an, PR China
| | - An Chongming
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, Chengdu, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
| | - Li Tingxin
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, Chengdu, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
| |
Collapse
|
13
|
Xu W, Zhuang L, Zhu H, Mao A, Zhou J, Wang L. TRIM14 Overexpression Induces Chemoresistance and Malignant Behaviors of Hepatocellular Carcinoma Cells by Activating the STAT3/HIF-1α Pathway. Int J Mol Sci 2023; 24:12589. [PMID: 37628777 PMCID: PMC10454020 DOI: 10.3390/ijms241612589] [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: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Members of the tripartite motif (TRIM)-containing protein family have been found to be involved in the progression of hepatocellular carcinoma (HCC). TRIM14 exerts a promotive impact on several cancers. This study aimed to explore the function and mechanism of TRIM14 in HCC. TRIM14 expression in HCC tissues and HCC cell lines was detected. The overexpression or knockdown model of TRIM14 was established in HCC cell lines. Cell Counting Kit-8 (CCK-8) assay, flow cytometry, Transwell assay, RT-PCR, Western blot, and immunofluorescence were performed to verify the influence of TRIM14 on cell proliferation, sensitivity to chemotherapy drugs, apoptosis, migration, invasion, and autophagy. A xenograft tumor model was used to confirm the impact of TRIM14 on tumor cell growth. As shown by the data, TRIM14 level was notably higher in the tumor tissues of HCC patients than in the adjacent tissues. The overall survival rate of patients with a high TRIM14 expression was relatively lower than that of patients with a low TRIM14 expression. TRIM14 upregulation enhanced the proliferation, autophagy, migration, and invasion of HCC cells and chemoresistant HCC cells and decreased apoptosis. TRIM14 knockdown contributed to the opposite effects. In in vivo experiments, TRIM14 upregulation bolstered tumor growth. Western blot analysis revealed that TRIM14 upregulation boosted signal transducer and activator of transcription3 (STAT3) and hypoxia-inducible factor-1alpha (HIF-1α) expression, and TRIM14 knockdown suppressed their expression. Moreover, repressing STAT3 and HIF-1α could mitigate the tumor-promoting role of TRIM14 in HCC cells. Overall, TRIM14 facilitated malignant HCC development and induced chemoresistance in HCC cells by activating the STAT3/HIF-1α axis.
Collapse
Affiliation(s)
- Weiqi Xu
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lihong Zhuang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Qingdao Institute, Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Qingdao 266500, China
| | - Hongxu Zhu
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Anrong Mao
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiamin Zhou
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lu Wang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Lu Y, Zhang S, Zhu X, Wang K, He Y, Liu C, Sun J, Pan J, Zheng L, Liu W, Li Y, Huang Y, Liu T. Aidi injection enhances the anti-tumor impact of doxorubicin in H22 tumor-containing mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115968. [PMID: 36473617 DOI: 10.1016/j.jep.2022.115968] [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: 09/06/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aidi injection (AD) is a traditional medical preparation that has a Chinese origin. It is extensively used particularly in combination with doxorubicin (DOX) for the management of hepatocellular carcinoma (HCC). However, the combination's synergistic mechanism has not yet been clarified. AIM OF THE STUDY To investigate the anti-tumor impact of AD in combination with DOX and their synergistic mechanism in HCC. MATERIALS AND METHODS An H22 mouse xenograft model was utilized to study the impact of AD, DOX, and their combination on HCC in vivo. Their effects on cell vitality, apoptosis, mitochondrial membrane potential, reactive oxygen species (ROS) production, caspase-3, and cleaved caspase-3 protein expression were also investigated in H22 cells in vitro. Subsequently, human umbilical vein endothelial cells (HUVECs) were utilized to investigate the impacts of AD, DOX, and their combination on cell viability, migration, invasion, tube formation, and vascular endothelial growth factor (VEGF) protein expression. RESULTS The study established that the tumor inhibition rate of AD combined with DOX reached 79.51%, which was significantly higher than that of AD (25.14%) or DOX (49.48%) alone. Additionally, the Q-value characterizing the synergy between AD and DOX was 1.72, demonstrating a strong synergistic effect. Furthermore, compared to AD or DOX administration alone, the combined administration group significantly decreased the alpha-fetoprotein (AFP) level in the serum, increased the tumor necrosis area, increased the Bax/Bcl-2, Cyt-c, caspase-9, Fas, Fasl, caspase-8, and caspase-3 protein expression, and significantly increased the CD31 and Ki67 protein expression in tumor tissue. Compared to AD or DOX alone, AD combined with DOX treatment had a synergistic effect on H22 cells (combination index values < 0.9), which inhibited cell viability, reduced mitochondrial membrane potential (MMP), induced apoptosis, promoted MMP loss, and increased ROS generation, cleaved caspase-3/caspase-3 levels, and caspase-3 activity. Moreover, combined administration showed a more pronounced inhibition of cell viability, migration, invasion, tube formation, and VEGF protein expression in HUVECs. CONCLUSIONS AD enhances the anti-tumor effect of DOX by promoting apoptosis and inhibiting angiogenesis and cell proliferation. The findings of this study lay experimental foundations for the clinical combination of AD and DOX.
Collapse
Affiliation(s)
- Yuan Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China; The Affiliated Hospital of Guizhou Medical University, 28# Guiyi Road, Guiyang, 550004, Guizhou, China
| | - Shuai Zhang
- The Affiliated Hospital of Guizhou Medical University, 28# Guiyi Road, Guiyang, 550004, Guizhou, China
| | - Xiaoqin Zhu
- School of Pharmacy, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Kailiang Wang
- School of Pharmacy, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yan He
- The Affiliated Hospital of Guizhou Medical University, 28# Guiyi Road, Guiyang, 550004, Guizhou, China
| | - Chunhua Liu
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Jie Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Lin Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Wen Liu
- The Affiliated Hospital of Guizhou Medical University, 28# Guiyi Road, Guiyang, 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yongjun Li
- School of Pharmacy, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550004, China; Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Yong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China.
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
15
|
Alassaf N, Attia H. Autophagy and necroptosis in cisplatin-induced acute kidney injury: Recent advances regarding their role and therapeutic potential. Front Pharmacol 2023; 14:1103062. [PMID: 36794281 PMCID: PMC9922871 DOI: 10.3389/fphar.2023.1103062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent, used to treat many different types of malignancies due to its high efficacy and low cost. However, its use is largely limited by acute kidney injury (AKI), which, if left untreated, may progress to cause irreversible chronic renal dysfunction. Despite substantial research, the exact mechanisms of CP-induced AKI are still so far unclear and effective therapies are lacking and desperately needed. In recent years, necroptosis, a novel subtype of regulated necrosis, and autophagy, a form of homeostatic housekeeping mechanism have witnessed a burgeoning interest owing to their potential to regulate and alleviate CP-induced AKI. In this review, we elucidate in detail the molecular mechanisms and potential roles of both autophagy and necroptosis in CP-induced AKI. We also explore the potential of targeting these pathways to overcome CP-induced AKI according to recent advances.
Collapse
Affiliation(s)
- Noha Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,*Correspondence: Noha Alassaf,
| | - Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,Department of Biochemistry, College of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Xia D, Li W, Tang C, Jiang J. Astragaloside IV, as a potential anticancer agent. Front Pharmacol 2023; 14:1065505. [PMID: 36874003 PMCID: PMC9981805 DOI: 10.3389/fphar.2023.1065505] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Cancer is a global intractable disease, and its morbidity and mortality are increasing year by year in developing countries. Surgery and chemotherapy are often used to treat cancer, but they result in unsatisfactory outcomes, such as severe side effects and drug resistance. With the accelerated modernization of traditional Chinese medicine (TCM), an increasing body of evidence has shown that several TCM components have significant anticancer activities. Astragaloside IV (AS-IV) is considered the main active ingredient of the dried root of Astragalus membranaceus. AS-IV exhibits various pharmacological effects, such as anti-inflammatory, hypoglycemic, antifibrotic, and anticancer activities. AS-IV possesses a wide range of activities, such as the modulation of reactive oxygen species-scavenging enzyme activities, participation in cell cycle arrest, induction of apoptosis and autophagy, and suppression of cancer cell proliferation, invasiveness, and metastasis. These effects are involved in the inhibition of different malignant tumors, such as lung, liver, breast, and gastric cancers. This article reviews the bioavailability, anticancer activity, and mechanism of AS-IV and provides suggestions for further research of this TCM.
Collapse
Affiliation(s)
- Dongqin Xia
- Chongqing University Cancer Hospital, Chongqing, China
| | - Wenjie Li
- Affiliated Hospital of Northwest Minzu University, Lanzhou, China
| | - Ce Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Jiang
- Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
17
|
Gui T, Chen Q, Li J, Lu K, Li C, Xu B, Chen Y, Men J, Kullak-Ublick GA, Wang W, Gai Z. Astragaloside IV alleviates 1-deoxysphinganine-induced mitochondrial dysfunction during the progression of chronic kidney disease through p62-Nrf2 antioxidant pathway. Front Pharmacol 2023; 14:1092475. [PMID: 37033627 PMCID: PMC10079923 DOI: 10.3389/fphar.2023.1092475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: Chronic kidney disease (CKD) can lead to significant elevation of 1-deoxysphingolipids (1-deoxySL). The increase of 1-deoxySL in turn can result in mitochondrial damage and oxidative stress, which can cause further progression of CKD. Methods: This study assessed the therapeutic effect of Astragaloside IV (AST) against 1-deoxySL-induced cytotoxicity in vitro and in rats with CKD. HK-2 cells were exposed to 1-deoxysphinganine (doxSA) or doxSA + AST. doxSA-induced mitochondrial dysfunction and oxidative stress were evaluated by immunostaining, real-time PCR, oxidative stress sensor, and transmission electron microscopy. The potential effects of AST on kidney damage were evaluated in a rat 5/6 nephrectomy (5/6 Nx) model of CKD. Results: The findings of in vitro experiments showed that doxSA induced mitochondrial damage, oxidative stress, and apoptosis. AST markedly reduced the level of mitochondrial reactive oxygen species, lowered apoptosis, and improved mitochondrial function. In addition, exposure to AST significantly induced the phosphorylation of p62 and the nuclear translocation of Nrf2 as well as its downstream anti-oxidant genes. p62 knock-down fully abolished Nrf2 nuclear translocation in cells after AST treatment. However, p62 knock-down did not affect TBHQ-induced Nrf2 nuclear translocation, indicating that AST can ameliorate doxSA-induced oxidative stress through modulation of p62 phosphorylation and Nrf2 nuclear translocation. Conclusion: The findings indicate that AST can activate Nrf2 antioxidant pathway in a p62 dependent manner. The anti-oxidative stress effect and the further mitochondrial protective effect of AST represent a promising therapeutic strategy for the progression of CKD.
Collapse
Affiliation(s)
- Ting Gui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingfa Chen
- Research Center of Basic Medicine, Jinan Central Hospital, Jinan, China
- Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Liaocheng, China
| | - Jiangsong Li
- Department of Urology, Liaocheng People’s Hospital, Liaocheng, China
| | - Ke Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingwen Men
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Mechanistic Safety, CMO and Patient Safety, Global Drug Development, Novartis Pharma, Basel, Switzerland
- *Correspondence: Zhibo Gai, ; Weihua Wang, ; Gerd A. Kullak-Ublick,
| | - Weihua Wang
- The Central Laboratory, Liaocheng People’s Hospital, Liaocheng, China
- *Correspondence: Zhibo Gai, ; Weihua Wang, ; Gerd A. Kullak-Ublick,
| | - Zhibo Gai
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic research, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Zhibo Gai, ; Weihua Wang, ; Gerd A. Kullak-Ublick,
| |
Collapse
|
18
|
Wei L, Wang Z, Jing N, Lu Y, Yang J, Xiao H, Guo H, Sun S, Li M, Zhao D, Li X, Qi W, Zhang Y. Frontier progress of the combination of modern medicine and traditional Chinese medicine in the treatment of hepatocellular carcinoma. Chin Med 2022; 17:90. [PMID: 35907976 PMCID: PMC9338659 DOI: 10.1186/s13020-022-00645-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/20/2022] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC, accounting for 90% of primary liver cancer) was the sixth most common cancer in the world and the third leading cause of cancer death in 2020. The number of new HCC patients in China accounted for nearly half of that in the world. HCC was of occult and complex onset, with poor prognosis. Clinically, at least 15% of patients with HCC had strong side effects of interventional therapy (IT) and have poor sensitivity to chemotherapy and targeted therapy. Traditional Chinese medicine (TCM), as a multi-target adjuvant therapy, had been shown to play an active anti-tumor role in many previous studies. This review systematically summarized the role of TCM combined with clinically commonly used drugs for the treatment of HCC (including mitomycin C, cyclophosphamide, doxorubicin, 5-fluorouracil, sorafenib, etc.) in the past basic research, and summarized the efficacy of TCM combined with surgery, IT and conventional therapy (CT) in clinical research. It was found that TCM, as an adjuvant treatment, played many roles in the treatment of HCC, including enhancing the tumor inhibition, reducing toxic and side effects, improving chemosensitivity and prolonging survival time of patients. This review summarized the advantages of integrated traditional Chinese and modern medicine in the treatment of HCC and provides a theoretical basis for clinical research.
Collapse
Affiliation(s)
- Lai Wei
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Niancai Jing
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Yi Lu
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Jili Yang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Hongyu Xiao
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Huanyu Guo
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Shoukun Sun
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Mingjing Li
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China.
| |
Collapse
|
19
|
Gao H, Song Y, Ma J, Zhai J, Zhang Y, Qu X. Untargeted metabolomics analysis of omeprazole-enhanced chemosensitivity to cisplatin in mice with non-small cell lung cancer. Chem Biol Interact 2022; 360:109933. [PMID: 35447140 DOI: 10.1016/j.cbi.2022.109933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/29/2022] [Accepted: 04/03/2022] [Indexed: 11/25/2022]
Abstract
Drug resistance of tumors remains a major barrier in cisplatin (CDDP)-based chemotherapy. Omeprazole (OME) is often utilized during chemotherapy to alleviate gastrointestinal symptoms. In a previous investigation, we demonstrated a protective effect of OME against CDDP-induced kidney injury. To further establish whether OME could enhance chemosensitivity to CDDP and the underlying mechanisms, an in vivo tumor-bearing mouse model with CDDP-resistant A549 non-small cell lung cancer (A549/CDDP) was established in the current study. A high-performance liquid chromatography-time of flight mass spectrometry (HPLC-TOF/MS)-based untargeted metabolomics approach for tumor tissue and serum was employed to explore the mechanisms underlying the enhanced therapeutic effects of co-administration of CDDP and OME. Notably, tumor weights of mice in the CDDP + OME group were significantly decreased compared with those treated with CDDP alone. HE and TUNEL staining revealed more significant apoptosis of tumor cells in the group co-administered CDDP + OME relative to CDDP alone. Overexpression of multidrug resistance-associated protein 2 in CDDP-resistant tumors was significantly reversed upon treatment with CDDP + OME. PCA score plots of the groups co-treated with CDDP + OME were clearly separated from those treated with CDDP alone in metabolomics analysis for tumor and serum samples, clearly suggesting that co-administration of OME enhances the antitumor effect of CDDP. Subsequently, 10 and 7 metabolites in CDDP + OME group with significant changes in tumor and serum compared with CDDP group, respectively, were identified. Pathway analysis both in tumor and serum samples revealed regulation of the metabolism of purines, several amino acids and riboflavin in enhanced chemotherapy with both OME and CDDP. The collective findings provide beneficial novel insights into drug-drug interactions, which could improve the application of CDDP in clinical practice.
Collapse
Affiliation(s)
- Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China
| | - Yanqing Song
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China
| | - Jie Ma
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China
| | - Jinghui Zhai
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China
| | - Yueming Zhang
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China
| | - Xiaoyu Qu
- Department of Pharmacy, The First Hospital of Jilin University, 130021, Changchun, China.
| |
Collapse
|
20
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J Adamcakova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | |
Collapse
|
21
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J ADAMCAKOVA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - D MOKRA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| |
Collapse
|
22
|
Fu K, Wang C, Ma C, Zhou H, Li Y. The Potential Application of Chinese Medicine in Liver Diseases: A New Opportunity. Front Pharmacol 2021; 12:771459. [PMID: 34803712 PMCID: PMC8600187 DOI: 10.3389/fphar.2021.771459] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Liver diseases have been a common challenge for people all over the world, which threatens the quality of life and safety of hundreds of millions of patients. China is a major country with liver diseases. Metabolic associated fatty liver disease, hepatitis B virus and alcoholic liver disease are the three most common liver diseases in our country, and the number of patients with liver cancer is increasing. Therefore, finding effective drugs to treat liver disease has become an urgent task. Chinese medicine (CM) has the advantages of low cost, high safety, and various biological activities, which is an important factor for the prevention and treatment of liver diseases. This review systematically summarizes the potential of CM in the treatment of liver diseases, showing that CM can alleviate liver diseases by regulating lipid metabolism, bile acid metabolism, immune function, and gut microbiota, as well as exerting anti-liver injury, anti-oxidation, and anti-hepatitis virus effects. Among them, Keap1/Nrf2, TGF-β/SMADS, p38 MAPK, NF-κB/IκBα, NF-κB-NLRP3, PI3K/Akt, TLR4-MyD88-NF-κB and IL-6/STAT3 signaling pathways are mainly involved. In conclusion, CM is very likely to be a potential candidate for liver disease treatment based on modern phytochemistry, pharmacology, and genomeproteomics, which needs more clinical trials to further clarify its importance in the treatment of liver diseases.
Collapse
Affiliation(s)
| | | | | | | | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
23
|
Zhang C, Li L, Hou S, Shi Z, Xu W, Wang Q, He Y, Gong Y, Fang Z, Yang Y. Astragaloside IV inhibits hepatocellular carcinoma by continually suppressing the development of fibrosis and regulating pSmad3C/3L and Nrf2/HO-1 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114350. [PMID: 34157326 DOI: 10.1016/j.jep.2021.114350] [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: 04/28/2021] [Revised: 06/02/2021] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus is a medicinal herb used in China for the prevention and treatment of diseases such as diabetes and cancer. As one of the main active ingredients of astragalus, Astragaloside IV (AS-IV) has a wide range of pharmacological effects, including anti-inflammation and anti-cancer effects. AIM OF THE STUDY Different phosphorylated forms of Smad3 differentially regulate the progression of hepatic carcinoma. The phosphorylation of the COOH-terminal of Smad3 (pSmad3C) and activation of the Nrf2/HO-1 pathway inhibits hepatic carcinoma, while phosphorylation of the linker region of Smad3 (pSmad3L) promotes progression. Thus, pSmad3C/3L and Nrf2/HO-1 pathways are potential targets for drug of anti-cancer development. AS-IV is anti-apoptotic and can inhibit hepatocellular carcinoma cell (HCC) proliferation, invasion, and tumor growth in nude mice. However, it is not clear whether AS-IV has a therapeutic effect on inhibiting the progression of primary liver cancer by regulating the pSmad3C/3L and Nrf2/HO-1 pathway. The purpose of this study is to investigate whether AS-IV inhibits hepatocellular carcinoma by regulating pSmad3C/3L and Nrf2/HO-1 pathway. MATERIALS AND METHODS primary liver cancer in mice induced by DEN/CCl4/C2H5OH (DCC) and HSC-T6/HepG2 cell models activated by TGF-β1 was investigated for the mechanisms of AS-IV. In vivo assays included liver biopsy, histopathology and post-mortem analysis included immunohistochemistry, immunofluorescent, and Western blotting analysis, and in vitro assays included immunofluorescent, and Western blotting analysis. RESULTS AS-IV significantly inhibited the development of primary liver cancer, reflecting improved liver biopsy, histopathology. The incidence and multiplicity of primary liver cancer were markedly decreased by AS-IV treatment at the 20th week. AS-IV had observable effects on the TGF-β1/Smad and Nrf2/HO-1 expression in vivo, especially up-regulated pSmad3C, pNrf2, HO-1, and NQO1, while it down-regulated pSmad2C, pSmad2L, pSmad3L, PAI-1, and α-SMA at the 12th week and the 20th week. Furthermore, in vitro analysis further confirmed that AS-IV regulated the expression of pSmad3C/3L and Nrf2/HO-1 pathway in HSC-T6 and HepG2 cells activated by TGF-β1. CONCLUSION AS-IV administration delays the occurrence of primary liver cancer by continually suppressing the development of fibrosis, the mechanism of the therapeutic effect involving the regulation of the pSmad3C/3L and Nrf2/HO-1 pathways, especially in regulation reversibility and antagonism of pSmad3C and pSmad3L and promoting the phosphorylation of Nrf2.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Lili Li
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Shu Hou
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Zhenghao Shi
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Wenjing Xu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Qin Wang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Yinghao He
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Yongfang Gong
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Zhirui Fang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Yan Yang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
24
|
Ding L, Li H, Wang Y. Application of Jianpi Xiaoai Recipe Combined with Cisplatin and Adriamycin in the Treatment of Endometrial Cancer and Its Effect on Disease Control Rate. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:2258183. [PMID: 34621319 PMCID: PMC8492281 DOI: 10.1155/2021/2258183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the application of Jianpi Xiaoai recipe combined with cisplatin and Adriamycin in the treatment of endometrial cancer (EC) and its effect on the disease control rate (DCR). METHODS The data of 120 EC patients treated in People's Hospital of Rizhao from February 2019 to February 2020 were retrospectively analyzed. They were equally split into experimental group and control group according to the order of admission. All patients were treated with neoadjuvant intra-arterial chemotherapy (continuous infusion of the uterine artery for 5 days before surgery, with 20 mg of cisplatin mixed with 2000 mg of normal saline and 10 mg of Adriamycin mixed with 500 ml of normal saline daily), while the experimental group was treated with Jianpi Xiaoai recipe at the same time to compare the short-term efficacy, immune function indexes, incidence of adverse reactions, and HEC-1-B (human endometrial adenocarcinoma cells) cell inhibition rates between the two groups. RESULTS The DCR and objective remission rate (ORR) in the experimental group were markedly higher compared with the control group (P < 0.05). The immune function indexes after treatment were remarkably better in the experimental group than in the control group (P < 0.05). Compared with the control group, the incidence of adverse reactions in the experimental group was notably lower (P < 0.05), while the HEC-1-B inhibition rates after treatment were obviously higher (P < 0.05). CONCLUSION Jianpi Xiaoai recipe combined with cisplatin and Adriamycin can increase the HEC-1-B cell inhibition rate in EC patients, improve their immune function, reduce the possibility of adverse reactions, and enhance the therapeutic effect, which is worthy of clinical application and popularization.
Collapse
Affiliation(s)
- Li Ding
- Department of Gynaecology, People's Hospital of Rizhao, Rizhao 276826, Shandong, China
| | - Hongyu Li
- Department of Radiology, People's Hospital of Lixia District, Jinan 250013, Shandong, China
| | - Yuping Wang
- Department of Postpartum Rehabilitation, Zibo City Maternal and Child Health Care Hospital, Zibo 250031, Shandong, China
| |
Collapse
|
25
|
NIR stimulus-responsive AstragalosideIV-Indocyanin green liposomes for chemo-photothermal therapy. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Balakrishnan B, Liang Q, Fenix K, Tamang B, Hauben E, Ma L, Zhang W. Combining the Anticancer and Immunomodulatory Effects of Astragalus and Shiitake as an Integrated Therapeutic Approach. Nutrients 2021; 13:nu13082564. [PMID: 34444724 PMCID: PMC8401741 DOI: 10.3390/nu13082564] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
Astragalus root (Huang Qi) and Shiitake mushrooms (Lentinus edodes) are both considered medicinal foods and are frequently used in traditional Chinese medicine due to their anticancer and immunomodulating properties. Here, the scientific literatures describing evidence for the anticancer and immunogenic properties of Shiitake and Astragalus were reviewed. Based on our experimental data, the potential to develop medicinal food with combined bioactivities was assessed using Shiitake mushrooms grown over Astragalus beds in a proprietary manufacturing process, as a novel cancer prevention approach. Notably, our data suggest that this new manufacturing process can result in transfer and increased bioavailability of Astragalus polysaccharides with therapeutic potential into edible Shiitake. Further research efforts are required to validate the therapeutic potential of this new Hengshan Astragalus Shiitake medicinal food.
Collapse
Affiliation(s)
- Biju Balakrishnan
- Centre for Marine Bioproducts Development, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (B.B.); (Q.L.); (B.T.)
- The Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5011, Australia;
| | - Qi Liang
- Centre for Marine Bioproducts Development, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (B.B.); (Q.L.); (B.T.)
- Shanxi University of Traditional Chinese Medicine, Taiyuan 030600, China
| | - Kevin Fenix
- The Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5011, Australia;
- Discipline of Surgery, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Bunu Tamang
- Centre for Marine Bioproducts Development, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (B.B.); (Q.L.); (B.T.)
| | - Ehud Hauben
- The Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5011, Australia;
- Discipline of Surgery, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
- AusHealth Corporate Pty Ltd., Adelaide, SA 5032, Australia
- Correspondence: (E.H.); (L.M.); (W.Z.); Tel.: +61-88132-7450 (E.H.); +61-7-3735-4175 (L.M.); +61-8-7221-8557 (W.Z.)
| | - Linlin Ma
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
- Correspondence: (E.H.); (L.M.); (W.Z.); Tel.: +61-88132-7450 (E.H.); +61-7-3735-4175 (L.M.); +61-8-7221-8557 (W.Z.)
| | - Wei Zhang
- Centre for Marine Bioproducts Development, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (B.B.); (Q.L.); (B.T.)
- Correspondence: (E.H.); (L.M.); (W.Z.); Tel.: +61-88132-7450 (E.H.); +61-7-3735-4175 (L.M.); +61-8-7221-8557 (W.Z.)
| |
Collapse
|
27
|
Gao P, Du X, Liu L, Xu H, Liu M, Guan X, Zhang C. Astragaloside IV Alleviates Tacrolimus-Induced Chronic Nephrotoxicity via p62-Keap1-Nrf2 Pathway. Front Pharmacol 2021; 11:610102. [PMID: 33536919 PMCID: PMC7848072 DOI: 10.3389/fphar.2020.610102] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Tacrolimus-induced chronic nephrotoxicity (TIN) hinders its long-term use in patients. However, there are no drugs available in the clinic to relieve it at present. Astragaloside IV (AS-IV) is a saponin extract of the Astragalus which is widely used in the treatment of kidney disease. This study aimed to investigate the effect of AS-IV on TIN and its underlying mechanism. Herein, C57BL/6 mice were treated with tacrolimus and/or AS-IV for 4 weeks, and then the renal function, fibrosis, oxidative stress and p62-Keap1-Nrf2 pathway were evaluated to ascertain the contribution of AS-IV and p62-Keap1-Nrf2 pathway to TIN. Our results demonstrated that AS-IV significantly improved renal function and alleviated tubulointerstitial fibrosis compared with the model group. The expression of fibrosis-related proteins, including TGF-β1, Collagen I and α-SMA, were also decreased by AS-IV. Furthermore, AS-IV relieved the inhibition of tacrolimus on antioxidant enzymes. The data in HK-2 cells also proved that AS-IV reduced tacrolimus-induced cell death and oxidative stress. Mechanistically, AS-IV markedly promoted the nuclear translocation of Nrf2 and the renal protective effects of AS-IV were abolished by Nrf2 inhibitor. Further researches showed that phosphorylated p62 was significantly increased after AS-IV pretreatment. Moreover, AS-IV failed to increase nuclear translocation of Nrf2 and subsequent anti-oxidative stress in HK-2 cells transfected with p62 siRNA. Collectively, these findings indicate that AS-IV relieve TIN by enhancing p62 phosphorylation, thereby increasing Nrf2 nuclear translocation, and then alleviating ROS accumulation and renal fibrosis.
Collapse
Affiliation(s)
- Ping Gao
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyi Du
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pediatrics, Maternal and Child Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Liu
- Department of Pathology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Xu
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maochang Liu
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinlei Guan
- Department of Pharmacy, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengliang Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|