1
|
Sang Y, Hu Y, Zhang Y, Chen L, Lu Y, Gao L, Lu Y, Cao X, Zhang Y, Chen G. Network pharmacology, molecular docking and biological verification to explore the potential anti-prostate cancer mechanisms of Tripterygium wilfordii Hook. F. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119071. [PMID: 39522845 DOI: 10.1016/j.jep.2024.119071] [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: 06/05/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii Hook. f. (TW) is extensively utilized in clinical practice for its effective anti-inflammatory and anti-cancer properties. AIM OF THE STUDY This study aims to elucidate the processes of TW in combating prostate cancer through a comprehensive strategy that integrates network pharmacology, molecular docking and molecular biology validation. MATERIALS AND METHODS A drug-target network and protein-protein interaction network were constructed established to predict the potential targets of TW for prostate cancer treatment. The interaction between active components and targets was confirmed using molecular docking. Moreover, prostate cancer cells were used to examine the anti-tumor effects of active ingredients in vitro. The xenograft animal model was constructed to evaluate the anti-tumor effect of triptonoterpene in vivo. RESULTS Twenty-nine active components interact with 226 corresponding targets, and 112 disease targets specifically related with prostate cancer were identified. The primary targets (AKT1, TP53, RELA) were chosen, and kaempferol, triptolide, and triptonoterpene exhibited probable binding affinity with these targets, respectively. Triptonoterpene was subsequently confirmed to inhibit the growth of prostate cancer cells and induce apoptosis in vitro and in vivo. CONCLUSION Overall, this study demonstrated that TW may serve as a viable therapeutic agent for prostate cancer. Triptonoterpene is a specific inhibitor of p-AKT1 and p65, making it an attractive contender for prostate cancer therapy.
Collapse
Affiliation(s)
- Yazhou Sang
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, 318000, Zhejiang, China; School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yue Hu
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, 318000, Zhejiang, China; Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China.
| | - Yueyue Zhang
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China.
| | - Luyi Chen
- Maternal Health Care Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yutian Lu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China.
| | - Lin Gao
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China.
| | - Yunyun Lu
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315048, Zhejiang, China.
| | - Xuan Cao
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, 318000, Zhejiang, China; Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China.
| | - Yaqiong Zhang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China.
| | - Guofu Chen
- Department of General Surgery, Affiliated Wenling First People's Hospital, Taizhou University, Taizhou, 318000, Zhejiang, China.
| |
Collapse
|
2
|
Zhang Z, Xiong R, Hu Q, Zhang Q, Wang S, Chen Y. Review on anti-tumor lipid nano drug delivery systems of traditional Chinese medicine. J Drug Target 2025:1-41. [PMID: 39743936 DOI: 10.1080/1061186x.2024.2448708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
In recent years, the use of traditional Chinese medicine (TCM) in the treatment of cancer has received widespread attention. Treatment of tumors using TCM can effectively reduce the side effects of anti-tumor drugs, meanwhile to improve the treatment efficacy of patients. However, most of the active ingredients in TCM, such as saponins, alkaloids, flavonoids, volatile oils, etc., have defects such as low bioavailability and poor solubility in clinical application, which seriously restrict the application of TCM. Meanwhile, the encapsulation of TCM into lipid nano-delivery systems for cancer therapy has received much attention. Lipid nano-delivery systems are obtained by using phospholipids as the base material and adding other auxiliary materials under a certain preparation process, including, for example, liposomes, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), microemulsions, and self-microemulsion drug delivery systems (SMEDDS), can resolve the application problems of TCM by improving the efficacy of active ingredients of TCM and reducing the toxicity of anti-tumor drugs. This paper focuses on the categories, development status, and research progress of lipid nano delivery system of TCM, aiming to provide a certain theoretical basis for further in-depth research and rational application of these systems.
Collapse
Affiliation(s)
- Ziwei Zhang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| | - Rui Xiong
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| | - Qiyan Hu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| | - Qiang Zhang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| | - Shaozhen Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| | - Yunyan Chen
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu 241002, China
| |
Collapse
|
3
|
Xu XH, Zhang JX, Liu HX, Zhao Z, Jiang JY. Intervention of inflammation associated with ankylosing spondylitis by triptolide promotes histone H3 Iys-27 trimethylation. Immunopharmacol Immunotoxicol 2024; 46:785-792. [PMID: 39307916 DOI: 10.1080/08923973.2024.2402911] [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] [Accepted: 08/24/2024] [Indexed: 11/19/2024]
Abstract
Objective: This study aims to explore the effects of Triptolide (TP) on the differentiation of Th17 cells in ankylosing spondylitis (AS). Methods: Peripheral blood mononuclear cells (PBMCs) collected from 10 patients with active AS patients were exposed to TP, GSK-J4 or vehicle. T lymphocyte subsets were analyzed using flow cytometry. ELISA was used to assess the level of IL-17. Western blot analysis and quantitative RT-PCR were used to measure the mRNA and protein levels of RORγt, JMJD3, EZH2, JAK2 and STAT3 in the JAK2/STAT3 signaling pathway. Results: We observed a tendency toward a greater percentage of IL-17-positive CD4+ T cells in peripheral blood mononuclear cells (PBMCs) from patients with active AS than in those from healthy controls. Triptolide (TP) and GSK-J4 significantly reduced IL-17 expression. In cultured PBMCs from patients with active AS, 24 h of treatment with TP or GSK-J4 decreased the expression of RORγt (p < 0.05), JAK2 and STAT3 (JAK2: p < 0.05; STAT3: p < 0.05). Furthermore, both triptolide and GSK-J4 increased the level of histone 3 with Lys 27 trimethylation (H3K27me3) in patient-derived PBMCs. H3K27me3 enrichment was detected at the promoters of the RORc, STAT3 and IL-17 genes. Consistent with this finding, triptolide upregulated the EZH2 gene and downregulated the JMJD3 gene. Conclusion: Triptolide inhibits Th17 cell differentiation via H3K27me3 upregulation and orchestrates changes in histone-modifying enzymes, including JMJD3 and EZH2. These findings support the clinical efficacy of triptolide for AS and may provide clues for identifying molecular targets for the development of novel treatments.
Collapse
Affiliation(s)
- Xiao-Han Xu
- Department of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Jin-Xu Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Hong-Xiao Liu
- Department of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing China
| | - Zhe Zhao
- Department of Rheumatology, The First Affiliated Hospital of Henan University of CM, Henan, Zhengzhou, China
| | - Jun-Yi Jiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
4
|
Bian Q, Li B, Zhang L, Sun Y, Zhao Z, Ding Y, Yu H. Molecular pathogenesis, mechanism and therapy of Cav1 in prostate cancer. Discov Oncol 2023; 14:196. [PMID: 37910338 PMCID: PMC10620365 DOI: 10.1007/s12672-023-00813-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
Prostate cancer is the second incidence of malignant tumors in men worldwide. Its incidence and mortality are increasing year by year. Enhanced expression of Cav1 in prostate cancer has been linked to both proliferation and metastasis of cancer cells, influencing disease progression. Dysregulation of the Cav1 gene shows a notable association with prostate cancer. Nevertheless, there is no systematic review to report about molecular signal mechanism of Cav1 and drug treatment in prostate cancer. This article reviews the structure, physiological and pathological functions of Cav1, the pathogenic signaling pathways involved in prostate cancer, and the current drug treatment of prostate cancer. Cav1 mainly affects the occurrence of prostate cancer through AKT/mTOR, H-RAS/PLCε, CD147/MMPs and other pathways, as well as substance metabolism including lipid metabolism and aerobic glycolysis. Baicalein, simvastatin, triptolide and other drugs can effectively inhibit the growth of prostate cancer. As a biomarker of prostate cancer, Cav1 may provide a potential therapeutic target for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Qiang Bian
- Department of Pathophysiology, Weifang Medicine University, Weifang, 261053, Shandong, People's Republic of China
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China
| | - Bei Li
- Department of Radiological Image, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, People's Republic of China
| | - Luting Zhang
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
| | - Yinuo Sun
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
| | - Zhankui Zhao
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China
| | - Yi Ding
- Department of Pathophysiology, Weifang Medicine University, Weifang, 261053, Shandong, People's Republic of China.
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China.
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China.
| |
Collapse
|
5
|
Hu L, Gao M, Jiang H, Zhuang L, Jiang Y, Xie S, Zhang H, Wang Q, Chen Q. Triptolide inhibits epithelial ovarian tumor growth by blocking the hedgehog/Gli pathway. Aging (Albany NY) 2023; 15:11131-11151. [PMID: 37851362 PMCID: PMC10637820 DOI: 10.18632/aging.205110] [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: 03/23/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023]
Abstract
Epithelial ovarian cancer (EOC), the most predominant subtype of ovarian cancer (OC), involves poor prognosis and exhibits high aggression. Triptolide (TPL), like other Chinese herbs, has historically played a significant role in modern medicine. The screening system based on Gli-dependent luciferase reporter activity assessed the effects of over 800 natural medicinal materials on hedgehog (Hh) signaling pathway activity and discovered that TPL had an excellent inhibitory effect on Hh signaling pathway activity. However, the significance and mechanism of TPL involvement in regulating the Hh pathway have not been well explored. Thus, this work aimed to understand better how TPL affects the Hh pathway activity, which, in turn, influences the biological behavior of EOC. Our findings observed that Smo agonist SAG-induced EOC cell proliferation, migration, and invasion were drastically reversed by TPL in a concentration-dependent pattern. Further evidence suggested that TPL promotes the degradation of Gli1 and Gli2 to inhibit the activity of the Hh signaling pathway by relying on Gli1 and Gli2 ubiquitination. Our in vivo studies also confirmed that TPL could significantly inhibit the tumor growth of EOC. Taken together, our results revealed that one of the antitumor mechanisms of TPL was the targeted inhibition of the Hh/Gli pathway.
Collapse
Affiliation(s)
- Lanyan Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Mai Gao
- Huankui Academy of Nanchang University, Nanchang 330036, Jiangxi, P.R. China
| | - Huifu Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Lingling Zhuang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Ying Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Siqi Xie
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qian Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
6
|
Ghosh S, Hazra J, Pal K, Nelson VK, Pal M. Prostate cancer: Therapeutic prospect with herbal medicine. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100034. [PMID: 34909665 PMCID: PMC8663990 DOI: 10.1016/j.crphar.2021.100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is a major cause of morbidity and mortality in men worldwide. A geographic variation on the burden of the disease suggested that the environment, genetic makeup, lifestyle, and food habits modulate one's susceptibility to the disease. Although it has been generally thought to be an older age disease, and awareness and timely execution of screening programs have managed to contain the disease in the older population over the last decades, the incidence is still increasing in the population younger than 50. Existing treatment is efficient for PCa that is localized and responsive to androgen. However, the androgen resistant and metastatic PCa are challenging to treat. Conventional radiation and chemotherapies are associated with severe side effects in addition to being exorbitantly expensive. Many isolated phytochemicals and extracts of plants used in traditional medicine are known for their safety and diverse healing properties, including many with varying levels of anti-PCa activities. Many of the phytochemicals discussed here, as shown by many laboratories, inhibit tumor cell growth and proliferation by interfering with the components in the pathways responsible for the enhanced proliferation, metabolism, angiogenesis, invasion, and metastasis in the prostate cells while upregulating the mechanisms of cell death and cell cycle arrest. Notably, many of these agents simultaneously target multiple cellular pathways. We analyzed the available literature and provided an update on this issue in this review article.
Collapse
Affiliation(s)
- Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | - Joyita Hazra
- Department of Biotechnology, Indian Institute of Technology Madras, Tamil Nadu, India
| | | | - Vinod K. Nelson
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Andhra Pradesh, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
7
|
Gao J, Zhang Y, Liu X, Wu X, Huang L, Gao W. Triptolide: pharmacological spectrum, biosynthesis, chemical synthesis and derivatives. Theranostics 2021; 11:7199-7221. [PMID: 34158845 PMCID: PMC8210588 DOI: 10.7150/thno.57745] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022] Open
Abstract
Triptolide, an abietane-type diterpenoid isolated from Tripterygium wilfordii Hook. F., has significant pharmacological activity. Research results show that triptolide has obvious inhibitory effects on many solid tumors. Therefore, triptolide has become one of the lead compounds candidates for being the next "blockbuster" drug, and multiple triptolide derivatives have entered clinical research. An increasing number of researchers have developed triptolide synthesis methods to meet the clinical need. To provide new ideas for researchers in different disciplines and connect different disciplines with researchers aiming to solve scientific problems more efficiently, this article reviews the research progress made with analyzes of triptolide pharmacological activity, biosynthetic pathways, and chemical synthesis pathways and reported in toxicological and clinical studies of derivatives over the past 20 years, which have laid the foundation for subsequent researchers to study triptolide in many ways.
Collapse
Affiliation(s)
- Jie Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yifeng Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xihong Liu
- Basic Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xiayi Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Luqi Huang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
8
|
Abstract
Caveolae are specialised and dynamic plasma membrane subdomains, involved in many cellular functions including endocytosis, signal transduction, mechanosensing and lipid storage, trafficking, and metabolism. Two protein families are indispensable for caveola formation and function, namely caveolins and cavins. Mutations of genes encoding these caveolar proteins cause serious pathological conditions such as cardiomyopathies, skeletal muscle diseases, and lipodystrophies. Deregulation of caveola-forming protein expression is associated with many types of cancers including prostate cancer. The distinct function of secretion of the prostatic fluid, and the unique metabolic phenotype of prostate cells relying on lipid metabolism as a main bioenergetic pathway further suggest a significant role of caveolae and caveolar proteins in prostate malignancy. Accumulating in vitro, in vivo, and clinical evidence showed the association of caveolin-1 with prostate cancer grade, stage, metastasis, and drug resistance. In contrast, cavin-1 was found to exhibit tumour suppressive roles. Studies on prostate cancer were the first to show the distinct function of the caveolar proteins depending on their localisation within the caveolar compartment or as cytoplasmic or secreted proteins. In this review, we summarise the roles of caveola-forming proteins in prostate cancer and the potential of exploiting them as therapeutic targets or biological markers.
Collapse
|
9
|
miR-4319 inhibited retinoblastoma cells proliferation, migration, invasion and EMT progress via suppressing CD147 mediated MMPs expression. J Mol Histol 2021; 52:269-277. [PMID: 33474692 DOI: 10.1007/s10735-020-09946-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/07/2020] [Indexed: 01/15/2023]
Abstract
Tumor migration is the critical step that lead to the migration in retinoblastoma (RB), in which microRNAs (miRNAs) play important roles. This study aimed to investigate the role of microRNA-4319 (miR-4319) in the development of retinoblastoma by identifying its targets, as well as its underlying regulatory mechanisms. Our data shown that miR-4319 was downregulated in RB tissues and RB cell lines. Enhanced miR-4319 suppressed cell proliferation, migration, invasion and EMT progress, promoted cell apoptosis in SO-RB50 and RB-Y79 cells. Of note, extracellular matrix metalloproteinase inducer (EMMPRI/CD147) was identified as a direct target gene for miR-4319. MMPs were regulated by CD147 and participated in the miR-4319 regulatory network in SO-RB50 cells. In addition, overexpression of CD147 abrogated the inhibitory effect of miR-4319 on RB cells. In summary, miR-4319 overexpression suppressed cell proliferation, migration and invasion may through suppressing the CD147 mediated MMPs expression, suggesting that miR-4319 may serve as a potential diagnostic biomarker and treatment target for RB.
Collapse
|
10
|
Fakhri S, Moradi SZ, Farzaei MH, Bishayee A. Modulation of dysregulated cancer metabolism by plant secondary metabolites: A mechanistic review. Semin Cancer Biol 2020; 80:276-305. [PMID: 32081639 DOI: 10.1016/j.semcancer.2020.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Several signaling pathways and basic metabolites are responsible for the control of metabolism in both normal and cancer cells. As emerging hallmarks of cancer metabolism, the abnormal activities of these pathways are of the most noticeable events in cancer. This altered metabolism expedites the survival and proliferation of cancer cells, which have attracted a substantial amount of interest in cancer metabolism. Nowadays, targeting metabolism and cross-linked signaling pathways in cancer has been a hot topic to investigate novel drugs against cancer. Despite the efficiency of conventional drugs in cancer therapy, their associated toxicity, resistance, and high-cost cause limitations in their application. Besides, considering the numerous signaling pathways cross-linked with cancer metabolism, discovery, and development of multi-targeted and safe natural compounds has been a high priority. Natural secondary metabolites have exhibited promising anticancer effects by targeting dysregulated signaling pathways linked to cancer metabolism. The present review reveals the metabolism and cross-linked dysregulated signaling pathways in cancer. The promising therapeutic targets in cancer, as well as the critical role of natural secondary metabolites for significant anticancer enhancements, have also been highlighted to find novel/potential therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
11
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 280] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
12
|
Lv H, Jiang L, Zhu M, Li Y, Luo M, Jiang P, Tong S, Zhang H, Yan J. The genus Tripterygium: A phytochemistry and pharmacological review. Fitoterapia 2019; 137:104190. [DOI: 10.1016/j.fitote.2019.104190] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022]
|
13
|
Zhang H, Li H, Liu Z, Ge A, Guo E, Liu S, Chen Z. Triptolide inhibits the proliferation and migration of medulloblastoma Daoy cells by upregulation of microRNA-138. J Cell Biochem 2018; 119:9866-9877. [PMID: 30156009 DOI: 10.1002/jcb.27307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/26/2018] [Indexed: 12/25/2022]
Abstract
Medulloblastoma is a primitive neuroectodermal-derived brain tumor and the most common malignant brain tumor in children. Triptolide (TPL) is the major active component extracted from Tripterygium wilfordii Hook F. This study aimed to explore the effects of TPL on medulloblastoma cell proliferation, migration, and apoptosis, as well as the underlying possible molecular mechanism. Viability, proliferation, and apoptosis of Daoy cells were measured using cell counting kit-8 assay, 5-bromo-2'-deoxyuridine incorporation assay, and Guava Nexin assay, respectively. Cell migration was detected using two-chamber transwell assay and wound healing assay. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to determine the relative expression of microRNA-138 (miR-138) in Daoy cells. Cell transfection was used to change the expression of miR-138 in cells. Western blot analysis was used to analyze the expression of key factors involved in cell apoptosis, cell migration, the phosphatidylinositol 3-kinase (PI3K)/protein kinase 3 (AKT) pathway, and the Notch pathway in Daoy cells. We found that TPL significantly inhibited the viability, proliferation, and migration of Daoy cells but promoted Daoy cell apoptosis. The expression levels of matrix metalloproteinases (MMP)-2 and MMP-9 after TPL treatment were decreased. The expression of miR-138 in Daoy cells after TPL treatment was increased. Suppression of miR-138 obviously reversed the TPL-induced Daoy cell proliferation, migration inhibition, and cell apoptosis enhancement, as well as the inactivation of the PI3K/AKT and Notch pathways. Cyclin-dependent kinase 6 (CDK6) was a direct target gene of miR-138, which might be involved in the antitumor effects of TPL on Daoy cells. In conclusion, our study verified that TPL exerted anticancer effects on medulloblastoma cells possibly via upregulating miR-138 and inactivating the PI3K/AKT and Notch pathways.
Collapse
Affiliation(s)
- Haifang Zhang
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Hui Li
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Zhenguo Liu
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Ang Ge
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Enyu Guo
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Shuxia Liu
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Zhiping Chen
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
14
|
Chai F, Li Y, Liu K, Li Q, Sun H. Caveolin enhances hepatocellular carcinoma cell metabolism, migration, and invasion in vitro via a hexokinase 2‐dependent mechanism. J Cell Physiol 2018; 234:1937-1946. [PMID: 30144070 DOI: 10.1002/jcp.27074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Fang Chai
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Yan Li
- Department of General Surgery The Fourth Affiliated Hospital of China Medical University Shenyang China
| | - Keyi Liu
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Qiang Li
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Hongzhi Sun
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| |
Collapse
|
15
|
Triptolide inhibits Epstein-Barr nuclear antigen 1 expression by increasing sensitivity of mitochondria apoptosis of nasopharyngeal carcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:192. [PMID: 30111354 PMCID: PMC6094928 DOI: 10.1186/s13046-018-0865-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/19/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV) is widely found in nasopharyngeal carcinoma (NPC) tissue and associated with poor prognosis of patients. EBV nuclear antigen 1 (EBNA1) is expressed in all NPC tumors and plays multiple biological roles in both virus and host cells. Triptolide is a natural product extracted from Tripterygium and shows anti-cancer activities. The goal of this work was to illustrate the anti-cancer effect of triptolide and elucidate a novel anti-apoptotic mechanism of EBNA1 in NPC cells encountered with triptolide. METHODS In the present study, a CCK-8 assay was used to analyze the proliferation of NPC cells treated with triptolide in a dose- and time-dependent ways. Effects of triptolide on NPC cell cycle and apoptosis were investigated by flow cytometric analysis. EBNA1 expression in mRNA and protein levels was determined by quantitative real-time PCR and Western blot, respectively. RESULTS Our results showed that triptolide effectively inhibited proliferation of NPC cells. Triptolide arrested NPC cell cycles in S phase and induced apoptosis through a caspase-9-dependent apoptosis pathway. Low-dose of triptolide reduced the half-life of EBNA1 and significantly decreased EBNA1 expression by promoting the process of proteasome-ubiquitin pathway. Over-expression of EBNA1, which was independent from EBV genome, effectively attenuated the apoptosis induced by triptolide. In addition, triptolide significantly inhibited proliferations of tumors induced by EBV-positive cells in vivo. Furthermore, EBNA1 were expressed in all NPC biopsies of Chinese patients. CONCLUSIONS In summary, our study provides the evidence that triptolide induces EBNA1 degradation and stimulates NPC apoptosis through mitochondria apoptotic pathway. In addition, EBNA1 assists NPC cells to resist triptolide-induced apoptosis through inhibiting caspase-9-dependent apoptotic pathway.
Collapse
|
16
|
Qiao S, Liu C, Xu W, AZhaTi W, Li C, Wang Z. Up-regulated expression of CD147 gene in malignant bone tumor and the possible induction mechanism during osteoclast formation. ACTA ACUST UNITED AC 2018; 51:e6948. [PMID: 30043854 PMCID: PMC6065812 DOI: 10.1590/1414-431x20186948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/14/2018] [Indexed: 01/17/2023]
Abstract
It is increasingly evident that the microenvironment of bone can influence cancer phenotype in many ways that favor growth in bone. CD147, a transmembrane protein of the immunoglobulin (Ig) superfamily, was identified independently in different species and has many designations across different species. However, expression levels of CD147 mRNA in bone cancer have not been described. In this study, we have used real-time fluorescence quantification (RT-PCR) to demonstrate CD147 expression in malignant bone cancer and benign bone tumor tissues. The results suggested that the expression of CD147 gene was significantly up-regulated in malignant bone cancer. Moreover, we found that over-expressed RANKL progressively enhanced osteoclast formation up to 48 h, which suggested that RANKL could promote the formation of osteoclast, indicating that both CD147 and RANKL play important roles in the formation of osteoclasts. Furthermore, the expressions of four osteoclast specific expression genes, including TRACP, MMP-2, MMP-9 and c-Src, were analyzed using RT-PCR. The results indicated that four osteoclast-specific expression genes were detectable in all osteoclast with different treatments. However, the highest expression level of these four osteoclast-specific expression genes appears in the CD147+ RANKL group and the lowest expression level of these four osteoclast-specific expression genes appears with si-RANKL treatment. Characterization of the role of CD147 in the development of tumors should lead to a better understanding of the changes occurring at the molecular level during the development and progression of primary human bone cancer.
Collapse
Affiliation(s)
- Suchi Qiao
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, China.,Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Chang Liu
- Department of Orthopedics, Fuzhou General Hospital, Fuzhou, China
| | - Weijie Xu
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - WuBuLi AZhaTi
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Cheng Li
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhiwei Wang
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
17
|
Triptolide antagonized the cisplatin resistance in human ovarian cancer cell line A2780/CP70 via hsa-mir-6751. Future Med Chem 2018; 10:1947-1955. [PMID: 29966441 DOI: 10.4155/fmc-2018-0108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
AIM We aimed to investigate the effect of triptolide on cisplatin resistance in ovarian cancer cell lines. METHODS The apoptosis of ovarian cancer cell lines A2780 and A2780/CP70 was determined by flow cytometry. Protein expression levels of Hexokinase 2 (HK2) were detected by western blot. Possible mRNAs which target HK 2 expression were predicted through online tools miRbase. RESULTS Triptolide-promoted cell apoptosis of cisplatin-resistant ovarian cancer cell line A2780/CP70. Triptolide treatment sensitized the effect of cisplatin in A2780/CP70, as evidenced by decreased survival fraction of A2780/CP70 cells. Mechanically, triptolide treatment inhibited HK 2 expression in A2780/CP70 cells. In addition, Hsa-mir-6751 expression level was upregulated in A2780/CP70 cells after triptolide treatment.
Collapse
|
18
|
Wang J, Zhang Z, Li R, Sun W, Chen J, Zhang H, Shu K, Lei T. Triptolide inhibits pituitary adenoma cell viability, migration and invasion via ADAM12/EGFR signaling pathway. Life Sci 2018; 194:150-156. [DOI: 10.1016/j.lfs.2017.12.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/29/2022]
|
19
|
Liu W, Yin NC, Liu H, Nan KJ. Cav-1 promote lung cancer cell proliferation and invasion through lncRNA HOTAIR. Gene 2018; 641:335-340. [DOI: 10.1016/j.gene.2017.10.070] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/23/2017] [Indexed: 12/24/2022]
|
20
|
Díaz-Valdivia NI, Calderón CC, Díaz JE, Lobos-González L, Sepulveda H, Ortíz RJ, Martinez S, Silva V, Maldonado HJ, Silva P, Wehinger S, Burzio VA, Torres VA, Montecino M, Leyton L, Quest AFG. Anti-neoplastic drugs increase caveolin-1-dependent migration, invasion and metastasis of cancer cells. Oncotarget 2017; 8:111943-111965. [PMID: 29340103 PMCID: PMC5762371 DOI: 10.18632/oncotarget.22955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022] Open
Abstract
Expression of the scaffolding protein Caveolin-1 (CAV1) enhances migration and invasion of metastatic cancer cells. Yet, CAV1 also functions as a tumor suppressor in early stages of cancer, where expression is suppressed by epigenetic mechanisms. Thus, we sought to identify stimuli/mechanisms that revert epigenetic CAV1 silencing in cancer cells and evaluate how this affects their metastatic potential. We reasoned that restricted tissue availability of anti-neoplastic drugs during chemotherapy might expose cancer cells to sub-therapeutic concentrations, which activate signaling pathways and the expression of CAV1 to favor the acquisition of more aggressive traits. Here, we used in vitro [2D, invasion] and in vivo (metastasis) assays, as well as genetic and biochemical approaches to address this question. Colon and breast cancer cells were identified where CAV1 levels were low due to epigenetic suppression and could be reverted by treatment with the methyltransferase inhibitor 5’-azacytidine. Exposure of these cells to anti-neoplastic drugs for short periods of time (24-48 h) increased CAV1 expression through ROS production and MEK/ERK activation. In colon cancer cells, increased CAV1 expression enhanced migration and invasion in vitro via pathways requiring Src-family kinases, as well as Rac-1 activity. Finally, elevated CAV1 expression in colon cancer cells following exposure in vitro to sub-cytotoxic drug concentrations increased their metastatic potential in vivo. Therefore exposure of cancer cells to anti-neoplastic drugs at non-lethal drug concentrations induces signaling events and changes in transcription that favor CAV1-dependent migration, invasion and metastasis. Importantly, this may occur in the absence of selection for drug-resistance.
Collapse
Affiliation(s)
- Natalia I Díaz-Valdivia
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Claudia C Calderón
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge E Díaz
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Hugo Sepulveda
- Gene Regulation Laboratory, Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Rina J Ortíz
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Universidad Bernardo O Higgins, Facultad de Salud, Departamento de Ciencias Químicas y Biológicas, Santiago, Chile
| | - Samuel Martinez
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Horacio J Maldonado
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Patricio Silva
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Sergio Wehinger
- Faculty of Health Sciences, University of Talca, Interdisciplinary Excellence Research Program Healthy Ageing (PIEI-ES), Talca, Chile
| | - Verónica A Burzio
- Fundación Ciencia & Vida, Santiago, Chile.,Faculty of Biological Sciences, Universidad Andrés Bello, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Martín Montecino
- Gene Regulation Laboratory, Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
21
|
Catalpol suppressed proliferation, growth and invasion of CT26 colon cancer by inhibiting inflammation and tumor angiogenesis. Biomed Pharmacother 2017; 95:68-76. [DOI: 10.1016/j.biopha.2017.08.049] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
|
22
|
Han F, Xue M, Chang Y, Li X, Yang Y, Sun B, Chen L. Triptolide Suppresses Glomerular Mesangial Cell Proliferation in Diabetic Nephropathy Is Associated with Inhibition of PDK1/Akt/mTOR Pathway. Int J Biol Sci 2017; 13:1266-1275. [PMID: 29104493 PMCID: PMC5666525 DOI: 10.7150/ijbs.20485] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023] Open
Abstract
Mesangial cell proliferation has been identified as a mainly contributing factor to glomerulosclerosis, which is typical of diabetic nephropathy. However, the specific mechanisms and therapies remain unclear. PDK1 is a critical regulator of cell proliferation, but the specific role of PDK1 in diabetic nephropathy has not been fully illuminated. In the current study, we demonstrated that triptolide (TP) ameliorated albuminuria in the high fat diet/STZ-induced diabetic rats. TP also suppressed the increased proliferating cell markers Ki-67 and PCNA in the kidney tissues. Our results of MTT and cell cycle analysis further confirmed that TP significantly inhibited mesangial cell proliferation, and the inhibition of PDK1/Akt/mTOR pathway might be the underlying mechanisms. In addition, we also found that the PDK1 activator (PS48) could reverse the cell proliferation inhibition role of TP. These data suggest that TP may be useful in prevention of diabetic glomerulosclerosis and that PDK1/Akt/mTOR pathway might be the underlying mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | - Bei Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University
| | - Liming Chen
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University
| |
Collapse
|