51
|
Wu C, Chen W, Fang M, Boye A, Tao X, Xu Y, Hou S, Yang Y. Compound Astragalus and Salvia miltiorrhiza extract inhibits hepatocellular carcinoma progression via miR-145/miR-21 mediated Smad3 phosphorylation. JOURNAL OF ETHNOPHARMACOLOGY 2019; 231:98-112. [PMID: 30412748 DOI: 10.1016/j.jep.2018.11.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/22/2018] [Accepted: 11/03/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Astragalus and Salvia miltiorrhiza extract (CASE), containing astragalosides, astragalus polysaccharide extracted from Astragalus membranaceus (Fisch.) Bge. and salvianolic acids from Salvia miltiorhiza Bge., has been found to inhibit hepatocarcinogenesis via mediating transforming growth factor-β (TGF-β)/Smad signaling, especially Smad3 phosphorylation. The crucial interaction between microRNA-145/microRNA-21 (miR-145/miR-21) and Smad3 phosphorylation is implicated in the pathogenesis and progression of hepatocellular carcinoma (HCC). However, effects of CASE on HCC progression involved in the expression of miR-145/miR-21 and their interaction with Smad3 phosphorylation downstream of TGF-β/MAPK/Smad pathway remain unclear. This study addressed above questions using in vitro (HepG2 cells) and in vivo (Xenografts of nude mice) models of HCC. MATERIALS AND METHODS In vivo [Diethylnitrosamine (DEN)-induced HCC in rats] and in vitro [TGF-β1-stimulated HepG2 cells] models of HCC were established and co-administrated using graded doses/concentrations CASE (60, 120, 240 mg/kg used in rats; 20, 40, 80 µg/ml used in HepG2 cells), miR-145 and miR-21 were measured. HepG2 cells were transfected with miR-145 antagomir, miR-21 agomir and Smad3C/L plasmids (Smad3 EPSM, Smad3 3S-A and Smad3 WT related to up-regulated expression of pSmad3C, pSmad3L and pSmad3C/3L respectively) and then treated by CASE (80 µg/ml). Similarly, HepG2 cell xenografted nude mice were administered with miR-145 antagomir, miR-21 agomir and CASE (310 mg/kg); Smad3 WT, Smad3 EPSM and Smad3 3S-A plasmids stably transfected HepG2 cell lines were constructed respectively and their xenografted nude mice were established, and then treated by CASE (310 mg/kg). Cell proliferation, migration, apoptosis, tumor growth and histopathologic characteristics of xenografts were assessed; also, domain-specific Smad3 phosphorylation isoforms (pSmad3C/pSmad3L), activated MAPKs (pERK1/2, pJNK1/2, pp38) and miR-145, miR-21 were measured. RESULTS CASE up-regulated miR-145 while down-regulated miR-21 expression in both rats with DEN-induced HCC and TGF-β1-stimulated HepG2 cells; CASE inhibited cell migration, proliferation and tumor growth while facilitated cell apoptosis in TGF-β1-stimulated HepG2 cells and xenografts of nude mice with miR-145 antagomir/miR-21 agomir treatment via increasing miR-145 and facilitating miR-145 modulated pSmad3L→pSmad3C signaling switch while decreasing miR-21 and inhibiting miR-21 modulated MAPK-dependent Smad3L phosphorylation. Also, up-regulated pSmad3C enhanced inhibited effect of CASE on tumor growth and facilitated effect of CASE on cell apoptosis involved in increased miR-145 while decreased miR-21 expression, however, inverse phenomena were observed when up-regulated pSmad3L. CONCLUSION Our results suggest that CASE inhibits HCC progression via mediating the interaction of miR-145/miR-21 and Smad3 phosphorylation, especially miR-145/miR-21 mediated Smad3 phosphorylation, which maybe provides an important theoretical foundation for CASE's anti-HCC therapy used for patients in a near future.
Collapse
Affiliation(s)
- Chao Wu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Weiyang Chen
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Meng Fang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Alex Boye
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Xiangming Tao
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Yuanyuan Xu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China
| | - Shu Hou
- Department of Pediatrics, First Affiliated Hospital, Anhui Medical University, Hefei 230022, China
| | - Yan Yang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
52
|
A Novel Discovery: Holistic Efficacy at the Special Organ Level of Pungent Flavored Compounds from Pungent Traditional Chinese Medicine. Int J Mol Sci 2019; 20:ijms20030752. [PMID: 30754631 PMCID: PMC6387020 DOI: 10.3390/ijms20030752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/25/2022] Open
Abstract
Pungent traditional Chinese medicines (TCMs) play a vital role in the clinical treatment of hepatobiliary disease, gastrointestinal diseases, cardiovascular diseases, diabetes, skin diseases and so on. Pungent TCMs have a vastness of pungent flavored (with pungent taste or smell) compounds. To elucidate the molecular mechanism of pungent flavored compounds in treating cardiovascular diseases (CVDs) and liver diseases, five pungent TCMs with the action of blood-activating and stasis-resolving (BASR) were selected. Here, an integrated systems pharmacology approach is presented for illustrating the molecular correlations between pungent flavored compounds and their holistic efficacy at the special organ level. First, we identified target proteins that are associated with pungent flavored compounds and found that these targets were functionally related to CVDs and liver diseases. Then, based on the phenotype that directly links human genes to the body parts they affect, we clustered target modules associated with pungent flavored compounds into liver and heart organs. We applied systems-based analysis to introduce a pungent flavored compound-target-pathway-organ network that clarifies mechanisms of pungent substances treating cardiovascular diseases and liver diseases by acting on the heart/liver organ. The systems pharmacology also suggests a novel systematic strategy for rational drug development from pungent TCMs in treating cardiovascular disease and associated liver diseases.
Collapse
|
53
|
Wu R, Dong S, Cai FF, Chen XL, Yang MD, Liu P, Su SB. Active Compounds Derived from Fuzheng Huayu Formula Protect Hepatic Parenchymal Cells from Apoptosis Based on Network Pharmacology and Transcriptomic Analysis. Molecules 2019; 24:molecules24020338. [PMID: 30669350 PMCID: PMC6358846 DOI: 10.3390/molecules24020338] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
Fuzheng huayu formula (FZHY), an antifibrotic traditional Chinese medicine, is frequently used for the treatment of liver fibrosis. In this study, network analysis, transcriptomic analysis, assays of cell apoptosis, viability and protein expression were used for investigating the effects and mechanisms of compounds derived from FZHY on hepatic parenchymal cell (HPC) protection and hepatic stellate cell activation. Network pharmacology analysis found that 6 major compounds and 39 potential targets were important network nodes. Our analysis predicted that the active compounds of FZHY, including hederagenin, luteolin and tanshinone IIA inhibited cell apoptosis (p < 0.05), increased PI3K expression and reduced cleaved caspase 3 expression and the Bax/Bcl-w ratio (p < 0.05) in L02 cells that had apoptosis induced by TNF-α. Few significant changes caused by FZHY, hederagenin, luteolin and tanshinone IIA were observed in hepatic stellate Lx2 cells upon TGF-β1 induction. These data suggest that FZHY is active against liver fibrosis, protects hepatic parenchymal cells from apoptosis, and recovers liver function, possibly through the effects of its active compounds hederagenin, luteolin and tanshinone IIA and is involved in the inhibition of apoptosis in HPCs, possibly through regulating the PI3K, ERK, cleaved caspase 3 and Bax/Bcl-w levels.
Collapse
Affiliation(s)
- Rong Wu
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Shu Dong
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Fei-Fei Cai
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiao-Le Chen
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Meng-Die Yang
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ping Liu
- E-institute of Shanghai Municipal Education Committee, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Shi-Bing Su
- Research Center for Traditional Chinese Medicine Complexity System, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
54
|
Wang S, Zhang C, Li Y, Li P, Zhang D, Li C. Anti-liver cancer effect and the mechanism of arsenic sulfide in vitro and in vivo. Cancer Chemother Pharmacol 2018; 83:519-530. [PMID: 30542770 DOI: 10.1007/s00280-018-3755-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE This study aimed at investigating the anti-tumor effect of arsenic sulfide (As2S2) against liver cancer both in vivo and in vitro and to elucidate its underlying mechanisms. METHODS Cell viability of the human hepatocellular carcinoma cell lines SMMC-7721, BEL-7402, HepG2 were measured by CCK-8 assay. The effects of As2S2 on cell proliferation and apoptosis of SMMC-7721 cells were investigated using Calcein-AM and PI staining, Hoechst 33258 staining, crystal violet staining, and JC-1 staining. Cell cycle and Annexin V/PI assay were analyzed via flow cytometry. The expression of apoptosis-related proteins, phosphorylation of PI3K and AKT were detected by Western blotting. H22-bearing mice model was established to evaluate the anti-tumor effect of As2S2 in vivo. HE staining, PCNA was observed via immunohistochemistry, and TUNEL assay was used to assess the anti-proliferation and pro-apoptotic effects of As2S2. RESULTS As2S2 significantly inhibited the growth of human hepatoma cells SMMC-7721, BEL-7402 and HepG2. As2S2 inhibited cell proliferation effectively by inducing G0/G1 cell cycle arrest in SMMC-7721 cells. As2S2 could increase Bax/Bcl-2 ratio, decrease mitochondrial membrane potential, promote the release of cytochrome c, increase the levels of cleaved caspase-3 and PARP, indicating that As2S2 induced apoptosis in SMMC-7721 cells via mitochondrial-mediated apoptosis pathway. Further research showed that As2S2 inhibited the PI3K/AKT signaling pathway leading to apoptotic cell death. In addition, As2S2 significantly inhibited tumor growth in H22-bearing mice and induced apoptosis by deactivating PI3K/AKT pathway, which was consistent with the in vitro results. CONCLUSION These findings suggested that As2S2 could induce apoptosis of liver cancer cells in vitro and in vivo, which was related to PI3K/AKT-mediated mitochondrial pathway and may provide a novel promising therapeutic agent for liver cancer treatment.
Collapse
Affiliation(s)
- Shudan Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yumei Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ping Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Dafang Zhang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Chaoying Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
55
|
A Network Pharmacology Analysis to Explore the Effect of Astragali Radix-Radix Angelica Sinensis on Traumatic Brain Injury. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3951783. [PMID: 30596090 PMCID: PMC6286735 DOI: 10.1155/2018/3951783] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/04/2018] [Accepted: 11/08/2018] [Indexed: 12/23/2022]
Abstract
Traumatic brain injury (TBI) is a critical public health and socioeconomic problem worldwide. The herb pair Astragali Radix (AR)-Radix Angelica Sinensis (RAS) is a common prescribed herbal formula or is added to other Chinese medicine prescriptions for traumatic brain injury (TBI) treatment. However, the underlying mechanisms are unclear. In this study, we aimed to explore the active ingredients and action targets of AR-RAS based on the combined methods of network pharmacology prediction and experimental verification. Furthermore, the corresponding potential mechanisms of “multicomponents, multitargets, and multipathways” were disclosed. Methods. A network pharmacology approach including ADME (absorption, distribution, metabolism, and excretion) filter analysis, target prediction, known therapeutic targets collection, Gene Ontology (GO), pathway enrichment analysis, and network construction was used in this study. Further verification experiments were performed to reveal the therapeutic effects of AR-RAS in a rat model of TBI. Results. The comprehensive systematic approach was to successfully identify 14 bioactive ingredients in AR-RAS, while 33 potential targets hit by these ingredients related to TBI. Based on GO annotation analysis, multiple biological processes were significantly regulated by AR-RAS. In addition, 89 novel signaling pathways (P<0.05) underlying the effects of AR-RAS for TBI treatment were identified by DAVID. The neurotrophin signaling pathway was suggested as the major related pathway targeted by AR-RAS to improve axonal growth. The animal experiment confirmed that AR-RAS significantly induced tissue recovery and improved neurological deficits on the 14th day (P<0.01). Treatment with AR-RAS markedly reduced the protein and mRNA expression level of NogoA in the hippocampus of TBI rats. Conclusion. Our work illuminates the “multicompounds, multitargets, and multipathways” curative action of AR-RAS in the treatment of TBI by network pharmacology. The animal experiment verifies the effects of AR-RAS on neurological function improvement and axonal outgrowth via downregulation of NogoA expression, providing a theoretical basis for further research on treatment of TBI.
Collapse
|
56
|
Systematic Investigation of Scutellariae Barbatae Herba for Treating Hepatocellular Carcinoma Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4365739. [PMID: 30584453 PMCID: PMC6280310 DOI: 10.1155/2018/4365739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/30/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
As the fifth most common type of malignant cancers globally, hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide. As a long-time medicinal herb in Traditional Chinese Medicine (TCM), Scutellariae Barbatae Herba (SBH) has also been used for treating various cancers including HCC, but its underlying mechanisms have not been completely clarified. Presently, an innovative network-pharmacology platform was introduced to systematically elucidate the pharmacological mechanisms of SBH against HCC, adopting active ingredients prescreening, target fishing, and network analysis. The results revealed that SBH appeared to work on HCC probably through regulating 4 molecular functions, 20 biological processes, and hitting on 21 candidate targets involved in 40 pathways. By in-depth analysis of the first-ranked signaling pathway and hit genes, only TTR was highly and specially expressed in the liver tissue. TTR might play a crucial role in neutrophil degranulation pathway during SBH against HCC. Hence, TTR might become a therapeutic target of HCC. The study investigated the anti-hepatoma mechanisms of SBH from a holistic perspective, which provided a theoretical foundation for further experimental research and rational clinical application of SBH.
Collapse
|
57
|
Lee SU, Ryu HW, Lee S, Shin IS, Choi JH, Lee JW, Lee J, Kim MO, Lee HJ, Ahn KS, Hong ST, Oh SR. Lignans Isolated From Flower Buds of Magnolia fargesii Attenuate Airway Inflammation Induced by Cigarette Smoke in vitro and in vivo. Front Pharmacol 2018; 9:970. [PMID: 30258361 PMCID: PMC6143820 DOI: 10.3389/fphar.2018.00970] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/06/2018] [Indexed: 12/22/2022] Open
Abstract
The flower buds of Magnolia fargesii, known traditionally as Xinyi, exert anti-inflammatory effects against inflammatory lung diseases such as COPD. Lignans isolated from Xinyi are an important group of plant-derived anti-inflammatory compounds. However, the mechanisms of action underlying their protective effects against COPD are not yet fully understood. Here, we showed that seven lignans (lignans 1–7) obtained from a CHCl3 fraction of Xinyi effectively suppress the inflammatory response in CSC-stimulated airway epithelial cells (in vitro) and in a mouse model of COPD established by exposure to CS and LPS. The CHCl3 fraction was found to inhibit CSC-induced IL-6 expression in human airway epithelial cells and to suppress the infiltration of inflammatory cells (neutrophils and macrophages) and secretion of inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the mouse model. Similarly, each of the seven lignans isolated from the CHCl3 fraction also suppressed the infiltration of inflammatory cells (neutrophils and macrophages) and secretion of inflammatory mediators such as reactive oxygen species (ROS), TNF-α, and IL-6 in vivo. Notably, all lignan compounds significantly suppressed both extracellular signal-related kinase (ERK) and Akt phosphorylation levels in CSC-stimulated human lung mucoepidermoid carcinoma (NCI-H292) cells. Of these, lignan 1 (dimethylpinoresinol) inhibited the expression of CSC-induced inflammatory cytokines (IL-1β, -6, and -8) in vitro in a dose-dependent manner by suppressing the activation of epidermal growth factor receptor (EGFR) and its downstream effectors, including ERK and Akt, in NCI-H292 cells. Our results show that the lignans isolated from Xinyi may prevent airway inflammatory diseases through the suppression of EGFR and its downstream effectors.
Collapse
Affiliation(s)
- Su-Ui Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Seoghyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea.,College of Bioscience and Biotechnology, Chungnam National University, Daejeon, South Korea
| | - In-Sik Shin
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, Gwangju, South Korea
| | - Ji-Hee Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Jinhyuk Lee
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Mun Ock Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Hyun-Jun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Sung-Tae Hong
- Department of Anatomy and Cell Biology, Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| |
Collapse
|
58
|
Zhu H, Hao J, Niu Y, Liu D, Chen D, Wu X. Molecular targets of Chinese herbs: a clinical study of metastatic colorectal cancer based on network pharmacology. Sci Rep 2018; 8:7238. [PMID: 29740162 PMCID: PMC5940835 DOI: 10.1038/s41598-018-25500-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/23/2018] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence has shown that Chinese herbal medicine (CHM) has promising therapeutic effects in colorectal cancer (CRC); however, the active ingredients and potential targets remain unclear. In this study, we aimed to investigate the relative molecular targets of the Chinese herbs that have been found effective in treating metastatic CRC (mCRC) based on clinical data and network pharmacology. In multivariate analysis CHM resulted an independent prognostic factor. The hazard ratio was 0.103 (95% confidence interval = 0.064-0.164; P < 0.001). Compared with the non-CHM group, the median survival time of the CHM group was also improved (40 versus 12 months; P < 0.001). Eighteen out of 295 herbs showed significant correlation with survival results (P < 0.05). Bioinformatics analysis indicated that the 18 herbs realize anti-CRC activity mainly through suppressing the proliferative activity of ERBB2, peroxisome proliferator-activated receptor gamma, and retinoid X receptor, suppressing angiogenesis via inhibition of VEGFR and VEGFA expression, inhibiting the phosphatidylinositol-3-kinase/AKT1 signaling pathway directly through SRC and AKT1, and reducing tumor necrosis factor-induced inflammation.
Collapse
MESH Headings
- Adult
- Aged
- Antineoplastic Agents, Phytogenic/therapeutic use
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/mortality
- Colorectal Neoplasms/pathology
- Computational Biology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks/drug effects
- Humans
- Lymphatic Metastasis
- Male
- Medicine, Chinese Traditional/methods
- Middle Aged
- Molecular Targeted Therapy
- Multivariate Analysis
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- PPAR gamma/antagonists & inhibitors
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Phosphatidylinositol 3-Kinase/genetics
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Receptors, Vascular Endothelial Growth Factor/genetics
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Retinoid X Receptors/antagonists & inhibitors
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Retrospective Studies
- Signal Transduction
- Survival Analysis
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Hongxu Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Jian Hao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yangyang Niu
- Tianjin Children's Hospital, Tianjin, 300134, China
| | - Dan Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Dan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, Qi-Xiang-Tai Road, Tianjin, 300070, China
| | - Xiongzhi Wu
- Zhong-Shan-Men Inpatient Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
59
|
Gao L, Wang KX, Zhang NN, Li JQ, Qin XM, Wang XL. 1H Nuclear Magnetic Resonance Based Metabolomics Approach Reveals the Metabolic Mechanism of (−)-5-Hydroxy-equol against Hepatocellular Carcinoma Cells in Vitro. J Proteome Res 2018; 17:1833-1843. [DOI: 10.1021/acs.jproteome.7b00853] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
| | - Ke-xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
- College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Nan-nan Zhang
- College of Life Sciences, Hebei Agricultural University, Baoding 071001, PR China
| | - Jia-qi Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
- College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Xue-mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
| | - Xiu-ling Wang
- College of Life Sciences, Hebei Agricultural University, Baoding 071001, PR China
| |
Collapse
|
60
|
Yang K, Liu G, Wang N, Zhang R, Yu J, Chen J, Zhou X. Heterogeneous network propagation for herb target identification. BMC Med Inform Decis Mak 2018; 18:17. [PMID: 29589568 PMCID: PMC5872392 DOI: 10.1186/s12911-018-0592-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Identifying targets of herbs is a primary step for investigating pharmacological mechanisms of herbal drugs in Traditional Chinese medicine (TCM). Experimental targets identification of herbs is a difficult and time-consuming work. Computational method for identifying herb targets is an efficient approach. However, how to make full use of heterogeneous network data about herbs and targets to improve the performance of herb targets prediction is still a dilemma. METHODS In our study, a random walk algorithm on the heterogeneous herb-target network (named heNetRW) has been proposed to identify protein targets of herbs. By building a heterogeneous herb-target network involving herbs, targets and their interactions and simulating random walk algorithm on the network, the candidate targets of the given herb can be predicted. RESULTS The experimental results on large-scale dataset showed that heNetRW had higher performance of targets prediction than PRINCE (improved F1-score by 0.08 and Hit@1 by 21.34% in one validation setting, and improved F1-score by 0.54 and Hit@1 by 69.08% in the other validation setting). Furthermore, we evaluated novel candidate targets of two herbs (rhizoma coptidis and turmeric), which showed our approach could generate potential targets that are valuable for further experimental investigations. CONCLUSIONS Compared with PRINCE algorithm, heNetRW algorithm can fuse more known information (such as, known herb-target associations and pathway-based similarities of protein pairs) to improve prediction performance. Experimental results also indicated heNetRW had higher performance than PRINCE. The prediction results not only can be used to guide the selection of candidate targets of herbs, but also help to reveal the molecule mechanisms of herbal drugs.
Collapse
Affiliation(s)
- Kuo Yang
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, 100044 China
| | - Guangming Liu
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, 100044 China
| | - Ning Wang
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, 100044 China
| | - Runshun Zhang
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Jian Yu
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, 100044 China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xuezhong Zhou
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, 100044 China
- Data Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700 China
| |
Collapse
|
61
|
Mao Y, Hao J, Jin ZQ, Niu YY, Yang X, Liu D, Cao R, Wu XZ. Network pharmacology-based and clinically relevant prediction of the active ingredients and potential targets of Chinese herbs in metastatic breast cancer patients. Oncotarget 2018; 8:27007-27021. [PMID: 28212580 PMCID: PMC5432314 DOI: 10.18632/oncotarget.15351] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/22/2017] [Indexed: 01/24/2023] Open
Abstract
Chinese Herbal Medicine (CHM) plays a significant role in breast cancer treatment. We conduct the study to ascertain the relative molecular targets of effective Chinese herbs in treating stage IV breast cancer.Survival benefit of CHM was verified by Kaplan-Meier method and Cox regression analysis. A bivariate correlation analysis was used to find and establish the effect of herbs in complex CHM formulas. A network pharmacological approach was adopted to explore the potential mechanisms of CHM.Patients in the CHM group had a median survival time of 55 months, which was longer than the 23 months of patients in the non-CHM group. Cox regression analysis indicated that CHM was an independent protective factor. Correlation analysis showed that 10 herbs were strongly correlated with favorable survival outcomes (P<0.01). Bioinformatics analyses suggested that the 10 herbs might achieve anti-breast cancer activity primarily through inhibiting HSP90, ERα and TOP-II related pathways.
Collapse
Affiliation(s)
- Yu Mao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Jian Hao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Zi-Qi Jin
- Tianjin Medical University, Tianjin, 300070, China
| | | | - Xue Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Dan Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Rui Cao
- Zhong-Shan-Men Inpatient Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xiong-Zhi Wu
- Zhong-Shan-Men Inpatient Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| |
Collapse
|
62
|
Gao L, Wang KX, Zhou YZ, Fang JS, Qin XM, Du GH. Uncovering the anticancer mechanism of Compound Kushen Injection against HCC by integrating quantitative analysis, network analysis and experimental validation. Sci Rep 2018; 8:624. [PMID: 29330507 PMCID: PMC5766629 DOI: 10.1038/s41598-017-18325-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/08/2017] [Indexed: 12/17/2022] Open
Abstract
Compound Kushen Injection (CKI) is a Traditional Chinese Medicine (TCM) preparation that has been clinically used in China to treat various types of solid tumours. Although several studies have revealed that CKI can inhibit the proliferation of hepatocellular carcinoma (HCC) cell lines, the active compounds, potential targets and pathways involved in these effects have not been systematically investigated. Here, we proposed a novel idea of “main active compound-based network pharmacology” to explore the anti-cancer mechanism of CKI. Our results showed that CKI significantly suppressed the proliferation and migration of SMMC-7721 cells. Four main active compounds of CKI (matrine, oxymatrine, sophoridine and N-methylcytisine) were confirmed by the integration of ultra-performance liquid chromatography/mass spectrometry (UPLC-MS) with cell proliferation assays. The potential targets and pathways involved in the anti-HCC effects of CKI were predicted by a network pharmacology approach, and some of the crucial proteins and pathways were further validated by western blotting and metabolomics approaches. Our results indicated that CKI exerted anti-HCC effects via the key targets MMP2, MYC, CASP3, and REG1A and the key pathways of glycometabolism and amino acid metabolism. These results provide insights into the mechanism of CKI by combining quantitative analysis of components, network pharmacology and experimental validation.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, PR China.
| | - Ke-Xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, PR China.,College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan, 030006, China
| | - Yu-Zhi Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, PR China
| | - Jian-Song Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, PR China.
| | - Guan-Hua Du
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, PR China.,Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
63
|
Xiang H, Zhang Q, Qi B, Tao X, Xia S, Song H, Qu J, Shang D. Chinese Herbal Medicines Attenuate Acute Pancreatitis: Pharmacological Activities and Mechanisms. Front Pharmacol 2017; 8:216. [PMID: 28487653 PMCID: PMC5403892 DOI: 10.3389/fphar.2017.00216] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/06/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is a commonly occurring gastrointestinal disorder. An increase in the annual incidence of AP has been observed, and it causes acute hospitalization and high mortality. The diagnosis and treatment guidelines for AP recommend conservative medical treatments focused on reducing pancreatic secretion and secondary injury, as a primary therapeutic approach. Unfortunately, the existing treatment options have limited impact on the incidence and severity of AP due to the complex and multifaceted pathological process of this disease. In recent decades, Chinese herbal medicines (CHMs) have been used as efficient therapeutic agents to attenuate AP in Asian countries. Despite early cell culture, animal models, and clinical trials, CHMs are capable of interacting with numerous molecular targets participating in the pathogenesis of AP; however, comprehensive, up-to-date communication in this field is not yet available. This review focuses on the pharmacological activities of CHMs against AP in vitro and in vivo and the underlying mechanisms. A computational prediction of few selected and promising plant-derived molecules (emodin, baicalin, resveratrol, curcumin, ligustrazine, and honokiol) to target numerous proteins or networks involved in AP was initially established based on a network pharmacology simulation. Moreover, we also summarized some potential toxic natural products for pancreas in order to more safe and reasonable medication. These breakthrough findings may have important implications for innovative drug research and the future development of treatments for AP.
Collapse
Affiliation(s)
- Hong Xiang
- College (Institute) of Integrative Medicine, Dalian Medical UniversityDalian, China
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Bing Qi
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical UniversityDalian, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Huiyi Song
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Jialin Qu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Dong Shang
- College (Institute) of Integrative Medicine, Dalian Medical UniversityDalian, China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| |
Collapse
|
64
|
Chen M, Yang F, Yang X, Lai X, Gao Y. Systematic Understanding of Mechanisms of a Chinese Herbal Formula in Treatment of Metabolic Syndrome by an Integrated Pharmacology Approach. Int J Mol Sci 2016; 17:ijms17122114. [PMID: 27999264 PMCID: PMC5187914 DOI: 10.3390/ijms17122114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MS) is becoming a worldwide health problem. Wendan decoction (WDD)—a famous traditional Chinese medicine formula—has been extensively employed to relieve syndromes related to MS in clinical practice in China. However, its pharmacological mechanisms still remain vague. In this study, a comprehensive approach that integrated chemomics, principal component analysis, molecular docking simulation, and network analysis was established to elucidate the multi-component and multi-target mechanism of action of WDD in treatment of MS. The compounds in WDD were found to possess chemical diversity, complexity and drug-likeness compared to MS drugs. Six nuclear receptors were obtained to have strong binding affinity with 217 compounds of five herbs in WDD. The importance roles of targets and herbs were also identified due to network parameters. Five compounds from Radix Glycyrrhizae Preparata can hit all six targets, which can assist in screening new MS drugs. The pathway network analysis demonstrated that the main pharmacological effects of WDD might lie in maintaining lipid and glucose metabolisms and anticancer activities as well as immunomodulatory and hepatoprotective effects. This study provided a comprehensive system approach for understanding the multi-component, multi-target and multi-pathway mechanisms of WDD during the treatment of MS.
Collapse
Affiliation(s)
- Meimei Chen
- College of Chemistry and Chemical Engineering, Fujian Normal University, Fuzhou 350007, China.
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Fafu Yang
- College of Chemistry and Chemical Engineering, Fujian Normal University, Fuzhou 350007, China.
| | - Xuemei Yang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Xinmei Lai
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Yuxing Gao
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
65
|
Xiang H, Wang G, Qu J, Xia S, Tao X, Qi B, Zhang Q, Shang D. Yin-Chen-Hao Tang Attenuates Severe Acute Pancreatitis in Rat: An Experimental Verification of In silico Network Target Prediction. Front Pharmacol 2016; 7:378. [PMID: 27790147 PMCID: PMC5061810 DOI: 10.3389/fphar.2016.00378] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/28/2016] [Indexed: 12/12/2022] Open
Abstract
Yin-Chen-Hao Tang (YCHT) is a classical Chinese medicine compound that has a long history of clinical use in China for the treatment of inflammatory diseases. However, the efficacy and mechanisms of YCHT for the treatment of severe acute pancreatitis (SAP) are not known. The current study investigated the pharmacological properties of YCHT against SAP and its underlying mechanisms. A computational prediction of potential targets of YCHT was initially established based on a network pharmacology simulation. The model suggested that YCHT attenuated SAP progress by apoptosis inducement, anti-inflammation, anti-oxidation and blood lipid regulation. These effects were validated in SAP rats. YCHT administration produced the following results: (1) significantly inhibited the secretion of pancreatic enzymes and protected pancreatic tissue; (2) obviously increased the number of in situ terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive cells and induced apoptosis; (3) markedly inhibited neutrophil infiltration to the impaired pancreas and reduced the inflammatory reaction; (4) notably enhanced the activities of antioxidant enzymes and decreased the nitric oxide synthase levels; (5) significantly reduced the levels of triglycerides, total cholesterol and low-density lipoprotein and increased high-density lipoprotein; and (6) significantly up-regulated peroxisome proliferator-activated receptor-γ (PPARγ) and down-regulated nuclear factor-kappa B (NF-κB). In summary, these results demonstrated that YCHT attenuated SAP progress by inducing apoptosis, repressing inflammation, alleviating oxidative stress and regulating lipid metabolism partially via regulation of the NF-κB/PPARγ signal pathway.
Collapse
Affiliation(s)
- Hong Xiang
- College (Institute) of Integrative Medicine, Dalian Medical University Dalian, China
| | - Guijun Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University Jinzhou, China
| | - Jialin Qu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University Dalian, China
| | - Bing Qi
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Dong Shang
- College (Institute) of Integrative Medicine, Dalian Medical UniversityDalian, China; Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| |
Collapse
|
66
|
Gai WT, Yu DP, Wang XS, Wang PT. Anti-cancer effect of ursolic acid activates apoptosis through ROCK/PTEN mediated mitochondrial translocation of cofilin-1 in prostate cancer. Oncol Lett 2016; 12:2880-2885. [PMID: 27698874 DOI: 10.3892/ol.2016.5015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 05/13/2016] [Indexed: 11/05/2022] Open
Abstract
Ursolic acid is a type of pentacyclic triterpene compound with multiple pharmacological activities including cancer resistance, protection from liver injury, antisepsis, anti-inflammation and antiviral activity. The present study aimed to investigate the anticancer effect of ursolic acid. Ursolic acid activates cell apoptosis and its pro-apoptotic mechanism remains to be fully elucidated. Cell Counting kit-8 assays, flow cytometric analysis and analysis of caspase-3 and caspase-9 activity were used to estimate the anticancer effect of ursolic acid on DU145 prostate cancer cells. The protein expression of cytochrome c, rho-associated protein kinase (ROCK), phosphatase and tensin homolog (PTEN) and cofilin-1 were examined using western blot analysis. In the present study, ursolic acid significantly suppressed cell growth and induced apoptosis, as well as increasing caspase-3 and caspase-9 activities of DU145 cells. Furthermore, cytoplasmic and mitochondrial cytochrome c protein expression was significantly activated and suppressed, respectively, by ursolic acid. Ursolic acid significantly suppressed the ROCK/PTEN signaling pathway and inhibited cofilin-1 protein expression in DU145 cells. The results of the present study indicate that the anticancer effect of ursolic acid activates cell apoptosis through ROCK/PTEN mediated mitochondrial translocation of cofilin-1 in prostate cancer.
Collapse
Affiliation(s)
- Wen-Tao Gai
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China; Department of Urology, Yantai Municipal Laiyang Central Hospital, Laiyang, Shandong 265200, P.R. China
| | - Da-Peng Yu
- Department of Urology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Xin-Sheng Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Pei-Tao Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|