51
|
Yu Q, Zhou X, Xia Q, Shen J, Yan J, Zhu J, Li X, Shu M. Long non-coding RNA CCAT1 that can be activated by c-Myc promotes pancreatic cancer cell proliferation and migration. Am J Transl Res 2016; 8:5444-5454. [PMID: 28078015 PMCID: PMC5209495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/14/2016] [Indexed: 06/06/2023]
Abstract
This study aimed to investigate the potential role of lncRNA CCAT1 in the progression of pancreatic cancer (PC) and to reveal its possible molecular mechanism. The expression of CCAT1 was analyzed in PC tissues and their adjacent normal tissues from patients diagnosed with PC and in two pancreas cancer cell lines, namely PANC-1 and Aspc-1 using real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. The effects of CCAT1 expression on cell proliferation, cell cycle, and migration were analyzed using MTT assay, flow cytometry, and transwell assay, respectively. The effects of c-Myc expression on the expression of CCAT1 and E-box were also analyzed using RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP) assays, respectively. The results showed that CCAT1 was highly expressed in PC tissues compared to the adjacent tissues (P<0.01) and was also overexpressed in PANC-1 and Aspc-1 cells (P<0.05). The silencing of CCAT1 significantly inhibited cell proliferation and migration (P<0.05), arrested cell cycle at G0/G1 stage, and decreased cyclin D1 expression (P<0.05). An increased expression of c-Myc was observed in the PC tissues compared to the adjacent tissues. We found that suppression of c-Myc altered CCAT1 expression by targeting its promoter at E-box. This study demonstrated that c-Myc-activated CCAT1 may contribute to tumorigenesis and metastasis of PC, which may serve as a potential target for the therapy of PC.
Collapse
Affiliation(s)
- Qiuyun Yu
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Xinfeng Zhou
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Qing Xia
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Jia Shen
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Jia Yan
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Jiuting Zhu
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Xiang Li
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| | - Ming Shu
- Department of Hepatopancreatobiliary Surgery, Ningbo NO.2 Hospital Ningbo 315010, Zhejiang, China
| |
Collapse
|
52
|
Synergic Effect of α-Mangostin on the Cytotoxicity of Cisplatin in a Cervical Cancer Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7981397. [PMID: 28053694 PMCID: PMC5178369 DOI: 10.1155/2016/7981397] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/03/2016] [Accepted: 11/08/2016] [Indexed: 11/17/2022]
Abstract
Cervical cancer is the second leading cause of death among Mexican women. The treatment with cis-diamminedichloroplatinum (II) (CDDP) has some serious side effects. Alpha-mangostin (α-M), has a protective effect against CDDP-induced nephrotoxicity, as well as antioxidant, antitumor, and anti-inflammatory properties. Hence, we explored the in vitro and in vivo effect of α-M on human cervical cancer cell proliferation when combined with CDDP. In vitro, The cytotoxic effect of α-M and/or CDDP was measured by the 3-(3,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium assay. Meanwhile, apoptosis, reactive oxygen species (ROS) production, and the cell cycle were determined with flow cytometry. For α-M+CDDP treatment, both a coincubation and preincubation scheme were employed. In vivo, xenotransplantation was performed in female athymic BALB/c (nu/nu) mice, and then tumor volume and body weight were measured weekly, whereas α-M interfered with the antiproliferative activity of CDDP in the coincubation scheme, with preincubation with α-M+CDDP showing significantly greater cytotoxicity than CDDP or α-M alone, significantly inhibiting average tumor volume and preventing nephrotoxicity. This effect was accompanied by increased apoptosis and ROS production by HeLa cervical cancer cells, as well as an arrest in the cell cycle. These results suggest that α-M may be useful as a neoadjuvant agent in cervical cancer therapy.
Collapse
|
53
|
Huang M, Wang YP, Zhu LQ, Cai Q, Li HH, Yang HF. MAPK pathway mediates epithelial-mesenchymal transition induced by paraquat in alveolar epithelial cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:1407-1414. [PMID: 25873302 DOI: 10.1002/tox.22146] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is believed to be involved in lung fibrosis process induced by paraquat (PQ); however, the molecular mechanism of this process has not been clearly established. The present study investigated the potential involvement of EMT after PQ poisoning. The expressions of EMT markers, such as E-cadherin and α-smooth muscle actin (α-SMA), at multiple time points after exposure to different concentrations of PQ were evaluated by western blot analysis. Following PQ treatment, EMT induction was observed under microscopy. Related fibrosis genes, including Matrix metalloproteinase 2 (MMP-2), Matrix metalloproteinase 9 (MMP-9), collagens type I (COL I), and type III (COL III), were also evaluated by measuring their mRNA levels using RT-PCR analysis. Signaling pathways were analyzed using selective pharmacological inhibitors for MAPK. Cell migration ability was evaluated by scratch wound and Transwell assays. The data showed that PQ-induced epithelial RLE-6NT cells to develop mesenchymal cell characteristics, as indicated by a significant decrease in the epithelial marker E-cadherin and a significant increase in the extracellular matrix (ECM) marker α-smooth muscle actin in a dose and time-dependent manner. Moreover, PQ-treated RLE-6NT cells had an EMT-like phenotype with elevated expression of MMP-2, MMP-9, and COL I and COL III and enhanced migration ability. Signal pathway analysis revealed that PQ-induced EMT led to ERK-1 and Smad2 phosphorylation through activation of the MAPK pathway. The results of the current study indicate that PQ-induced pulmonary fibrosis occurs via EMT, which is mediated by the MAPK pathway. This implies that the MAPK pathway is a promising therapeutic target in alveolar epithelial cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1407-1414, 2016.
Collapse
Affiliation(s)
- Min Huang
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Ya-Peng Wang
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Ling-Qin Zhu
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Qian Cai
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Hong-Hui Li
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Hui-Fang Yang
- The Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, 750004, People's Republic of China.
| |
Collapse
|
54
|
Berberine suppressed epithelial mesenchymal transition through cross-talk regulation of PI3K/AKT and RARα/RARβ in melanoma cells. Biochem Biophys Res Commun 2016; 479:290-296. [DOI: 10.1016/j.bbrc.2016.09.061] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 11/24/2022]
|
55
|
Verma RK, Yu W, Shrivastava A, Shankar S, Srivastava RK. α-Mangostin-encapsulated PLGA nanoparticles inhibit pancreatic carcinogenesis by targeting cancer stem cells in human, and transgenic (Kras(G12D), and Kras(G12D)/tp53R270H) mice. Sci Rep 2016; 6:32743. [PMID: 27624879 PMCID: PMC5021984 DOI: 10.1038/srep32743] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022] Open
Abstract
Activation of sonic hedgehog (Shh) in cancer stem cell (CSC) has been demonstrated with aggressiveness of pancreatic cancer. In order to enhance the biological activity of α-mangostin, we formulated mangostin-encapsulated PLGA nanoparticles (Mang-NPs) and examined the molecular mechanisms by which they inhibit human and KC mice (PdxCre;LSL-KrasG12D) pancreatic CSC characteristics in vitro, and pancreatic carcinogenesis in KPC (PdxCre;LSLKrasG12D;LSL-Trp53R172H) mice. Mang-NPs inhibited human and KrasG12D mice pancreatic CSC characteristics in vitro. Mang-NPs also inhibited EMT by up-regulating E-cadherin and inhibiting N-cadherin and transcription factors Slug, and pluripotency maintaining factors Nanog, c-Myc, and Oct4. Furthermore, Mang-NPs inhibited the components of Shh pathway and Gli targets. In vivo, Mang-NPs inhibited the progression of pancreatic intraneoplasia to pancreatic ductal adenocarcinoma and liver metastasis in KPC mice. The inhibitory effects of Mang-NPs on carcinogenesis in KPC mice were associated with downregulation of pluripotency maintaining factors (c-Myc, Nanog and Oct4), stem cell markers (CD24 and CD133), components of Shh pathway (Gli1, Gli2, Patched1/2, and Smoothened), Gli targets (Bcl-2, XIAP and Cyclin D1), and EMT markers and transcription factors (N-cadherin, Slug, Snail and Zeb1), and upregulation of E-cadherin. Overall, our data suggest that Mang-NPs can inhibit pancreatic cancer growth, development and metastasis by targeting Shh pathway.
Collapse
Affiliation(s)
- Raj Kumar Verma
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Wei Yu
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Anju Shrivastava
- St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pathology and Laboratory Medicine, University of Missouri Kansas City, MO, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
56
|
Wei S, Wang L, Zhang L, Li B, Li Z, Zhang Q, Wang J, Chen L, Sun G, Li Q, Xu H, Zhang D, Xu Z. ZNF143 enhances metastasis of gastric cancer by promoting the process of EMT through PI3K/AKT signaling pathway. Tumour Biol 2016; 37:12813-12821. [PMID: 27449034 DOI: 10.1007/s13277-016-5239-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022] Open
Abstract
The zinc finger protein 143 (ZNF143) is a transcription factor, which regulates many cell cycle-associated genes. ZNF143 expressed strongly in multiple solid tumors. However, the influence of ZNF143 on gastric cancer (GC) remains largely unknown. In this study, we investigated the ZNF143 mRNA level in GC tissues and cells by quantitative real-time PCR (qRT-PCR). The protein expression of ZNF143 in GC cells, and the signaling pathway proteins were detected by Western blotting. Transwell assay and wound healing assay were performed to explore the effects of ZNF143 for the migration ability of GC cells in vitro. We also performed the tail vein injection in nude mice with GC cells to explore the impact of ZNF143 on GC metastasis in vivo. ZNF143 was overexpressed in specimens of GC compared with adjacent normal tissues and increased more significantly in GC tissues of patients who had lymph node metastasis. Ectopic overexpression of ZNF143 enhanced GC migration, whereas ZNF143 knockdown suppressed this effect in vitro. In vivo, ZNF143 knockdown reduced distant metastasis of GC cells in nude mice. In addition, overexpression of ZNF143 reduced the expression of epithelial cell marker (E-cadherin) and induced the expression of mesenchymal cell marker (N-cadherin,Vimentin), Snail and Slug. We also found that ZNF143 enhanced GC cell migration by promoting the process of EMT through PI3K/AKT signaling pathway. In general, our findings show that ZNF143 expressed strongly in GC and enhanced migration of GC cells in vitro and in vivo. It is conceivable that ZNF143 could be a therapeutic genetic target for GC treatment.
Collapse
Affiliation(s)
- Song Wei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjun Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qun Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiwei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liang Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guangli Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
57
|
α-Mangostin Induces Apoptosis and Cell Cycle Arrest in Oral Squamous Cell Carcinoma Cell. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5352412. [PMID: 27478478 PMCID: PMC4960343 DOI: 10.1155/2016/5352412] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/05/2016] [Accepted: 06/14/2016] [Indexed: 01/20/2023]
Abstract
Mangosteen has long been used as a traditional medicine and is known to have antibacterial, antioxidant, and anticancer effects. Although the effects of α-mangostin, a natural compound extracted from the pericarp of mangosteen, have been investigated in many studies, there is limited data on the effects of the compound in human oral squamous cell carcinoma (OSCC). In this study, α-mangostin was assessed as a potential anticancer agent against human OSCC cells. α-Mangostin inhibited cell proliferation and induced cell death in OSCC cells in a dose- and time-dependent manner with little to no effect on normal human PDLF cells. α-Mangostin treatment clearly showed apoptotic evidences such as nuclear fragmentation and accumulation of annexin V and PI-positive cells on OSCC cells. α-Mangostin treatment also caused the collapse of mitochondrial membrane potential and the translocation of cytochrome c from the mitochondria into the cytosol. The expressions of the mitochondria-related proteins were activated by α-mangostin. Treatment with α-mangostin also induced G1 phase arrest and downregulated cell cycle-related proteins (CDK/cyclin). Hence, α-mangostin specifically induces cell death and inhibits proliferation in OSCC cells via the intrinsic apoptosis pathway and cell cycle arrest at the G1 phase, suggesting that α-mangostin may be an effective agent for the treatment of OSCC.
Collapse
|
58
|
Xia Y, Li Y, Westover KD, Sun J, Chen H, Zhang J, Fisher DE. Inhibition of Cell Proliferation in an NRAS Mutant Melanoma Cell Line by Combining Sorafenib and α-Mangostin. PLoS One 2016; 11:e0155217. [PMID: 27152946 PMCID: PMC4859503 DOI: 10.1371/journal.pone.0155217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/26/2016] [Indexed: 12/19/2022] Open
Abstract
α-Mangostin is a natural product commonly used in Asia for cosmetic and medicinal applications including topical treatment of acne and skin cancer. Towards finding new pharmacological strategies that overcome NRAS mutant melanoma, we performed a cell proliferation-based combination screen using a collection of well-characterized small molecule kinase inhibitors and α-Mangostin. We found that α-Mangostin significantly enhances Sorafenib pharmacological efficacy against an NRAS mutant melanoma cell line. The synergistic effects of α-Mangostin and Sorafenib were associated with enhanced inhibition of activated AKT and ERK, induced ER stress, and reduced autophagy, eventually leading to apoptosis. The structure of α-Mangostin resembles several inhibitors of the Retinoid X receptor (RXR). MITF expression, which is regulated by RXR, was modulated by α-Mangostin. Molecular docking revealed that α-Mangostin can be accommodated by the ligand binding pocket of RXR and may thereby compete with RXR-mediated control of MITF expression. In summary, these data demonstrate an unanticipated synergy between α-Mangostin and sorafenib, with mechanistic actions that convert a known safe natural product to a candidate combinatorial therapeutic agent.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - Ying Li
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
- Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, No.7 Front Kangfu ST, Zhengzhou 450052, China
| | - Kenneth D. Westover
- Departments of Biochemistry and Radiation Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongxiang Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - Jianming Zhang
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
| | - David E. Fisher
- Cutaneous Biology Research Center, Massachusetts General Hospital, 149 Building 13th ST, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
59
|
Xia Y, Chen J, Gong C, Chen H, Sun J. α-Mangostin, a Natural Agent, Enhances the Response of NRAS Mutant Melanoma to Retinoic Acid. Med Sci Monit 2016; 22:1360-7. [PMID: 27104669 PMCID: PMC4844330 DOI: 10.12659/msm.898204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The identification and use of novel compounds alone or in combination hold promise for the fight against NRAS mutant melanoma. MATERIAL AND METHODS We screened a kinase-specific inhibitor library through combining it with α-Mangostin in NRAS mutant melanoma cell line, and verified the enhancing effect of α-Mangostin through inhibition of the tumorigenesis pathway. RESULTS Within the kinase inhibitors, retinoic acid showed a significant synergistic effect with α-Mangostin. α-Mangostin also can reverse the drug resistance of retinoic acid in RARa siRNA-transduced sk-mel-2 cells. Colony assay, TUNEL staining, and the expressions of several apoptosis-related genes revealed that a-Mangostin enhanced the effect of retinoic acid-induced apoptosis. The combination treatment resulted in marked induction of ROS generation and inhibition of the AKT/S6 pathway. CONCLUSIONS These results indicate that the combination of these novel natural agents with retinoid acid may be clinically effective in NRAS mutant melanoma.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jing Chen
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Chongwen Gong
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| |
Collapse
|
60
|
Yu P, Zhang Z, Li S, Wen X, Quan W, Tian Q, Chen J, Zhang J, Jiang R. Progesterone modulates endothelial progenitor cell (EPC) viability through the CXCL12/CXCR4/PI3K/Akt signalling pathway. Cell Prolif 2016; 49:48-57. [PMID: 26818151 DOI: 10.1111/cpr.12231] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/06/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Progesterone treatment can effectively increase levels of circulating endothelial progenitor cells (EPCs) and improve neurological functional outcome in a traumatic brain injury (TBI) rat model. However, the mechanisms of progesterone's effects on EPC viability remain elusive. The CXCL12/CXCR4 (CXC chemokine ligand 12/CXC chemokine receptor 4) signalling pathway regulates cell proliferation; we hypothesize that it mediates progesterone-induced EPC viability. MATERIALS AND METHODS EPCs were isolated from bone marrow-derived mononuclear cells (BM-MNCs) and treated with progesterone (5, 10 and 100 nm). MTS assay was used to investigate EPC viability. Protein expression was examined by Western blotting, ELISA assay and flow cytometry. Cell membrane and cytoplasm proteins were extracted with membrane and cytoplasm protein extraction kits. CXCR4 antagonist (AMD3100) and phosphatidylinositol 3-kinases (PI3K) antagonist (LY294002) were used to characterize underlying mechanisms. RESULTS Progesterone-induced EPC viability was time- and dose-dependent. Administration of progesterone facilitated EPC viability and increased expression of CXCL12 and phosphorylated Akt (also known as protein kinase B, pAkt) activity (P < 0.05). Progesterone did not regulate CXCR4 protein expression in cultured EPC membranes or cytoplasm. However, progesterone-induced EPC viability was significantly attenuated by AMD3100 or LY294002. Inhibition of the signalling pathway with AMD3100 and LY294002 subsequently reduced progesterone-induced CXCL12/CXCR4/PI3K/pAkt signalling activity. CONCLUSIONS The CXCL12/CXCR4/PI3K/pAkt signalling pathway increased progesterone-induced EPC viability.
Collapse
Affiliation(s)
- Peng Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Zhifei Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Shengjie Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Xiaolong Wen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Wei Quan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Qilong Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-Neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| |
Collapse
|
61
|
Wang Y, Gan Y, Tan Z, Zhou J, Kitazawa R, Jiang X, Tang Y, Yang J. TDRG1 functions in testicular seminoma are dependent on the PI3K/Akt/mTOR signaling pathway. Onco Targets Ther 2016; 9:409-20. [PMID: 26855590 PMCID: PMC4725695 DOI: 10.2147/ott.s97294] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human testis development-related gene 1 (TDRG1) is a recently identified gene that is expressed exclusively in the testes and promotes the development of testicular germ cell tumors. In this study, the role of TDRG1 in the development of testicular seminoma, which is the most common testicular germ cell tumor, was further investigated. Based on polymerase chain reaction, Western blotting, and immunohistochemistry tests, both gene and protein expression levels of TDRG1 were significantly upregulated in testicular seminoma tissues compared with normal testicular tissues. Additionally, the levels of phosphoinositide-3 kinase (PI3K)/p110 and Akt phosphorylation were dramatically upregulated in testicular seminoma tissues. Accordingly, in our cell experiment, seminoma TCam-2 cells were subjected to different treatments: the TDRG1 knockout, TDRG1 overexpression, PI3K inhibition (LY294002 administration), or PI3K activation (insulin-like growth factor-1 administration). Cell proliferation, the proliferation index, apoptosis rate, cell adhesive capacity, and cell invasion capability were assessed. Cells with both TDRG1 knockout and PI3K inhibition exhibited decreased cell proliferation, proliferation indexes, cell adhesion capacity, and cell invasion capability and increased apoptosis rates. Most of these effects were reversed by TDRG1 overexpression or PI3K activation, indicating that both TDRG1- and PI3K-mediated signaling promote proliferation and invasion of testicular seminoma cells. The knockout of TDRG1 significantly decreased the phosphorylation levels of PI3K/p85, PI3K/p110, Akt, and mammalian target of rapamycin (mTOR; Ser2448). Except for PI3K/p110, TDRG1 overexpression had the opposite effects on phosphorylation levels. Phosphorylated mTOR at Ser2481 and Thr2446 was not affected by TDRG1 or PI3K in our tests. Thus, these results indicate that TDRG1 promotes the development and migration of seminoma cells via the regulation of the PI3K/Akt/mTOR signaling pathway; this contributes to an understanding of the precise mechanisms underlying the development and migration of seminomas and lays a theoretical foundation for the development of appropriate therapies.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Yu Gan
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zhengyu Tan
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Jun Zhou
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Riko Kitazawa
- Department of Diagnostic Pathology, Ehime University Hospital, Shitsukawa, Tōon, Ehime Perfecture, Japan
| | - Xianzhen Jiang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Yuxin Tang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Jianfu Yang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
62
|
Zhang L, Wang X, Lai M. Modulation of epithelial-to-mesenchymal cancerous transition by natural products. Fitoterapia 2015; 106:247-55. [PMID: 26386389 DOI: 10.1016/j.fitote.2015.09.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/11/2022]
Abstract
Metastasis is mainly responsible for poor prognosis of cancer, and epithelial-to-mesenchymal transition (EMT) is a significant process often activated during cancer invasion and metastasis. Therefore EMT could be an effective target of chemotherapy to inhibit cancer metastasis and improve prognosis. Considering that many chemotherapeutics are plant-based, we reviewed recent reports about natural products extracted from plants and cancer EMT prevention.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Xue Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Maode Lai
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China; Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
63
|
Nugitrangson P, Puthong S, Iempridee T, Pimtong W, Pornpakakul S, Chanchao C. In vitro and in vivo characterization of the anticancer activity of Thai stingless bee (Tetragonula laeviceps) cerumen. Exp Biol Med (Maywood) 2015; 241:166-76. [PMID: 26290139 DOI: 10.1177/1535370215600102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/16/2015] [Indexed: 11/16/2022] Open
Abstract
Tetragonula laeviceps cerumen was sequentially extracted with 80% (v/v) methanol, dichloromethane, and hexane and also in the reverse order. By the MTT assay and the respective 50% inhibition concentration value, the most active fraction was further purified to apparent homogeneity by bioassay-guided silica gel column chromatography. α-Mangostin was identified by high-resolution electrospray ionization mass spectrometry and nuclear magnetic resonance analyses. It had a potent cytotoxicity against the BT474, Chago, Hep-G2, KATO-III, and SW620 cell lines (IC50 values of 1.22 ± 0.03, 2.25 ± 0.20, 0.94 ± 0.01, 0.88 ± 0.16, and 1.50 ± 0.39 µmol/L, respectively). The in vitro cytotoxicity of α-mangostin against the five human cancer cell lines and primary fibroblasts was further characterized by real-time impedance-based analysis. Interestingly, α-mangostin was more cytotoxic against the cancer-derived cell lines than against the primary fibroblasts. Later, the migration assay was performed by continuously measuring the attachment of cells to the plate electrodes at the bottom of the transwell membrane. The combined caspase-3 and -7 activities were assayed by the Caspase-Glo® 3/7 kit. It showed that the cytotoxic mechanism involved caspase-independent apoptosis, while at low (non-toxic) concentrations α-mangostin did not significantly alter cell migration. Furthermore, the in vivo cytotoxicity and angiogenesis were determined by alkaline phosphatase staining in zebrafish embryos along with monitoring changes in the transcript expression level of two genes involved in angiogenesis (vegfaa and vegfr2) by quantitative real-time reverse transcriptase- polymerase chain reaction. It was found that the in vivo cytotoxicity of α-mangostin against zebrafish embryos had a 50% lethal concentration of 9.4 µM, but no anti-angiogenic properties were observed in zebrafish embryos at 9 and 12 µM even though it downregulated the expression of vegfaa and vegfr2 transcripts. Thus, α-mangostin is a major active compound with a potential anticancer activity in T. laeviceps cerumen in Thailand.
Collapse
Affiliation(s)
- Pongvit Nugitrangson
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Songchan Puthong
- Institute of Biotechnology and Genetic Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tawin Iempridee
- National Nanotechnology Center, National Science and Technology Development Agency, Thanon Phahonyothin, Tambon Khlong Nueng, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Wittaya Pimtong
- National Nanotechnology Center, National Science and Technology Development Agency, Thanon Phahonyothin, Tambon Khlong Nueng, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Surachai Pornpakakul
- Research Centre for Bioorganic Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chanpen Chanchao
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
64
|
Xu W, Yang Z, Lu N. A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adh Migr 2015; 9:317-24. [PMID: 26241004 DOI: 10.1080/19336918.2015.1016686] [Citation(s) in RCA: 438] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tumor metastasis is not only a sign of disease severity but also a major factor causing treatment failure and cancer-related death. Therefore, studies on the molecular mechanisms of tumor metastasis are critical for the development of treatments and for the improvement of survival. The epithelial-mesenchymal transition (EMT) is an orderly, polygenic biological process that plays an important role in tumor cell invasion, metastasis and chemoresistance. The complex, multi-step process of EMT involves multiple regulatory mechanisms. Specifically, the PI3K/Akt signaling pathway can affect the EMT in a variety of ways to influence tumor aggressiveness. A better understanding of the regulatory mechanisms related to the EMT can provide a theoretical basis for the early prediction of tumor progression as well as targeted therapy.
Collapse
Key Words
- CK, cytokeratin
- ECM, extracellular matrix
- EMT
- EMT, epithelial-mesenchymal transition
- FGF, fibroblast growth factor
- GSK-3β, glycogen synthase kinase 3 β
- ILK, integrin-linked kinase
- MDR, multidrug resistance
- MET, mesenchymal-epithelial transition
- PDGF, platelet-derived growth factor
- PDK1, 3-phosphoinositide-dependent protein kinase 1
- PI3K, phosphatidylinositol-3-kinase
- PI3K/Akt signaling pathway
- PKA, protein kinase A
- PKB, protein kinase B
- PKC, protein kinase C
- TGF-β, transforming growth factor-β
- TNF-α, tumor necrosis factor-α
- YB-1, Y-box binding protein-1
- anti-cancer therapy
- bHLH, basic helix-loop-helix protein
- extracellular matrix
- transcription factors
- tumor aggressiveness
Collapse
Affiliation(s)
- Wenting Xu
- a Department of Gastroenterology ; The First Affiliated Hospital of Nanchang University ; Nanchang , Jiangxi , China
| | | | | |
Collapse
|
65
|
Yamamoto T, Uemura K, Moriyama K, Mitamura K, Taga A. Inhibitory effect of maple syrup on the cell growth and invasion of human colorectal cancer cells. Oncol Rep 2015; 33:1579-84. [PMID: 25647359 PMCID: PMC4358083 DOI: 10.3892/or.2015.3777] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/20/2014] [Indexed: 12/27/2022] Open
Abstract
Maple syrup is a natural sweetener consumed by individuals of all ages throughout the world. Maple syrup contains not only carbohydrates such as sucrose but also various components such as organic acids, amino acids, vitamins and phenolic compounds. Recent studies have shown that these phenolic compounds in maple syrup may possess various activities such as decreasing the blood glucose level and an anticancer effect. In this study, we examined the effect of three types of maple syrup, classified by color, on the cell proliferation, migration and invasion of colorectal cancer (CRC) cells in order to investigate whether the maple syrup is suitable as a phytomedicine for cancer treatment. CRC cells that were administered maple syrup showed significantly lower growth rates than cells that were administered sucrose. In addition, administration of maple syrup to CRC cells caused inhibition of cell invasion, while there was no effect on cell migration. Administration of maple syrup clearly inhibited AKT phosphorylation, while there was no effect on ERK phosphorylation. These data suggest that maple syrup might inhibit cell proliferation and invasion through suppression of AKT activation and be suitable as a phytomedicine for CRC treatment, with fewer adverse effects than traditional chemotherapy.
Collapse
Affiliation(s)
- Tetsushi Yamamoto
- Pathological and Biomolecule Analyses Laboratory, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kentaro Uemura
- Pathological and Biomolecule Analyses Laboratory, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kaho Moriyama
- Pathological and Biomolecule Analyses Laboratory, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kuniko Mitamura
- Pathological and Biomolecule Analyses Laboratory, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Osaka 577-8502, Japan
| | - Atsushi Taga
- Pathological and Biomolecule Analyses Laboratory, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|