14 results on '"Epithelial-mesenchymal-transition"'
Search Results
2. SOS1 regulates HCC cell epithelial-mesenchymal transition via the PI3K/AKT/mTOR pathway.
- Author
-
Li, Yonghe, Yin, Yaolin, He, Yi, He, Kun, and Li, Jingdong
- Subjects
- *
EPITHELIAL-mesenchymal transition , *LIVER cancer , *HEPATOCELLULAR carcinoma , *CANCER patients , *WESTERN immunoblotting , *NUDITY , *CELL migration - Abstract
The influence of son of sevenless homolog 1 (SOS1) on invasion and metastasis of hepatocellular carcinoma (HCC) cells was investigated. HCC cells were transfected with siRNA and lentivirus to achieve SOS1 knock down/overexpression and changes in RNA and protein levels analyzed by q-PCR and Western blotting (WB). Transwell assay was utilized to assess variations in cell invasion and migration in vitro and by a lung metastasis model of liver cancer in vivo. High expression of SOS1 was observed in most human liver cancers, which indicated a worse prognosis. SOS1 knockout in HepG2 cells significantly decreased cell invasion and migration. SOS1 knockout also reduced the number of metastatic foci in a lung metastasis model of HCC established in nude mice. SOS1 knockout inhibited the epithelial-mesenchymal transition (EMT) in HepG2 cells as well as the PI3K/AKT/mTOR pathway. Overexpression of SOS1 in Huh7 cells had the opposite effect. To conclude, SOS1 may induce the EMT by the activation of the PI3K/AKT/mTOR pathway, thereby enhancing invasion, migration and metastasis of HCC cells. These findings may expose SOS1 as a new HCC therapeutic target. • SOS1 gene is generally overexpressed in patients with liver cancer, which is associated with poor prognosis. • Invasion and migration of hepatocellular carcinoma cells are regulated by SOS1 gene. • SOS1 regulates epithelial mesenchymal transformation of hepatocellular carcinoma cells through PI3K/AKT/mTOR signal pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
3. RETRACTED: Corrigendum: Hepatoma Cell-Derived Extracellular Vesicles Promote Liver Cancer Metastasis by Inducing the Differentiation of Bone Marrow Stem Cells Through microRNA-181d-5p and the FAK/Src Pathway.
- Subjects
BONE marrow cells ,LIVER cancer ,EXTRACELLULAR vesicles ,METASTASIS ,HEPATOCELLULAR carcinoma - Abstract
This document is a corrigendum, or correction, for an article titled "Hepatoma Cell-Derived Extracellular Vesicles Promote Liver Cancer Metastasis by Inducing the Differentiation of Bone Marrow Stem Cells Through microRNA-181d-5p and the FAK/Src Pathway." The correction addresses errors in the corresponding author's name and email address, as well as errors in the institutional affiliations listed for some of the authors. The authors apologize for these errors and state that they do not affect the scientific conclusions of the article. The original article has been updated. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
4. CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis
- Author
-
Xiao-Lu Ma, Min-Na Shen, Bo Hu, Bei-Li Wang, Wen-Jing Yang, Li-Hua Lv, Hao Wang, Yan Zhou, An-Li Jin, Yun-Fan Sun, Chuan-Yan Zhang, Shuang-Jian Qiu, Bai-Shen Pan, Jian Zhou, Jia Fan, Xin-Rong Yang, and Wei Guo
- Subjects
Hepatocellular carcinoma ,CD73 ,Epithelial-mesenchymal-transition ,Prognosis ,PI3K/AKT ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide because of rapid progression and high incidence of metastasis or recurrence. Accumulating evidence shows that CD73-expressing tumor cell is implicated in development of several types of cancer. However, the role of CD73 in HCC cell has not been systematically investigated and its underlying mechanism remains elusive. Methods CD73 expression in HCC cell was determined by RT-PCR, Western blot, and immunohistochemistry staining. Clinical significance of CD73 was evaluated by Cox regression analysis. Cell counting kit-8 and colony formation assays were used for proliferation evaluation. Transwell assays were used for motility evaluations. Co-immunoprecipitation, cytosolic and plasma membrane fractionation separation, and ELISA were applied for evaluating membrane localization of P110β and its catalytic activity. NOD/SCID/γc(null) (NOG) mice model was used to investigate the in vivo functions of CD73. Results In the present study, we demonstrate that CD73 was crucial for epithelial-mesenchymal transition (EMT), progression and metastasis in HCC. CD73 expression is increased in HCC cells and correlated with aggressive clinicopathological characteristics. Clinically, CD73 is identified as an independent poor prognostic indicator for both time to recurrence and overall survival. CD73 knockdown dramatically inhibits HCC cells proliferation, migration, invasion, and EMT in vitro and hinders tumor growth and metastasis in vivo. Opposite results could be observed when CD73 is overexpressed. Mechanistically, adenosine produced by CD73 binds to adenosine A2A receptor (A2AR) and activates Rap1, which recruits P110β to the plasma membrane and triggers PIP3 production, thereby promoting AKT phosphorylation in HCC cells. Notably, a combination of anti-CD73 and anti-A2AR achieves synergistic depression effects on HCC growth and metastasis than single agent alone. Conclusions CD73 promotes progression and metastasis through activating PI3K/AKT signaling, indicating a novel prognostic biomarker for HCC. Our data demonstrate the importance of CD73 in HCC in addition to its immunosuppressive functions and revealed that co-targeting CD73 and A2AR strategy may be a promising novel therapeutic strategy for future HCC management.
- Published
- 2019
- Full Text
- View/download PDF
5. Tumor cells derived-extracellular vesicles transfer miR-3129 to promote hepatocellular carcinoma metastasis by targeting TXNIP.
- Author
-
Yang, Yang, Mao, Feifei, Guo, Lei, Shi, Jie, Wu, Mengchao, Cheng, Shuqun, and Guo, Weixing
- Abstract
Hepatocellular carcinoma (HCC) is the most predominant primary liver cancer. Extracellular vesicles (EV)-mediated microRNA (miRNA) delivery is critical in cancer metastasis. We aimed to identify the mechanism of HCC cell-derived EVs-mediated miR-3129 in HCC. After EVs isolation and identification, miR-3129 expression in plasma EVs was evaluated and its diagnostic efficiency was analyzed. miR-3129 inhibitor was transfected into HepG2 and SMMC7721 cells, and cell malignant episodes were assessed. HCC cells were incubated with EVs from MHCC-97H cells and transfected with miR-3129 inhibitor and/or TXNIP. The nude mice were injected with MHCC-97H cells-EV or MHCC-97H cells-EV/miR-3129 inhibitor, and HCC growth and metastasis were assessed. miR-3129 was highly expressed in plasma EVs from HCC patients, which was the essential diagnostic biomarker for HCC. miR-3129 downregulation inhibited the malignant episodes of HCC cells. MHCC-97H cell-EVs were absorbed by HCC cells and transferred miR-3129 to HCC cells. EVs-carried miR-3129 promoted malignant episodes of HCC cells, which were weakened by miR-3129 inhibition in EVs. miR-3129 targeted TXNIP. TXNIP overexpression averted the effect of EVs-carried miR-3129 in HCC. In vivo, MHCC-97H cell-EVs transferred miR-3129 to facilitate HCC growth and metastasis. MHCC-97H cell-EVs transferred miR-3129 to promote HCC metastasis by targeting TXNIP. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
6. NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells
- Author
-
Bo Hu, Guang-Yu Ding, Pei-Yao Fu, Xiao-Dong Zhu, Yuan Ji, Guo-Ming Shi, Ying-Hao Shen, Jia-Bin Cai, Zhen Yang, Jian Zhou, Jia Fan, Hui-Chuan Sun, Ming Kuang, and Cheng Huang
- Subjects
Hepatocellular carcinoma ,NLRX1 ,Epithelial-mesenchymal-transition ,Tumor suppressor ,Senescence ,Transition ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background This study was performed to investigate the role of nucleotide-binding oligomerization domain (NOD)-like receptor X1 (NLRX1) in regulating hepatocellular carcinoma (HCC) progression. Methods Expression levels of NLRX1 in clinical specimens and cell lines were determined by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Transwell assays were conducted to evaluate the effect of NLRX1 on cell invasion, and flow cytometry was used to assess apoptosis. Expression patterns of key molecules in the phosphoinositide 3-kinase (PI3K)-AKT pathways were determined via WB. The effect of NLRX1 on cell senescence was evaluated with β-galactosidase assays. Kaplan-Meier analyses and Cox regression models were used for prognostic evaluation. Results NLRX1 was downregulated in tumor tissue compared with adjacent normal liver tissue. Low tumor NLRX1 expression was identified as an independent indicator for HCC prognosis (recurrence: hazard ratio [HR] 1.87, 95% confidence interval [CI] 1.26–2.76, overall survival [OS] 2.26, 95% CI 1.44–3.56). NLRX1 over-expression (OE) significantly inhibited invasiveness ability and induced apoptosis in HCC cells. In vivo experiments showed that NLRX1 knock-down (KD) significantly promoted HCC growth. Mechanistically, NLRX1 exhibited a suppressor function by decreasing phosphorylation of AKT and thus downregulating Snail1 expression, which inhibited epithelial-mesenchymal-transition (EMT) in HCC cells. Moreover, NLRX1 OE could induce cell senescence via an AKT-P21-dependent manner. Conclusions NLRX1 acted as a tumor suppressor in HCC by inducing apoptosis, promoting senescence, and decreasing invasiveness by repressing PI3K-AKT signaling pathway. Future investigations will focus on restoring expression of NLRX1 to provide new insights into HCC treatment.
- Published
- 2018
- Full Text
- View/download PDF
7. Down-regulation of Transducin-Like Enhancer of Split protein 4 in hepatocellular carcinoma promotes cell proliferation and epithelial-Mesenchymal-Transition.
- Author
-
Wu, Xiao-cai, Xiao, Cui-cui, Li, Hua, Tai, Yan, Zhang, Qi, and Yang, Yang
- Subjects
- *
LIVER cancer , *DOWNREGULATION , *TRANSDUCIN , *GENE enhancers , *CANCER cell proliferation , *NEOPLASTIC cell transformation , *EPITHELIAL cells , *MESENCHYMAL stem cells , *GENETICS - Abstract
Background Transducin-Like Enhancer of Split protein 4 (TLE4) has been reported to be involved in some subsets of acute myeloid leukemia and colorectal cancer. In the present study, we aimed to explore the role of TLE4 in tumorigenesis and cancer progression in hepatocellular carcinoma (HCC). Methods The expression pattern of TLE4 in HCC was determined by Western-blot and qRT-PCR, gain-of-function and loss-of-function was used to explore the biological role of TLE4 in HCC cells. A xenograft model was established to confirm its effects on proliferation. Results The protein expression levels of TLE4 were significantly down-regulated in HCC tissues compared to matched adjacent normal liver tissues. In vitro , down-regulation of TLE4 in Huh7 or SMMC-7721 promoted cell proliferation and ectopical expression of TLE4 in Hep3B or Bel-7404 suppressed cell proliferation. In addition, the cell colony formation ability was enhanced after down-regulation of TLE4 expression in Huh-7 but suppressed after over-expression in Hep3B. Furthermore, down-regulation of TLE4 increased the cell invasion ability, as well as increased the expression level of Vimentin and decreased that of E-cadherin, indicating a phenotype of epithelial-mesenchymal transition (EMT) in HCC cells. On the contrary, ectopical expression of TLE4 in HCC cells decreased the cell invasion ability and inhibited EMT. In vivo , compared to control group, xenograft tumor volumes were significantly decreased in TLE4 overexpression group. Conclusions These results demonstrated that TLE4 might play important regulatory roles in cellular proliferation and EMT process in HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
8. Long noncoding RNA lncTCF7, induced by IL-6/STAT3 transactivation, promotes hepatocellular carcinoma aggressiveness through epithelial-mesenchymal transition.
- Author
-
Jun Wu, Jun Zhang, Bin Shen, Kai Yin, Jianwei Xu, Wencan Gao, and Lihong Zhang
- Subjects
- *
NON-coding RNA , *LIVER cancer , *INTERLEUKIN-6 , *POLYMERASE chain reaction , *EPITHELIAL cells - Abstract
Background: Accumulating evidence suggests the pro-inflammatory cytokine interleukin-6 (IL-6) in tumor microenvironment may promote the development of hepatocellular carcinoma (HC). However, the underlying mechanism remains largely unknown. Methods: The expression and promoter activity of lncTCF7 were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay. The function of the STAT3 binding site in the lncTCF7 promoter region was tested by luciferase reporter assay with nucleotide substitutions. The binding of STAT3 to the lncTCF7 promoter was confirmed by chromatin immunoprecipitation assay (CHIP) in vivo. The effects of decreasing STAT3 with small interference RNA and inhibiting STAT3 activation by small molecular inhibitor on lncTCF7 expression were also determined. Results: We demonstrate that IL-6 could induce lncTCF7 expression in a time- and dose-dependent manner, and we showed that IL-6 transcriptionally activated the expression of lncTCF7 in HC cells by activating STAT3, a transcription activator which binds to promoter regions of lncTCF7. Furthermore, knocking-down STAT3 and inhibiting STAT3 activation reduced lncTCF7 expression. Importantly, RNA interference-based attenuation of lncTCF7 prevented IL-6-induced EMT and cell invasion. Conclusion: Thus, these data provides evidence to the existence of an aberrant IL-6/STAT3/ lncTCF7 signaling axis that leads to HC aggressiveness through EMT induction, which could be novel therapeutic targets in malignancies. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
9. NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells
- Author
-
Guang-Yu Ding, Jia Fan, Jia-Bin Cai, Ying-Hao Shen, Xiao-Dong Zhu, Yuan Ji, Ming Kuang, Bo Hu, Zhen Yang, Pei-Yao Fu, Jian Zhou, Hui-Chuan Sun, Cheng Huang, and Guo-Ming Shi
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Hepatocellular carcinoma ,Cell ,Mice, Nude ,Senescence ,lcsh:RC254-282 ,Flow cytometry ,Mitochondrial Proteins ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,NLRX1 ,Epithelial–mesenchymal transition ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cellular Senescence ,medicine.diagnostic_test ,Chemistry ,lcsh:RC633-647.5 ,Research ,Liver Neoplasms ,Tumor suppressor ,Hematology ,lcsh:Diseases of the blood and blood-forming organs ,Middle Aged ,Prognosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,Cell culture ,Epithelial-mesenchymal-transition ,Transition ,Cancer research ,Female ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Background This study was performed to investigate the role of nucleotide-binding oligomerization domain (NOD)-like receptor X1 (NLRX1) in regulating hepatocellular carcinoma (HCC) progression. Methods Expression levels of NLRX1 in clinical specimens and cell lines were determined by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Transwell assays were conducted to evaluate the effect of NLRX1 on cell invasion, and flow cytometry was used to assess apoptosis. Expression patterns of key molecules in the phosphoinositide 3-kinase (PI3K)-AKT pathways were determined via WB. The effect of NLRX1 on cell senescence was evaluated with β-galactosidase assays. Kaplan-Meier analyses and Cox regression models were used for prognostic evaluation. Results NLRX1 was downregulated in tumor tissue compared with adjacent normal liver tissue. Low tumor NLRX1 expression was identified as an independent indicator for HCC prognosis (recurrence: hazard ratio [HR] 1.87, 95% confidence interval [CI] 1.26–2.76, overall survival [OS] 2.26, 95% CI 1.44–3.56). NLRX1 over-expression (OE) significantly inhibited invasiveness ability and induced apoptosis in HCC cells. In vivo experiments showed that NLRX1 knock-down (KD) significantly promoted HCC growth. Mechanistically, NLRX1 exhibited a suppressor function by decreasing phosphorylation of AKT and thus downregulating Snail1 expression, which inhibited epithelial-mesenchymal-transition (EMT) in HCC cells. Moreover, NLRX1 OE could induce cell senescence via an AKT-P21-dependent manner. Conclusions NLRX1 acted as a tumor suppressor in HCC by inducing apoptosis, promoting senescence, and decreasing invasiveness by repressing PI3K-AKT signaling pathway. Future investigations will focus on restoring expression of NLRX1 to provide new insights into HCC treatment. Electronic supplementary material The online version of this article (10.1186/s13045-018-0573-9) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
10. CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis
- Author
-
Ma, Xiao-Lu, Shen, Min-Na, Hu, Bo, Wang, Bei-Li, Yang, Wen-Jing, Lv, Li-Hua, Wang, Hao, Zhou, Yan, Jin, An-Li, Sun, Yun-Fan, Zhang, Chuan-Yan, Qiu, Shuang-Jian, Pan, Bai-Shen, Zhou, Jian, Fan, Jia, Yang, Xin-Rong, and Guo, Wei
- Published
- 2019
- Full Text
- View/download PDF
11. NEDD9 may regulate hepatocellular carcinoma cell metastasis by promoting epithelial-mesenchymal-transition and stemness via repressing Smad7
- Author
-
Shaojun Zhu, Shuqiang Yue, Zhipeng Wang, Peng Lu, Xiao Li, and Min Shen
- Subjects
Male ,0301 basic medicine ,Pathology ,MMP2 ,NEDD9 ,epithelial-mesenchymal-transition ,Metastasis ,Mice ,0302 clinical medicine ,Cell Movement ,Tumor Cells, Cultured ,Medicine ,AC133 Antigen ,Liver Neoplasms ,hepatocellular carcinoma ,Proto-Oncogene Proteins c-crk ,Extracellular Matrix ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Neoplastic Stem Cells ,Matrix Metalloproteinase 2 ,Research Paper ,Signal Transduction ,cancer stem cell ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Proto-Oncogene Proteins pp60(c-src) ,Mice, Nude ,Smad7 Protein ,03 medical and health sciences ,Downregulation and upregulation ,Cancer stem cell ,Spheroids, Cellular ,Precursor cell ,Cell Adhesion ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Adaptor Proteins, Signal Transducing ,business.industry ,developmentally downregulated 9 ,Aldehyde Dehydrogenase ,Phosphoproteins ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Focal Adhesion Kinase 1 ,Cancer research ,business - Abstract
// Zhipeng Wang 1, * , Min Shen 2, * , Peng Lu 3, * , Xiao Li 4 , Shaojun Zhu 5 , Shuqiang Yue 4 1 School of Pharmacy, Fourth Military Medical University, Xi’an, China 2 Department of Cardiovascular diseases, Xijing Hospital, Xi’an, China 3 Department of Hepatobiliary Surgery, Hainan Branch of Chinese PLA general Hospital, Sanya, China 4 Department of Hepatobiliary Surgery, Xijing Hospital, Xi’an, China 5 Department of Pathology, Tangdu Hospital, Xian, China * These authors have contributed equally to this work Correspondence to: Shuqiang Yue, email: shuqiangyue@126.com Keywords: hepatocellular carcinoma, cancer stem cell, epithelial-mesenchymal-transition, developmentally downregulated 9 Received: March 21, 2016 Accepted: November 08, 2016 Published: December 10, 2016 ABSTRACT Overexpression of neural precursor cell expressed, developmentally downregulated 9 (NEDD9) is a prognostic marker of many cancers, including hepatocellular carcinoma (HCC). However, the functions and mechanisms of NEDD9 are unclear. We found that upregulation of NEDD9 promoted migration, invasion and cell-to-extracellular matrix adhesion of HCC cells. NEDD9 also induced the epithelial-mesenchymal transition (EMT) and expression of matrix metalloprotein 2 (MMP2). Increased aldehyde dehydrogenase (ALDH) activity and CD133-positive cells were observed in HCC cells with high expression of NEDD9, corresponding to greater sphere formation in cancer stem cells (CSCs). NEDD9 deregulated Smad7 expression to inhibit Smad signaling and binding to the FAK-Src-Crk complex. We propose that this is the mechanism by which NEDD9 induced CSC properties.
- Published
- 2016
- Full Text
- View/download PDF
12. NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells
- Author
-
Hu, Bo, Ding, Guang-Yu, Fu, Pei-Yao, Zhu, Xiao-Dong, Ji, Yuan, Shi, Guo-Ming, Shen, Ying-Hao, Cai, Jia-Bin, Yang, Zhen, Zhou, Jian, Fan, Jia, Sun, Hui-Chuan, Kuang, Ming, and Huang, Cheng
- Published
- 2018
- Full Text
- View/download PDF
13. Knockdown of CD44 inhibits the invasion and metastasis of hepatocellular carcinoma bothin vitroandin vivoby reversing epithelial-mesenchymal transition
- Author
-
Xia Li, Bai Ruan, Fuqing Zhang, Weihui Liu, Yuan Gao, Zhuochao Zhang, Rui Ding, Kefeng Dou, Jianlin Wang, Xisheng Yang, Juanli Duan, and Kaishan Tao
- Subjects
MAPK/ERK pathway ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Hepatocellular carcinoma ,Mice, Nude ,epithelial-mesenchymal-transition ,Metastasis ,Small hairpin RNA ,Mice ,Cancer stem cell ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Neoplasm Metastasis ,CD44 ,Mice, Inbred BALB C ,Gene knockdown ,biology ,business.industry ,Liver Neoplasms ,Hep G2 Cells ,medicine.disease ,Hyaluronan Receptors ,Snail ,Oncology ,Cell culture ,Gene Knockdown Techniques ,Cancer research ,biology.protein ,Heterografts ,business ,Research Paper - Abstract
Mounting evidence has shown that induction of epithelial-mesenchymal transition (EMT) contributes to the the expression of CSC (cancer stem cell) markers. However, whether and how CSC markers could be involved in regulating EMT has rarely been reported. CD44, being one of the most commonly used CSC markers in hepatocellular carcinoma (HCC), has been demonstrated to act as a multidomain, transmembrane platform that serves to integrate a wide variety of extracellular signals. Therefore, we determined to seek whether CD44 is necessary for the EMT process in HCC. First, we noticed that CD44 expression was associated with the mesenchymal phenotype in HCC cell lines, and knocking down CD44 with lentivirus-mediated shRNA in HCC cell lines resulted in the mesenchymal-epithelial-transition (MET) and the subsequent impaired migration and invasion in vitro. Moreover, in a metastatic mice model established by tail vein injection of luciferase labelled MHCC97-H cells, we confirmed that CD44 knockdown resulted in the decreased metastasis of HCC cells. Furthermore, we found that the induction of MET by CD44 inhibition might be achieved, at least in part, by repressing the ERK/Snail pathway.
- Published
- 2015
- Full Text
- View/download PDF
14. Long noncoding RNA lncTCF7, induced by IL-6/STAT3 transactivation, promotes hepatocellular carcinoma aggressiveness through epithelial-mesenchymal transition
- Author
-
Lihong Zhang, Jianwei Xu, Jun-Xiang Wu, Bin Shen, Kai-sheng Yin, Jun Zhang, and Wencan Gao
- Subjects
STAT3 Transcription Factor ,Transcriptional Activation ,Cancer Research ,Chromatin Immunoprecipitation ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Hepatocellular carcinoma ,Biology ,Polymerase Chain Reaction ,Transactivation ,RNA interference ,Cell Movement ,Cell Line, Tumor ,T Cell Transcription Factor 1 ,Tumor Microenvironment ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Binding site ,RNA, Small Interfering ,Promoter Regions, Genetic ,Regulation of gene expression ,Binding Sites ,Interleukin-6 ,Research ,Liver Neoplasms ,RNA ,Promoter ,Gene Expression Regulation, Neoplastic ,Oncology ,Epithelial-mesenchymal-transition ,Cancer research ,RNA Interference ,RNA, Long Noncoding ,Chromatin immunoprecipitation ,Long noncoding RNA - Abstract
Background Accumulating evidence suggests the pro-inflammatory cytokine interleukin-6 (IL-6) in tumor microenvironment may promote the development of hepatocellular carcinoma (HCC). However, the underlying mechanism remains largely unknown. Methods The expression and promoter activity of lncTCF7 were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay. The function of the STAT3 binding site in the lncTCF7 promoter region was tested by luciferase reporter assay with nucleotide substitutions. The binding of STAT3 to the lncTCF7 promoter was confirmed by chromatin immunoprecipitation assay (CHIP) in vivo. The effects of decreasing STAT3 with small interference RNA and inhibiting STAT3 activation by small molecular inhibitor on lncTCF7 expression were also determined. Results We demonstrate that IL-6 could induce lncTCF7 expression in a time- and dose-dependent manner, and we showed that IL-6 transcriptionally activated the expression of lncTCF7 in HCC cells by activating STAT3, a transcription activator which binds to promoter regions of lncTCF7. Furthermore, knocking-down STAT3 and inhibiting STAT3 activation reduced lncTCF7 expression. Importantly, RNA interference-based attenuation of lncTCF7 prevented IL-6-induced EMT and cell invasion. Conclusion Thus, these data provides evidence to the existence of an aberrant IL-6/STAT3/ lncTCF7 signaling axis that leads to HCC aggressiveness through EMT induction, which could be novel therapeutic targets in malignancies.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.