scholarly journals A Novel Long Non-coding RNA, ENSG00000236199, Inhibits Proliferation and Metastasis through NRIP1 by Sponging miR-576-5p in Hepatocellular Carcinoma

Author(s):  
Yifeng Cui ◽  
Han Lin ◽  
Yunzheng Zhao ◽  
Jiaming Liu ◽  
Chengpeng Zhang ◽  
...  

Abstract Background: Hepatocellular carcinoma (HCC) is a common malignancy of the digestive system. A novel long non-coding RNA, ENSG00000236199 (lncRNA-199) , whose role in tumors has not been reported, especially in HCC. Methods: The expression of lncRNA-199 and miR-576-5p were detected by Real-time quantitative polymerase chain reaction (RT-qPCR), NRIP1 was measured by Western blotting. HCC cell proliferation and metastasis of HCC were examined using functional tests. Luciferase reporter assay was performed to confirm the relationship between lncRNA-199 and miR-576-5p, between miR-576-5p and NRIP1. Results: LncRNA-199 was frequently down-regulated in HCC and this down-regulation was negatively correlated with prognosis. Overexpression of lncRNA-199 could inhibit the growth and metastasis of HCC, while knockdown lncRNA-199 had the opposite effect. Down-regulated lncRNA-199 also could induce epithelial-mesenchymal transformation (EMT). Luciferase reporter assay demonstrated lncRNA-199 could modulate NRIP1 by sponge miR-576-5p. Conclusion: LncRNA-199 inhibited growth and metastasis through miR-576-5p/NRIP1 axis in HCC. This study revealed a novel unknown lncRNA function in malignant tumors, especially in HCC. At the same time, the regulatory mechanism of lncRNA-199 in HCC was clarified.

2022 ◽  
Vol 12 (4) ◽  
pp. 747-755
Author(s):  
Shengyong Liu ◽  
Xiangcheng Li

Background: Hepatocellular carcinoma (HCC) is a common malignant tumor worldwide with a poor prognosis. Amounting studies revealed that long non-coding RNAs (lncRNAs) show important roles in various biological processes. The purpose of this study was to explore the biological function and potential molecular mechanism of CASC7 in HCC. Methods: CASC7 expression in HCC cell lines was detected by qRT-PCR. The expressions of CASC7 and miR-340-5p were changed by transfection of miR-340-5p mimic, the CASC7 overexpression and knockdown plasmids. The interaction between CASC7 and miR-340-5p was assessed by a Dual-Luciferase reporter assay. The biological functions of CASC7 were evaluated by CCK-8, colony formation assay, ROS assay kit, immunofluorescence and flow cytometry (FCM). Results: CASC7 was upregulated in HCC cell lines. CASC7 overexpression significantly promoted cell proliferation, as well as inhibited apoptosis and oxidative stress. In contrast, CASC7 knockdown could reverse these above changes. The result of the Dual-luciferase reporter assay revealed that CASC7 directly targeted miR-340-5p and negatively regulated its expression. In addition, CASC7 promoted proliferation and inhibited apoptosis of HCC cells through activating Nrf2 pathway by downregulating miR-340-5p. Conclusions: In summary, CASC7 promotes HCC tumorigenesis and progression through the Nrf2 pathway by targeting miR-340-5p, which may provide a new target for therapy of HCC.


2020 ◽  
Author(s):  
Bo Lin ◽  
Enyi Shi ◽  
Qiu Jin ◽  
Wenhui Zhao ◽  
Juan Wang ◽  
...  

Abstract Background:Dysregulation of miRNAs is involved in carcinogenesis of breast and may be used as prognostic biomarkers and therapeutic targets during cancer process. The purpose of this study was to explore the effect of miR-105-3p on tumourigenicity of breast cancer and its underlying molecular mechanisms.Methods:Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to detect the expression of miR-105-3p in breast cancer tissues and cell lines. The impacts of miR-105-3p on proliferation, migration, invasion and apoptosis of human breast cancer cells (MCF-7 and ZR-75-30) were evaluated by CCK-8, transwell chamber assay, TUNEL assay and western blot assay, respectively. Besides, bioinformatics and luciferase reporter assay were used to find out the target genes of miR-105-3p.Results:The expression of miR-105-3p was elevated in breast cancer tissues and increased along with tumor severity. Downregulation of miR-105-3p could inhibit cell proliferation, suppress cell migration/invasion, and promote cell apoptosis in MCF-7 and ZR-75-30 cells. Furthermore, Golgi integral membrane protein 4 (GOLIM4) was identified to be the direct target gene of miR-105-3p by bioinformatics and luciferase reporter assay. In addition, silencing of GOLIM4 could restore the anti-breast cancer effects induced by miR-105-3p downregulation.Conclusions:miR-105-3p acts an oncogene to promote proliferation and metastasis of breast cancer cell by targeting GOLIM4, which provides a new target for the prevention and treatment of breast cancer.


2021 ◽  
Author(s):  
Bo Lin ◽  
Enyi Shi ◽  
Qiu Jin ◽  
Wenhui Zhao ◽  
Juan Wang ◽  
...  

Abstract Background: Dysregulation of miRNAs is involved in carcinogenesis of breast and may be used as prognostic biomarkers and therapeutic targets during cancer process. The purpose of this study was to explore the effect of miR-105-3p on tumourigenicity of breast cancer and its underlying molecular mechanisms.Methods: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to detect the expression of miR-105-3p in breast cancer tissues and cell lines. The impacts of miR-105-3p on proliferation, migration, invasion and apoptosis of human breast cancer cells (MCF-7 and ZR-75-30) were evaluated by CCK-8, transwell chamber assay, TUNEL assay and western blot assay, respectively. Besides, bioinformatics and luciferase reporter assay were used to find out the target genes of miR-105-3p.Results: The expression of miR-105-3p was elevated in breast cancer tissues and increased along with tumor severity. Downregulation of miR-105-3p could inhibit cell proliferation, suppress cell migration/invasion, and promote cell apoptosis in MCF-7 and ZR-75-30 cells. Furthermore, Golgi integral membrane protein 4 (GOLIM4) was identified to be the direct target gene of miR-105-3p by bioinformatics and luciferase reporter assay. In addition, silencing of GOLIM4 could restore the anti-breast cancer effects induced by miR-105-3p downregulation.Conclusions: MiR-105-3p acts an oncogene to promote proliferation and metastasis of breast cancer cell by targeting GOLIM4, which provides a new target for the prevention and treatment of breast cancer.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Qingyun Pan ◽  
Ying Huang ◽  
Yirui Wang ◽  
Deke Li ◽  
Changjiang Lei

Abstract Background Long non-coding RNA is considered to be essential to modulate the development and progression of human malignant cancers. And long non-coding RNA can act as crucial modulators by sponging the corresponding microRNA in tumorigenesis. We aimed to elucidate the function of ACTA2-AS1 and its molecular mechanism in colon adenocarcinoma. Materials and methods The expression of ACTA2-AS1, miR-4428 and BCL2L11 in colon adenocarcinoma tissues were detected via qRT-PCR. SW480 and HT29 cells were transfected with shRNA ACTA2-AS1, OE ACTA2-AS1, miRNA mimics of miR-4428, miR-4428 inhibitor, si-BCL2L11 and over-expression of si-BCL2L11. Cell proliferation, colony formation and apoptosis were respectively assessed using CCK-8 assay, colony assay and flow cytometry. Luciferase reporter assay was performed to verify the targets of ACTA2-AS1 and miR-4428. Tumor subcutaneous xenograft mode was constructed to explore tumor growth in vivo. Results ACTA2-AS1 was obviously downregulated in human colon adenocarcinoma tissues and colon adenocarcinoma cell lines. Silence or over-expression of ACTA2-AS1 promoted or inhibited cell proliferation and colony formation abilities, and regulated apoptosis. The silence of ACTA2-AS1 resulted in the decrease of Bax and increase of Bal2, while restored in OE ACTA2-AS1 group when compared with the control transfected cells. In addition, luciferase reporter assay revealed that ACTA2-AS1 interacted with miR-4428 and suppressed its expression. miR-4428 could bind to 3ʹ untranslated region of BCL2L11 and modulated the expression of BCL2L11 negatively. Knockdown of ACTA2-AS1 and over-expression of BCL2L11 reversed the biological function that ACTA2-AS1 mediated by knockdown ACTA2-AS1 alone. Conclusion Our data demonstrated that ACTA2-AS1 could suppress colon adenocarcinoma progression via sponging miR-4428 to regulate BCL2L11 expression.


2019 ◽  
Vol 25 (1) ◽  
Author(s):  
Ming Yang ◽  
Shijuan Sun ◽  
Yao Guo ◽  
Junjie Qin ◽  
Guangming Liu

Abstract Background Pancreatic cancer (PC) is a type of malignant gastrointestinal tumor. Long non-coding RNA MCM3AP antisense RNA 1 (MCM3AP-AS1) has been reported to stimulate proliferation, migration and invasion in several types of tumors. However, the role of MCM3AP-AS1 in PC remains unclear. Methods MCM3AP-AS1, microRNA miR-138-5p (miR-138-5p) and FOXK1 levels were detected using quantitative real time PCR. Cell proliferation, migration and invasion were analyzed. Dual luciferase reporter assay was used to confirm the relationship between MCM3AP-AS1 and miR-138-5p, between miR-138-5p and FOXK1. Protein levels were identified using western blot analysis. Results MCM3AP-AS1 overexpression promoted proliferation, migration and invasion in PC cells. MCM3AP-AS1 silencing showed a suppressive effect on cell growth in PC cells. Moreover, MCM3AP-AS1 knockdown suppressed tumor growth in mice. Dual luciferase reporter assay demonstrated MCM3AP-AS1 could sponge microRNA-138-5p (miR-138-5p), and FOXK1 could bind with miR-138-5p. Positive correlation between MCM3AP-AS1 and FOXK1 was testified, as well as negative correlation between miR-138-5p and FOXK1. MCM3AP-AS1 promoted FOXK1 expression by targeting miR-138-5p, and MCM3AP-AS1 facilitated growth and invasion in PC cells by FOXK1. Conclusion MCM3AP-AS1 promoted growth and migration through modulating miR-138-5p/FOXK1 axis in PC, providing insights into MCM3AP-AS1/miR-138-5p/FOXK1 axis as novel candidates for PC therapy from bench to clinic.


Author(s):  
Haiyan Jia ◽  
Hong Wang ◽  
Yanfen Yao ◽  
Chunlei Wang ◽  
Pibao Li

MicroRNAs (miRNAs) play a vital role in regulating tumor progression. Dysregulated miR-136 expression was linked to the development of various human cancers. In the present study, we investigated the expression and relationship of miR-136 and COX2 in hepatocellular carcinoma (HCC) using relevant experiments, involving CCK-8, Transwell assay, and luciferase reporter assay. We demonstrated that miR-136 expression is obviously decreased in HCC tissues and cells, and negatively correlated with the expression of COX2 mRNA. In vitro assay revealed that overexpression of miR-136 significantly changed the expression of proliferation- and metastasis-related proteins and inhibited the proliferation, migration, and invasion of HepG2 and Hep3B cells. Dual-luciferase reporter assay validated that the 3′-untranslated region (3′-UTR) of COX2 is a direct target of miR-136. Furthermore, COX2 siRNA partially enhanced the miR-136 overexpression-induced inhibitory effects. In conclusion, miR-136 was vital in the regulation of HCC cell proliferation and metastasis by targeting COX2. Thus, our findings provided novel evidence that miR-136 might be recommended as a potential target for the diagnosis and treatment of HCC patients.


Author(s):  
Baiyin Mu ◽  
Chenlan Lv ◽  
Qingli Liu ◽  
Hong Yang

Abstract There is emerging evidence that dysregulation of long non-coding RNAs (lncRNAs) is associated with hepatocellular carcinoma (HCC). Zinc finger E-box binding homeobox 1 antisense 1 (ZEB1-AS1) functions as an oncogenic regulator in various malignancies. Nonetheless, the potential role of ZEB1-AS1 in HCC remains poorly elucidated. Herein, qRT-PCR was employed for examining ZEB1-AS1, miR-299-3p and E2F1 mRNA expressions in HCC cells and tissues. MTT assay was performed to evaluate cell proliferation. Transwell assay was utilized for evaluating cancer cell migration and invasion. Western blot was employed for measuring E2F1 protein expression. What’s more, dual-luciferase reporter assay was utilized for verifying the targeting relationships between ZEB1-AS1 and miR-299-3p, as well as E2F1 and miR-299-3p. It was demonstrated that, in HCC tissues and cells, ZEB1-AS1 expression was markedly increased, and meanwhile, its high expression level is related to the unfavorable clinicopathologic indicators. ZEB1-AS1 overexpression facilitated HCC cell proliferation, migration and invasion, while its knockdown led to the opposite effects. In terms of mechanism, we discovered that ZEB1-AS1 could decoy miR-299-3p and up-regulate E2F1 expression. This work reveals the functions and mechanism of ZEB1-AS1 in HCC tumorigenesis and progression, which provides novel biomarkers and therapeutic targets for HCC.


2020 ◽  
Author(s):  
Bo Lin ◽  
Enyi Shi ◽  
Qiu Jin ◽  
Wenhui Zhao ◽  
Juan Wang ◽  
...  

Abstract Background: Dysregulation of miRNAs is involved in carcinogenesis of breast cancer and may be used to prognostic biomarkers and therapeutic target during cancer process. The purpose of this study was to explore the effects of miR-105-3p on tumourigenicity of breast cancer and its underlying molecular mechanisms.Methods: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to detect the miR-105-3p in breast cancer tissues. The impacts of miR-105-3p on proliferation, migration, invasion and apoptosis of human breast cancer lines (MCF-7 and ZR-75-30) were evaluated by CCK-8, transwell chamber assay, TUNEL assay and western blot assay, respectively. Besides, bioinformatics and luciferase reporter assay were used find out the targeted gene of miR-105-3p.Results: The expression of miR-105-3p was elevated in breast cancer tissues and increased with tumor severity. Downregulation of miR-105-3p could inhibit cell proliferation, suppress cell migration/invasion, and promote cell apoptosis in MCF-7 and ZR-75-30 cells. Furthermore, Golgi integral membrane protein 4 (GOLIM4) was identified to be the direct target gene of miR-105-3p by bioinformatics and luciferase reporter assay. In addition, silencing of GOLIM4 could restore the anti-breast cancer effects induced by miR-105-3p downregulation.Conclusions: miR-105-3p acts an oncogene to promote proliferation and metastasis of breast cancer cell by targeting GOLIM4, which provides a new target for the prevention and treatment of breast cancer.


Author(s):  
Haitao Xie ◽  
Hui Zhou ◽  
Yan Jiang ◽  
Wenqian Xu ◽  
Leping Zeng ◽  
...  

IntroductionLong non-coding RNA LINC00641 has been reported to regulate tumor progression in several cancers. However, the expression and function of LINC00641 in hepatocellular carcinoma (HCC) is still unclear.Material and methodsIn this study, we measured the expression of LINC00641 in 79 pairs of HCC and adjacent normal liver tissues. The clinical significance of LINC00641 in HCC was explored. We also investigated the function of LINC00641 in HCC proliferation and invasion.ResultsWe observed that LINC00641 expression was significantly increased in HCC relative to normal tissues (P < 0.0001). High expression of LINC00641 was significantly associated with vascular invasion, advanced TNM stage, and reduced overall survival in HCC patients. Knockdown of LINC00641 inhibited the proliferation, colony formation, and invasion of HCC cells. In contrast, overexpression of LINC00641 promoted HCC cell growth and invasiveness. In vivo studies confirmed that knockdown of LINC00641 restrained tumorigenesis of HCC cells. Mechanistic studies revealed that LINC00641 inhibited the expression of miR-501-3p, which has been previously reported to act as a tumor suppressor in HCC. Furthermore, luciferase reporter assays validated that LINC00641 harbored a target site for miR-501-3p. Rescue experiments demonstrated that LINC00641-induced proliferation and invasion of HCC cells was reversed by co-expression of miR-501-3p.ConclusionsTaken together, LINC00641 contributes to aggressive phenotype of HCC cells by sponging miR-501-3p and represents a promising therapeutic target for this disease.


Gene ◽  
2018 ◽  
Vol 678 ◽  
pp. 129-136 ◽  
Author(s):  
Zhangjun Cheng ◽  
Zhengqing Lei ◽  
Pinghua Yang ◽  
Anfeng Si ◽  
Daimin Xiang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document