scholarly journals MiR-133a-3p Functions as a Tumor Suppressor in Colorectal Cancer by Targeting PFKFB3

Author(s):  
Yanjie Zhou ◽  
Lu Jiang ◽  
Yuanjun Cai ◽  
Wendong Tang ◽  
Ke Wang

Abstract Background: Recent studies reveal that PFKFB3 plays an important role in tumorigenesis and tumor progression. Our study aims to identify an novel microRNA which can suppress the expression of PFKFB3 and to provide a potential target for tumor therapy.Methods: Bioinformatics methods were implemented to explore the expression and clinical significance of PFKFB3 and miR-133a-3p in colorectal cancer (CRC). qRT-PCR was performed to detect PFKFB3, miR-133a-3p, KI67 and MMP9 mRNA expression, while western bot was carried out for the detection of protein expression of PFKFB3, miR-133a-3p, KI67 and MMP9. Bioinformatics analysis was used to predict the binding sites of miR-133a-3p on PFKFB3 3’UTR, while dual-luciferase assay was conducted to validate their binding relationship. CCK-8 assay, KI67 detection, Transwell assay and MMP9 detection were employed to measure CRC cell proliferative and invasive abilities. Results: PFKFB3 is up-regulated in CRC and significantly associated with poor prognosis. Overexpressed PFKFB3 promotes CRC cell proliferation and invasion. miR-133a-3p is down-regulated and has diagnostic value in CRC. Dual-luciferase assay confirmed that there was a binding relationship between miR-133a-3p and PFKFB3.Overexpressed miR-133a-3p remarkably reduced PFKFB3 expression in CRC cells, weakened the promoting effect of PFKFB3 on cell proliferation and invasion.Conclusions: Our study suggests that miR-133a-3p functions as a novel tumor suppressor in colorectal cancer by targeting PFKFB3.

2021 ◽  
Author(s):  
Yanjie Zhou ◽  
Lu Jiang ◽  
Yuanjun Cai ◽  
Wendong Tang ◽  
Ke Wang

Abstract Background Recent studies reveal that PFKFB3 plays an important role in tumorigenesis and tumor progression. Our study aims to identify an novel microRNA which can suppress the expression of PFKFB3 and to provide a potential target for tumor therapy. Methods Bioinformatics methods were implemented to explore the expression and clinical significance of PFKFB3 and miR-133a-3p in colorectal cancer (CRC). qRT-PCR was performed to detect PFKFB3, miR-133a-3p, KI67 and MMP9 mRNA expression, while western bot was carried out for the detection of protein expression of PFKFB3, miR-133a-3p, KI67 and MMP9. Bioinformatics analysis was used to predict the binding sites of miR-133a-3p on PFKFB3 3’UTR, while dual-luciferase assay was conducted to validate their binding relationship. CCK-8 assay, KI67 detection, Transwell assay and MMP9 detection were employed to measure CRC cell proliferative and invasive abilities. Results PFKFB3 expression is up-regulated in colorectal cancer, and is significantly associated with poor prognosis. Silencing PFKFB3 could inhibit the proliferation and invasion of colorectal cancer cells. miR-133a-3p is down regulated in colorectal cancer, which has diagnostic value for colorectal cancer. Dual luciferase assay confirmed that PFKFB3 was the direct acting site of miR-133a-3p. Overexpression of miR-133a-3p could significantly reduce the expression of PFKFB3 and inhibit the effect of PFKFB3 on the proliferation and invasion of colorectal cancer cells. Conclusions Our study suggested that miR-133a-3p functions as a novel tumor suppressor in colorectal cancer by targeting PFKFB3.


2020 ◽  
Vol 20 ◽  
Author(s):  
Wenbin Wu ◽  
Yangmei Zhang ◽  
Xiaowu Li ◽  
Xiang Wang ◽  
Yuan Yuan

Objective: The purpose of this study was to explore the mechanism of the miR-375/XPR1 axis in esophageal squamous cell carcinoma (ESCC) and provide a new idea for targeted therapy of ESCC. Methods: Differentially expressed genes in GEO and TCGA databases were analyzed by bioinformatics. The expression levels of miR-375 and XPR1 mRNA were detected by qRT-PCR. Protein expression of XPR1 was detected by western blot. Bioinformatics analysis and dual luciferase assay were conducted to confirm the targeting relationship between miR-375 and XPR1. The viability, proliferation, migration and invasion of cells in each treatment group were detected by CCK-8, colony formation, wound healing and Transwell assays. Results: Significantly down-regulated miR-375 and remarkably up-regulated XPR1 were observed in ESCC tissue and cells. Overexpression of miR-375 inhibited proliferation, invasion and migration of ESCC cells, and greatly reduced the promoting effect of XPR1 overexpression on cell proliferation, migration and invasion. Dual luciferase assay confirmed that miR-375 targeted and inhibited XPR1 expression in ESCC. Conclusion: These results demonstrate the regulatory role of the miR-375/XPR1 axis in ESCC cells and provide a new potential target for the precise treatment of patients with ESCC.


2021 ◽  
Vol 12 (16) ◽  
pp. 4958-4970
Author(s):  
Xiaoyuan Wang ◽  
Peng Yang ◽  
Dongsheng Zhang ◽  
Ming Lu ◽  
Chi Zhang ◽  
...  

2019 ◽  
Vol 60 (5) ◽  
pp. 414 ◽  
Author(s):  
Jin'e Wan ◽  
Jian Yang ◽  
Cuixia Qiao ◽  
Xiaomei Sun ◽  
Aiting Di ◽  
...  

Author(s):  
Luciana Petti ◽  
Giulia Rizzo ◽  
Federica Rubbino ◽  
Sudharshan Elangovan ◽  
Piergiuseppe Colombo ◽  
...  

Abstract Background Sphingosine-1-phosphate receptor 2 (S1PR2) mediates pleiotropic functions encompassing cell proliferation, survival, and migration, which become collectively de-regulated in cancer. Information on whether S1PR2 participates in colorectal carcinogenesis/cancer is scanty, and we set out to fill the gap. Methods We screened expression changes of S1PR2 in human CRC and matched normal mucosa specimens [N = 76]. We compared CRC arising in inflammation-driven and genetically engineered models in wild-type (S1PR2+/+) and S1PR2 deficient (S1PR2−/−) mice. We reconstituted S1PR2 expression in RKO cells and assessed their growth in xenografts. Functionally, we mimicked the ablation of S1PR2 in normal mucosa by treating S1PR2+/+ organoids with JTE013 and characterized intestinal epithelial stem cells isolated from S1PR2−/−Lgr5-EGFP- mice. Results S1PR2 expression was lost in 33% of CRC; in 55%, it was significantly decreased, only 12% retaining expression comparable to normal mucosa. Both colitis-induced and genetic Apc+/min mouse models of CRC showed a higher incidence in size and number of carcinomas and/or high-grade adenomas, with increased cell proliferation in S1PR2−/− mice compared to S1PR2+/+ controls. Loss of S1PR2 impaired mucosal regeneration, ultimately promoting the expansion of intestinal stem cells. Whereas its overexpression attenuated cell cycle progression, it reduced the phosphorylation of AKT and augmented the levels of PTEN. Conclusions In normal colonic crypts, S1PR2 gains expression along with intestinal epithelial cells differentiation, but not in intestinal stem cells, and contrasts intestinal tumorigenesis by promoting epithelial differentiation, preventing the expansion of stem cells and braking their malignant transformation. Targeting of S1PR2 may be of therapeutic benefit for CRC expressing high Lgr5. Graphical Abstract. Schematic drawing of the role of S1PR2 in normal mucosa and colorectal cancer. In the normal mucosa, S1PR2 is highly expressed by differentiated cells at the upper region of both colon and intestinal crypts (S1PR2 ON), but not by the undifferentiated stem cell at the base of the crypts (S1PR2 OFF), in which acts as a negative proliferative regulator promoting epithelial differentiation. Its loss leads to the expansion of stem cells and reduced levels of PTEN and Axin-2, two negative regulators respectively of PI3K/AKT and Wnt signaling that control β-catenin signaling. The translocation of β-catenin into the nucleus promotes the transcription of target genes involved in the proliferation and malignant transformation. Thereby, S1PR2 works in the intestine as a tumor suppressor


2017 ◽  
Vol 41 (2) ◽  
pp. 635-644 ◽  
Author(s):  
Jian Xu ◽  
Rui Zhang ◽  
Jian Zhao

Background/Aims: The novel long noncoding RNA (lncRNA) tumor suppressor candidate 7 (TUSC7) has been reported as a potential tumor suppressor, while the functional role of TUSC7 is still unknown in colorectal cancer (CRC). Here, we characterized TUSC7 expression profile in CRC patients and investigated its biological function and potential molecular mechanism. Methods: RNA isolation, qRT-PCR, cell counter kit-8 assay, cell cycle assay, EdU assay, and western blot were performed. Statistical analyses were performed using SPSS 18.0 software and p value < 0.05 was considered as statistically significant. Results: In a cohort of CRC patients, we found TUSC7 was significantly downregulated in CRC tissues compared with adjacent non-tumor tissues (P < 0.01). Patients with high expression of TUSC7 had better survival than those with low expression of TUSC7 (HR = 0.342, 95% CI: 0.120-0.972, P = 0.044). Cell count kit 8 and EdU assays showed that ectopic expression of TUSC7 in HCT116 and SW480 cells significantly inhibited cell proliferation rate. After silence of TUSC7 with small interfering RNA, cell proliferation rate increased. Flow cytometry analyses revealed cycles were arrested at G1 phase after TUSC7 overexpression. We found there were 2 binding sites of miR-211-3p within the sequence of TUSC7 and TUSC7 expression level was negatively correlated with miR-211-3p. TUSC7 overexpression increased the expression level of CDK6, which is a downstream target of miR-211-3p, in both RNA and protein level. Furthermore, luciferase reporter assay indicated that TUSC7 could sponge miR-211-3p. Conclusion: To summary, we demonstrated that TUSC7 is a potential tumor suppressor in CRC, and TUSC7 could inhibit CRC cell proliferation by completely sponging miR-211-3p.


Sign in / Sign up

Export Citation Format

Share Document