miR-516a-3p inhibits breast cancer cell growth and EMT by blocking the Pygo2/Wnt signalling pathway.
miR-516a-3p has been reported to play a suppressive role in several types of human tumours. However, the expression level, biological function and fundamental mechanisms of miR-516a-3p in breast cancer remain unclear. In the present study, we found that miR-516a-3p expression was down-regulated and Pygopus2 (Pygo2) expression was up-regulated in human breast cancer tissues and cells. Through analysing the clinicopathological characteristics, we demonstrated that low miR-516a-3p expression or positive Pygo2 expression was a predictor of poor prognosis for patients with breast cancer. The results of a dual luciferase reporter assay and Western blot analysis indicated that Pygo2 was a target gene of miR-516a-3p. Moreover, overexpression of miR-516a-3p inhibited cell growth, migration and invasion as well as epithelial-mesenchymal transition (EMT) of breast cancer cells, whereas reduced miR-516a-3p expression promoted breast cancer cell growth, migration, invasion and EMT. Furthermore, we showed that miR-516a-3p suppressed cell proliferation, metastasis and EMT of breast cancer cells by inhibiting Pygo2 expression. We confirmed that miR-516a-3p exerted an anti-tumour effect by inhibiting the activation of the Wnt/β-catenin pathway. Finally, xenograft tumour models were used to show that miR-516a-3p inhibited breast cancer cell growth and EMT via suppressing the Pygo2/Wnt signalling pathway. Taken together, these results show that miR-516a-3p inhibits breast cancer cell growth, metastasis and EMT by blocking the Pygo2/ Wnt/β-catenin pathway.
Chi Y
,Wang F
,Zhang T
,Xu H
,Zhang Y
,Shan Z
,Wu S
,Fan Q
,Sun Y
... -
《-》
MiR-19a-3p regulates the Forkhead box F2-mediated Wnt/β-catenin signaling pathway and affects the biological functions of colorectal cancer cells.
Colorectal cancer (CRC) is one of the most common malignancies worldwide.
To explore the expression of microRNA miR-19a-3p and Forkhead box F2 (FOXF2) in patients with CRC and the relevant mechanisms.
Sixty-two CRC patients admitted to the hospital were enrolled into the study group, and sixty healthy people from the same period were assigned to the control group. Elbow venous blood was sampled from the patients and healthy individuals, and blood serum was saved for later analysis. MiR-19a-3p mimics, miR-19a-3p inhibitor, miR-negative control, small interfering-FOXF2, and short hairpin-FOXF2 were transfected into HT29 and HCT116 cells. Then quantitative polymerase chain reaction was performed to quantify the expression of miR-19a-3p and FOXF2 in HT29 and HCT116 cells, and western blot (WB) analysis was conducted to evaluate the levels of FOXF2, glycogen synthase kinase 3 beta (GSK-3β), phosphorylated GSK-3β (p-GSK-3β), β-catenin, p-β-catenin, α-catenin, N-cadherin, E-cadherin, and vimentin. The MTT, Transwell, and wound healing assays were applied to analyze cell proliferation, invasion, and migration, respectively, and the dual luciferase reporter assay was used to determine the correlation of miR-19a-3p with FOXF2.
The patients showed high serum levels of miR-19a-3p and low levels of FOXF2, and the area under the curves of miR-19a-3p and FOXF2 were larger than 0.8. MiR-19a-3p and FOXF2 were related to sex, tumor size, age, tumor-node-metastasis staging, lymph node metastasis, and differentiation of CRC patients. Silencing of miR-19a-3p and overexpression of FOXF2 suppressed the epithelial-mesenchymal transition, invasion, migration, and proliferation of cells. WB analysis revealed that silencing of miR-19a-3p and FOXF2 overexpression significantly suppressed the expression of p-GSK-3β, β-catenin, N-cadherin, and vimentin; and increased the levels of GSK-3β, p-β-catenin, α-catenin, and E-cadherin. The dual luciferase reporter assay confirmed that there was a targeted correlation of miR-19a-3p with FOXF2. In addition, a rescue experiment revealed that there were no differences in cell proliferation, invasion, and migration in HT29 and HCT116 cells co-transfected with miR-19a-3p-mimics+sh-FOXF2 and miR-19a-3p-inhibitor+si-FOXF2 compared to the miR-negative control group.
Inhibiting miR-19a-3p expression can upregulate the FOXF2-mediated Wnt/β-catenin signaling pathway, thereby affecting the epithelial-mesenchymal transition, proliferation, invasion, and migration of cells. Thus, miR-19a-3p is likely to be a therapeutic target in CRC.
Yu FB
,Sheng J
,Yu JM
,Liu JH
,Qin XX
,Mou B
... -
《-》
LncRNA HOXD-AS1 promotes epithelial ovarian cancer cells proliferation and invasion by targeting miR-133a-3p and activating Wnt/β-catenin signaling pathway.
Long non-coding RNA (lncRNA) HOXD cluster antisense RNA 1 (HOXD-AS1) functions as a crucial regulator in the progression and development of tumors. The aim of this study is to unravel the underlying mechanisms of HOXD-AS1 on epithelial ovarian cancer (EOC).
43 paired EOC tissues and adjacent non-tumor tissues were collected postoperatively from patients. QRT-PCR was used to explore HOXD-AS1 expression in both EOC tissues and cell lines. Cell proliferation and invasion were monitored by MTT assay and transwell invasion assay.
In the current study, we found that the expression of HOXD-AS1 was upregulated in EOC tissues and cell lines. High HOXD-AS1 expression was correlated with advanced FIGO stage, lymph node metastasis, and poor overall survival in EOC patients. We also showed that HOXD-AS1 promoted cell proliferation, invasion, and epithelial-mesenchymal transition (EMT) via activating Wnt/β-catenin signaling in EOC cells. Furthermore, we found that miR-133a-3p was a direct downstream target of HOXD-AS1 in EOC. HOXD-AS1 promoted cell proliferation, invasion, and EMT process through sponging miR-133a-3p in EOC cells.
Our study indicated that lncRNA HOXD-AS1 promoted the proliferation, invasion, and EMT process of EOC cells via targeting miR-133a-3p and activating Wnt/β-catenin signaling pathway.
Zhang Y
,Dun Y
,Zhou S
,Huang XH
... -
《-》
MiR-675 is frequently overexpressed in gastric cancer and enhances cell proliferation and invasion via targeting a potent anti-tumor gene PITX1.
Gastric cancer (GC) is a common malignancy around the world. Irregular expression of microRNAs (miRNAs) contributes to the progression of malignancies. Our study illustrated that miR-675 facilitates GC proliferation and invasion via targeting paired-like homeodomain transcription factor 1 (PITX1) and promoting epithelial-mesenchymal transition (EMT) as well as Wnt/β-catenin signaling pathway.
We collected the RNA-seq data of GC and normal stomach tissues from TCGA database to analyze the expression of miR-675 and PITX1. Kaplan-Meier plotter on line tool was used to analyze the association between miR-675 or PITX1 expression and the overall survival of GC patients. The biological function of miR-675 in GC cells was evaluated via altering its expression using miR-675 agomiR or antamiR. Dual-luciferase reporter assay was applied for verifying whether miR-675 could direct bind to 3'UTR of PITX1. Rescue assays were applied for characterizing the effects of miR-675/PITX1 axis on GC growth and invasion. Western blot was performed to evaluate the protein expression levels of PITX1, EMT-related and Wnt signaling-related proteins.
Our results showed that miR-675 is up-regulated and predictive of worse prognosis in GC patients. Overexpression of miR-675 in AGS cells notably promoted cell proliferation, migration and invasion, whilst down-regulation of miR-675 in SGC-7901 cells gained the opposite results. PITX1 is down-regulated in GC and identified as a direct target of miR-675. Overexpression of PITX1 in AGS cells reverses cell viability and invasion that enhanced by miR-675 up-regulation. Conversely, depletion of PITX1 in SGC-7901 cells rescues cell viability and invasion that inhibited by miR-675 down-regulation. Western bolt results revealed that miR-675 positively regulated EMT and Wnt/β-catenin signaling pathway in GC cells via targeting PITX1.
Our study emphasized the functional mechanism of miR-675 in GC and intimated that miR-675/PITX1 axis possibly affects proliferative and invasive properties of GC cells via regulating EMT and Wnt/β-catenin signaling pathway. Furthermore, miR-675 and PITX1 may be served as early diagnostic markers as well as therapeutic targets for GC.
Liu L
,Tian YC
,Mao G
,Zhang YG
,Han L
... -
《-》