Abstract

BackgroundThe present study aimed to compare the protein profiles between paired samples of cervical cancer and paracancerous tissue and to identify a series of differentially expressed proteins (DEPs) through quantitative proteomics. MethodsThe DEPs were identified through proteomic profiles of three paired samples of cervical cancer (CC) and paracancerous tissue and through IHC examination in the CC tissue arrays. The lentiviral particles containing DUSP7 gene were transfected into SIHA cells (DUSP7-SIHA). CCK8 assay, colony formation assay, wound healing assay and transwell migration assay were used to evaluate the ability of cell proliferation, cell clone formation, cell migration and invasion. Immunofluorescence assay was used to detect the expression of E-cadherin and Vimentin in the target cells. ResultsThe decreased expression of DUSP7 (P=0.045 and 0.044, respectively) and increased expression of PLD1 (P=0.046 and 0.028, respectively) were significantly associated with a tumor size >2cm and parametrial infiltration. The decreased expression of DUSP7, and increased expression of PLD1 were adversely related to patients’ relapse (P=0.003, 0.040, and 0.001, respectively) and survival (P=0.034, 0.001, and 0.006, respectively). The DUSP7-SIHA cells proliferated slower than NC-SIHA cells, based on a clear delay in the doubling time (47.72±1.14h vs. 23.99±0.47h, P=0.0001). Cell cycle analysis indicated that the DUSP7-SIHA cells displayed a concomitant decrease in the percentage of cells in S phase (37.71±0.53% vs. 46.96±0.59%, P<0.0001) and a significant increase in the percentage of cells in G0/G1 phase (52.50±3.49% vs. 44.04±0.71%, P=0.0473) suggesting that inhibited proliferation of DUSP7-SIHA cells might be due to the inactivation of DNA synthesis. E-cadherin increased but Vimentin was reduced in DUSP7-SIHA cells, which demonstrated that overexpression of DUSP7 had a potential role in inhibiting EMT of SIHA cells. ConclusionsThe biological function of DUSP7 was possibly achieved through dephosphorylation of the ERK1/2 and inactivation of the RAS pathway. Legal entity responsible for the studyBeijing Chaoyang Hospital. FundingHas not received any funding. DisclosureAll authors have declared no conflicts of interest.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call