Abstract

BackgroundAberrant endocytic recycling has fundamental functions on plasma membrane component turnover. Recent studies have identified an uncharacterized protein, CCDC132, in the endosome-associated recycling protein complex. Besides, our preliminary data first showed that CCDC132 was elevated in malignant neoplasms, especially in esophagus/stomach cancers. However, the functions and the underlying mechanisms of CCDC132 in gastric cancer (GC) biology remain unclear.MethodsThe CCDC132 mRNA expression in 4 GC cell lines and normal gastric epithelial cell lines was detected by qRT-PCR. Then, CCDC132 was downregulated in AGS and MGC-803 cells by lentivirus-induced RNA interfere, and cell viability assay, clone formation assay and apoptosis assay were carried out. The mechanism of CCDC132 on cell proliferation and apoptosis activation was explored using PathScan® Stress, apoptosis signaling arrays and Western blot. We further investigated the pro-oncogenesis of CCDC132 in vivo. Meanwhile, immunohistochemistry was utilized to analyze the association between CCDC132 expression and clinicopathological features and prognosis. Finally, the correlation between CCDC132 and p53 was analyzed by Spearman’s rank correlation analysis.ResultsIn this study, knockdown of CCDC132 significantly decreased cell proliferation and clone formation ability and facilitated apoptosis, and increased phosphorylation of p53 and Chk2 and protein levels of γ-H2AX, 53BP1, cleaved Caspase 3 and cleaved PARP. Additionally, knockdown of CCDC132 attenuated tumorigenesis and tumor growth of MGC-803 cell xenografts. CCDC132 expression was significantly higher in GC tissues compared with that in adjacent normal tissues and was positively correlated with nodal metastasis and TNM stage and negatively associated with prognosis. The survival rate of CCDC132 positive patients was lower than that of CCDC132-negative patients. Furthermore, CCDC132 expression was negatively related to p53.ConclusionThis study unravels that knockdown of CCDC132 attenuates GC cell proliferation and tumorigenesis by facilitating DNA damage signaling, indicating that CCDC132 may serve as a potential target for GC therapy.

Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.