Original Article


Integrated analysis and knockdown of RAB23 indicate the role of RAB23 in gastric adenocarcinoma

Hui Chen, Dun Pan, Zhihuang Yang, Liangqing Li

Abstract

Background: The present study aimed to identify key differentially expressed genes (DEGs) and miRNAs (DEmiRNAs) in gastric adenocarcinoma.
Methods: We performed integrated analysis to determine DEGs and DEmiRNAs of gastric adenocarcinoma based on the GEO database. A DEmiRNA-target interaction network was established. GO and KEGG pathway enrichment analyses were utilized. Then, MKN45 cells were transfected with shRNA-RAB23 to knock down the expression of RAB23. CCK-8, transwell and flow cytometry assays were utilized to measure the capacities for cell proliferation, migration and apoptosis, and the apoptosis-related gene and protein levels were measured by using polymerase chain reaction (PCR) and Western blot, respectively. Colocalization analysis of Snc1 with the vesicular protein VAMP3 and the endoplasmic reticulum protein Calnexin was performed to assess the influence of RAB23 on vesicle transport. Finally, we performed metabolomic analysis by using gas chromatography mass spectrometry (GC-MS).
Results: We performed MMIA of gastric adenocarcinoma based on two miRNA datasets and two mRNA datasets. A total of 4,586 DEmRNAs and 30 DEmiRNAs were obtained. The DEmRNAs of gastric adenocarcinoma were significantly enriched in PI3K/Akt signaling. We identified three interactions, hsa- miR-23a-3p-PTPN4, hsa-miR-20b-5p (hsa-miR-130a-3p)-TNFRSF10B, and hsa-miR-130a-3p (hsa-miR- 363-3p)-RAB23, that may be related to the pathogenesis of gastric adenocarcinoma. The growth of MKN45 cells was inhibited by RAB23 knockdown via shRNA-RAB23 transfection. Metabolic analysis of three groups revealed a number of significantly altered metabolites, including glycerol, niacinamide, and nonadecanoic acid methylester.
Conclusions: RAB23 might be a target gene of hsa-miR-130a-3p and hsa-miR-363-3p. In gastric adenocarcinoma cells, knockdown of RAB23 inhibited cell proliferation, migration, and invasion and increased apoptosis by downregulating the PI3K/Akt pathway.

Download Citation