Your browser doesn't support javascript.
loading
ZEB1-AS1 initiates a miRNA-mediated ceRNA network to facilitate gastric cancer progression.
Ma, Ming-Hui; An, Jia-Xiang; Zhang, Cheng; Liu, Jie; Liang, Yu; Zhang, Chun-Dong; Zhang, Zhen; Dai, Dong-Qiu.
Afiliação
  • Ma MH; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • An JX; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • Zhang C; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • Liu J; 2Science Experiment Center, China Medical University, Shenyang, 110122 China.
  • Liang Y; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • Zhang CD; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • Zhang Z; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
  • Dai DQ; 1Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 China.
Cancer Cell Int ; 19: 27, 2019.
Article em En | MEDLINE | ID: mdl-30774556
ABSTRACT

BACKGROUND:

Currently, cancer-related competing endogenous RNA (ceRNA) networks are attracting significant interest. As long noncoding RNA ZEB1-AS1 has been reported to function as an oncogene due to sponging microRNAs (miRNAs) in several cancers, we hypothesized that it could interact with specific miRNAs to form regulatory networks and facilitate the growth of gastric cancer (GC).

METHODS:

MiRNAs interacting with ZEB1-AS1 were screened for and selected by bioinformatics analysis. Overexpression or repression of ZEB1-AS1 was performed to determine whether it could regulate selected miRNAs. Quantitative real-time polymerase chain reactions (qPCR) validated the expression profiles of ZEB1-AS1 and miR-149-3p in GC cell lines and tissue. Statistical analysis determined the clinical significance of ZEB1-AS1 in relation to miR-149-3p. Cell counting, wound healing and transwell assays were performed to assess cell proliferation, migration and invasion. A luciferase reporter assay was utilized to confirm the putative miR-149-3p-binding sites in ZEB1-AS1.

RESULTS:

Briefly, bioinformatics analysis inferred that ZEB1-AS1 interacts with miR-204, miR-610, and miR-149. Gain- or loss-of function assays suggested that ZEB1-AS1 negatively regulates miR-149-3p, miR-204-5p and miR-610 in GC cells. Validated by qPCR, ZEB1-AS1 was up-regulated and miR-149-3p down-regulated in GC cells and tissue. Data analyses indicated that ZEB1-AS1 and miR-149-3p are associated with the independent diagnosis and prognosis of GC. Functional assays support the theory that miR-149-3p hinders GC proliferation, migration and invasion, whereas its overexpression abrogates the corresponding effects induced by ZEB1-AS1. Lastly, dissection of the molecular mechanisms involved indicated that ZEB1-AS1 can regulate GC partly via a ZEB1-AS1/miR-149-3p axis.

CONCLUSIONS:

ZEB1-AS1 can interact with specific miRNAs, forming a miRNA-mediated ceRNA network and promoting GC progress, partly through a ZEB1-AS1/miR-149-3p axis.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article