Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
Eur Rev Med Pharmacol Sci ; 24(5): 2428-2441, 2020 03.
Article in English | MEDLINE | ID: mdl-32196594

ABSTRACT

OBJECTIVE: Gastric cancer (GC) remains a serious disease to human health with high mortality worldwide. Evolving evidence implied that long non-coding RNA Opa interacting protein 5-antisense RNA1 (OIP5-AS1) went in for the pathological progress of GC. Nevertheless, the potential molecular mechanism of OIP5-AS1 needed to be further investigated. MATERIALS AND METHODS: Levels of OIP5-AS1, microRNA (miR)-153-3p, and zinc finger and BTB domain containing 2 (ZBTB2) were assessed using quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot assays. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) was implemented to detect cell proliferation in vitro. Cell apoptosis was evaluated by flow cytometry. Besides, transwell assay was conducted to examine cell migration and invasion in AGS and MKN45 cells. The interaction between miR-153-3p and OIP5-AS1 or ZBTB2 was validated utilizing dual-luciferase reporter assay. Lastly, the role of OIP5-AS1 in tumor growth was researched through adopting xenograft tumor model. RESULTS: OIP5-AS1 and ZBTB2 were strongly higher in GC tissues than noncancerous samples. OIP5-AS1 silencing remarkably curbed cell proliferation, migration and invasion, and elevated cell apoptosis in both AGS and MKN45 cells. Functional analysis indicated that OIP5-AS1 regulated ZBTB2 expression via binding to miR-153-3p. Moreover, the role of miR-153-3p in cell growth and metastasis was abrogated by ZBTB2 overexpression. Above all, OIP5-AS1 could reduce the growth of xenograft tumor in vivo. CONCLUSIONS: OIP5-AS1 exerted its role via miR-153-3p/ZBTB2 axis in the progression of GC cells. These findings might supply a biomarker for the diagnosis and therapy of GC clinically.


Subject(s)
MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Repressor Proteins/metabolism , Stomach Neoplasms/metabolism , Animals , Cell Proliferation , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Stomach Neoplasms/pathology , Tumor Cells, Cultured
2.
Eur Rev Med Pharmacol Sci ; 23(22): 9890-9899, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31799657

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is the fourth common cancer worldwide. Long non-coding RNA TOB1 antisense RNA 1 (TOB1-AS1) has been found to participate in the process of GC, while the precise role of TOB1-AS1 is still not understood in GC progression. MATERIALS AND METHODS: We collected 21-paired GC and para-carcinoma tissue specimens, and the levels of TOB1-AS1 and lysosomal sialidase (NEU1) were detected by quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). The protein expression levels of NEU1, b-catenin, c-Myc, Cyclin D1, N-cadherin were determined via Western blot. Cell proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Flow cytometry was performed to evaluate cell proliferation. Besides, GC cell migration and invasion capacities were identified by transwell assay. Dual-Luciferase reporter assay was employed to examine the interrelation between miR-23a and TOB1-AS1 or NEU1. Finally, the role of TOB1-AS1 was verified in vivo. RESULTS: The levels of TOB1-AS1 were decreased in GC tissues and cell lines. Either TOB1-AS1 or NEU1 upregulation accelerated GC cell apoptosis, hampered proliferation, migration, and invasion. Further, the role of TOB1-AS1 silencing on cell behaviors was abrogated by NEU1 upregulation. TOB1-AS1 and NEU1 exerted their roles via Wnt/b-catenin signaling pathway. Overexpression of TOB1-AS1 blocked GC development in vivo. Mechanically, miR-23a was targeted by TOB1-AS1, but directly targeted NEU1. CONCLUSIONS: TOB1-AS1/miR-23a/NEU1 axis regulated proliferation, apoptosis, migration, and invasion of GC cells via Wnt/b-catenin pathway, providing the evidence for serving TOB1-AS1 as an underlying therapeutic target in human GC treatment.


Subject(s)
MicroRNAs/genetics , Neuraminidase/genetics , Neuraminidase/metabolism , RNA, Long Noncoding/genetics , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Transplantation , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL