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.
Mol Carcinog ; 57(7): 854-865, 2018 07.
Article in English | MEDLINE | ID: mdl-29566283

ABSTRACT

As a long non-coding RNA, C5orf66-AS1 is located at 5q31.1. Downregulation and aberrant hypermethylation of C5orf66-AS1 have been detected in a limited several tumors. However, the biological role and distribution of methylated CpG sites of C5orf66-AS1 in gastric cardia adenocarcinoma (GCA) development and prognosis are poorly clarified. The present study was to investigate the expression status and function of C5orf66-AS1 in GCA, and to detect the distribution of methylated CpG sites within the three CpG islands of the promoter and gene body of C5orf66-AS1, further to clarify its prognostic value in GCA patients. C5orf66-AS1 was significantly downregulated in GCA tissues and cell lines, and the expression level was associated with TNM stage, pathological differentiation, lymph node metastasis, and distant metastasis or recurrence. The expression level of C5orf66-AS1 was significantly increased in cancer cells after treated with 5-Aza-dC. Further methylation analysis demonstrated that the aberrant hypermethylation of the regions around the transcription start site of C5orf66-AS1 was more tumor specific and was associated with its expression. Moreover, Sp1 may upregulate C5orf66-AS1 expression and CpG sites hypermethylation within the binding sites may abrogate Sp1 binding. In addition, C5orf66-AS1 inhibited gastric cancer cell proliferation and invasion, and the dysregulation and hypermethylation of the regions around the transcription start site of C5orf66-AS1 were associated with poorer GCA patients' survival. These findings suggest that aberrant hypermethylation-mediated downregulation of C5orf66-AS1 may play important roles in GCA tumorigenesis and C5orf66-AS1 may serve as a potential prognostic marker in predicting GCA patients' survival.


Subject(s)
Adenocarcinoma/genetics , Cardia/pathology , DNA Methylation/genetics , Down-Regulation/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Adult , Aged , Binding Sites/genetics , Carcinogenesis/genetics , Cell Proliferation/genetics , CpG Islands/genetics , Female , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Promoter Regions, Genetic/genetics , Stomach Neoplasms/pathology , Transcription Initiation Site/physiology , Up-Regulation/genetics
2.
Mol Cancer Res ; 15(7): 800-810, 2017 07.
Article in English | MEDLINE | ID: mdl-28539329

ABSTRACT

Maternally expressed gene 3 (MEG3), a long non-coding RNA (lncRNA), has tumor-suppressor properties and its expression is lost in several human tumors. However, its biological role in esophageal squamous cell carcinoma (ESCC) tumorigenesis is poorly defined. The present study determined the role and methylation status of MEG3 in esophageal cancer cells and ESCC clinical specimens, and further observed the competing endogenous RNA (ceRNA) activity of MEG3 in the pathogenesis and development of ESCC. Significant downregulation of MEG3 was detected in esophageal cancer cells and ESCC tissues and the expression level of MEG3 was significantly increased in cancer cells after treated with the DNA methyltransferase inhibitor 5-Aza-dC. Upregulation of MEG3 led to the inhibition of proliferation and invasiveness of the cancer cells. The aberrant promoter hypermethylation of MEG3 indicates silencing of its expression. Furthermore, MEG3 acts as a ceRNA to regulate the expression of E-cadherin and FOXO1 by binding hsa-miR-9. Upregulation of miR-9 was detected in esophageal cancer cell lines and ESCC tissues, and miR-9 promoted esophageal cancer cell proliferation and invasion. Finally, downregulation and hypermethylation of MEG3 was associated with ESCC patients' survival.Implications: MEG3 functions as a tumor-suppressive lncRNA and aberrant promoter hypermethylation is critical for MEG3 gene silencing in ESCC. In addition, MEG3 acts as a ceRNA to regulate expression of E-cadherin and FOXO1 by competitively binding miR-9 and may be used as a potential biomarker in predicting ESCC patients' progression and prognosis. Mol Cancer Res; 15(7); 800-10. ©2017 AACR.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Forkhead Box Protein O1/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Adult , Aged , Cell Proliferation/genetics , DNA Methylation/genetics , DNA Modification Methylases/genetics , Disease-Free Survival , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Genes, Tumor Suppressor , Humans , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , RNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...