Cargando…

The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway

BACKGROUND: Microspherule protein 1 (MCRS1) is a candidate oncogene and participates in various cellular processes, including growth, migration, senescence and transformation. MCRS1 is overexpressed in non-small cell lung cancer (NSCLC) and promotes the growth of cancer cells. However, the mechanism...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Minxia, Zhou, Kecheng, Huang, Yunchao, Cao, Yi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4606992/
https://www.ncbi.nlm.nih.gov/pubmed/26467212
http://dx.doi.org/10.1186/s13046-015-0235-5
_version_ 1782395450561658880
author Liu, Minxia
Zhou, Kecheng
Huang, Yunchao
Cao, Yi
author_facet Liu, Minxia
Zhou, Kecheng
Huang, Yunchao
Cao, Yi
author_sort Liu, Minxia
collection PubMed
description BACKGROUND: Microspherule protein 1 (MCRS1) is a candidate oncogene and participates in various cellular processes, including growth, migration, senescence and transformation. MCRS1 is overexpressed in non-small cell lung cancer (NSCLC) and promotes the growth of cancer cells. However, the mechanisms driving these processes are not fully understood. METHODS: Retrovirus-mediated RNA interference was employed to knockdown MCRS1 expression in cell lines. Cell proliferation assays and animal experiments were respectively performed to evaluate the growth of NSCLC cells in vitro and in vivo. Microarray analysis was carried out for mRNA profiling. Luciferase reporter assay and microRNA (miRNA) transfection were used to investigate the interaction between miRNA and gene. RESULTS: Stably knocking down MCRS1 expression inhibited the proliferation of NSCLC cells in vitro and in vivo. By comparing the mRNA expression profiles of NSCLC cells with or without MCRS1 silencing, we found that MCRS1 regulated expressions of various genes related to cell proliferation, including Rb1, TP53, cell cycle-related genes, MYC, E2F2, PCNA, and Ki67. However, MCRS1 did not directly bind to these differentially expressed genes. Here, we confirmed that Rb1, an important tumor suppression gene (TSG), is a direct target of miR-155 which is directly up-regulated by MCRS1. Furthermore, the level of Rb1 expression in NSCLC tissues was inversely correlated with those of miR-155 and MCRS1, and MCRS1 regulated expression of Rb1 via miR-155. Additionally, we found that the DNA copy number of the MCRS1 gene played a role in MCRS1 overexpression in NSCLCs. CONCLUSION: MCRS1 overexpression induced NSCLC proliferation through the miR-155–Rb1 pathway and DNA copy-number amplification is one of the mechanisms underlying MCRS1 overexpression in NSCLC. Moreover, we put forward the hypothesis that there are regulatory relationships between oncogenes and TSGs apart from the functional synergy of both; the oncogene-miRNA-TSG networks are one of mechanisms among the regulatory relationships; the regulatory relationships and the networks might play active roles in the development and progression of cancer. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13046-015-0235-5) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4606992
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-46069922015-10-16 The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway Liu, Minxia Zhou, Kecheng Huang, Yunchao Cao, Yi J Exp Clin Cancer Res Research BACKGROUND: Microspherule protein 1 (MCRS1) is a candidate oncogene and participates in various cellular processes, including growth, migration, senescence and transformation. MCRS1 is overexpressed in non-small cell lung cancer (NSCLC) and promotes the growth of cancer cells. However, the mechanisms driving these processes are not fully understood. METHODS: Retrovirus-mediated RNA interference was employed to knockdown MCRS1 expression in cell lines. Cell proliferation assays and animal experiments were respectively performed to evaluate the growth of NSCLC cells in vitro and in vivo. Microarray analysis was carried out for mRNA profiling. Luciferase reporter assay and microRNA (miRNA) transfection were used to investigate the interaction between miRNA and gene. RESULTS: Stably knocking down MCRS1 expression inhibited the proliferation of NSCLC cells in vitro and in vivo. By comparing the mRNA expression profiles of NSCLC cells with or without MCRS1 silencing, we found that MCRS1 regulated expressions of various genes related to cell proliferation, including Rb1, TP53, cell cycle-related genes, MYC, E2F2, PCNA, and Ki67. However, MCRS1 did not directly bind to these differentially expressed genes. Here, we confirmed that Rb1, an important tumor suppression gene (TSG), is a direct target of miR-155 which is directly up-regulated by MCRS1. Furthermore, the level of Rb1 expression in NSCLC tissues was inversely correlated with those of miR-155 and MCRS1, and MCRS1 regulated expression of Rb1 via miR-155. Additionally, we found that the DNA copy number of the MCRS1 gene played a role in MCRS1 overexpression in NSCLCs. CONCLUSION: MCRS1 overexpression induced NSCLC proliferation through the miR-155–Rb1 pathway and DNA copy-number amplification is one of the mechanisms underlying MCRS1 overexpression in NSCLC. Moreover, we put forward the hypothesis that there are regulatory relationships between oncogenes and TSGs apart from the functional synergy of both; the oncogene-miRNA-TSG networks are one of mechanisms among the regulatory relationships; the regulatory relationships and the networks might play active roles in the development and progression of cancer. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13046-015-0235-5) contains supplementary material, which is available to authorized users. BioMed Central 2015-10-14 /pmc/articles/PMC4606992/ /pubmed/26467212 http://dx.doi.org/10.1186/s13046-015-0235-5 Text en © Liu et al. 2015 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Liu, Minxia
Zhou, Kecheng
Huang, Yunchao
Cao, Yi
The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title_full The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title_fullStr The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title_full_unstemmed The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title_short The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR–155–Rb1 pathway
title_sort candidate oncogene (mcrs1) promotes the growth of human lung cancer cells via the mir–155–rb1 pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4606992/
https://www.ncbi.nlm.nih.gov/pubmed/26467212
http://dx.doi.org/10.1186/s13046-015-0235-5
work_keys_str_mv AT liuminxia thecandidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT zhoukecheng thecandidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT huangyunchao thecandidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT caoyi thecandidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT liuminxia candidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT zhoukecheng candidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT huangyunchao candidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway
AT caoyi candidateoncogenemcrs1promotesthegrowthofhumanlungcancercellsviathemir155rb1pathway