Cargando…

Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation

BACKGROUND: The preferential use of aerobic glycolysis by tumor cells lead to high accumulation of lactate in tumor microenvironment. Clinical evidence has linked elevated lactate concentration with cancer outcomes. However, the role and molecular mechanisms of lactate in cellular senescence and tum...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Xiangrui, Zhang, Zhijian, Zhang, Yao, Cao, Yuxiang, Wei, Huijun, Wu, Zhihao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5828408/
https://www.ncbi.nlm.nih.gov/pubmed/29482580
http://dx.doi.org/10.1186/s13046-018-0701-y
_version_ 1783302639154364416
author Li, Xiangrui
Zhang, Zhijian
Zhang, Yao
Cao, Yuxiang
Wei, Huijun
Wu, Zhihao
author_facet Li, Xiangrui
Zhang, Zhijian
Zhang, Yao
Cao, Yuxiang
Wei, Huijun
Wu, Zhihao
author_sort Li, Xiangrui
collection PubMed
description BACKGROUND: The preferential use of aerobic glycolysis by tumor cells lead to high accumulation of lactate in tumor microenvironment. Clinical evidence has linked elevated lactate concentration with cancer outcomes. However, the role and molecular mechanisms of lactate in cellular senescence and tumor progression remain elusive. METHODS: The function of Snail in lactate-induced EMT in lung cancer cells was explored by wound healing assay and cell invasion assay. The qRT-PCR and dual luciferase reporter assay were performed to investigate how lactate regulates Snail expression. The level of TGF-β1 in culture supernatant of cells was measured by ELISA for its correlation with extracellular levels of lactate. Ras activity assay and SA-β-gal activity assay were established to determine the effect of lactate on oncogene-induced senescence in human lung epithelial cells. ChIP assays were conducted to determine the binding of snail to p16(INK4a) promoter. Two TCGA data sets (TCGA-LUAD and TCGA-LUSC) were used to explore the correlations between SNAI1 and CDKN2A expression. RESULTS: In this study, we showed the invasive and migratory potential of lung cancer cells was significantly enhanced by lactate and was directly linked to snail activity. We also demonstrated that extracellular acidification itself is a direct cause of the increased snail expression and physiologically coupled to LDHA-dependent conversion of pyruvate to lactate. Mechanistically, lactate exerts its central function in induction of snail and EMT by directly remodeling ECM and releasing activated TGF-β1. We also demonstrated that Snail help premalignant cells to escape the oncogene-induced senescence by directly targeting and inhibiting p16(INK4a) expression. CONCLUSIONS: Our study extends the understanding of EMT in tumorigenesis by uncovering the role of snail in cellular senescence. This study also reveals lactate may be a potent tumor-promoting factor and provides the basis for the development of lactate-targeted therapy.
format Online
Article
Text
id pubmed-5828408
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-58284082018-02-28 Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation Li, Xiangrui Zhang, Zhijian Zhang, Yao Cao, Yuxiang Wei, Huijun Wu, Zhihao J Exp Clin Cancer Res Research BACKGROUND: The preferential use of aerobic glycolysis by tumor cells lead to high accumulation of lactate in tumor microenvironment. Clinical evidence has linked elevated lactate concentration with cancer outcomes. However, the role and molecular mechanisms of lactate in cellular senescence and tumor progression remain elusive. METHODS: The function of Snail in lactate-induced EMT in lung cancer cells was explored by wound healing assay and cell invasion assay. The qRT-PCR and dual luciferase reporter assay were performed to investigate how lactate regulates Snail expression. The level of TGF-β1 in culture supernatant of cells was measured by ELISA for its correlation with extracellular levels of lactate. Ras activity assay and SA-β-gal activity assay were established to determine the effect of lactate on oncogene-induced senescence in human lung epithelial cells. ChIP assays were conducted to determine the binding of snail to p16(INK4a) promoter. Two TCGA data sets (TCGA-LUAD and TCGA-LUSC) were used to explore the correlations between SNAI1 and CDKN2A expression. RESULTS: In this study, we showed the invasive and migratory potential of lung cancer cells was significantly enhanced by lactate and was directly linked to snail activity. We also demonstrated that extracellular acidification itself is a direct cause of the increased snail expression and physiologically coupled to LDHA-dependent conversion of pyruvate to lactate. Mechanistically, lactate exerts its central function in induction of snail and EMT by directly remodeling ECM and releasing activated TGF-β1. We also demonstrated that Snail help premalignant cells to escape the oncogene-induced senescence by directly targeting and inhibiting p16(INK4a) expression. CONCLUSIONS: Our study extends the understanding of EMT in tumorigenesis by uncovering the role of snail in cellular senescence. This study also reveals lactate may be a potent tumor-promoting factor and provides the basis for the development of lactate-targeted therapy. BioMed Central 2018-02-26 /pmc/articles/PMC5828408/ /pubmed/29482580 http://dx.doi.org/10.1186/s13046-018-0701-y Text en © The Author(s). 2018 Open Access This 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
Li, Xiangrui
Zhang, Zhijian
Zhang, Yao
Cao, Yuxiang
Wei, Huijun
Wu, Zhihao
Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title_full Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title_fullStr Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title_full_unstemmed Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title_short Upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(INK4a) inactivation
title_sort upregulation of lactate-inducible snail protein suppresses oncogene-mediated senescence through p16(ink4a) inactivation
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5828408/
https://www.ncbi.nlm.nih.gov/pubmed/29482580
http://dx.doi.org/10.1186/s13046-018-0701-y
work_keys_str_mv AT lixiangrui upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation
AT zhangzhijian upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation
AT zhangyao upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation
AT caoyuxiang upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation
AT weihuijun upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation
AT wuzhihao upregulationoflactateinduciblesnailproteinsuppressesoncogenemediatedsenescencethroughp16ink4ainactivation