Cargando…

Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling

BACKGROUND: Metallothionein 1M (MT1M) functions to regulate cell proliferation and cancer metastasis. This study assessed the effects of MT1M overexpression and mouse double minute 2 homolog (MDM2) knockdown on the regulation of non-small cell lung cancer A549 cell viability, migration, and protein...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Wei, Jiang, Guo-Jun, Shi, Guo-Zhen, Chen, Ming-Zhi, Ma, Tie-Liang, Tan, Yong-Fei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8798298/
https://www.ncbi.nlm.nih.gov/pubmed/35117630
http://dx.doi.org/10.21037/tcr.2020.02.61
_version_ 1784641769480126464
author Xu, Wei
Jiang, Guo-Jun
Shi, Guo-Zhen
Chen, Ming-Zhi
Ma, Tie-Liang
Tan, Yong-Fei
author_facet Xu, Wei
Jiang, Guo-Jun
Shi, Guo-Zhen
Chen, Ming-Zhi
Ma, Tie-Liang
Tan, Yong-Fei
author_sort Xu, Wei
collection PubMed
description BACKGROUND: Metallothionein 1M (MT1M) functions to regulate cell proliferation and cancer metastasis. This study assessed the effects of MT1M overexpression and mouse double minute 2 homolog (MDM2) knockdown on the regulation of non-small cell lung cancer A549 cell viability, migration, and protein expression in vitro and explored the underlying molecular events. METHODS: A549 cells were stably infected with lentivirus carrying MT1M cDNA or transiently transfected MDM2 siRNA and/or treated with the p53 inhibitor for the assessment of changes in cell viability, wound healing, Transwell migration, and qRT-PCR and Western blot assays. Luciferase reporter assay was performed to investigate p53 binding to the MT1M promoter. RESULTS: The data showed that MT1M overexpression inhibited A549 cell viability and migration capacity in vitro, whereas the p53 inhibitor reversed the inhibition of A549 cell viability and migration caused by MT1M overexpression as well as the expression of MMP2, MMP9, and MMP14. Furthermore, knockdown of MDM2, an upstream inhibitor of p53 activity, was able to reduce A549 cell viability, migration, and protein expression. Thus, MDM2 knockdown had synergistic effects with MT1M overexpression on the suppression of A549 cell viability, migration, and protein expression. CONCLUSIONS: In conclusion, MDM2 can bind to and phosphorylate p53 protein to inactivate the protein, thereby reducing MT1M expression and leading to tumor cell proliferation and migration.
format Online
Article
Text
id pubmed-8798298
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-87982982022-02-02 Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling Xu, Wei Jiang, Guo-Jun Shi, Guo-Zhen Chen, Ming-Zhi Ma, Tie-Liang Tan, Yong-Fei Transl Cancer Res Original Article BACKGROUND: Metallothionein 1M (MT1M) functions to regulate cell proliferation and cancer metastasis. This study assessed the effects of MT1M overexpression and mouse double minute 2 homolog (MDM2) knockdown on the regulation of non-small cell lung cancer A549 cell viability, migration, and protein expression in vitro and explored the underlying molecular events. METHODS: A549 cells were stably infected with lentivirus carrying MT1M cDNA or transiently transfected MDM2 siRNA and/or treated with the p53 inhibitor for the assessment of changes in cell viability, wound healing, Transwell migration, and qRT-PCR and Western blot assays. Luciferase reporter assay was performed to investigate p53 binding to the MT1M promoter. RESULTS: The data showed that MT1M overexpression inhibited A549 cell viability and migration capacity in vitro, whereas the p53 inhibitor reversed the inhibition of A549 cell viability and migration caused by MT1M overexpression as well as the expression of MMP2, MMP9, and MMP14. Furthermore, knockdown of MDM2, an upstream inhibitor of p53 activity, was able to reduce A549 cell viability, migration, and protein expression. Thus, MDM2 knockdown had synergistic effects with MT1M overexpression on the suppression of A549 cell viability, migration, and protein expression. CONCLUSIONS: In conclusion, MDM2 can bind to and phosphorylate p53 protein to inactivate the protein, thereby reducing MT1M expression and leading to tumor cell proliferation and migration. AME Publishing Company 2020-04 /pmc/articles/PMC8798298/ /pubmed/35117630 http://dx.doi.org/10.21037/tcr.2020.02.61 Text en 2020 Translational Cancer Research. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
spellingShingle Original Article
Xu, Wei
Jiang, Guo-Jun
Shi, Guo-Zhen
Chen, Ming-Zhi
Ma, Tie-Liang
Tan, Yong-Fei
Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title_full Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title_fullStr Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title_full_unstemmed Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title_short Metallothionein 1M (MT1M) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through MDM2/p53/MT1M signaling
title_sort metallothionein 1m (mt1m) inhibits lung adenocarcinoma cell viability, migration, and expression of cell mobility-related proteins through mdm2/p53/mt1m signaling
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8798298/
https://www.ncbi.nlm.nih.gov/pubmed/35117630
http://dx.doi.org/10.21037/tcr.2020.02.61
work_keys_str_mv AT xuwei metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling
AT jiangguojun metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling
AT shiguozhen metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling
AT chenmingzhi metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling
AT matieliang metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling
AT tanyongfei metallothionein1mmt1minhibitslungadenocarcinomacellviabilitymigrationandexpressionofcellmobilityrelatedproteinsthroughmdm2p53mt1msignaling