Cargando…

Mitochondrial dynamics regulates migration and invasion of breast cancer cells

Mitochondria are highly dynamic and undergo constant fusion and fission that are essential for maintaining physiological functions of cells. Although dysfunction of mitochondria has been implicated in tumorigenesis, little is known about the roles of mitochondrial dynamics in metastasis, the major c...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Jing, Zhang, Juan, Yu, Meifang, Xie, Yan, Huang, Youguo, Wolff, Dennis W., Abel, Peter W., Tu, Yaping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3911914/
https://www.ncbi.nlm.nih.gov/pubmed/23128392
http://dx.doi.org/10.1038/onc.2012.494
_version_ 1782302012241608704
author Zhao, Jing
Zhang, Juan
Yu, Meifang
Xie, Yan
Huang, Youguo
Wolff, Dennis W.
Abel, Peter W.
Tu, Yaping
author_facet Zhao, Jing
Zhang, Juan
Yu, Meifang
Xie, Yan
Huang, Youguo
Wolff, Dennis W.
Abel, Peter W.
Tu, Yaping
author_sort Zhao, Jing
collection PubMed
description Mitochondria are highly dynamic and undergo constant fusion and fission that are essential for maintaining physiological functions of cells. Although dysfunction of mitochondria has been implicated in tumorigenesis, little is known about the roles of mitochondrial dynamics in metastasis, the major cause of cancer death. In the present study, we found a marked upregulation of mitochondrial fission protein dynamin-related protein 1 (Drp1) expression in human invasive breast carcinoma and metastases to lymph nodes. Compared to non-metastatic breast cancer cells, mitochondria also were more fragmented in metastatic breast cancer cells that express higher levels of total and active Drp1 and less mitochondrial fusion protein 1 (Mfn1). Silencing Drp1 or overexpression of Mfn1 resulted in mitochondria elongation or clusters, respectively, and significantly suppressed metastatic abilities of breast cancer cells. In contrast, silencing Mfn proteins led to mitochondrial fragmentation and enhanced metastatic abilities of breast cancer cells. Interestingly, these manipulations of mitochondrial dynamics altered the subcellular distribution of mitochondria in breast cancer cells. For example, silencing Drp1 or overexpression of Mfn1 inhibited lamellipodia formation, a key step for cancer metastasis, and suppressed chemoattractant-induced recruitment of mitochondria to lamellipodial regions. Conversely, silencing Mfn proteins resulted in more cell spreading and lamellipodia formation, causing accumulation of more mitochondria in lamollipodia regions. More importantly, treatment with a mitochondrial uncoupling agent or ATP synthesis inhibitor reduced lamellipodia formation and decreased breast cancer cell migration and invasion, suggesting a functional importance of mitochondria in breast cancer metastasis. Together, our findings show a new role and mechanism for regulation of cancer cell migration and invasion by mitochondrial dynamics. Thus targeting dysregulated Drp1-dependent mitochondrial fission may provide a novel strategy for suppressing breast cancer metastasis.
format Online
Article
Text
id pubmed-3911914
institution National Center for Biotechnology Information
language English
publishDate 2012
record_format MEDLINE/PubMed
spelling pubmed-39119142014-04-01 Mitochondrial dynamics regulates migration and invasion of breast cancer cells Zhao, Jing Zhang, Juan Yu, Meifang Xie, Yan Huang, Youguo Wolff, Dennis W. Abel, Peter W. Tu, Yaping Oncogene Article Mitochondria are highly dynamic and undergo constant fusion and fission that are essential for maintaining physiological functions of cells. Although dysfunction of mitochondria has been implicated in tumorigenesis, little is known about the roles of mitochondrial dynamics in metastasis, the major cause of cancer death. In the present study, we found a marked upregulation of mitochondrial fission protein dynamin-related protein 1 (Drp1) expression in human invasive breast carcinoma and metastases to lymph nodes. Compared to non-metastatic breast cancer cells, mitochondria also were more fragmented in metastatic breast cancer cells that express higher levels of total and active Drp1 and less mitochondrial fusion protein 1 (Mfn1). Silencing Drp1 or overexpression of Mfn1 resulted in mitochondria elongation or clusters, respectively, and significantly suppressed metastatic abilities of breast cancer cells. In contrast, silencing Mfn proteins led to mitochondrial fragmentation and enhanced metastatic abilities of breast cancer cells. Interestingly, these manipulations of mitochondrial dynamics altered the subcellular distribution of mitochondria in breast cancer cells. For example, silencing Drp1 or overexpression of Mfn1 inhibited lamellipodia formation, a key step for cancer metastasis, and suppressed chemoattractant-induced recruitment of mitochondria to lamellipodial regions. Conversely, silencing Mfn proteins resulted in more cell spreading and lamellipodia formation, causing accumulation of more mitochondria in lamollipodia regions. More importantly, treatment with a mitochondrial uncoupling agent or ATP synthesis inhibitor reduced lamellipodia formation and decreased breast cancer cell migration and invasion, suggesting a functional importance of mitochondria in breast cancer metastasis. Together, our findings show a new role and mechanism for regulation of cancer cell migration and invasion by mitochondrial dynamics. Thus targeting dysregulated Drp1-dependent mitochondrial fission may provide a novel strategy for suppressing breast cancer metastasis. 2012-11-05 2013-10 /pmc/articles/PMC3911914/ /pubmed/23128392 http://dx.doi.org/10.1038/onc.2012.494 Text en Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms
spellingShingle Article
Zhao, Jing
Zhang, Juan
Yu, Meifang
Xie, Yan
Huang, Youguo
Wolff, Dennis W.
Abel, Peter W.
Tu, Yaping
Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title_full Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title_fullStr Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title_full_unstemmed Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title_short Mitochondrial dynamics regulates migration and invasion of breast cancer cells
title_sort mitochondrial dynamics regulates migration and invasion of breast cancer cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3911914/
https://www.ncbi.nlm.nih.gov/pubmed/23128392
http://dx.doi.org/10.1038/onc.2012.494
work_keys_str_mv AT zhaojing mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT zhangjuan mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT yumeifang mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT xieyan mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT huangyouguo mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT wolffdennisw mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT abelpeterw mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells
AT tuyaping mitochondrialdynamicsregulatesmigrationandinvasionofbreastcancercells