Cargando…

Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy

The MuvB multiprotein complex, together with B-MYB and FOXM1 (MMB-FOXM1), plays an essential role in cell cycle progression by regulating the transcription of genes required for mitosis and cytokinesis. In many tumors, B-MYB and FOXM1 are overexpressed as part of the proliferation signature. However...

Descripción completa

Detalles Bibliográficos
Autores principales: Wolter, Patrick, Hanselmann, Steffen, Pattschull, Grit, Schruf, Eva, Gaubatz, Stefan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5355254/
https://www.ncbi.nlm.nih.gov/pubmed/28061449
http://dx.doi.org/10.18632/oncotarget.14466
_version_ 1782515512522047488
author Wolter, Patrick
Hanselmann, Steffen
Pattschull, Grit
Schruf, Eva
Gaubatz, Stefan
author_facet Wolter, Patrick
Hanselmann, Steffen
Pattschull, Grit
Schruf, Eva
Gaubatz, Stefan
author_sort Wolter, Patrick
collection PubMed
description The MuvB multiprotein complex, together with B-MYB and FOXM1 (MMB-FOXM1), plays an essential role in cell cycle progression by regulating the transcription of genes required for mitosis and cytokinesis. In many tumors, B-MYB and FOXM1 are overexpressed as part of the proliferation signature. However, the transcriptional targets that are important for oncogenesis have not been identified. Given that mitotic kinesins are highly expressed in cancer cells and that selected kinesins have been reported as target genes of MMB-FOXM1, we sought to determine which mitotic kinesins are directly regulated by MMB-FOXM1. We demonstrate that six mitotic kinesins and two microtubule-associated non-motor proteins (MAPs) CEP55 and PRC1 are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cells. Suppression of KIF23 and PRC1 strongly suppressed proliferation of MDA-MB-231 cells. The set of MMB-FOXM1 regulated kinesins genes and 4 additional kinesins which we referred to as the mitotic kinesin signature (MKS) is linked to poor outcome in breast cancer patients. Thus, mitotic kinesins could be used as prognostic biomarker and could be potential therapeutic targets for the treatment of breast cancer.
format Online
Article
Text
id pubmed-5355254
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-53552542017-04-26 Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy Wolter, Patrick Hanselmann, Steffen Pattschull, Grit Schruf, Eva Gaubatz, Stefan Oncotarget Research Paper The MuvB multiprotein complex, together with B-MYB and FOXM1 (MMB-FOXM1), plays an essential role in cell cycle progression by regulating the transcription of genes required for mitosis and cytokinesis. In many tumors, B-MYB and FOXM1 are overexpressed as part of the proliferation signature. However, the transcriptional targets that are important for oncogenesis have not been identified. Given that mitotic kinesins are highly expressed in cancer cells and that selected kinesins have been reported as target genes of MMB-FOXM1, we sought to determine which mitotic kinesins are directly regulated by MMB-FOXM1. We demonstrate that six mitotic kinesins and two microtubule-associated non-motor proteins (MAPs) CEP55 and PRC1 are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cells. Suppression of KIF23 and PRC1 strongly suppressed proliferation of MDA-MB-231 cells. The set of MMB-FOXM1 regulated kinesins genes and 4 additional kinesins which we referred to as the mitotic kinesin signature (MKS) is linked to poor outcome in breast cancer patients. Thus, mitotic kinesins could be used as prognostic biomarker and could be potential therapeutic targets for the treatment of breast cancer. Impact Journals LLC 2017-01-03 /pmc/articles/PMC5355254/ /pubmed/28061449 http://dx.doi.org/10.18632/oncotarget.14466 Text en Copyright: © 2017 Wolter et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Wolter, Patrick
Hanselmann, Steffen
Pattschull, Grit
Schruf, Eva
Gaubatz, Stefan
Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title_full Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title_fullStr Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title_full_unstemmed Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title_short Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy
title_sort central spindle proteins and mitotic kinesins are direct transcriptional targets of muvb, b-myb and foxm1 in breast cancer cell lines and are potential targets for therapy
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5355254/
https://www.ncbi.nlm.nih.gov/pubmed/28061449
http://dx.doi.org/10.18632/oncotarget.14466
work_keys_str_mv AT wolterpatrick centralspindleproteinsandmitotickinesinsaredirecttranscriptionaltargetsofmuvbbmybandfoxm1inbreastcancercelllinesandarepotentialtargetsfortherapy
AT hanselmannsteffen centralspindleproteinsandmitotickinesinsaredirecttranscriptionaltargetsofmuvbbmybandfoxm1inbreastcancercelllinesandarepotentialtargetsfortherapy
AT pattschullgrit centralspindleproteinsandmitotickinesinsaredirecttranscriptionaltargetsofmuvbbmybandfoxm1inbreastcancercelllinesandarepotentialtargetsfortherapy
AT schrufeva centralspindleproteinsandmitotickinesinsaredirecttranscriptionaltargetsofmuvbbmybandfoxm1inbreastcancercelllinesandarepotentialtargetsfortherapy
AT gaubatzstefan centralspindleproteinsandmitotickinesinsaredirecttranscriptionaltargetsofmuvbbmybandfoxm1inbreastcancercelllinesandarepotentialtargetsfortherapy