Cargando…

HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis

Tumor metastasis depends on the dynamic balance of the actomyosin cytoskeleton. As a key component of actomyosin filaments, non-muscle myosin-IIA disassembly contributes to tumor cell spreading and migration. However, its regulatory mechanism in tumor migration and invasion is poorly understood. Her...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Lu, Zhou, Xiaolei, Liu, Bowen, Shi, Xuhe, Li, Xianmeng, Xu, Feifei, Fu, Xueli, Wang, Xue, Ye, Kai, Jin, Tianzhi, Sun, Huimin, Li, Qianqian, Zhang, Weiying, Ye, Lihong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10031283/
https://www.ncbi.nlm.nih.gov/pubmed/36970214
http://dx.doi.org/10.1016/j.apsb.2022.11.025
_version_ 1784910572236570624
author Zhang, Lu
Zhou, Xiaolei
Liu, Bowen
Shi, Xuhe
Li, Xianmeng
Xu, Feifei
Fu, Xueli
Wang, Xue
Ye, Kai
Jin, Tianzhi
Sun, Huimin
Li, Qianqian
Zhang, Weiying
Ye, Lihong
author_facet Zhang, Lu
Zhou, Xiaolei
Liu, Bowen
Shi, Xuhe
Li, Xianmeng
Xu, Feifei
Fu, Xueli
Wang, Xue
Ye, Kai
Jin, Tianzhi
Sun, Huimin
Li, Qianqian
Zhang, Weiying
Ye, Lihong
author_sort Zhang, Lu
collection PubMed
description Tumor metastasis depends on the dynamic balance of the actomyosin cytoskeleton. As a key component of actomyosin filaments, non-muscle myosin-IIA disassembly contributes to tumor cell spreading and migration. However, its regulatory mechanism in tumor migration and invasion is poorly understood. Here, we found that oncoprotein hepatitis B X-interacting protein (HBXIP) blocked the myosin-IIA assemble state promoting breast cancer cell migration. Mechanistically, mass spectrometry analysis, co-immunoprecipitation assay and GST-pull down assay proved that HBXIP directly interacted with the assembly-competent domain (ACD) of non-muscle heavy chain myosin-IIA (NMHC-IIA). The interaction was enhanced by NMHC-IIA S1916 phosphorylation via HBXIP-recruited protein kinase PKCβII. Moreover, HBXIP induced the transcription of PRKCB, encoding PKCβII, by coactivating Sp1, and triggered PKCβII kinase activity. Interestingly, RNA sequencing and mouse metastasis model indicated that the anti-hyperlipidemic drug bezafibrate (BZF) suppressed breast cancer metastasis via inhibiting PKCβII-mediated NMHC-IIA phosphorylation in vitro and in vivo. We reveal a novel mechanism by which HBXIP promotes myosin-IIA disassembly via interacting and phosphorylating NMHC-IIA, and BZF can serve as an effective anti-metastatic drug in breast cancer.
format Online
Article
Text
id pubmed-10031283
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-100312832023-03-23 HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis Zhang, Lu Zhou, Xiaolei Liu, Bowen Shi, Xuhe Li, Xianmeng Xu, Feifei Fu, Xueli Wang, Xue Ye, Kai Jin, Tianzhi Sun, Huimin Li, Qianqian Zhang, Weiying Ye, Lihong Acta Pharm Sin B Original Article Tumor metastasis depends on the dynamic balance of the actomyosin cytoskeleton. As a key component of actomyosin filaments, non-muscle myosin-IIA disassembly contributes to tumor cell spreading and migration. However, its regulatory mechanism in tumor migration and invasion is poorly understood. Here, we found that oncoprotein hepatitis B X-interacting protein (HBXIP) blocked the myosin-IIA assemble state promoting breast cancer cell migration. Mechanistically, mass spectrometry analysis, co-immunoprecipitation assay and GST-pull down assay proved that HBXIP directly interacted with the assembly-competent domain (ACD) of non-muscle heavy chain myosin-IIA (NMHC-IIA). The interaction was enhanced by NMHC-IIA S1916 phosphorylation via HBXIP-recruited protein kinase PKCβII. Moreover, HBXIP induced the transcription of PRKCB, encoding PKCβII, by coactivating Sp1, and triggered PKCβII kinase activity. Interestingly, RNA sequencing and mouse metastasis model indicated that the anti-hyperlipidemic drug bezafibrate (BZF) suppressed breast cancer metastasis via inhibiting PKCβII-mediated NMHC-IIA phosphorylation in vitro and in vivo. We reveal a novel mechanism by which HBXIP promotes myosin-IIA disassembly via interacting and phosphorylating NMHC-IIA, and BZF can serve as an effective anti-metastatic drug in breast cancer. Elsevier 2023-03 2022-11-25 /pmc/articles/PMC10031283/ /pubmed/36970214 http://dx.doi.org/10.1016/j.apsb.2022.11.025 Text en © 2022 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting by Elsevier B.V. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Article
Zhang, Lu
Zhou, Xiaolei
Liu, Bowen
Shi, Xuhe
Li, Xianmeng
Xu, Feifei
Fu, Xueli
Wang, Xue
Ye, Kai
Jin, Tianzhi
Sun, Huimin
Li, Qianqian
Zhang, Weiying
Ye, Lihong
HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title_full HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title_fullStr HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title_full_unstemmed HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title_short HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis
title_sort hbxip blocks myosin-iia assembly by phosphorylating and interacting with nmhc-iia in breast cancer metastasis
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10031283/
https://www.ncbi.nlm.nih.gov/pubmed/36970214
http://dx.doi.org/10.1016/j.apsb.2022.11.025
work_keys_str_mv AT zhanglu hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT zhouxiaolei hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT liubowen hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT shixuhe hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT lixianmeng hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT xufeifei hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT fuxueli hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT wangxue hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT yekai hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT jintianzhi hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT sunhuimin hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT liqianqian hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT zhangweiying hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis
AT yelihong hbxipblocksmyosiniiaassemblybyphosphorylatingandinteractingwithnmhciiainbreastcancermetastasis