Cargando…

ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA

BACKGROUND: The regulatory roles of human epidermal growth factor receptor [erb-b2 receptor tyrosine kinase 4 (ERBB)] family in tumors was received widespread attention. Although ERBB4 was crucial regulator in metastasis of malignant tumors, the exact mechanism of ERBB4 in inflammatory breast cancer...

Descripción completa

Detalles Bibliográficos
Autores principales: Gong, Ni, Wu, Runliu, Ding, Boni, Wu, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8798440/
https://www.ncbi.nlm.nih.gov/pubmed/35117693
http://dx.doi.org/10.21037/tcr-19-2132
_version_ 1784641807035924480
author Gong, Ni
Wu, Runliu
Ding, Boni
Wu, Wei
author_facet Gong, Ni
Wu, Runliu
Ding, Boni
Wu, Wei
author_sort Gong, Ni
collection PubMed
description BACKGROUND: The regulatory roles of human epidermal growth factor receptor [erb-b2 receptor tyrosine kinase 4 (ERBB)] family in tumors was received widespread attention. Although ERBB4 was crucial regulator in metastasis of malignant tumors, the exact mechanism of ERBB4 in inflammatory breast cancer (IBC) remains unclarified. METHODS: In this study, we collected IBC tissues and cell lines, and explored the expression levels of ERBB4 and platelet-derived growth factor receptor alpha (PDGFRA) using real-time quantitative polymerase chain reaction (RT-PCR), immunohistochemistry (IHC) and western blot assays. Furthermore, cell viability with ERBB4 silencing in SUM149 cells was examined by MTT assay and cell migration and invasion were detected by transwell assay. RESULTS: The data indicated thatERBB4abolishing dramatically depressed capacity of proliferation, migration and invasion of IBC cells. Moreover, PDGFRA was an important factor for the function of ERBB4 and PDGFRA overexpression could, at least, partly rescue the ERBB4 silencing-mediated inhibition in proliferation and metastasis of IBC cells. CONCLUSIONS: Take together, we verified the first time that ERBB4 promoted the progression of IBC through regulating PDGFRA. Thus, inhibition of ERBB4 might be a novel therapeutic candidate against IBC.
format Online
Article
Text
id pubmed-8798440
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-87984402022-02-02 ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA Gong, Ni Wu, Runliu Ding, Boni Wu, Wei Transl Cancer Res Original Article BACKGROUND: The regulatory roles of human epidermal growth factor receptor [erb-b2 receptor tyrosine kinase 4 (ERBB)] family in tumors was received widespread attention. Although ERBB4 was crucial regulator in metastasis of malignant tumors, the exact mechanism of ERBB4 in inflammatory breast cancer (IBC) remains unclarified. METHODS: In this study, we collected IBC tissues and cell lines, and explored the expression levels of ERBB4 and platelet-derived growth factor receptor alpha (PDGFRA) using real-time quantitative polymerase chain reaction (RT-PCR), immunohistochemistry (IHC) and western blot assays. Furthermore, cell viability with ERBB4 silencing in SUM149 cells was examined by MTT assay and cell migration and invasion were detected by transwell assay. RESULTS: The data indicated thatERBB4abolishing dramatically depressed capacity of proliferation, migration and invasion of IBC cells. Moreover, PDGFRA was an important factor for the function of ERBB4 and PDGFRA overexpression could, at least, partly rescue the ERBB4 silencing-mediated inhibition in proliferation and metastasis of IBC cells. CONCLUSIONS: Take together, we verified the first time that ERBB4 promoted the progression of IBC through regulating PDGFRA. Thus, inhibition of ERBB4 might be a novel therapeutic candidate against IBC. AME Publishing Company 2020-05 /pmc/articles/PMC8798440/ /pubmed/35117693 http://dx.doi.org/10.21037/tcr-19-2132 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
Gong, Ni
Wu, Runliu
Ding, Boni
Wu, Wei
ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title_full ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title_fullStr ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title_full_unstemmed ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title_short ERBB4 promotes the progression of inflammatory breast cancer through regulating PDGFRA
title_sort erbb4 promotes the progression of inflammatory breast cancer through regulating pdgfra
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8798440/
https://www.ncbi.nlm.nih.gov/pubmed/35117693
http://dx.doi.org/10.21037/tcr-19-2132
work_keys_str_mv AT gongni erbb4promotestheprogressionofinflammatorybreastcancerthroughregulatingpdgfra
AT wurunliu erbb4promotestheprogressionofinflammatorybreastcancerthroughregulatingpdgfra
AT dingboni erbb4promotestheprogressionofinflammatorybreastcancerthroughregulatingpdgfra
AT wuwei erbb4promotestheprogressionofinflammatorybreastcancerthroughregulatingpdgfra