Cargando…

FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation

BACKGROUND: Increasing evidence suggests that FBXW7 has a high frequency of mutations in esophageal squamous cell carcinoma (ESCC). However, the function of FBXW7, especially the mutations, is not clear. This study was designed to investigate the functional significance of FBXW7 loss of function and...

Descripción completa

Detalles Bibliográficos
Autores principales: Pan, Yunzhi, Liu, Jing, Gao, Yingyin, Guo, Yuqing, Wang, Changxing, Liang, Zhipan, Wu, Meiying, Qian, Yulan, Li, Yinyan, Shen, Jingyi, Lu, Chenchen, Ma, Sai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10054043/
https://www.ncbi.nlm.nih.gov/pubmed/36991467
http://dx.doi.org/10.1186/s13046-023-02630-3
_version_ 1785015558002966528
author Pan, Yunzhi
Liu, Jing
Gao, Yingyin
Guo, Yuqing
Wang, Changxing
Liang, Zhipan
Wu, Meiying
Qian, Yulan
Li, Yinyan
Shen, Jingyi
Lu, Chenchen
Ma, Sai
author_facet Pan, Yunzhi
Liu, Jing
Gao, Yingyin
Guo, Yuqing
Wang, Changxing
Liang, Zhipan
Wu, Meiying
Qian, Yulan
Li, Yinyan
Shen, Jingyi
Lu, Chenchen
Ma, Sai
author_sort Pan, Yunzhi
collection PubMed
description BACKGROUND: Increasing evidence suggests that FBXW7 has a high frequency of mutations in esophageal squamous cell carcinoma (ESCC). However, the function of FBXW7, especially the mutations, is not clear. This study was designed to investigate the functional significance of FBXW7 loss of function and underlying mechanism in ESCC. METHODS: Immunofluorescence was applied to clarify the localization and main isoform of FBXW7 in ESCC cells. Sanger sequencing were performed to explore mutations of FBXW7 in ESCC tissues. Proliferation, colony, invasion and migration assays were performed to examine the functional roles of FBXW7 in ESCC cells in vitro and in vivo. Real-time RT-PCR, immunoblotting, GST-pulldown, LC–MS/MS and co-immunoprecipitation assay were used to explore the molecular mechanism underlying the actions of FBXW7 functional inactivation in ESCC cells. Immunohistochemical staining were used to explore the expression of FBXW7 and MAP4 in ESCC tissues. RESULTS: The main FBXW7 isoform in ESCC cells was the β transcript in the cytoplasm. Functional inactivation of FBXW7 led to activation of the MAPK signaling pathway and upregulation of the downstream MMP3 and VEGFA, which enhanced tumor proliferation cell invasion and migration. Among the five mutation forms screened, S327X (X means truncated mutation) had an effect similar to the FBXW7 deficiency and led to the inactivation of FBXW7 in ESCC cells. Three other point mutations, S382F, D400N and R425C, attenuated but did not eliminate FBXW7 function. The other truncating mutation, S598X, which was located outside of the WD40 domain, revealed a tiny attenuation of FBXW7 in ESCC cells. Notably, MAP4 was identified as a potential target of FBXW7. The threonine T521 of MAP4, which was phosphorylated by CHEK1, played a key role in the FBXW7-related degradation system. Immunohistochemical staining indicated that FBXW7 loss of function was associated with tumor stage and shorter survival of patients with ESCC. Univariate and multivariate Cox proportional hazards regression analyses showed that high FBXW7 and low MAP4 was an independent prognostic indicator and prospective longer survival. Moreover, a combination regimen that included MK-8353 to inhibit the phosphorylation of ERK and bevacizumab to inhibit VEGFA produced potent inhibitory effects on the growth of FBXW7 inactivation xenograft tumors in vivo. CONCLUSIONS: This study provided evidence that FBXW7 loss of function promoted ESCC via MAP4 overexpression and ERK phosphorylation, and this novel FBXW7/MAP4/ERK axis may be an efficient target for ESCC treatment. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02630-3.
format Online
Article
Text
id pubmed-10054043
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100540432023-03-30 FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation Pan, Yunzhi Liu, Jing Gao, Yingyin Guo, Yuqing Wang, Changxing Liang, Zhipan Wu, Meiying Qian, Yulan Li, Yinyan Shen, Jingyi Lu, Chenchen Ma, Sai J Exp Clin Cancer Res Research BACKGROUND: Increasing evidence suggests that FBXW7 has a high frequency of mutations in esophageal squamous cell carcinoma (ESCC). However, the function of FBXW7, especially the mutations, is not clear. This study was designed to investigate the functional significance of FBXW7 loss of function and underlying mechanism in ESCC. METHODS: Immunofluorescence was applied to clarify the localization and main isoform of FBXW7 in ESCC cells. Sanger sequencing were performed to explore mutations of FBXW7 in ESCC tissues. Proliferation, colony, invasion and migration assays were performed to examine the functional roles of FBXW7 in ESCC cells in vitro and in vivo. Real-time RT-PCR, immunoblotting, GST-pulldown, LC–MS/MS and co-immunoprecipitation assay were used to explore the molecular mechanism underlying the actions of FBXW7 functional inactivation in ESCC cells. Immunohistochemical staining were used to explore the expression of FBXW7 and MAP4 in ESCC tissues. RESULTS: The main FBXW7 isoform in ESCC cells was the β transcript in the cytoplasm. Functional inactivation of FBXW7 led to activation of the MAPK signaling pathway and upregulation of the downstream MMP3 and VEGFA, which enhanced tumor proliferation cell invasion and migration. Among the five mutation forms screened, S327X (X means truncated mutation) had an effect similar to the FBXW7 deficiency and led to the inactivation of FBXW7 in ESCC cells. Three other point mutations, S382F, D400N and R425C, attenuated but did not eliminate FBXW7 function. The other truncating mutation, S598X, which was located outside of the WD40 domain, revealed a tiny attenuation of FBXW7 in ESCC cells. Notably, MAP4 was identified as a potential target of FBXW7. The threonine T521 of MAP4, which was phosphorylated by CHEK1, played a key role in the FBXW7-related degradation system. Immunohistochemical staining indicated that FBXW7 loss of function was associated with tumor stage and shorter survival of patients with ESCC. Univariate and multivariate Cox proportional hazards regression analyses showed that high FBXW7 and low MAP4 was an independent prognostic indicator and prospective longer survival. Moreover, a combination regimen that included MK-8353 to inhibit the phosphorylation of ERK and bevacizumab to inhibit VEGFA produced potent inhibitory effects on the growth of FBXW7 inactivation xenograft tumors in vivo. CONCLUSIONS: This study provided evidence that FBXW7 loss of function promoted ESCC via MAP4 overexpression and ERK phosphorylation, and this novel FBXW7/MAP4/ERK axis may be an efficient target for ESCC treatment. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02630-3. BioMed Central 2023-03-29 /pmc/articles/PMC10054043/ /pubmed/36991467 http://dx.doi.org/10.1186/s13046-023-02630-3 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Pan, Yunzhi
Liu, Jing
Gao, Yingyin
Guo, Yuqing
Wang, Changxing
Liang, Zhipan
Wu, Meiying
Qian, Yulan
Li, Yinyan
Shen, Jingyi
Lu, Chenchen
Ma, Sai
FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title_full FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title_fullStr FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title_full_unstemmed FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title_short FBXW7 loss of function promotes esophageal squamous cell carcinoma progression via elevating MAP4 and ERK phosphorylation
title_sort fbxw7 loss of function promotes esophageal squamous cell carcinoma progression via elevating map4 and erk phosphorylation
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10054043/
https://www.ncbi.nlm.nih.gov/pubmed/36991467
http://dx.doi.org/10.1186/s13046-023-02630-3
work_keys_str_mv AT panyunzhi fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT liujing fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT gaoyingyin fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT guoyuqing fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT wangchangxing fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT liangzhipan fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT wumeiying fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT qianyulan fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT liyinyan fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT shenjingyi fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT luchenchen fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation
AT masai fbxw7lossoffunctionpromotesesophagealsquamouscellcarcinomaprogressionviaelevatingmap4anderkphosphorylation