Cargando…

KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression

BACKGROUND: Histone deacetylases (HDACs) engage in the regulation of various cellular processes by controlling global gene expression. The dysregulation of HDACs leads to carcinogenesis, making HDACs ideal targets for cancer therapy. However, the use of HDAC inhibitors (HDACi) as single agents has b...

Descripción completa

Detalles Bibliográficos
Autores principales: Wei, Haoran, Ma, Wenhao, Lu, Xiaofei, Liu, Haiying, Lin, Kashuai, Wang, Yinghui, Ye, Zijian, Sun, Linchong, Huang, Zhitong, Pan, Tingting, Zhou, Zilong, Cheng, Eric Y., Zhang, Huafeng, Gao, Ping, Zhong, Xiuying
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8441056/
https://www.ncbi.nlm.nih.gov/pubmed/34146461
http://dx.doi.org/10.1002/cac2.12180
_version_ 1783752798218747904
author Wei, Haoran
Ma, Wenhao
Lu, Xiaofei
Liu, Haiying
Lin, Kashuai
Wang, Yinghui
Ye, Zijian
Sun, Linchong
Huang, Zhitong
Pan, Tingting
Zhou, Zilong
Cheng, Eric Y.
Zhang, Huafeng
Gao, Ping
Zhong, Xiuying
author_facet Wei, Haoran
Ma, Wenhao
Lu, Xiaofei
Liu, Haiying
Lin, Kashuai
Wang, Yinghui
Ye, Zijian
Sun, Linchong
Huang, Zhitong
Pan, Tingting
Zhou, Zilong
Cheng, Eric Y.
Zhang, Huafeng
Gao, Ping
Zhong, Xiuying
author_sort Wei, Haoran
collection PubMed
description BACKGROUND: Histone deacetylases (HDACs) engage in the regulation of various cellular processes by controlling global gene expression. The dysregulation of HDACs leads to carcinogenesis, making HDACs ideal targets for cancer therapy. However, the use of HDAC inhibitors (HDACi) as single agents has been shown to have limited success in treating solid tumors in clinical studies. This study aimed to identify a novel downstream effector of HDACs to provide a potential target for combination therapy. METHODS: Transcriptome sequencing and bioinformatics analysis were performed to screen for genes responsive to HDACi in breast cancer cells. The effects of HDACi on cell viability were detected using the MTT assay. The mRNA and protein levels of genes were determined by quantitative reverse transcription‐PCR (qRT‐PCR) and Western blotting. Cell cycle distribution and apoptosis were analyzed by flow cytometry. The binding of CREB1 (cAMP‐response element binding protein 1) to the promoter of the KDELR (The KDEL (Lys‐Asp‐Glu‐Leu) receptor) gene was validated by the ChIP (chromatin immunoprecipitation assay). The association between KDELR2 and protein of centriole 5 (POC5) was detected by immunoprecipitation. A breast cancer‐bearing mouse model was employed to analyze the effect of the HDAC3‐KDELR2 axis on tumor growth. RESULTS: KDELR2 was identified as a novel target of HDAC3, and its aberrant expression indicated the poor prognosis of breast cancer patients. We found a strong correlation between the protein expression patterns of HADC3 and KDELR2 in tumor tissues from breast cancer patients. The results of the ChIP assay and qRT‐PCR analysis validated that HDAC3 transactivated KDELR2 via CREB1. The HDAC3‐KDELR2 axis accelerated the cell cycle progression of cancer cells by protecting the centrosomal protein POC5 from proteasomal degradation. Moreover, the HDAC3‐KDELR2 axis promoted breast cancer cell proliferation and tumorigenesis in vitro and in vivo. CONCLUSION: Our results uncovered a previously unappreciated function of KDELR2 in tumorigenesis, linking a critical Golgi‐the endoplasmic reticulum traffic transport protein to HDAC‐controlled cell cycle progression on the path of cancer development and thus revealing a potential therapeutical target for breast cancer.
format Online
Article
Text
id pubmed-8441056
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-84410562021-09-15 KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression Wei, Haoran Ma, Wenhao Lu, Xiaofei Liu, Haiying Lin, Kashuai Wang, Yinghui Ye, Zijian Sun, Linchong Huang, Zhitong Pan, Tingting Zhou, Zilong Cheng, Eric Y. Zhang, Huafeng Gao, Ping Zhong, Xiuying Cancer Commun (Lond) Original Articles BACKGROUND: Histone deacetylases (HDACs) engage in the regulation of various cellular processes by controlling global gene expression. The dysregulation of HDACs leads to carcinogenesis, making HDACs ideal targets for cancer therapy. However, the use of HDAC inhibitors (HDACi) as single agents has been shown to have limited success in treating solid tumors in clinical studies. This study aimed to identify a novel downstream effector of HDACs to provide a potential target for combination therapy. METHODS: Transcriptome sequencing and bioinformatics analysis were performed to screen for genes responsive to HDACi in breast cancer cells. The effects of HDACi on cell viability were detected using the MTT assay. The mRNA and protein levels of genes were determined by quantitative reverse transcription‐PCR (qRT‐PCR) and Western blotting. Cell cycle distribution and apoptosis were analyzed by flow cytometry. The binding of CREB1 (cAMP‐response element binding protein 1) to the promoter of the KDELR (The KDEL (Lys‐Asp‐Glu‐Leu) receptor) gene was validated by the ChIP (chromatin immunoprecipitation assay). The association between KDELR2 and protein of centriole 5 (POC5) was detected by immunoprecipitation. A breast cancer‐bearing mouse model was employed to analyze the effect of the HDAC3‐KDELR2 axis on tumor growth. RESULTS: KDELR2 was identified as a novel target of HDAC3, and its aberrant expression indicated the poor prognosis of breast cancer patients. We found a strong correlation between the protein expression patterns of HADC3 and KDELR2 in tumor tissues from breast cancer patients. The results of the ChIP assay and qRT‐PCR analysis validated that HDAC3 transactivated KDELR2 via CREB1. The HDAC3‐KDELR2 axis accelerated the cell cycle progression of cancer cells by protecting the centrosomal protein POC5 from proteasomal degradation. Moreover, the HDAC3‐KDELR2 axis promoted breast cancer cell proliferation and tumorigenesis in vitro and in vivo. CONCLUSION: Our results uncovered a previously unappreciated function of KDELR2 in tumorigenesis, linking a critical Golgi‐the endoplasmic reticulum traffic transport protein to HDAC‐controlled cell cycle progression on the path of cancer development and thus revealing a potential therapeutical target for breast cancer. John Wiley and Sons Inc. 2021-06-19 /pmc/articles/PMC8441056/ /pubmed/34146461 http://dx.doi.org/10.1002/cac2.12180 Text en © 2021 The Authors. Cancer Communications published by John Wiley & Sons Australia, Ltd. on behalf of Sun Yat‐sen University Cancer Center https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Original Articles
Wei, Haoran
Ma, Wenhao
Lu, Xiaofei
Liu, Haiying
Lin, Kashuai
Wang, Yinghui
Ye, Zijian
Sun, Linchong
Huang, Zhitong
Pan, Tingting
Zhou, Zilong
Cheng, Eric Y.
Zhang, Huafeng
Gao, Ping
Zhong, Xiuying
KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title_full KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title_fullStr KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title_full_unstemmed KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title_short KDELR2 promotes breast cancer proliferation via HDAC3‐mediated cell cycle progression
title_sort kdelr2 promotes breast cancer proliferation via hdac3‐mediated cell cycle progression
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8441056/
https://www.ncbi.nlm.nih.gov/pubmed/34146461
http://dx.doi.org/10.1002/cac2.12180
work_keys_str_mv AT weihaoran kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT mawenhao kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT luxiaofei kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT liuhaiying kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT linkashuai kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT wangyinghui kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT yezijian kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT sunlinchong kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT huangzhitong kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT pantingting kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT zhouzilong kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT chengericy kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT zhanghuafeng kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT gaoping kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression
AT zhongxiuying kdelr2promotesbreastcancerproliferationviahdac3mediatedcellcycleprogression