Cargando…

Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()

Krüppel-like factor 5 (KLF5) regulates multiple biologic processes. Its function in tumorigenesis appears contradictory though, showing both tumor suppressor and tumor promoting activities. In this study, we examined whether and how Klf5 functions in prostatic tumorigenesis using mice with prostate-...

Descripción completa

Detalles Bibliográficos
Autores principales: Xing, Changsheng, Ci, Xinpei, Sun, Xiaodong, Fu, Xiaoying, Zhang, Zhiqian, Dong, Eric N., Hao, Zhao-Zhe, Dong, Jin-Tang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Neoplasia Press 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4240924/
https://www.ncbi.nlm.nih.gov/pubmed/25425963
http://dx.doi.org/10.1016/j.neo.2014.09.006
_version_ 1782345799690092544
author Xing, Changsheng
Ci, Xinpei
Sun, Xiaodong
Fu, Xiaoying
Zhang, Zhiqian
Dong, Eric N.
Hao, Zhao-Zhe
Dong, Jin-Tang
author_facet Xing, Changsheng
Ci, Xinpei
Sun, Xiaodong
Fu, Xiaoying
Zhang, Zhiqian
Dong, Eric N.
Hao, Zhao-Zhe
Dong, Jin-Tang
author_sort Xing, Changsheng
collection PubMed
description Krüppel-like factor 5 (KLF5) regulates multiple biologic processes. Its function in tumorigenesis appears contradictory though, showing both tumor suppressor and tumor promoting activities. In this study, we examined whether and how Klf5 functions in prostatic tumorigenesis using mice with prostate-specific deletion of Klf5 and phosphatase and tensin homolog (Pten), both of which are frequently inactivated in human prostate cancer. Histologic analysis demonstrated that when one Pten allele was deleted, which causes mouse prostatic intraepithelial neoplasia (mPIN), Klf5 deletion accelerated the emergence and progression of mPIN. When both Pten alleles were deleted, which causes prostate cancer, Klf5 deletion promoted tumor growth, increased cell proliferation, and caused more severe morphologic and molecular alterations. Homozygous deletion of Klf5 was more effective than hemizygous deletion. Unexpectedly, while Pten deletion alone expanded basal cell population in a tumor as reported, Klf5 deletion in the Pten-null background clearly reduced basal cell population while expanding luminal cell population. Global gene expression profiling, pathway analysis, and experimental validation indicate that multiple mechanisms could mediate the tumor-promoting effect of Klf5 deletion, including the up-regulation of epidermal growth factor and its downstream signaling molecules AKT and ERK and the inactivation of the p15 cell cycle inhibitor. KLF5 also appears to cooperate with several transcription factors, including CREB1, Sp1, Myc, ER and AR, to regulate gene expression. These findings validate the tumor suppressor function of KLF5. They also yield a mouse model that shares two common genetic alterations with human prostate cancer—mutation/deletion of Pten and deletion of Klf5.
format Online
Article
Text
id pubmed-4240924
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Neoplasia Press
record_format MEDLINE/PubMed
spelling pubmed-42409242014-11-25 Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()() Xing, Changsheng Ci, Xinpei Sun, Xiaodong Fu, Xiaoying Zhang, Zhiqian Dong, Eric N. Hao, Zhao-Zhe Dong, Jin-Tang Neoplasia Article Krüppel-like factor 5 (KLF5) regulates multiple biologic processes. Its function in tumorigenesis appears contradictory though, showing both tumor suppressor and tumor promoting activities. In this study, we examined whether and how Klf5 functions in prostatic tumorigenesis using mice with prostate-specific deletion of Klf5 and phosphatase and tensin homolog (Pten), both of which are frequently inactivated in human prostate cancer. Histologic analysis demonstrated that when one Pten allele was deleted, which causes mouse prostatic intraepithelial neoplasia (mPIN), Klf5 deletion accelerated the emergence and progression of mPIN. When both Pten alleles were deleted, which causes prostate cancer, Klf5 deletion promoted tumor growth, increased cell proliferation, and caused more severe morphologic and molecular alterations. Homozygous deletion of Klf5 was more effective than hemizygous deletion. Unexpectedly, while Pten deletion alone expanded basal cell population in a tumor as reported, Klf5 deletion in the Pten-null background clearly reduced basal cell population while expanding luminal cell population. Global gene expression profiling, pathway analysis, and experimental validation indicate that multiple mechanisms could mediate the tumor-promoting effect of Klf5 deletion, including the up-regulation of epidermal growth factor and its downstream signaling molecules AKT and ERK and the inactivation of the p15 cell cycle inhibitor. KLF5 also appears to cooperate with several transcription factors, including CREB1, Sp1, Myc, ER and AR, to regulate gene expression. These findings validate the tumor suppressor function of KLF5. They also yield a mouse model that shares two common genetic alterations with human prostate cancer—mutation/deletion of Pten and deletion of Klf5. Neoplasia Press 2014-11-20 /pmc/articles/PMC4240924/ /pubmed/25425963 http://dx.doi.org/10.1016/j.neo.2014.09.006 Text en © 2014 Neoplasia Press, Inc. Published by Elsevier Inc. http://creativecommons.org/licenses/by-nc-nd/3.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
spellingShingle Article
Xing, Changsheng
Ci, Xinpei
Sun, Xiaodong
Fu, Xiaoying
Zhang, Zhiqian
Dong, Eric N.
Hao, Zhao-Zhe
Dong, Jin-Tang
Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title_full Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title_fullStr Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title_full_unstemmed Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title_short Klf5 Deletion Promotes Pten Deletion–Initiated Luminal-Type Mouse Prostate Tumors through Multiple Oncogenic Signaling Pathways()()
title_sort klf5 deletion promotes pten deletion–initiated luminal-type mouse prostate tumors through multiple oncogenic signaling pathways()()
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4240924/
https://www.ncbi.nlm.nih.gov/pubmed/25425963
http://dx.doi.org/10.1016/j.neo.2014.09.006
work_keys_str_mv AT xingchangsheng klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT cixinpei klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT sunxiaodong klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT fuxiaoying klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT zhangzhiqian klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT dongericn klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT haozhaozhe klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways
AT dongjintang klf5deletionpromotesptendeletioninitiatedluminaltypemouseprostatetumorsthroughmultipleoncogenicsignalingpathways