Cargando…

ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65

The NF-κB is found in almost all animal cell types and is involved in a myriad of cellular responses. Aberrant expression of NF-κB has been linked to cancer, inflammatory diseases and improper development. Little is known about transcriptional regulation of the NF-κB family member gene RelA/p65. Sp1...

Descripción completa

Detalles Bibliográficos
Autores principales: Kim, Min-Young, Koh, Dong-In, Choi, Won-Il, Jeon, Bu-Nam, Jeong, Deok-yoon, Kim, Kyung-Sup, Kim, Kunhong, Kim, Se-Hoon, Hur, Man-Wook
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4330387/
https://www.ncbi.nlm.nih.gov/pubmed/25609694
http://dx.doi.org/10.1093/nar/gkv026
_version_ 1782357578542481408
author Kim, Min-Young
Koh, Dong-In
Choi, Won-Il
Jeon, Bu-Nam
Jeong, Deok-yoon
Kim, Kyung-Sup
Kim, Kunhong
Kim, Se-Hoon
Hur, Man-Wook
author_facet Kim, Min-Young
Koh, Dong-In
Choi, Won-Il
Jeon, Bu-Nam
Jeong, Deok-yoon
Kim, Kyung-Sup
Kim, Kunhong
Kim, Se-Hoon
Hur, Man-Wook
author_sort Kim, Min-Young
collection PubMed
description The NF-κB is found in almost all animal cell types and is involved in a myriad of cellular responses. Aberrant expression of NF-κB has been linked to cancer, inflammatory diseases and improper development. Little is known about transcriptional regulation of the NF-κB family member gene RelA/p65. Sp1 plays a key role in the expression of the RelA/p65 gene. ZBTB2 represses transcription of the gene by inhibiting Sp1 binding to a Sp1-binding GC-box in the RelA/p65 proximal promoter (bp, −31 to −21). Moreover, recent studies revealed that RelA/p65 directly binds to the peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α) to decrease transcriptional activation of the PGC1α target gene PDK4, whose gene product inhibits pyruvate dehydrogenase (PDH), a key regulator of TCA cycle flux. Accordingly, we observed that RelA/p65 repression by ZBTB2 indirectly results in increased PDK4 expression, which inhibits PDH. Consequently, in cells with ectopic ZBTB2, the concentrations of pyruvate and lactate were higher than those in normal cells, indicating changes in glucose metabolism flux favoring glycolysis over the TCA cycle. Knockdown of ZBTB2 in mouse xenografts decreased tumor growth. ZBTB2 may increase cell proliferation by reprogramming glucose metabolic pathways to favor glycolysis by upregulating PDK4 expression via repression of RelA/p65 expression.
format Online
Article
Text
id pubmed-4330387
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-43303872015-03-18 ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65 Kim, Min-Young Koh, Dong-In Choi, Won-Il Jeon, Bu-Nam Jeong, Deok-yoon Kim, Kyung-Sup Kim, Kunhong Kim, Se-Hoon Hur, Man-Wook Nucleic Acids Res Gene regulation, Chromatin and Epigenetics The NF-κB is found in almost all animal cell types and is involved in a myriad of cellular responses. Aberrant expression of NF-κB has been linked to cancer, inflammatory diseases and improper development. Little is known about transcriptional regulation of the NF-κB family member gene RelA/p65. Sp1 plays a key role in the expression of the RelA/p65 gene. ZBTB2 represses transcription of the gene by inhibiting Sp1 binding to a Sp1-binding GC-box in the RelA/p65 proximal promoter (bp, −31 to −21). Moreover, recent studies revealed that RelA/p65 directly binds to the peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α) to decrease transcriptional activation of the PGC1α target gene PDK4, whose gene product inhibits pyruvate dehydrogenase (PDH), a key regulator of TCA cycle flux. Accordingly, we observed that RelA/p65 repression by ZBTB2 indirectly results in increased PDK4 expression, which inhibits PDH. Consequently, in cells with ectopic ZBTB2, the concentrations of pyruvate and lactate were higher than those in normal cells, indicating changes in glucose metabolism flux favoring glycolysis over the TCA cycle. Knockdown of ZBTB2 in mouse xenografts decreased tumor growth. ZBTB2 may increase cell proliferation by reprogramming glucose metabolic pathways to favor glycolysis by upregulating PDK4 expression via repression of RelA/p65 expression. Oxford University Press 2015-02-18 2015-01-21 /pmc/articles/PMC4330387/ /pubmed/25609694 http://dx.doi.org/10.1093/nar/gkv026 Text en © The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research. http://creativecommons.org/licenses/by-nc/4.0/ This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
spellingShingle Gene regulation, Chromatin and Epigenetics
Kim, Min-Young
Koh, Dong-In
Choi, Won-Il
Jeon, Bu-Nam
Jeong, Deok-yoon
Kim, Kyung-Sup
Kim, Kunhong
Kim, Se-Hoon
Hur, Man-Wook
ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title_full ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title_fullStr ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title_full_unstemmed ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title_short ZBTB2 increases PDK4 expression by transcriptional repression of RelA/p65
title_sort zbtb2 increases pdk4 expression by transcriptional repression of rela/p65
topic Gene regulation, Chromatin and Epigenetics
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4330387/
https://www.ncbi.nlm.nih.gov/pubmed/25609694
http://dx.doi.org/10.1093/nar/gkv026
work_keys_str_mv AT kimminyoung zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT kohdongin zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT choiwonil zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT jeonbunam zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT jeongdeokyoon zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT kimkyungsup zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT kimkunhong zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT kimsehoon zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65
AT hurmanwook zbtb2increasespdk4expressionbytranscriptionalrepressionofrelap65