Cargando…

Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling

Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the...

Descripción completa

Detalles Bibliográficos
Autores principales: Lin, Chuwen, Yao, Erica, Wang, Kevin, Nozawa, Yoko, Shimizu, Hirohito, Johnson, Jeffrey R., Chen, Jau-Nian, Krogan, Nevan J., Chuang, Pao-Tien
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory Press 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4233246/
https://www.ncbi.nlm.nih.gov/pubmed/25403183
http://dx.doi.org/10.1101/gad.249425.114
_version_ 1782344701171466240
author Lin, Chuwen
Yao, Erica
Wang, Kevin
Nozawa, Yoko
Shimizu, Hirohito
Johnson, Jeffrey R.
Chen, Jau-Nian
Krogan, Nevan J.
Chuang, Pao-Tien
author_facet Lin, Chuwen
Yao, Erica
Wang, Kevin
Nozawa, Yoko
Shimizu, Hirohito
Johnson, Jeffrey R.
Chen, Jau-Nian
Krogan, Nevan J.
Chuang, Pao-Tien
author_sort Lin, Chuwen
collection PubMed
description Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the nucleus, highlighting critical functions of Sufu in the nucleus. Through a proteomic approach, we identified several Sufu-interacting proteins, including p66β (a member of the NuRD [nucleosome remodeling and histone deacetylase] repressor complex) and Mycbp (a Myc-binding protein). p66β negatively and Mycbp positively regulate Hh signaling in cell-based assays and zebrafish. They function downstream from the membrane receptors, Patched and Smoothened, and the primary cilium. Sufu, p66β, Mycbp, and Gli are also detected on the promoters of Hh targets in a dynamic manner. Our results support a new model of Hh signaling in the nucleus. Sufu recruits p66β to block Gli-mediated Hh target gene expression. Meanwhile, Mycbp forms a complex with Gli and Sufu without Hh stimulation but remains inactive. Hh pathway activation leads to dissociation of Sufu/p66β from Gli, enabling Mycbp to promote Gli protein activity and Hh target gene expression. These studies provide novel insight into how Sufu controls Hh signaling in the nucleus.
format Online
Article
Text
id pubmed-4233246
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Cold Spring Harbor Laboratory Press
record_format MEDLINE/PubMed
spelling pubmed-42332462015-05-15 Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling Lin, Chuwen Yao, Erica Wang, Kevin Nozawa, Yoko Shimizu, Hirohito Johnson, Jeffrey R. Chen, Jau-Nian Krogan, Nevan J. Chuang, Pao-Tien Genes Dev Research Paper Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the nucleus, highlighting critical functions of Sufu in the nucleus. Through a proteomic approach, we identified several Sufu-interacting proteins, including p66β (a member of the NuRD [nucleosome remodeling and histone deacetylase] repressor complex) and Mycbp (a Myc-binding protein). p66β negatively and Mycbp positively regulate Hh signaling in cell-based assays and zebrafish. They function downstream from the membrane receptors, Patched and Smoothened, and the primary cilium. Sufu, p66β, Mycbp, and Gli are also detected on the promoters of Hh targets in a dynamic manner. Our results support a new model of Hh signaling in the nucleus. Sufu recruits p66β to block Gli-mediated Hh target gene expression. Meanwhile, Mycbp forms a complex with Gli and Sufu without Hh stimulation but remains inactive. Hh pathway activation leads to dissociation of Sufu/p66β from Gli, enabling Mycbp to promote Gli protein activity and Hh target gene expression. These studies provide novel insight into how Sufu controls Hh signaling in the nucleus. Cold Spring Harbor Laboratory Press 2014-11-15 /pmc/articles/PMC4233246/ /pubmed/25403183 http://dx.doi.org/10.1101/gad.249425.114 Text en © 2014 Lin et al.; Published by Cold Spring Harbor Laboratory Press http://creativecommons.org/licenses/by-nc/4.0/ This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.
spellingShingle Research Paper
Lin, Chuwen
Yao, Erica
Wang, Kevin
Nozawa, Yoko
Shimizu, Hirohito
Johnson, Jeffrey R.
Chen, Jau-Nian
Krogan, Nevan J.
Chuang, Pao-Tien
Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title_full Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title_fullStr Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title_full_unstemmed Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title_short Regulation of Sufu activity by p66β and Mycbp provides new insight into vertebrate Hedgehog signaling
title_sort regulation of sufu activity by p66β and mycbp provides new insight into vertebrate hedgehog signaling
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4233246/
https://www.ncbi.nlm.nih.gov/pubmed/25403183
http://dx.doi.org/10.1101/gad.249425.114
work_keys_str_mv AT linchuwen regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT yaoerica regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT wangkevin regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT nozawayoko regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT shimizuhirohito regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT johnsonjeffreyr regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT chenjaunian regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT krogannevanj regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling
AT chuangpaotien regulationofsufuactivitybyp66bandmycbpprovidesnewinsightintovertebratehedgehogsignaling