Cargando…

Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation

In cancer, many genes are mutated by genome rearrangement, but our understanding of the functional consequences of this remains rudimentary. Here we report the F-box protein encoded by FBXL17 is disrupted in the region of the gene that encodes its substrate-binding leucine rich repeat (LRR) domain....

Descripción completa

Detalles Bibliográficos
Autores principales: Mason, Bethany, Flach, Susanne, Teixeira, Felipe R., Manzano Garcia, Raquel, Rueda, Oscar M., Abraham, Jean E., Caldas, Carlos, Edwards, Paul A. W., Laman, Heike
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer International Publishing 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7320043/
https://www.ncbi.nlm.nih.gov/pubmed/31560077
http://dx.doi.org/10.1007/s00018-019-03306-y
_version_ 1783551168027295744
author Mason, Bethany
Flach, Susanne
Teixeira, Felipe R.
Manzano Garcia, Raquel
Rueda, Oscar M.
Abraham, Jean E.
Caldas, Carlos
Edwards, Paul A. W.
Laman, Heike
author_facet Mason, Bethany
Flach, Susanne
Teixeira, Felipe R.
Manzano Garcia, Raquel
Rueda, Oscar M.
Abraham, Jean E.
Caldas, Carlos
Edwards, Paul A. W.
Laman, Heike
author_sort Mason, Bethany
collection PubMed
description In cancer, many genes are mutated by genome rearrangement, but our understanding of the functional consequences of this remains rudimentary. Here we report the F-box protein encoded by FBXL17 is disrupted in the region of the gene that encodes its substrate-binding leucine rich repeat (LRR) domain. Truncating Fbxl17 LRRs impaired its association with the other SCF holoenzyme subunits Skp1, Cul1 and Rbx1, and decreased ubiquitination activity. Loss of the LRRs also differentially affected Fbxl17 binding to its targets. Thus, genomic rearrangements in FBXL17 are likely to disrupt SCF(Fbxl17)-regulated networks in cancer cells. To investigate the functional effect of these rearrangements, we performed a yeast two-hybrid screen to identify Fbxl17-interacting proteins. Among the 37 binding partners Uap1, an enzyme involved in O-GlcNAcylation of proteins was identified most frequently. We demonstrate that Fbxl17 binds to UAP1 directly and inhibits its phosphorylation, which we propose regulates UAP1 activity. Knockdown of Fbxl17 expression elevated O-GlcNAcylation in breast cancer cells, arguing for a functional role for Fbxl17 in this metabolic pathway. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s00018-019-03306-y) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-7320043
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Springer International Publishing
record_format MEDLINE/PubMed
spelling pubmed-73200432020-07-01 Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation Mason, Bethany Flach, Susanne Teixeira, Felipe R. Manzano Garcia, Raquel Rueda, Oscar M. Abraham, Jean E. Caldas, Carlos Edwards, Paul A. W. Laman, Heike Cell Mol Life Sci Original Article In cancer, many genes are mutated by genome rearrangement, but our understanding of the functional consequences of this remains rudimentary. Here we report the F-box protein encoded by FBXL17 is disrupted in the region of the gene that encodes its substrate-binding leucine rich repeat (LRR) domain. Truncating Fbxl17 LRRs impaired its association with the other SCF holoenzyme subunits Skp1, Cul1 and Rbx1, and decreased ubiquitination activity. Loss of the LRRs also differentially affected Fbxl17 binding to its targets. Thus, genomic rearrangements in FBXL17 are likely to disrupt SCF(Fbxl17)-regulated networks in cancer cells. To investigate the functional effect of these rearrangements, we performed a yeast two-hybrid screen to identify Fbxl17-interacting proteins. Among the 37 binding partners Uap1, an enzyme involved in O-GlcNAcylation of proteins was identified most frequently. We demonstrate that Fbxl17 binds to UAP1 directly and inhibits its phosphorylation, which we propose regulates UAP1 activity. Knockdown of Fbxl17 expression elevated O-GlcNAcylation in breast cancer cells, arguing for a functional role for Fbxl17 in this metabolic pathway. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s00018-019-03306-y) contains supplementary material, which is available to authorized users. Springer International Publishing 2019-09-27 2020 /pmc/articles/PMC7320043/ /pubmed/31560077 http://dx.doi.org/10.1007/s00018-019-03306-y Text en © The Author(s) 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
spellingShingle Original Article
Mason, Bethany
Flach, Susanne
Teixeira, Felipe R.
Manzano Garcia, Raquel
Rueda, Oscar M.
Abraham, Jean E.
Caldas, Carlos
Edwards, Paul A. W.
Laman, Heike
Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title_full Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title_fullStr Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title_full_unstemmed Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title_short Fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global O-GlcNAcylation
title_sort fbxl17 is rearranged in breast cancer and loss of its activity leads to increased global o-glcnacylation
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7320043/
https://www.ncbi.nlm.nih.gov/pubmed/31560077
http://dx.doi.org/10.1007/s00018-019-03306-y
work_keys_str_mv AT masonbethany fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT flachsusanne fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT teixeirafeliper fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT manzanogarciaraquel fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT ruedaoscarm fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT abrahamjeane fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT caldascarlos fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT edwardspaulaw fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation
AT lamanheike fbxl17isrearrangedinbreastcancerandlossofitsactivityleadstoincreasedglobaloglcnacylation