A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response

Abstract p53-mediated cell cycle arrest during DNA damage is dependent on the induction of p21 protein, encoded by the CDKN1A gene. p21 inhibits cyclin-dependent kinases required for cell cycle progression to guarantee accurate repair of DNA lesions. Hence, fine-tuning of p21 levels is crucial to pr...

Full description

Bibliographic Details
Main Authors: Bella M. Ben-Oz, Feras E. Machour, Marian Nicola, Amir Argoetti, Galia Polyak, Rawad Hanna, Oded Kleifeld, Yael Mandel-Gutfreund, Nabieh Ayoub
Format: Article
Language:English
Published: Nature Portfolio 2023-11-01
Series:Nature Communications
Online Access:https://doi.org/10.1038/s41467-023-43495-6
_version_ 1797452101074288640
author Bella M. Ben-Oz
Feras E. Machour
Marian Nicola
Amir Argoetti
Galia Polyak
Rawad Hanna
Oded Kleifeld
Yael Mandel-Gutfreund
Nabieh Ayoub
author_facet Bella M. Ben-Oz
Feras E. Machour
Marian Nicola
Amir Argoetti
Galia Polyak
Rawad Hanna
Oded Kleifeld
Yael Mandel-Gutfreund
Nabieh Ayoub
author_sort Bella M. Ben-Oz
collection DOAJ
description Abstract p53-mediated cell cycle arrest during DNA damage is dependent on the induction of p21 protein, encoded by the CDKN1A gene. p21 inhibits cyclin-dependent kinases required for cell cycle progression to guarantee accurate repair of DNA lesions. Hence, fine-tuning of p21 levels is crucial to preserve genomic stability. Currently, the multilayered regulation of p21 levels during DNA damage is not fully understood. Herein, we identify the human RNA binding motif protein 42 (RBM42) as a regulator of p21 levels during DNA damage. Genome-wide transcriptome and interactome analysis reveals that RBM42 alters the expression of p53-regulated genes during DNA damage. Specifically, we demonstrate that RBM42 facilitates CDKN1A splicing by counteracting the splicing inhibitory effect of RBM4 protein. Unexpectedly, we also show that RBM42, underpins translation of various splicing targets, including CDKN1A. Concordantly, transcriptome-wide mapping of RBM42-RNA interactions using eCLIP further substantiates the dual function of RBM42 in regulating splicing and translation of its target genes, including CDKN1A. Collectively, our data show that RBM42 couples splicing and translation machineries to fine-tune gene expression during DNA damage response.
first_indexed 2024-03-09T15:03:54Z
format Article
id doaj.art-5d2786df41464164a9f134fb00067f8c
institution Directory Open Access Journal
issn 2041-1723
language English
last_indexed 2024-03-09T15:03:54Z
publishDate 2023-11-01
publisher Nature Portfolio
record_format Article
series Nature Communications
spelling doaj.art-5d2786df41464164a9f134fb00067f8c2023-11-26T13:46:07ZengNature PortfolioNature Communications2041-17232023-11-0114111810.1038/s41467-023-43495-6A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage responseBella M. Ben-Oz0Feras E. Machour1Marian Nicola2Amir Argoetti3Galia Polyak4Rawad Hanna5Oded Kleifeld6Yael Mandel-Gutfreund7Nabieh Ayoub8Department of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyDepartment of Biology, Technion - Israel Institute of TechnologyAbstract p53-mediated cell cycle arrest during DNA damage is dependent on the induction of p21 protein, encoded by the CDKN1A gene. p21 inhibits cyclin-dependent kinases required for cell cycle progression to guarantee accurate repair of DNA lesions. Hence, fine-tuning of p21 levels is crucial to preserve genomic stability. Currently, the multilayered regulation of p21 levels during DNA damage is not fully understood. Herein, we identify the human RNA binding motif protein 42 (RBM42) as a regulator of p21 levels during DNA damage. Genome-wide transcriptome and interactome analysis reveals that RBM42 alters the expression of p53-regulated genes during DNA damage. Specifically, we demonstrate that RBM42 facilitates CDKN1A splicing by counteracting the splicing inhibitory effect of RBM4 protein. Unexpectedly, we also show that RBM42, underpins translation of various splicing targets, including CDKN1A. Concordantly, transcriptome-wide mapping of RBM42-RNA interactions using eCLIP further substantiates the dual function of RBM42 in regulating splicing and translation of its target genes, including CDKN1A. Collectively, our data show that RBM42 couples splicing and translation machineries to fine-tune gene expression during DNA damage response.https://doi.org/10.1038/s41467-023-43495-6
spellingShingle Bella M. Ben-Oz
Feras E. Machour
Marian Nicola
Amir Argoetti
Galia Polyak
Rawad Hanna
Oded Kleifeld
Yael Mandel-Gutfreund
Nabieh Ayoub
A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
Nature Communications
title A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
title_full A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
title_fullStr A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
title_full_unstemmed A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
title_short A dual role of RBM42 in modulating splicing and translation of CDKN1A/p21 during DNA damage response
title_sort dual role of rbm42 in modulating splicing and translation of cdkn1a p21 during dna damage response
url https://doi.org/10.1038/s41467-023-43495-6
work_keys_str_mv AT bellambenoz adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT ferasemachour adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT mariannicola adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT amirargoetti adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT galiapolyak adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT rawadhanna adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT odedkleifeld adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT yaelmandelgutfreund adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT nabiehayoub adualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT bellambenoz dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT ferasemachour dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT mariannicola dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT amirargoetti dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT galiapolyak dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT rawadhanna dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT odedkleifeld dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT yaelmandelgutfreund dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse
AT nabiehayoub dualroleofrbm42inmodulatingsplicingandtranslationofcdkn1ap21duringdnadamageresponse