Cargando…

Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation

Small nuclear RNAs (snRNAs) are structural and functional cores of the spliceosome. In metazoan genomes, each snRNA has multiple copies/variants, up to hundreds in mammals. However, the expressions and functions of each copy/variant in one organism have not been systematically studied. Focus on U1 s...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Mei, Liang, An-Min, Zhou, Zhen-Zhen, Pang, Ting-Lin, Fan, Yu-Jie, Xu, Yong-Zhen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10645366/
https://www.ncbi.nlm.nih.gov/pubmed/37917726
http://dx.doi.org/10.1371/journal.pgen.1011021
_version_ 1785134738541903872
author Wang, Mei
Liang, An-Min
Zhou, Zhen-Zhen
Pang, Ting-Lin
Fan, Yu-Jie
Xu, Yong-Zhen
author_facet Wang, Mei
Liang, An-Min
Zhou, Zhen-Zhen
Pang, Ting-Lin
Fan, Yu-Jie
Xu, Yong-Zhen
author_sort Wang, Mei
collection PubMed
description Small nuclear RNAs (snRNAs) are structural and functional cores of the spliceosome. In metazoan genomes, each snRNA has multiple copies/variants, up to hundreds in mammals. However, the expressions and functions of each copy/variant in one organism have not been systematically studied. Focus on U1 snRNA genes, we investigated all five copies in Drosophila melanogaster using two series of constructed strains. Analyses of transgenic flies that each have a U1 promoter-driven gfp revealed that U1:21D is the major and ubiquitously expressed copy, and the other four copies have specificities in developmental stages and tissues. Mutant strains that each have a precisely deleted copy of U1-gene exhibited various extents of defects in fly morphology or mobility, especially deletion of U1:82Eb. Interestingly, splicing was changed at limited levels in the deletion strains, while large amounts of differentially-expressed genes and alternative polyadenylation events were identified, showing preferences in the down-regulation of genes with 1–2 introns and selection of proximal sites for 3’-end polyadenylation. In vitro assays suggested that Drosophila U1 variants pulled down fewer SmD2 proteins compared to the canonical U1. This study demonstrates that all five U1-genes in Drosophila have physiological functions in development and play regulatory roles in transcription and 3’-end formation.
format Online
Article
Text
id pubmed-10645366
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-106453662023-11-02 Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation Wang, Mei Liang, An-Min Zhou, Zhen-Zhen Pang, Ting-Lin Fan, Yu-Jie Xu, Yong-Zhen PLoS Genet Research Article Small nuclear RNAs (snRNAs) are structural and functional cores of the spliceosome. In metazoan genomes, each snRNA has multiple copies/variants, up to hundreds in mammals. However, the expressions and functions of each copy/variant in one organism have not been systematically studied. Focus on U1 snRNA genes, we investigated all five copies in Drosophila melanogaster using two series of constructed strains. Analyses of transgenic flies that each have a U1 promoter-driven gfp revealed that U1:21D is the major and ubiquitously expressed copy, and the other four copies have specificities in developmental stages and tissues. Mutant strains that each have a precisely deleted copy of U1-gene exhibited various extents of defects in fly morphology or mobility, especially deletion of U1:82Eb. Interestingly, splicing was changed at limited levels in the deletion strains, while large amounts of differentially-expressed genes and alternative polyadenylation events were identified, showing preferences in the down-regulation of genes with 1–2 introns and selection of proximal sites for 3’-end polyadenylation. In vitro assays suggested that Drosophila U1 variants pulled down fewer SmD2 proteins compared to the canonical U1. This study demonstrates that all five U1-genes in Drosophila have physiological functions in development and play regulatory roles in transcription and 3’-end formation. Public Library of Science 2023-11-02 /pmc/articles/PMC10645366/ /pubmed/37917726 http://dx.doi.org/10.1371/journal.pgen.1011021 Text en © 2023 Wang et al https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Wang, Mei
Liang, An-Min
Zhou, Zhen-Zhen
Pang, Ting-Lin
Fan, Yu-Jie
Xu, Yong-Zhen
Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title_full Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title_fullStr Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title_full_unstemmed Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title_short Deletions of singular U1 snRNA gene significantly interfere with transcription and 3’-end mRNA formation
title_sort deletions of singular u1 snrna gene significantly interfere with transcription and 3’-end mrna formation
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10645366/
https://www.ncbi.nlm.nih.gov/pubmed/37917726
http://dx.doi.org/10.1371/journal.pgen.1011021
work_keys_str_mv AT wangmei deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation
AT lianganmin deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation
AT zhouzhenzhen deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation
AT pangtinglin deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation
AT fanyujie deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation
AT xuyongzhen deletionsofsingularu1snrnagenesignificantlyinterferewithtranscriptionand3endmrnaformation