Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Cell ; 76(4): 590-599.e4, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31522989

RESUMO

Full-length transcription in the majority of human genes depends on U1 snRNP (U1) to co-transcriptionally suppress transcription-terminating premature 3' end cleavage and polyadenylation (PCPA) from cryptic polyadenylation signals (PASs) in introns. However, the mechanism of this U1 activity, termed telescripting, is unknown. Here, we captured a complex, comprising U1 and CPA factors (U1-CPAFs), that binds intronic PASs and suppresses PCPA. U1-CPAFs are distinct from U1-spliceosomal complexes; they include CPA's three main subunits, CFIm, CPSF, and CstF; lack essential splicing factors; and associate with transcription elongation and mRNA export complexes. Telescripting requires U1:pre-mRNA base-pairing, which can be disrupted by U1 antisense oligonucleotide (U1 AMO), triggering PCPA. U1 AMO remodels U1-CPAFs, revealing changes, including recruitment of CPA-stimulating factors, that explain U1-CPAFs' switch from repressive to activated states. Our findings outline this U1 telescripting mechanism and demonstrate U1's unique role as central regulator of pre-mRNA processing and transcription.


Assuntos
Núcleo Celular/metabolismo , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Clivagem do RNA , Precursores de RNA/biossíntese , RNA Mensageiro/biossíntese , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Transcrição Gênica , Regiões 3' não Traduzidas , Transporte Ativo do Núcleo Celular , Sítios de Ligação , Núcleo Celular/genética , Fator de Especificidade de Clivagem e Poliadenilação/genética , Fator Estimulador de Clivagem/genética , Fator Estimulador de Clivagem/metabolismo , Células HeLa , Humanos , Complexos Multiproteicos , Poli A/metabolismo , Ligação Proteica , Precursores de RNA/genética , RNA Mensageiro/genética , Ribonucleoproteína Nuclear Pequena U1/genética
2.
Cell ; 146(3): 384-95, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816274

RESUMO

The SMN complex mediates the assembly of heptameric Sm protein rings on small nuclear RNAs (snRNAs), which are essential for snRNP function. Specific Sm core assembly depends on Sm proteins and snRNA recognition by SMN/Gemin2- and Gemin5-containing subunits, respectively. The mechanism by which the Sm proteins are gathered while preventing illicit Sm assembly on non-snRNAs is unknown. Here, we describe the 2.5 Å crystal structure of Gemin2 bound to SmD1/D2/F/E/G pentamer and SMN's Gemin2-binding domain, a key assembly intermediate. Remarkably, through its extended conformation, Gemin2 wraps around the crescent-shaped pentamer, interacting with all five Sm proteins, and gripping its bottom and top sides and outer perimeter. Gemin2 reaches into the RNA-binding pocket, preventing RNA binding. Interestingly, SMN-Gemin2 interaction is abrogated by a spinal muscular atrophy (SMA)-causing mutation in an SMN helix that mediates Gemin2 binding. These findings provide insight into SMN complex assembly and specificity, linking snRNP biogenesis and SMA pathogenesis.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Proteínas do Complexo SMN/metabolismo , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas de Ligação a RNA/genética , Alinhamento de Sequência
3.
Nucleic Acids Res ; 47(2): 929-940, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30418624

RESUMO

Aminoacyl-tRNA synthetases catalyze the attachment of cognate amino acids onto tRNAs. To avoid mistranslation, editing mechanisms evolved to maintain tRNA aminoacylation fidelity. For instance, while rejecting the majority of non-cognate amino acids via discrimination in the synthetic active site, prolyl-tRNA synthetase (ProRS) misactivates and mischarges Ala and Cys, which are similar in size to cognate Pro. Ala-tRNAPro is specifically hydrolyzed by the editing domain of ProRS in cis, while YbaK, a free-standing editing domain, clears Cys-tRNAPro in trans. ProXp-ala is another editing domain that clears Ala-tRNAPro in trans. YbaK does not appear to possess tRNA specificity, readily deacylating Cys-tRNACysin vitro. We hypothesize that YbaK binds to ProRS to gain specificity for Cys-tRNAPro and avoid deacylation of Cys-tRNACys in the cell. Here, in vivo evidence for ProRS-YbaK interaction was obtained using a split-green fluorescent protein assay. Analytical ultracentrifugation and native mass spectrometry were used to investigate binary and ternary complex formation between ProRS, YbaK, and tRNAPro. Our combined results support the hypothesis that the specificity of YbaK toward Cys-tRNAPro is determined by the formation of a three-component complex with ProRS and tRNAPro and establish the stoichiometry of a 'triple-sieve' editing complex for the first time.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Escherichia coli/metabolismo , Aminoacil-RNA de Transferência/metabolismo , RNA de Transferência de Prolina/metabolismo , Ligação Competitiva , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Substâncias Luminescentes , Espectrometria de Massas , Ultracentrifugação
4.
Mol Cell ; 33(5): 654-60, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19285947

RESUMO

Faithful translation of the genetic code depends on the GTPase EF-Tu delivering correctly charged aminoacyl-tRNAs to the ribosome for pairing with cognate codons. The accurate coupling of cognate amino acids and tRNAs by the aminoacyl-tRNA synthetases is achieved through a combination of substrate specificity and product editing. Once released by aminoacyl-tRNA synthetases, both cognate and near-cognate aminoacyl-tRNAs were considered to be committed to ribosomal protein synthesis through their association with EF-Tu. Here we show instead that aminoacyl-tRNAs in ternary complex with EF-Tu*GTP can readily dissociate and rebind to aminoacyl-tRNA synthetases. For mischarged species, this allows resampling by the product editing pathway, leading to a reduction in the overall error rate of aminoacyl-tRNA synthesis. Resampling of mischarged tRNAs was shown to increase the accuracy of translation over ten fold during in vitro protein synthesis, supporting the presence of an additional quality control step prior to translation elongation.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Guanosina Trifosfato/metabolismo , Elongação Traducional da Cadeia Peptídica , Fator Tu de Elongação de Peptídeos/metabolismo , RNA de Transferência/metabolismo , Aminoacilação de RNA de Transferência , Sítios de Ligação , Escherichia coli/genética , Código Genético , Cinética , Leucina-tRNA Ligase/metabolismo , Conformação de Ácido Nucleico , Peptídeos/metabolismo , Fenilalanina-tRNA Ligase/metabolismo , RNA Bacteriano , RNA de Transferência/química , Especificidade por Substrato , Tirosina-tRNA Ligase/metabolismo
5.
J Biol Chem ; 286(36): 31810-20, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21768119

RESUMO

Aminoacyl-tRNA synthetases attach specific amino acids to cognate tRNAs. Prolyl-tRNA synthetases are known to mischarge tRNA(Pro) with the smaller amino acid alanine and with cysteine, which is the same size as proline. Quality control in proline codon translation is partly ensured by an editing domain (INS) present in most bacterial prolyl-tRNA synthetases that hydrolyzes smaller Ala-tRNA(Pro) and excludes Pro-tRNA(Pro). In contrast, Cys-tRNA(Pro) is cleared by a freestanding INS domain homolog, YbaK. Here, we have investigated the molecular mechanism of catalysis and substrate recognition by Hemophilus influenzae YbaK using site-directed mutagenesis, enzymatic assays of isosteric substrates and functional group analogs, and computational modeling. These studies together with mass spectrometric characterization of the YbaK-catalyzed reaction products support a novel substrate-assisted mechanism of Cys-tRNA(Pro) deacylation that prevents nonspecific Pro-tRNA(Pro) hydrolysis. Collectively, we propose that the INS and YbaK domains co-evolved distinct mechanisms involving steric exclusion and thiol-specific chemistry, respectively, to ensure accurate decoding of proline codons.


Assuntos
Aminoacil-tRNA Sintetases/genética , Proteínas de Bactérias/genética , Códon , Haemophilus influenzae/genética , Prolina/genética , Biossíntese de Proteínas , Acilação , Biocatálise , Catálise , Hidrólise , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Aminoacil-RNA de Transferência , Especificidade por Substrato
6.
Methods Enzymol ; 655: 325-347, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34183128

RESUMO

Full-length transcription in the majority of protein-coding and other genes transcribed by RNA polymerase II in complex eukaryotes requires U1 snRNP (U1) to co-transcriptionally suppress transcription-terminating premature 3'-end cleavage and polyadenylation (PCPA) from cryptic polyadenylation signals (PASs). This U1 activity, termed telescripting, requires U1 to base-pair with the nascent RNA and inhibit usage of a downstream PAS. Here we describe experimental methods to determine the mechanism of U1 telescripting, involving mapping of U1 and CPA factors (CPAFs) binding locations in relation to PCPA sites, and identify U1 and CPAFs interactomes. The methods which utilizes rapid reversible protein-RNA and protein-protein chemical crosslinking, immunoprecipitations (XLIPs) of components of interest, and RNA-seq and quantitative proteomic mass spectrometry, captured U1-CPAFs complexes in cells, providing important insights into telescripting mechanism. XLIP profiling can be used for comprehensive molecular definition of diverse RNPs.


Assuntos
Ribonucleoproteína Nuclear Pequena U1 , Fatores de Poliadenilação e Clivagem de mRNA , Poliadenilação , Proteômica , RNA , Ribonucleoproteína Nuclear Pequena U1/genética , Ribonucleoproteína Nuclear Pequena U1/metabolismo
7.
Nat Commun ; 11(1): 1, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31911652

RESUMO

Stimulated cells and cancer cells have widespread shortening of mRNA 3'-untranslated regions (3'UTRs) and switches to shorter mRNA isoforms due to usage of more proximal polyadenylation signals (PASs) in introns and last exons. U1 snRNP (U1), vertebrates' most abundant non-coding (spliceosomal) small nuclear RNA, silences proximal PASs and its inhibition with antisense morpholino oligonucleotides (U1 AMO) triggers widespread premature transcription termination and mRNA shortening. Here we show that low U1 AMO doses increase cancer cells' migration and invasion in vitro by up to 500%, whereas U1 over-expression has the opposite effect. In addition to 3'UTR length, numerous transcriptome changes that could contribute to this phenotype are observed, including alternative splicing, and mRNA expression levels of proto-oncogenes and tumor suppressors. These findings reveal an unexpected role for U1 homeostasis (available U1 relative to transcription) in oncogenic and activated cell states, and suggest U1 as a potential target for their modulation.


Assuntos
Movimento Celular , Neoplasias/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/patologia , Neoplasias/fisiopatologia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Ribonucleoproteína Nuclear Pequena U1/genética
8.
Artigo em Inglês | MEDLINE | ID: mdl-32651264

RESUMO

This summary of the 84th Cold Spring Harbor Laboratory (CSHL) Symposium on Quantitative Biology: RNA Control and Regulation, held in May 2019, highlights key emerging themes in this field, which now impacts nearly every aspect of biology and medicine. Recent discoveries accelerated by technological developments reveal enormous diversity of RNAs and RNA-binding proteins (RBPs) with ever-increasing roles in eukaryotes. Atomic structures and live-cell imaging of transcription, RNA splicing, 3'-end processing, modifications, and degradation machineries provide mechanistic insights, explaining hundreds of diseases caused by their perturbations. This great progress uncovered numerous targets for therapies, some of which have already been successfully exploited, and many opportunities for pharmacological intervention and RNA-guided genome engineering. Myriad unexplained RNAs and RBPs leave the RNA field open for many more exciting discoveries.

9.
Artigo em Inglês | MEDLINE | ID: mdl-32518092

RESUMO

Telescripting is a fundamental cotranscriptional gene regulation process that relies on U1 snRNP (U1) to suppress premature 3'-end cleavage and polyadenylation (PCPA) in RNA polymerase II (Pol II) transcripts, which is necessary for full-length transcription of thousands of protein-coding (pre-mRNAs) and long noncoding (lncRNA) genes. Like U1 role in splicing, telescripting requires U1 snRNA base-pairing with nascent transcripts. Inhibition of U1 base-pairing with U1 snRNA antisense morpholino oligonucleotide (U1 AMO) mimics widespread PCPA from cryptic polyadenylation signals (PASs) in human tissues, including PCPA in introns and last exons' 3'-untranslated regions (3' UTRs). U1 telescripting-PCPA balance changes generate diverse RNAs depending on where in a gene it occurs. Long genes are highly U1-telescripting-dependent because of PASs in introns compared to short genes. Enrichment of cell cycle control, differentiation, and developmental functions in long genes, compared to housekeeping and acute cell stress response genes in short genes, reveals a gene size-function relationship in mammalian genomes. This polarization increased in metazoan evolution by previously unexplained intron expansion, suggesting that U1 telescripting could shift global gene expression priorities. We show that that modulating U1 availability can profoundly alter cell phenotype, such as cancer cell migration and invasion, underscoring the critical role of U1 homeostasis and suggesting it as a potential target for therapies. We describe a complex of U1 with cleavage and polyadenylation factors that silences PASs in introns and 3' UTR, which gives insights into U1 telescripting mechanism and transcription elongation regulation.

10.
Artigo em Inglês | MEDLINE | ID: mdl-30709878

RESUMO

Recent observations showed that nascent RNA polymerase II transcripts, pre-mRNAs, and noncoding RNAs are highly susceptible to premature 3'-end cleavage and polyadenylation (PCPA) from numerous intronic cryptic polyadenylation signals (PASs). The importance of this in gene regulation was not previously appreciated as PASs, despite their prevalence, were thought to be active in terminal exons at gene ends. Unexpectedly, antisense oligonucleotide interference with U1 snRNA base-pairing to 5' splice sites, which is necessary for U1 snRNP's (U1) function in splicing, caused widespread PCPA in metazoans. This uncovered U1's PCPA suppression activity, termed telescripting, as crucial for full-length transcription in thousands of vertebrate genes, providing a general role in transcription elongation control. Progressive intron-size expansion in metazoan evolution greatly increased PCPA vulnerability and dependence on U1 telescripting. We describe how these observations unfolded and discuss U1 telescripting's role in shaping the transcriptome.


Assuntos
Regulação da Expressão Gênica/fisiologia , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Terminação da Transcrição Genética , Animais , Humanos , Íntrons , RNA Nuclear Pequeno/genética
11.
Nat Struct Mol Biol ; 24(11): 993-999, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28967884

RESUMO

U1 snRNP (U1) functions in splicing introns and telescripting, which suppresses premature cleavage and polyadenylation (PCPA). Using U1 inhibition in human cells, we show that U1 telescripting is selectively required for sustaining long-distance transcription elongation in introns of large genes (median 39 kb). Evidence of widespread PCPA in the same locations in normal tissues reveals that large genes incur natural transcription attrition. Underscoring the importance of U1 telescripting as a gene-size-based mRNA-regulation mechanism, small genes were not sensitive to PCPA, and the spliced-mRNA productivity of ∼1,000 small genes (median 6.8 kb) increased upon U1 inhibition. Notably, these small, upregulated genes were enriched in functions related to acute stimuli and cell-survival response, whereas genes subject to PCPA were enriched in cell-cycle progression and developmental functions. This gene size-function polarization increased in metazoan evolution by enormous intron expansion. We propose that telescripting adds an overarching layer of regulation to size-function-stratified genomes, leveraged by selective intron expansion to rapidly shift gene expression priorities.


Assuntos
Regulação da Expressão Gênica , Genoma Humano , Poliadenilação , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Transcrição Gênica , Humanos
12.
Nat Struct Mol Biol ; 23(3): 225-30, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26828962

RESUMO

Despite equal snRNP stoichiometry in spliceosomes, U1 snRNP (U1) is typically the most abundant vertebrate snRNP. Mechanisms regulating U1 overabundance and snRNP repertoire are unknown. In Sm-core assembly, a key snRNP-biogenesis step mediated by the SMN complex, the snRNA-specific RNA-binding protein (RBP) Gemin5 delivers pre-snRNAs, which join SMN-Gemin2-recruited Sm proteins. We show that the human U1-specific RBP U1-70K can bridge pre-U1 to SMN-Gemin2-Sm, in a Gemin5-independent manner, thus establishing an additional and U1-exclusive Sm core-assembly pathway. U1-70K hijacks SMN-Gemin2-Sm, enhancing Sm-core assembly on U1s and inhibiting that on other snRNAs, thereby promoting U1 overabundance and regulating snRNP repertoire. SMN-Gemin2's ability to facilitate transactions between different RBPs and RNAs explains its multi-RBP valency and the myriad transcriptome perturbations associated with SMN deficiency in neurodegenerative spinal muscular atrophy. We propose that SMN-Gemin2 is a versatile hub for RNP exchange that functions broadly in RNA metabolism.


Assuntos
Redes e Vias Metabólicas , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Humanos , Modelos Moleculares , Ribonucleoproteína Nuclear Pequena U1/química , Proteínas do Complexo SMN/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA