Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
1.
Cell Res ; 34(6): 428-439, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38658629

RESUMEN

Spliceosome is often assembled across an exon and undergoes rearrangement to span a neighboring intron. Most states of the intron-defined spliceosome have been structurally characterized. However, the structure of a fully assembled exon-defined spliceosome remains at large. During spliceosome assembly, the pre-catalytic state (B complex) is converted from its precursor (pre-B complex). Here we report atomic structures of the exon-defined human spliceosome in four sequential states: mature pre-B, late pre-B, early B, and mature B. In the previously unknown late pre-B state, U1 snRNP is already released but the remaining proteins are still in the pre-B state; unexpectedly, the RNAs are in the B state, with U6 snRNA forming a duplex with 5'-splice site and U5 snRNA recognizing the 3'-end of the exon. In the early and mature B complexes, the B-specific factors are stepwise recruited and specifically recognize the exon 3'-region. Our study reveals key insights into the assembly of the exon-defined spliceosomes and identifies mechanistic steps of the pre-B-to-B transition.


Asunto(s)
Exones , ARN Nuclear Pequeño , Empalmosomas , Humanos , Empalmosomas/metabolismo , Exones/genética , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Empalme del ARN , Intrones/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/genética , Sitios de Empalme de ARN/genética , Modelos Moleculares
2.
Front Cell Infect Microbiol ; 14: 1354410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38415010

RESUMEN

Background: Monkeypox or mpox virus (mpox) is a double-stranded DNA virus that poses a significant threat to global public health security. The F3 protein, encoded by mpox, is an apoenzyme believed to possess a double-stranded RNA-binding domain (dsRBD). However, limited research has been conducted on its function. In this study, we present data on the transcriptomics and proteomics of F3L-transfected HEK293T cells, aiming to enhance our comprehension of F3L. Methods: The gene expression profiles of pCAGGS-HA-F3L transfected HEK293T cells were analyzed using RNA-seq. Proteomics was used to identify and study proteins that interact with F3L. Real-time PCR was used to detect mRNA levels of several differentially expressed genes (DEGs) in HEK293T cells (or Vero cells) after the expression of F3 protein. Results: A total of 14,822 genes were obtained in cells by RNA-Seq and 1,672 DEGs were identified, including 1,156 up-regulated genes and 516 down-regulated genes. A total of 27 cellular proteins interacting with F3 proteins were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS), and 19 cellular proteins with large differences in abundance ratios were considered to be candidate cellular proteins. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that the DEGs were significantly enriched in immune-related pathways, including type I interferon signaling pathway, response to virus, RIG-I-like receptor signaling pathway, NOD-like receptor signaling pathway, etc. Moreover, some selected DEGs were further confirmed by real-time PCR and the results were consistent with the transcriptome data. Proteomics data show that cellular proteins interacting with F3 proteins are mainly related to RNA splicing and protein translation. Conclusions: Our analysis of transcriptomic and proteomic data showed that (1) F3L up-regulates the transcript levels of key genes in the innate immune signaling pathway, such as RIGI, MDA5, IRF5, IRF7, IRF9, ISG15, IFNA14, and elicits a broad spectrum of antiviral immune responses in the host. F3L also increases the expression of the FOS and JNK genes while decreasing the expression of TNFR2, these factors may ultimately induce apoptosis. (2) F3 protein interacts with host proteins involved in RNA splicing and protein translation, such as SNRNP70, POLR2H, HNRNPA1, DDX17, etc. The findings of this study shed light on the function of the F3 protein.


Asunto(s)
Mpox , Transcriptoma , Animales , Chlorocebus aethiops , Humanos , Monkeypox virus/genética , Células Vero , Cromatografía Liquida , Células HEK293 , Proteómica , Espectrometría de Masas en Tándem , Perfilación de la Expresión Génica/métodos , Ribonucleoproteína Nuclear Pequeña U1/genética
3.
ACS Nano ; 17(23): 23331-23346, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37971502

RESUMEN

Synthetic riboswitches are promising regulatory devices due to their small size, lack of immunogenicity, and ability to fine-tune gene expression in the absence of exogenous trans-acting factors. Based on a gene inhibitory system developed at our lab, termed U1snRNP interference (U1i), we developed tetracycline (TC)-inducible riboswitches that modulate mRNA polyadenylation through selective U1 snRNP recruitment. First, we engineered different TC-U1i riboswitches, which repress gene expression unless TC is added, leading to inductions of gene expression of 3-to-4-fold. Second, we developed a technique called Systematic Evolution of Riboswitches by Exponential Enrichment (SEREX), to isolate riboswitches with enhanced U1 snRNP binding capacity and activity, achieving inducibilities of up to 8-fold. Interestingly, by multiplexing riboswitches we increased inductions up to 37-fold. Finally, we demonstrated that U1i-based riboswitches are dose-dependent and reversible and can regulate the expression of reporter and endogenous genes in culture cells and mouse models, resulting in attractive systems for gene therapy applications. Our work probes SEREX as a much-needed technology for the in vitro identification of riboswitches capable of regulating gene expression in vivo.


Asunto(s)
Riboswitch , Animales , Ratones , Riboswitch/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Tetraciclina/farmacología , Antibacterianos , Mamíferos/genética , Expresión Génica
4.
J Cell Biol ; 222(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37956386

RESUMEN

Current models posit that nuclear speckles (NSs) serve as reservoirs of splicing factors and facilitate posttranscriptional mRNA processing. Here, we discovered that ribotoxic stress induces a profound reorganization of NSs with enhanced recruitment of factors required for splice-site recognition, including the RNA-binding protein TIAR, U1 snRNP proteins and U2-associated factor 65, as well as serine 2 phosphorylated RNA polymerase II. NS reorganization relies on the stress-activated p38 mitogen-activated protein kinase (MAPK) pathway and coincides with splicing activation of both pre-existing and newly synthesized pre-mRNAs. In particular, ribotoxic stress causes targeted excision of retained introns from pre-mRNAs of immediate early genes (IEGs), whose transcription is induced during the stress response. Importantly, enhanced splicing of the IEGs ZFP36 and FOS is accompanied by relocalization of the corresponding nuclear mRNA foci to NSs. Our study reveals NSs as a dynamic compartment that is remodeled under stress conditions, whereby NSs appear to become sites of IEG transcription and efficient cotranscriptional splicing.


Asunto(s)
Genes Inmediatos-Precoces , Motas Nucleares , Empalme del ARN , Intrones , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Humanos
5.
RNA ; 29(8): 1140-1165, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37137667

RESUMEN

Removal of introns during pre-mRNA splicing, which is central to gene expression, initiates by base pairing of U1 snRNA with a 5' splice site (5'SS). In mammals, many introns contain weak 5'SSs that are not efficiently recognized by the canonical U1 snRNP, suggesting alternative mechanisms exist. Here, we develop a cross-linking immunoprecipitation coupled to a high-throughput sequencing method, BCLIP-seq, to identify NRDE2 (nuclear RNAi-defective 2), and CCDC174 (coiled-coil domain-containing 174) as novel RNA-binding proteins in mouse ES cells that associate with U1 snRNA and 5'SSs. Both proteins bind directly to U1 snRNA independently of canonical U1 snRNP-specific proteins, and they are required for the selection and effective processing of weak 5'SSs. Our results reveal that mammalian cells use noncanonical splicing factors bound directly to U1 snRNA to effectively select suboptimal 5'SS sequences in hundreds of genes, promoting proper splice site choice, and accurate pre-mRNA splicing.


Asunto(s)
Precursores del ARN , Sitios de Empalme de ARN , Animales , Ratones , Sitios de Empalme de ARN/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Interferencia de ARN , Empalme del ARN , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Empalme Alternativo , Mamíferos/genética
6.
J Biol Chem ; 299(7): 104854, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37224962

RESUMEN

Functional depletion of the U1 small nuclear ribonucleoprotein (snRNP) with a 25 nt U1 AMO (antisense morpholino oligonucleotide) may lead to intronic premature cleavage and polyadenylation of thousands of genes, a phenomenon known as U1 snRNP telescripting; however, the underlying mechanism remains elusive. In this study, we demonstrated that U1 AMO could disrupt U1 snRNP structure both in vitro and in vivo, thereby affecting the U1 snRNP-RNAP polymerase II interaction. By performing chromatin immunoprecipitation sequencing for phosphorylation of Ser2 and Ser5 of the C-terminal domain of RPB1, the largest subunit of RNAP polymerase II, we showed that transcription elongation was disturbed upon U1 AMO treatment, with a particular high phosphorylation of Ser2 signal at intronic cryptic polyadenylation sites (PASs). In addition, we showed that core 3'processing factors CPSF/CstF are involved in the processing of intronic cryptic PAS. Their recruitment accumulated toward cryptic PASs upon U1 AMO treatment, as indicated by chromatin immunoprecipitation sequencing and individual-nucleotide resolution CrossLinking and ImmunoPrecipitation sequencing analysis. Conclusively, our data suggest that disruption of U1 snRNP structure mediated by U1 AMO provides a key for understanding the U1 telescripting mechanism.


Asunto(s)
Morfolinos , Oligonucleótidos Antisentido , Precursores del ARN , Ribonucleoproteína Nuclear Pequeña U1 , Morfolinos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Antisentido/farmacología , Poliadenilación , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Precursores del ARN/metabolismo , Humanos , Células HeLa , Técnicas de Silenciamiento del Gen , Factor de Especificidad de Desdoblamiento y Poliadenilación , Factor de Estimulación del Desdoblamiento/metabolismo , Transcripción Genética/efectos de los fármacos
7.
Cancer Res ; 83(12): 2000-2015, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37057875

RESUMEN

Dysregulation of RNA-binding proteins (RBP) is one of the characteristics of cancer. Investigating the biological functions and molecular mechanisms of abnormal RBPs can help uncover new cancer biomarkers and treatment strategies. To identify oncogenic RBPs in triple-negative breast cancer (TNBC), we employed an in vivo CRISPR screen and a TNBC progression model, which revealed small nuclear ribonucleoprotein polypeptide C (SNRPC), a subunit of the U1 small nuclear ribonucleoprotein particle (U1 snRNP), as a key modulator of TNBC progression. SNRPC was frequently upregulated, which corresponded to poor prognosis in patients with TNBC. SNRPC ablation significantly impaired the proliferation, migration, and invasion of TNBC cells in vitro and in vivo. In addition, SNRPC was essential for the stability of U1 snRNP and contributed to the RNA Pol II-controlled transcriptional program. Knockdown of SNRPC decreased RNA Pol II enrichment on a subset of oncogenes (TNFAIP2, E2F2, and CDK4) and reduced their expression levels. Furthermore, SNRPC deletion was confirmed to inhibit TNBC progression partially through regulation of the TNFAIP2-Rac1-ß-catenin signaling pathway. Taken together, this data suggests that SNRPC plays an oncogenic role in TNBC, is a marker of poor prognosis, and may be a valuable therapeutic target for patients with intractable TNBC. SIGNIFICANCE: A functional CRISPR screen identifies SNRPC as an RNA-binding protein that promotes the aggressiveness of breast cancer by facilitating Pol II-controlled transcription of oncogenes.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Pronóstico , ARN Polimerasa II/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
8.
Mol Cell ; 83(8): 1264-1279.e10, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36965480

RESUMEN

The expansion of introns within mammalian genomes poses a challenge for the production of full-length messenger RNAs (mRNAs), with increasing evidence that these long AT-rich sequences present obstacles to transcription. Here, we investigate RNA polymerase II (RNAPII) elongation at high resolution in mammalian cells and demonstrate that RNAPII transcribes faster across introns. Moreover, we find that this acceleration requires the association of U1 snRNP (U1) with the elongation complex at 5' splice sites. The role of U1 to stimulate elongation rate through introns reduces the frequency of both premature termination and transcriptional arrest, thereby dramatically increasing RNA production. We further show that changes in RNAPII elongation rate due to AT content and U1 binding explain previous reports of pausing or termination at splice junctions and the edge of CpG islands. We propose that U1-mediated acceleration of elongation has evolved to mitigate the risks that long AT-rich introns pose to transcript completion.


Asunto(s)
ARN Polimerasa II , Ribonucleoproteína Nuclear Pequeña U1 , Animales , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Transcripción Genética , Empalmosomas/genética , Intrones/genética , Sitios de Empalme de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Empalme del ARN , Precursores del ARN/genética , Mamíferos/metabolismo
9.
RNA ; 29(5): 531-550, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36737103

RESUMEN

Premessenger RNA splicing is catalyzed by the spliceosome, a multimegadalton RNA-protein complex that assembles in a highly regulated process on each intronic substrate. Most studies of splicing and spliceosomes have been carried out in human or S. cerevisiae model systems. There exists, however, a large diversity of spliceosomes, particularly in organisms with reduced genomes, that suggests a means of analyzing the essential elements of spliceosome assembly and regulation. In this review, we characterize changes in spliceosome composition across phyla, describing those that are most frequently observed and highlighting an analysis of the reduced spliceosome of the red alga Cyanidioschyzon merolae We used homology modeling to predict what effect splicing protein loss would have on the spliceosome, based on currently available cryo-EM structures. We observe strongly correlated loss of proteins that function in the same process, for example, in interacting with the U1 snRNP (which is absent in C. merolae), regulation of Brr2, or coupling transcription and splicing. Based on our observations, we predict splicing in C. merolae to be inefficient, inaccurate, and post-transcriptional, consistent with the apparent trend toward its elimination in this lineage. This work highlights the striking flexibility of the splicing pathway and the spliceosome when viewed in the context of eukaryotic diversity.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Empalmosomas , Humanos , Empalmosomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Empalme del ARN , Intrones , Ribonucleoproteína Nuclear Pequeña U1/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Helicasas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
10.
J Mol Cell Biol ; 14(8)2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36073763

RESUMEN

In eukaryotic cells, both alternative splicing and alternative polyadenylation (APA) play essential roles in the gene regulation network. U1 small ribonucleoprotein particle (U1 snRNP) is a major component of spliceosome, and U1 snRNP complex can suppress proximal APA sites through crosstalking with 3' end processing factors. However, here we show that both knockdown and overexpression of SNRPA, SNRPC, SNRNP70, and SNRPD2, the U1 snRNP proteins, promote the usage of proximal APA sites at the transcriptome level. SNRNP70 can drive the phase transition of PABPN1 from droplet to aggregate, which may reduce the repressive effects of PABPN1 on the proximal APA sites. Additionally, SNRNP70 can also promote the proximal APA sites by recruiting CPSF6, suggesting that the function of CPSF6 on APA is related with other RNA-binding proteins and cell context-dependent. Consequently, these results reveal that, on the contrary to U1 snRNP complex, the free proteins of U1 snRNP complex can promote proximal APA sites through the interaction with 3' end processing machinery.


Asunto(s)
Poliadenilación , Ribonucleoproteína Nuclear Pequeña U1 , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Empalme Alternativo/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Empalme del ARN
11.
Plant Cell ; 34(12): 4920-4935, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36087009

RESUMEN

In plants, microRNA (miRNA) biogenesis involves cotranscriptional processing of RNA polymerase II (RNAPII)-generated primary transcripts by a multi-protein complex termed the microprocessor. Here, we report that Arabidopsis (Arabidopsis thaliana) PRE-MRNA PROCESSING PROTEIN 40 (PRP40), the U1 snRNP auxiliary protein, positively regulates the recruitment of SERRATE, a core component of the plant microprocessor, to miRNA genes. The association of DICER-LIKE1 (DCL1), the microprocessor endoribonuclease, with chromatin was altered in prp40ab mutant plants. Impaired cotranscriptional microprocessor assembly was accompanied by RNAPII accumulation at miRNA genes and retention of miRNA precursors at their transcription sites in the prp40ab mutant plants. We show that cotranscriptional microprocessor assembly, regulated by AtPRP40, positively affects RNAPII transcription of miRNA genes and is important to reach the correct levels of produced miRNAs.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , MicroARNs , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Microcomputadores , Cromatina/genética , Cromatina/metabolismo , Procesamiento Postranscripcional del ARN/genética
12.
Methods Mol Biol ; 2537: 247-262, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35895269

RESUMEN

Alternative RNA splicing is an essential part of gene expression that not only increases the protein diversity of metazoan but also provides an additional layer of gene expression regulation. The U1 small ribonucleoparticle (U1 snRNP) plays an essential role in seeding spliceosome assembly and its binding on weak 5'-splice sites is regulated by transient interactions with splicing factors. Recent progress in allele specific splicing correction has shown the therapeutic potential offered by small molecule splicing modifiers that specifically promotes the recruitment of U1 snRNP to modulate alternative splicing and gene expression. Here, we described a method to reconstitute U1 snRNP in vitro and to study labile interactions with protein or synthetic splicing factors using solution state NMR spectroscopy. This approach allowed us to validate direct interactions between splicing regulators and U1 snRNP and could also be useful for the screening of small molecules acting on splicing regulation.


Asunto(s)
Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U1 , Empalme Alternativo , Animales , Espectroscopía de Resonancia Magnética , Proteínas/metabolismo , Precursores del ARN/genética , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo
13.
RNA Biol ; 19(1): 943-960, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35866748

RESUMEN

In Eukarya, immature mRNA transcripts (pre-mRNA) often contain coding sequences, or exons, interleaved by non-coding sequences, or introns. Introns are removed upon splicing, and further regulation of the retained exons leads to alternatively spliced mRNA. The splicing reaction requires the stepwise assembly of the spliceosome, a macromolecular machine composed of small nuclear ribonucleoproteins (snRNPs). This review focuses on the early stage of spliceosome assembly, when U1 snRNP defines each intron 5'-splice site (5'ss) in the pre-mRNA. We first introduce the splicing reaction and the impact of alternative splicing on gene expression regulation. Thereafter, we extensively discuss splicing descriptors that influence the 5'ss selection by U1 snRNP, such as sequence determinants, and interactions mediated by U1-specific proteins or U1 small nuclear RNA (U1 snRNA). We also include examples of diseases that affect the 5'ss selection by U1 snRNP, and discuss recent therapeutic advances that manipulate U1 snRNP 5'ss selectivity with antisense oligonucleotides and small-molecule splicing switches.


Asunto(s)
Precursores del ARN , Ribonucleoproteína Nuclear Pequeña U1 , Empalme Alternativo , Precursores del ARN/genética , Sitios de Empalme de ARN , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo
14.
Nucleic Acids Res ; 50(14): 8262-8278, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35871302

RESUMEN

We recently reported that serine-arginine-rich (SR) protein-mediated pre-mRNA structural remodeling generates a pre-mRNA 3D structural scaffold that is stably recognized by the early spliceosomal components. However, the intermediate steps between the free pre-mRNA and the assembled early spliceosome are not yet characterized. By probing the early spliceosomal complexes in vitro and RNA-protein interactions in vivo, we show that the SR proteins bind the pre-mRNAs cooperatively generating a substrate that recruits U1 snRNP and U2AF65 in a splice signal-independent manner. Excess U1 snRNP selectively displaces some of the SR protein molecules from the pre-mRNA generating the substrate for splice signal-specific, sequential recognition by U1 snRNP, U2AF65 and U2AF35. Our work thus identifies a novel function of U1 snRNP in mammalian splicing substrate definition, explains the need for excess U1 snRNP compared to other U snRNPs in vivo, demonstrates how excess SR proteins could inhibit splicing, and provides a conceptual basis to examine if this mechanism of splicing substrate definition is employed by other splicing regulatory proteins.


Asunto(s)
Precursores del ARN , Empalme del ARN , Empalmosomas , Animales , Mamíferos/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Empalmosomas/metabolismo , Factor de Empalme U2AF/genética , Factor de Empalme U2AF/metabolismo
15.
Methods Mol Biol ; 2442: 713-726, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35320554

RESUMEN

Fractionation of HeLa cell nuclear extracts by glycerol gradient centrifugation separates endogenous uracil-rich small nuclear ribonucleoprotein complexes (U snRNP) into numerous particles sedimenting from 7S to greater than 60S. Complexes sedimenting at 10S contain a single U snRNP (U1 snRNP) and galectin-3. Addition of antibodies specific for galectin-3 to fractions containing these 10S complexes coprecipitates U1 snRNP, indicating that a fraction of the U1 snRNP is associated with this galectin. Galectin-3 has been shown by depletion-reconstitution studies to be an integral splicing component involved both in spliceosome assembly and splicing activity. The first step in initiation of spliceosome assembly is binding of U1 snRNP to the 5' splice site of the premessenger RNA substrate. The finding that U1 snRNP and galectin-3 are associated in splicing extracts hints that this complex affords a potential entry point for galectin-3 into the splicing pathway. Addition of U1 snRNP-galectin-3 complexes immunoselected from the 10S region of glycerol gradients to a U1-depleted nuclear extract initiates splicing activity with the formation of splicing intermediates and mature mRNA. This chapter describes the materials and methods for these experiments that document galectin-3-U1 snRNP complexes initiate the splicing reaction in a U1-depleted nuclear extract.


Asunto(s)
Galectina 3 , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U1 , Empalmosomas , Fraccionamiento Celular , Galectina 3/genética , Galectina 3/metabolismo , Células HeLa/metabolismo , Humanos , Espacio Intranuclear/química , Espacio Intranuclear/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/fisiología , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Empalmosomas/metabolismo , Uracilo/análisis , Uracilo/metabolismo
16.
RNA ; 28(6): 878-894, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35351812

RESUMEN

Quality control of mRNA represents an important regulatory mechanism for gene expression in eukaryotes. One component of this quality control is the nuclear retention and decay of misprocessed RNAs. Previously, we demonstrated that mature mRNAs containing a 5' splice site (5'SS) motif, which is typically found in misprocessed RNAs such as intronic polyadenylated (IPA) transcripts, are nuclear retained and degraded. Using high-throughput sequencing of cellular fractions, we now demonstrate that IPA transcripts require the zinc finger protein ZFC3H1 for their nuclear retention and degradation. Using reporter mRNAs, we demonstrate that ZFC3H1 promotes the nuclear retention of mRNAs with intact 5'SS motifs by sequestering them into nuclear speckles. Furthermore, we find that U1-70K, a component of the spliceosomal U1 snRNP, is also required for the nuclear retention of these reporter mRNAs and likely functions in the same pathway as ZFC3H1. Finally, we show that the disassembly of nuclear speckles impairs the nuclear retention of reporter mRNAs with 5'SS motifs. Our results highlight a splicing independent role of U1 snRNP and indicate that it works in conjunction with ZFC3H1 in preventing the nuclear export of misprocessed mRNAs by sequestering them into nuclear speckles.


Asunto(s)
Sitios de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U1 , Motas Nucleares , Sitios de Empalme de ARN/genética , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Empalmosomas/genética , Empalmosomas/metabolismo
17.
Gene ; 818: 146203, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35101583

RESUMEN

Amyotrophic lateral sclerosis (ALS) has been considered as one of the progressive neurodegenerative diseases. Numerous genetic factors in divergent molecular pathways have been identified as causative factors of ALS. However, the underlying molecular mechanism that causes this disease remains undetermined; as a result, this has driven the search to find consensus disease-specific hallmarks. In this study, we focused on the alteration of the ratio of two specific gene-splicing events in the SNRNP70 gene from RNA-seq data derived from patients with ALS and control subjects. The splicing profile was significantly and specifically changed in one previously identified ALS subtype. Conversely, the gene expression profile of other ALS cases containing a splicing alteration in the SNRNP70 gene was similar to that of the subtype, whereas ALS cases without this change have exhibited less similarity. These results indicate that this splicing event in the SNRNP70 gene could represent a novel and broadly applicable molecular hallmark of a subtype of ALS.


Asunto(s)
Empalme del ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Regiones no Traducidas 3'/genética , Esclerosis Amiotrófica Lateral/clasificación , Esclerosis Amiotrófica Lateral/genética , Exones/genética , Predisposición Genética a la Enfermedad , Humanos , Estrés Oxidativo , Análisis de Componente Principal
18.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35101980

RESUMEN

In mammals, the structural basis for the interaction between U1 and U2 small nuclear ribonucleoproteins (snRNPs) during the early steps of splicing is still elusive. The binding of the ubiquitin-like (UBL) domain of SF3A1 to the stem-loop 4 of U1 snRNP (U1-SL4) contributes to this interaction. Here, we determined the 3D structure of the complex between the UBL of SF3A1 and U1-SL4 RNA. Our crystallography, NMR spectroscopy, and cross-linking mass spectrometry data show that SF3A1-UBL recognizes, sequence specifically, the GCG/CGC RNA stem and the apical UUCG tetraloop of U1-SL4. In vitro and in vivo mutational analyses support the observed intermolecular contacts and demonstrate that the carboxyl-terminal arginine-glycine-glycine-arginine (RGGR) motif of SF3A1-UBL binds sequence specifically by inserting into the RNA major groove. Thus, the characterization of the SF3A1-UBL/U1-SL4 complex expands the repertoire of RNA binding domains and reveals the capacity of RGG/RG motifs to bind RNA in a sequence-specific manner.


Asunto(s)
Factores de Empalme de ARN/química , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U2/química , Cristalografía por Rayos X , Humanos , Resonancia Magnética Nuclear Biomolecular , Motivos de Nucleótidos , Factores de Empalme de ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U2/genética
19.
Biochem Genet ; 60(5): 1809-1824, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35098410

RESUMEN

Circular RNA (circRNA) has been proved to be a key regulator of gastric cancer (GC) progression. Circ_0009910 was found to be highly expressed in GC and related to GC progression, but its role and mechanism in GC progression need to be further improved. Our study aims to further reveal circ_0009910 roles in GC progression and elucidate its potential molecular mechanism. The expression of circ_0009910, microRNA (miR)-361-3p, and small nuclear ribonucleoprotein polypeptide A (SNRPA) mRNA was measured by quantitative real-time PCR. Protein expression was determined using western blot analysis. Cell proliferation, migration, invasion, and apoptosis were evaluated using EDU staining, transwell assay, and flow cytometry. Cell glycolysis were assessed by detecting glucose consumption, lactate production, and glycolysis-related markers protein expression. The relationship between miR-361-3p and circ_0009910 or SNRPA was confirmed by RNA pull-down assay and dual-luciferase reporter assay. In vivo experiments were performed to explore the effect of circ_0009910 silencing on GC tumorigenesis. Circ_0009910 and SNRPA were upregulated in GC tumor tissues and cells. Knockdown of circ_0009910 or SNRPA could inhibit GC cell proliferation, migration, invasion, glycolysis, and promote apoptosis. Circ_0009910 could sponge miR-361-3p, and miR-361-3p could target SNRPA. Further experiments confirmed that circ_0009910 positively regulated SNRPA by sponging miR-361-3p. Additionally, SNRPA overexpression abolished the negative regulation of circ_0009910 silencing on GC progression. Furthermore, silenced circ_0009910 also reduced GC tumorigenesis in vivo. Our data showed that circ_0009910 might be a target for GC treatment, which could promote GC proliferation, metastasis, and glycolysis by the miR-361-3p/SNRPA axis.


Asunto(s)
MicroARNs , ARN Circular , Ribonucleoproteína Nuclear Pequeña U1 , Neoplasias Gástricas , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Glucólisis , Humanos , MicroARNs/genética , Péptidos/metabolismo , ARN Circular/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Neoplasias Gástricas/patología
20.
Ann Rheum Dis ; 81(1): 56-67, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34625402

RESUMEN

OBJECTIVES: To characterise splicing machinery (SM) alterations in leucocytes of patients with rheumatoid arthritis (RA), and to assess its influence on their clinical profile and therapeutic response. METHODS: Leucocyte subtypes from 129 patients with RA and 29 healthy donors (HD) were purified, and 45 selected SM elements (SME) were evaluated by quantitative PCR-array based on microfluidic technology (Fluidigm). Modulation by anti-tumour necrosis factor (TNF) therapy and underlying regulatory mechanisms were assessed. RESULTS: An altered expression of several SME was found in RA leucocytes. Eight elements (SNRNP70, SNRNP200, U2AF2, RNU4ATAC, RBM3, RBM17, KHDRBS1 and SRSF10) were equally altered in all leucocytes subtypes. Logistic regressions revealed that this signature might: discriminate RA and HD, and anti-citrullinated protein antibodies (ACPAs) positivity; classify high-disease activity (disease activity score-28 (DAS28) >5.1); recognise radiological involvement; and identify patients showing atheroma plaques. Furthermore, this signature was altered in RA synovial fluid and ankle joints of K/BxN-arthritic mice. An available RNA-seq data set enabled to validate data and identified distinctive splicing events and splicing variants among patients with RA expressing high and low SME levels. 3 and 6 months anti-TNF therapy reversed their expression in parallel to the reduction of the inflammatory profile. In vitro, ACPAs modulated SME, at least partially, by Fc Receptor (FcR)-dependent mechanisms. Key inflammatory cytokines further altered SME. Lastly, induced SNRNP70-overexpression and KHDRBS1-overexpression reversed inflammation in lymphocytes, NETosis in neutrophils and adhesion in RA monocytes and influenced activity of RA synovial fibroblasts. CONCLUSIONS: Overall, we have characterised for the first time a signature comprising eight dysregulated SME in RA leucocytes from both peripheral blood and synovial fluid, linked to disease pathophysiology, modulated by ACPAs and reversed by anti-TNF therapy.


Asunto(s)
Empalme Alternativo , Artritis Reumatoide/sangre , Artritis Reumatoide/genética , ARN/sangre , Empalmosomas , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Empalme Alternativo/efectos de los fármacos , Animales , Anticuerpos Antiproteína Citrulinada/farmacología , Antirreumáticos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Células Cultivadas , Citrulinación , Citocinas/farmacología , Proteínas de Unión al ADN/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Linfocitos , Masculino , Ratones , Persona de Mediana Edad , Monocitos , Neutrófilos , ARN/metabolismo , Factores de Empalme de ARN/genética , ARN Nuclear Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Análisis de Secuencia de ARN , Factores de Empalme Serina-Arginina/genética , Factor de Empalme U2AF/genética , Líquido Sinovial/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA