Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.778
Filtrar
1.
Mol Cell ; 84(19): 3656-3666, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39366353

RESUMEN

Proper gene expression requires the collaborative effort of multiple macromolecular machines to produce functional messenger RNA. As RNA polymerase II (RNA Pol II) transcribes DNA, the nascent pre-messenger RNA is heavily modified by other complexes such as 5' capping enzymes, the spliceosome, the cleavage, and polyadenylation machinery as well as RNA-modifying/editing enzymes. Recent evidence has demonstrated that pre-mRNA splicing and 3' end cleavage can occur on similar timescales as transcription and significantly cross-regulate. In this review, we discuss recent advances in co-transcriptional processing and how it contributes to gene regulation. We highlight how emerging areas-including coordinated splicing events, physical interactions between the RNA synthesis and modifying machinery, rapid and delayed splicing, and nuclear organization-impact mRNA isoforms. Coordination among RNA-processing choices yields radically different mRNA and protein products, foreshadowing the likely regulatory importance of co-transcriptional RNA folding and co-transcriptional modifications that have yet to be characterized in detail.


Asunto(s)
Precursores del ARN , Empalme del ARN , ARN Mensajero , Empalmosomas , Transcripción Genética , Precursores del ARN/metabolismo , Precursores del ARN/genética , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Animales , Empalmosomas/metabolismo , Empalmosomas/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Procesamiento Postranscripcional del ARN , Regulación de la Expresión Génica
2.
Invest Ophthalmol Vis Sci ; 65(12): 11, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39374010

RESUMEN

Purpose: Unfortunately, treatment of patients with uveal melanoma (UM) with metastatic disease is limited. Twenty percent of patients with UM harbor a mutation in the splicing factor gene SF3B1, suggesting that aberrant spliceosome function plays a vital role in tumorigenesis. Splicing inhibitors exploit the preferential sensitivity of spliceosome-compromised leukemic cells to these compounds. Methods: We studied the effect of the splicing inhibitor E7107 using two UM cell lines and ex vivo cultured SF3B1- and BAP1-mutated primary UM tumor slices. These UM cell lines and ex vivo tumor slices were exposed for 24 hours to different concentrations of E7107. Tumor slices were stained with hematoxylin and eosin (H&E) and incubated with BAP1, MelanA, MIB-1, and caspase-3 antisera. Results: The E7107-exposed UM cell lines exhibited decreased cell viability and increased apoptosis, with the greatest effect on SF3B1-mutated UM cells. A similar effect on UM tumor slices was observed upon exposure to E7107. Additionally, RNA was isolated for differential isoform expression analysis. No significant difference in isoform usage was found genome-wide. However, specific genes were differentially expressed after E7107 treatment in the SF3B1-mutated samples. Moreover, E7107 had the greatest effect on intron retention. Conclusions: This study indicates/suggests that mutated SF3B1 UM cells are more sensitive to the splicing inhibitor E7107 than wild-type SF3B1 UM cells.


Asunto(s)
Apoptosis , Melanoma , Mutación , Fosfoproteínas , Factores de Empalme de ARN , Empalmosomas , Neoplasias de la Úvea , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología , Humanos , Melanoma/genética , Melanoma/tratamiento farmacológico , Melanoma/patología , Melanoma/metabolismo , Empalmosomas/genética , Empalmosomas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Supervivencia Celular , Línea Celular Tumoral , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Células Tumorales Cultivadas , Ribonucleoproteína Nuclear Pequeña U2/genética , Empalme del ARN , Regulación Neoplásica de la Expresión Génica , Compuestos Epoxi , Macrólidos , Proteínas Supresoras de Tumor
3.
Toxins (Basel) ; 16(9)2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39330852

RESUMEN

Epsilon toxin (ETX), a potential agent of biological and toxic warfare, causes the death of many ruminants and threatens human health. It is crucial to understand the toxic mechanism of such a highly lethal and rapid course toxin. In this study, we detected the effects of ETX on the proteome and phosphoproteome of MDCK cells after 10 min and 30 min. A total of 44 differentially expressed proteins (DEPs) and 588 differentially phosphorylated proteins (DPPs) were screened in the 10 min group, while 73 DEPs and 489 DPPs were screened in the 30 min group. ETX-induced proteins and phosphorylated proteins were mainly located in the nucleus, cytoplasm, and mitochondria, and their enrichment pathways were related to transcription and translation, virus infection, and intercellular junction. Meanwhile, the protein-protein interaction network screened out several hub proteins, including SRSF1/2/6/7/11, SF3B1/2, NOP14/56, ANLN, GTPBP4, THOC2, and RRP1B. Almost all of these proteins were present in the spliceosome pathway, indicating that the spliceosome pathway is involved in ETX-induced cell death. Next, we used RNAi lentiviruses and inhibitors of several key proteins to verify whether these proteins play a critical role. The results confirmed that SRSF1, SF3B2, and THOC2 were the key proteins involved in the cytotoxic effect of ETX. In addition, we found that the common upstream kinase of these key proteins was SRPK1, and a reduction in the level of SRPK1 could also reduce ETX-induced cell death. This result was consistent with the phosphorylated proteomics analysis. In summary, our study demonstrated that ETX induces phosphorylation of SRSF1, SF3B2, THOC2, and SRPK1 proteins on the spliceosome pathway, which inhibits normal splicing of mRNA and leads to cell death.


Asunto(s)
Toxinas Bacterianas , Fosfoproteínas , Proteoma , Animales , Perros , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Células de Riñón Canino Madin Darby , Toxinas Bacterianas/toxicidad , Fosforilación , Proteómica , Mapas de Interacción de Proteínas , Empalmosomas/metabolismo , Empalmosomas/efectos de los fármacos
4.
Int J Mol Sci ; 25(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39273537

RESUMEN

Heart failure (HF) is associated with global changes in gene expression. Alternative mRNA splicing (AS) is a key regulatory mechanism underlying these changes. However, the whole status of molecules involved in the splicing process in human HF is unknown. Therefore, we analysed the spliceosome transcriptome in cardiac tissue (n = 36) from control subjects and HF patients (with ischaemic (ICM) and dilated (DCM) cardiomyopathies) using RNA-seq. We found greater deregulation of spliceosome machinery in ICM. Specifically, we showed widespread upregulation of the E and C complex components, highlighting an increase in SNRPD2 (FC = 1.35, p < 0.05) and DHX35 (FC = 1.34, p < 0.001) mRNA levels. In contrast, we observed generalised downregulation of the A complex and cardiac-specific AS factors, such as the multifunctional protein PCBP2 (FC = -1.29, p < 0.001) and the RNA binding proteins QKI (FC = -1.35, p < 0.01). In addition, we found a relationship between SNPRD2 (an E complex component) and the left ventricular mass index in ICM patients (r = 0.779; p < 0.01). On the other hand, we observed the specific underexpression of DDX46 (FC = -1.29), RBM17 (FC = -1.33), SDE2 (FC = -1.35) and RBFOX1 (FC = -1.33), p < 0.05, in DCM patients. Therefore, these aetiology-related alterations may indicate the differential involvement of the splicing process in the development of ICM and DCM.


Asunto(s)
Empalme Alternativo , Insuficiencia Cardíaca , Factores de Empalme de ARN , Empalmosomas , Transcriptoma , Humanos , Empalmosomas/metabolismo , Empalmosomas/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Factores de Empalme de ARN/metabolismo , Factores de Empalme de ARN/genética , Anciano , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Miocardio/metabolismo , Miocardio/patología , Perfilación de la Expresión Génica , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
5.
Cell Rep ; 43(9): 114751, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39276346

RESUMEN

Triple negative breast cancer (TNBC) is an aggressive type of breast cancer. While most TNBCs are initially sensitive to chemotherapy, a substantial fraction acquires resistance to treatments and progresses to more advanced stages. Here, we identify the spliceosome U2 small nuclear ribonucleoprotein particle (snRNP) complex as a modulator of chemotherapy efficacy in TNBC. Transient U2 snRNP inhibition induces persistent DNA damage in TNBC cells and organoids, regardless of their homologous recombination proficiency. U2 snRNP inhibition pervasively deregulates genes involved in the DNA damage response (DDR), an effect relying on their genomic structure characterized by a high number of small exons. Furthermore, a pulse of splicing inhibition elicits long-lasting repression of DDR proteins and enhances the cytotoxic effect of platinum-based drugs and poly(ADP-ribose) polymerase inhibitors (PARPis) in multiple TNBC models. These findings identify the U2 snRNP as an actionable target that can be exploited to enhance chemotherapy efficacy in TNBCs.


Asunto(s)
Daño del ADN , Empalme del ARN , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Humanos , Femenino , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Línea Celular Tumoral , Animales , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ratones , Empalmosomas/metabolismo , Empalmosomas/efectos de los fármacos
6.
Sci Total Environ ; 952: 175967, 2024 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-39226955

RESUMEN

Abnormal biosynthesis of spliceosomes and ribosomes can lead to their dysfunction, which in turn disrupts protein synthesis and results in various diseases. While genetic factors have been extensively studied, our understanding of how environmental compounds interfere with spliceosome and ribosome biosynthesis remains limited. In the present study, we employed a Reduced Transcriptome Analysis (RTA) approach, integrating large-scale transcriptome data sets of zebrafish and compiling a specific zebrafish gene panel focusing on the spliceosome and ribosome, to elucidate the potential disruptors targeting their biosynthesis. Transcriptomic data sets for 118 environmental substances and 1400 related gene expression profiles were integrated resulting in 513 exposure signatures. Among these substances, several categories including PCB126, transition metals Lanthanum (La) and praseodymium (Pr), heavy metals Cd2+ and AgNO3 and atrazine were highlighted for inducing the significant transcriptional alterations. Furthermore, we found that the transcriptional patterns were distinct between categories, yet overlapping patterns were generally observed within each group. For instance, over 82 % differentially expressed ribosomal genes were shared between La and Pr within the equivalent concentration range. Additionally, transcriptional complexities were also evident across various organs and developmental stages of zebrafish, with notable differences in the inhibition of the transcription of various spliceosome subunits. Overall, our results provide novel insights into the understanding of the adverse effects of environmental compounds, thereby contributing to their environmental risk assessments.


Asunto(s)
Ribosomas , Empalmosomas , Transcriptoma , Pez Cebra , Pez Cebra/genética , Animales , Empalmosomas/metabolismo , Empalmosomas/efectos de los fármacos , Ribosomas/metabolismo , Perfilación de la Expresión Génica , Contaminantes Químicos del Agua/toxicidad
7.
Cancer Lett ; 604: 217263, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39313128

RESUMEN

Approximately 50 % of poor prognosis neuroblastomas arise due to MYCN over-expression. We previously demonstrated that MYCN and PRMT5 proteins interact and PRMT5 knockdown led to apoptosis of MYCN-amplified (MNA) neuroblastoma. Here we evaluate the highly selective first-in-class PRMT5 inhibitor GSK3203591 and its in vivo analogue GSK3326593 as targeted therapeutics for MNA neuroblastoma. Cell-line analyses show MYCN-dependent growth inhibition and apoptosis, with approximately 200-fold greater sensitivity of MNA neuroblastoma lines. RNA sequencing of three MNA neuroblastoma lines treated with GSK3203591 reveal deregulated MYCN transcriptional programmes and altered mRNA splicing, converging on key regulatory pathways such as DNA damage response, epitranscriptomics and cellular metabolism. Stable isotope labelling experiments in the same cell lines demonstrate that glutamine metabolism is impeded following GSK3203591 treatment, linking with disruption of the MLX/Mondo nutrient sensors via intron retention of MLX mRNA. Interestingly, glutaminase (GLS) protein decreases after GSK3203591 treatment despite unchanged transcript levels. We demonstrate that the RNA methyltransferase METTL3 and cognate reader YTHDF3 proteins are lowered following their mRNAs undergoing GSK3203591-induced splicing alterations, indicating epitranscriptomic regulation of GLS; accordingly, we observe decreases of GLS mRNA m6A methylation following GSK3203591 treatment, and decreased GLS protein following YTHDF3 knockdown. In vivo efficacy of GSK3326593 is confirmed by increased survival of Th-MYCN mice, with drug treatment triggering splicing events and protein decreases consistent with in vitro data. Together our study demonstrates the PRMT5-dependent spliceosomal vulnerability of MNA neuroblastoma and identifies the epitranscriptome and glutamine metabolism as critical determinants of this sensitivity.


Asunto(s)
Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Proteína-Arginina N-Metiltransferasas , Empalmosomas , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/metabolismo , Humanos , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Línea Celular Tumoral , Empalmosomas/metabolismo , Empalmosomas/genética , Apoptosis , Regulación Neoplásica de la Expresión Génica , Epigénesis Genética , Animales , Transcriptoma , Metabolómica/métodos , Glutaminasa/genética , Glutaminasa/metabolismo , Ratones , Empalme del ARN , Proliferación Celular
8.
Curr Opin Struct Biol ; 88: 102907, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39168044

RESUMEN

Splicing is a critical processing step during pre-mRNA maturation in eukaryotes. The correct selection of splice sites during the early steps of spliceosome assembly is highly important and crucial for the regulation of alternative splicing. Splice site recognition and alternative splicing depend on cis-regulatory sequence elements in the RNA and trans-acting splicing factors that recognize these elements and crosstalk with the canonical splicing machinery. Structural mechanisms involving early spliceosome complexes are governed by dynamic RNA structures, protein-RNA interactions and conformational flexibility of multidomain RNA binding proteins. Here, we highlight structural studies and integrative structural biology approaches, which provide complementary information from cryo-EM, NMR, small angle scattering, and X-ray crystallography to elucidate mechanisms in the regulation of early spliceosome assembly and quality control, highlighting the role of conformational dynamics.


Asunto(s)
Empalmosomas , Empalmosomas/metabolismo , Empalmosomas/química , Humanos , Empalme del ARN , Unión Proteica , Animales , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Modelos Moleculares
9.
Biophys Chem ; 314: 107307, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39173313

RESUMEN

The two transesterification reactions of pre-mRNA splicing require highly complex yet well-controlled rearrangements of small nuclear RNAs and proteins (snRNP) in the spliceosome. The efficiency and accuracy of these reactions are critical for gene expression, as almost all human genes pass through pre-mRNA splicing. Key parameters that determine the splicing outcome are the length of the intron, the strengths of its splicing signals and gaps between them, and the presence of splicing controlling elements. In particular, the gap between the branchpoint (BP) and the 3' splice site (ss) of introns is a major determinant of the splicing efficiency. This distance falls within a small range across the introns of an organism. The constraints exist possibly because BP and 3'ss are recognized by BP-binding proteins, U2 snRNP and U2 accessory factors (U2AF) in a coordinated manner. Furthermore, varying distances between the two signals may also affect the second transesterification reaction since the intervening RNA needs to be accurately positioned within the complex RNP machinery. Splicing such pre-mRNAs requires cis-acting elements in the RNA and many trans-acting splicing regulators. Regulated pre-mRNA splicing with BP-distant 3'ss adds another layer of control to gene expression and promotes alternative splicing.


Asunto(s)
Intrones , Sitios de Empalme de ARN , Empalme del ARN , Humanos , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalmosomas/metabolismo , Empalmosomas/genética , Animales
10.
RNA ; 30(11): 1397-1407, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39187383

RESUMEN

The spliceosome is a eukaryotic multimegadalton RNA-protein complex that removes introns from transcripts. The spliceosome ensures the selection of each exon-intron boundary through multiple recognition events. Initially, the 5' splice site (5' SS) and branch site (BS) are bound by the U1 small nuclear ribonucleoprotein (snRNP) and the U2 snRNP, respectively, while the 3' SS is mostly determined by proximity to the branch site. A large number of splicing factors recognize the splice sites and recruit the snRNPs before the stable binding of the snRNPs occurs by base-pairing the snRNA to the transcript. Fidelity of this process is crucial, as mutations in splicing factors and U2 snRNP components are associated with many diseases. In recent years, major advances have been made in understanding how splice sites are selected in Saccharomyces cerevisiae and humans. Here, I review and discuss the current understanding of the recognition of splice sites by the spliceosome with a focus on recognition and binding of the branch site by the U2 snRNP in humans.


Asunto(s)
Sitios de Empalme de ARN , Empalme del ARN , Saccharomyces cerevisiae , Empalmosomas , Empalmosomas/metabolismo , Empalmosomas/genética , Humanos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Intrones , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/genética , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/química , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/química , Unión Proteica
11.
Genomics ; 116(5): 110917, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39147335

RESUMEN

Alternative splicing (AS) has significant clinical relevance with cancers and is a potential source of neoepitopes. In this study, RNA-seq data of 94 solid tumor and matched adjacent normal tissues from 47 clinical patients covering nine cancer types were comprehensively analyzed using SUVA developed by ourselves. The results identified highly conserved pan-cancer differential alternative splicing (DAS) events and cancer-specific DAS events in a series of tumor samples, which in turn revealed the heterogeneity of AS post-transcriptional regulation across different cancers. The co-disturbed network between spliceosome factors (SFs) and common cancer-associated DAS was further constructed, suggesting the potential possibility of the regulation of differentially expressed SFs on DAS. Finally, the common cancer-associated DAS events were fully validated using the TCGA dataset, confirming the significant correlation between cancer-associated DAS and prognosis. Briefly, our study elucidates new insights into conservatived and specific DAS in cancer, providing valuable resources for cancer therapeutic targets.


Asunto(s)
Empalme Alternativo , Neoplasias , Humanos , Neoplasias/genética , Empalmosomas/metabolismo , Empalmosomas/genética
12.
Mol Cell ; 84(14): 2618-2633.e10, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39025073

RESUMEN

The twenty-three Fanconi anemia (FA) proteins cooperate in the FA/BRCA pathway to repair DNA interstrand cross-links (ICLs). The cell division cycle and apoptosis regulator 1 (CCAR1) protein is also a regulator of ICL repair, though its possible function in the FA/BRCA pathway remains unknown. Here, we demonstrate that CCAR1 plays a unique upstream role in the FA/BRCA pathway and is required for FANCA protein expression in human cells. Interestingly, CCAR1 co-immunoprecipitates with FANCA pre-mRNA and is required for FANCA mRNA processing. Loss of CCAR1 results in retention of a poison exon in the FANCA transcript, thereby leading to reduced FANCA protein expression. A unique domain of CCAR1, the EF hand domain, is required for interaction with the U2AF heterodimer of the spliceosome and for excision of the poison exon. Taken together, CCAR1 is a splicing modulator required for normal splicing of the FANCA mRNA and other mRNAs involved in various cellular pathways.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Proteínas de Ciclo Celular , Proteína del Grupo de Complementación A de la Anemia de Fanconi , Anemia de Fanconi , Empalme del ARN , Factor de Empalme U2AF , Humanos , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Proteína BRCA2/metabolismo , Proteína BRCA2/genética , Reparación del ADN , Endodesoxirribonucleasas , Exones , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Células HEK293 , Células HeLa , Unión Proteica , Precursores del ARN/metabolismo , Precursores del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Empalmosomas/metabolismo , Empalmosomas/genética , Factor de Empalme U2AF/metabolismo , Factor de Empalme U2AF/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo
13.
Plant Sci ; 347: 112199, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39038708

RESUMEN

U6 snRNA is one of the uridine-rich non-coding RNAs, abundant and stable in various cells, function as core particles in the intron-lariat spliceosome (ILS) complex. The Increased Level of Polyploidy1-1D (ILP1) and NTC-related protein 1 (NTR1), two conserved disassembly factors of the ILS complex, facilitates the disintegration of the ILS complex after completing intron splicing. The functional impairment of ILP1 and NTR1 lead to increased U6 levels, while other snRNAs comprising the ILS complex remained unaffected. We revealed that ILP1 and NTR1 had no impact on the transcription, 3' end phosphate structure or oligo(U) tail of U6 snRNA. Moreover, we uncovered that the mutation of ILP1 and NTR1 resulted in the accumulation of ILS complexes, impeding the dissociation of U6 from splicing factors, leading to an extended half-life of U6 and ultimately causing an elevation in U6 snRNA levels. Our findings broaden the understanding of the functions of ILS disassembly factors ILP1 and NTR1, and providing insights into the dynamic disassembly between U6 and ILS.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , ARN Nuclear Pequeño , Empalmosomas , Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Empalme del ARN , Estabilidad del ARN/genética , ARN de Planta/metabolismo , ARN de Planta/genética , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/genética , Empalmosomas/metabolismo
14.
Nucleic Acids Res ; 52(17): 10668-10682, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39011894

RESUMEN

RNA helicases are involved in RNA metabolism in an ATP-dependent manner. Although many RNA helicases unwind the RNA structure and/or remove proteins from the RNA, some can load their interacting proteins onto RNAs. Here, we developed an in vitro strategy to identify the ATP-dependent factors involved in spliceosomal uridine-rich small nuclear RNA (U snRNA) export. We identified the RNA helicase UAP56/DDX39B, a component of the mRNA export complex named the transcription-export (TREX) complex, and its closely related RNA helicase URH49/DDX39A as the factors that stimulated RNA binding of PHAX, an adapter protein for U snRNA export. ALYREF, another TREX component, acted as a bridge between PHAX and UAP56/DDX39B. We also showed that UAP56/DDX39B and ALYREF participate in U snRNA export through a mechanism distinct from that of mRNA export. This study describes a novel aspect of the TREX components for U snRNP biogenesis and highlights the loading activity of RNA helicases.


Asunto(s)
Transporte Activo de Núcleo Celular , ARN Helicasas DEAD-box , ARN Nuclear Pequeño , Empalmosomas , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/genética , Empalmosomas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Células HeLa , Núcleo Celular/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Células HEK293 , Fosfoproteínas , Factores de Transcripción
15.
Protein Sci ; 33(8): e5117, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39023093

RESUMEN

In eukaryotes, pre-mRNA splicing is vital for RNA processing and orchestrated by the spliceosome, whose assembly starts with the interaction between U1-70K and SR proteins. Despite the significance of the U1-70K/SR interaction, the dynamic nature of the complex and the challenges in obtaining soluble U1-70K have impeded a comprehensive understanding of the interaction at the structural level for decades. We overcome the U1-70K solubility issues, enabling us to characterize the interaction between U1-70K and SRSF1, a representative SR protein. We unveil specific interactions: phosphorylated SRSF1 RS with U1-70K BAD1, and SRSF1 RRM1 with U1-70K RRM. The RS/BAD1 interaction plays a dominant role, whereas the interaction between the RRM domains further enhances the stability of the U1-70K/SRSF1 complex. The RRM interaction involves the C-terminal extension of U1-70K RRM and the conserved acid patches on SRSF1 RRM1 that is involved in SRSF1 phase separation. Our circular dichroism spectra reveal that BAD1 adapts an α-helical conformation and RS is intrinsically disordered. Intriguingly, BAD1 undergoes a conformation switch from α-helix to ß-strand and random coil upon RS binding. In addition to the regulatory mechanism via SRSF1 phosphorylation, the U1-70K/SRSF1 interaction is also regulated by U1-70K BAD1 phosphorylation. We find that U1-70K phosphorylation inhibits the U1-70K and SRSF1 interaction. Our structural findings are validated through in vitro splicing assays and in-cell saturated domain scanning using the CRISPR method, providing new insights into the intricate regulatory mechanisms of pre-mRNA splicing.


Asunto(s)
Ribonucleoproteína Nuclear Pequeña U1 , Factores de Empalme Serina-Arginina , Empalmosomas , Factores de Empalme Serina-Arginina/metabolismo , Factores de Empalme Serina-Arginina/química , Factores de Empalme Serina-Arginina/genética , Fosforilación , Empalmosomas/metabolismo , Empalmosomas/química , Humanos , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/genética , Empalme del ARN , Unión Proteica , Precursores del ARN/metabolismo , Precursores del ARN/genética , Precursores del ARN/química
16.
BMC Biol ; 22(1): 153, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38982460

RESUMEN

Pre-mRNA splicing is a significant step for post-transcriptional modifications and functions in a wide range of physiological processes in plants. Human NHP2L binds to U4 snRNA during spliceosome assembly; it is involved in RNA splicing and mediates the development of human tumors. However, no ortholog has yet been identified in plants. Therefore, we report At4g12600 encoding the ortholog NHP2L protein, and AtSNU13 associates with the component of the spliceosome complex; the atsnu13 mutant showed compromised resistance in disease resistance, indicating that AtSNU13 is a positive regulator of plant immunity. Compared to wild-type plants, the atsnu13 mutation resulted in altered splicing patterns for defense-related genes and decreased expression of defense-related genes, such as RBOHD and ALD1. Further investigation shows that AtSNU13 promotes the interaction between U4/U6.U5 tri-snRNP-specific 27 K and the motif in target mRNAs to regulate the RNA splicing. Our study highlights the role of AtSNU13 in regulating plant immunity by affecting the pre-mRNA splicing of defense-related genes.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Inmunidad de la Planta , Precursores del ARN , Empalme del ARN , Arabidopsis/genética , Arabidopsis/inmunología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Regulación de la Expresión Génica de las Plantas , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/inmunología , Inmunidad de la Planta/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalmosomas/metabolismo , Empalmosomas/genética
17.
Adv Sci (Weinh) ; 11(35): e2403387, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39018261

RESUMEN

RNA splicing is a dynamic molecular process in response to environmental stimuli and is strictly regulated by the spliceosome. Sm proteins, constituents of the spliceosome, are key components that mediate splicing reactions; however, their potential role in hepatocellular carcinoma (HCC) is poorly understood. In the study, SNRPD2 (PD2) is found to be the most highly upregulated Sm protein in HCC and to act as an oncogene. PD2 modulates DDX39A intron retention together with HNRNPL to sustain the DDX39A short variant (39A_S) expression. Mechanistically, 39A_S can mediate MYC mRNA nuclear export to maintain high MYC protein expression, while MYC in turn potentiates PD2 transcription. Importantly, digitoxin can directly interact with PD2 and has a notable cancer-suppressive effect on HCC. The study reveals a novel mechanism by which DDX39A senses oncogenic MYC signaling and undergoes splicing via PD2 to form a positive feedback loop in HCC, which can be targeted by digitoxin.


Asunto(s)
Carcinoma Hepatocelular , ARN Helicasas DEAD-box , Intrones , Neoplasias Hepáticas , Empalme del ARN , Empalmosomas , Animales , Humanos , Masculino , Ratones , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Intrones/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Empalme del ARN/genética , Empalmosomas/genética , Empalmosomas/metabolismo
18.
Nat Commun ; 15(1): 5752, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982135

RESUMEN

The early-life organ development and maturation shape the fundamental blueprint for later-life phenotype. However, a multi-organ proteome atlas from infancy to adulthood is currently not available. Herein, we present a comprehensive proteomic analysis of ten mouse organs (brain, heart, lung, liver, kidney, spleen, stomach, intestine, muscle and skin) at three crucial developmental stages (1-, 4- and 8-weeks after birth) acquired using data-independent acquisition mass spectrometry. We detect and quantify 11,533 protein groups across the ten organs and obtain 115 age-related differentially expressed protein groups that are co-expressed in all organs from infancy to adulthood. We find that spliceosome proteins prevalently play crucial regulatory roles in the early-life development of multiple organs, and detect organ-specific expression patterns and sexual dimorphism. This multi-organ proteome atlas provides a fundamental resource for understanding the molecular mechanisms underlying early-life organ development and maturation.


Asunto(s)
Proteoma , Proteómica , Animales , Proteoma/metabolismo , Ratones , Femenino , Masculino , Proteómica/métodos , Riñón/metabolismo , Riñón/crecimiento & desarrollo , Empalmosomas/metabolismo , Especificidad de Órganos , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Hígado/metabolismo , Pulmón/metabolismo , Pulmón/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Caracteres Sexuales , Bazo/metabolismo , Bazo/crecimiento & desarrollo
19.
Nature ; 632(8024): 261-262, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39048865
20.
Nat Commun ; 15(1): 6348, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39068178

RESUMEN

The spliceosome executes pre-mRNA splicing through four sequential stages: assembly, activation, catalysis, and disassembly. Activation of the spliceosome, namely remodeling of the pre-catalytic spliceosome (B complex) into the activated spliceosome (Bact complex) and the catalytically activated spliceosome (B* complex), involves major flux of protein components and structural rearrangements. Relying on a splicing inhibitor, we have captured six intermediate states between the B and B* complexes: pre-Bact, Bact-I, Bact-II, Bact-III, Bact-IV, and post-Bact. Their cryo-EM structures, together with an improved structure of the catalytic step I spliceosome (C complex), reveal how the catalytic center matures around the internal stem loop of U6 snRNA, how the branch site approaches 5'-splice site, how the RNA helicase PRP2 rearranges to bind pre-mRNA, and how U2 snRNP undergoes remarkable movement to facilitate activation. We identify a previously unrecognized key role of PRP2 in spliceosome activation. Our study recapitulates a molecular choreography of the human spliceosome during its catalytic activation.


Asunto(s)
Microscopía por Crioelectrón , Precursores del ARN , Empalme del ARN , ARN Nuclear Pequeño , Empalmosomas , Empalmosomas/metabolismo , Humanos , Precursores del ARN/metabolismo , Precursores del ARN/genética , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/genética , Modelos Moleculares , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , Dominio Catalítico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...