Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 574(7777): 273-277, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31578525

RESUMEN

Transcription and pre-mRNA splicing are key steps in the control of gene expression and mutations in genes regulating each of these processes are common in leukaemia1,2. Despite the frequent overlap of mutations affecting epigenetic regulation and splicing in leukaemia, how these processes influence one another to promote leukaemogenesis is not understood and, to our knowledge, there is no functional evidence that mutations in RNA splicing factors initiate leukaemia. Here, through analyses of transcriptomes from 982 patients with acute myeloid leukaemia, we identified frequent overlap of mutations in IDH2 and SRSF2 that together promote leukaemogenesis through coordinated effects on the epigenome and RNA splicing. Whereas mutations in either IDH2 or SRSF2 imparted distinct splicing changes, co-expression of mutant IDH2 altered the splicing effects of mutant SRSF2 and resulted in more profound splicing changes than either mutation alone. Consistent with this, co-expression of mutant IDH2 and SRSF2 resulted in lethal myelodysplasia with proliferative features in vivo and enhanced self-renewal in a manner not observed with either mutation alone. IDH2 and SRSF2 double-mutant cells exhibited aberrant splicing and reduced expression of INTS3, a member of the integrator complex3, concordant with increased stalling of RNA polymerase II (RNAPII). Aberrant INTS3 splicing contributed to leukaemogenesis in concert with mutant IDH2 and was dependent on mutant SRSF2 binding to cis elements in INTS3 mRNA and increased DNA methylation of INTS3. These data identify a pathogenic crosstalk between altered epigenetic state and splicing in a subset of leukaemias, provide functional evidence that mutations in splicing factors drive myeloid malignancy development, and identify spliceosomal changes as a mediator of IDH2-mutant leukaemogenesis.


Asunto(s)
Empalme Alternativo/genética , Carcinogénesis/genética , Epigénesis Genética , Leucemia Mieloide Aguda/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Mutación/genética , ARN Polimerasa II/metabolismo , Factores de Empalme Serina-Arginina/genética , Transcriptoma
3.
Nature ; 510(7505): 412-6, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24814343

RESUMEN

The global shortening of messenger RNAs through alternative polyadenylation (APA) that occurs during enhanced cellular proliferation represents an important, yet poorly understood mechanism of regulated gene expression. The 3' untranslated region (UTR) truncation of growth-promoting mRNA transcripts that relieves intrinsic microRNA- and AU-rich-element-mediated repression has been observed to correlate with cellular transformation; however, the importance to tumorigenicity of RNA 3'-end-processing factors that potentially govern APA is unknown. Here we identify CFIm25 as a broad repressor of proximal poly(A) site usage that, when depleted, increases cell proliferation. Applying a regression model on standard RNA-sequencing data for novel APA events, we identified at least 1,450 genes with shortened 3' UTRs after CFIm25 knockdown, representing 11% of significantly expressed mRNAs in human cells. Marked increases in the expression of several known oncogenes, including cyclin D1, are observed as a consequence of CFIm25 depletion. Importantly, we identified a subset of CFIm25-regulated APA genes with shortened 3' UTRs in glioblastoma tumours that have reduced CFIm25 expression. Downregulation of CFIm25 expression in glioblastoma cells enhances their tumorigenic properties and increases tumour size, whereas CFIm25 overexpression reduces these properties and inhibits tumour growth. These findings identify a pivotal role of CFIm25 in governing APA and reveal a previously unknown connection between CFIm25 and glioblastoma tumorigenicity.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/fisiopatología , Poliadenilación , ARN Mensajero/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Regiones no Traducidas 3' , Animales , Carcinogénesis/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Xenoinjertos , Humanos , Masculino , Ratones , Análisis de Regresión
4.
Nucleic Acids Res ; 46(8): 4241-4255, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29471365

RESUMEN

Integrator (INT) is a transcriptional regulatory complex associated with RNA polymerase II that is required for the 3'-end processing of both UsnRNAs and enhancer RNAs. Integrator subunits 9 (INTS9) and INTS11 constitute the catalytic core of INT and are paralogues of the cleavage and polyadenylation specificity factors CPSF100 and CPSF73. While CPSF73/100 are known to associate with a third protein called Symplekin, there is no paralog of Symplekin within INT raising the question of how INTS9/11 associate with the other INT subunits. Here, we have identified that INTS4 is a specific and conserved interaction partner of INTS9/11 that does not interact with either subunit individually. Although INTS4 has no significant homology with Symplekin, it possesses N-terminal HEAT repeats similar to Symplekin but also contains a ß-sheet rich C-terminal region, both of which are important to bind INTS9/11. We assess three functions of INT including UsnRNA 3'-end processing, maintenance of Cajal body structural integrity, and formation of histone locus bodies to conclude that INTS4/9/11 are the most critical of the INT subunits for UsnRNA biogenesis. Altogether, these results indicate that INTS4/9/11 compose a heterotrimeric complex that likely represents the Integrator 'cleavage module' responsible for its endonucleolytic activity.


Asunto(s)
Endorribonucleasas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Drosophila/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/química , ARN Nuclear Pequeño/metabolismo , Técnicas del Sistema de Dos Híbridos
5.
Proc Natl Acad Sci U S A ; 114(17): 4394-4399, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396433

RESUMEN

The metazoan Integrator complex (INT) has important functions in the 3'-end processing of noncoding RNAs, including the uridine-rich small nuclear RNA (UsnRNA) and enhancer RNA (eRNA), and in the transcription of coding genes by RNA polymerase II. The INT contains at least 14 subunits, but its molecular mechanism of action is poorly understood, because currently there is little structural information about its subunits. The endonuclease activity of INT is mediated by its subunit 11 (IntS11), which belongs to the metallo-ß-lactamase superfamily and is a paralog of CPSF-73, the endonuclease for pre-mRNA 3'-end processing. IntS11 forms a stable complex with Integrator complex subunit 9 (IntS9) through their C-terminal domains (CTDs). Here, we report the crystal structure of the IntS9-IntS11 CTD complex at 2.1-Å resolution and detailed, structure-based biochemical and functional studies. The complex is composed of a continuous nine-stranded ß-sheet with four strands from IntS9 and five from IntS11. Highly conserved residues are located in the extensive interface between the two CTDs. Yeast two-hybrid assays and coimmunoprecipitation experiments confirm the structural observations on the complex. Functional studies demonstrate that the IntS9-IntS11 interaction is crucial for the role of INT in snRNA 3'-end processing.


Asunto(s)
Endorribonucleasas/metabolismo , Cristalización , Endorribonucleasas/genética , Escherichia coli/metabolismo , Regulación Enzimológica de la Expresión Génica , Inmunoprecipitación , Modelos Moleculares , Unión Proteica , Conformación Proteica , ARN Nuclear Pequeño/metabolismo , Técnicas del Sistema de Dos Híbridos , Difracción de Rayos X
6.
RNA ; 18(12): 2148-56, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23097424

RESUMEN

Formation of the 3' end of RNA polymerase II-transcribed snRNAs requires a poorly understood group of proteins called the Integrator complex. Here we used a fluorescence-based read-through reporter that expresses GFP in response to snRNA misprocessing and performed a genome-wide RNAi screen in Drosophila S2 cells to identify novel factors required for snRNA 3'-end formation. In addition to the known Integrator complex members, we identified Asunder and CG4785 as additional Integrator subunits. Functional and biochemical experiments revealed that Asunder and CG4785 are additional core members of the Integrator complex. We also identified a conserved requirement in both fly and human snRNA 3'-end processing for cyclin C and Cdk8 that is distinct from their function in the Mediator Cdk8 module. Moreover, we observed biochemical association between Integrator proteins and cyclin C/Cdk8, and that overexpression of a kinase-dead Cdk8 causes snRNA misprocessing. These data functionally define the Drosophila Integrator complex and demonstrate an additional function for cyclin C/Cdk8 unrelated to its function in Mediator.


Asunto(s)
Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ciclina C/metabolismo , Quinasa 8 Dependiente de Ciclina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Animales , Línea Celular , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Complejo Mediador/genética , Complejo Mediador/metabolismo , Procesamiento de Término de ARN 3' , Interferencia de ARN , Especificidad de la Especie
7.
J Hematol Oncol ; 9: 30, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-27025456

RESUMEN

The t(11;14) translocation resulting in constitutive cyclin D1 expression is an early event in mantle cell lymphoma (MCL) transformation. Patients with a highly proliferative phenotype produce cyclin D1 transcripts with truncated 3'UTRs that evade miRNA regulation. Here, we report the recurrence of a novel gene fusion in MCL cell lines and MCL patient isolates that consists of the full protein coding region of cyclin D1 (CCND1) and a 3'UTR consisting of sequences from both the CCND1 3'UTR and myotonic dystrophy kinase-related Cdc42-binding kinase's (MRCK) intron one. The resulting CCND1/MRCK mRNA is resistant to CCND1-targeted miRNA regulation, and targeting the MRCK region of the chimeric 3'UTR with siRNA results in decreased CCND1 levels.


Asunto(s)
Ciclina D1/genética , Fusión Génica , Linfoma de Células del Manto/genética , Proteína Quinasa de Distrofia Miotónica/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Ciclina D1/metabolismo , Células HEK293 , Células HeLa , Humanos , Intrones/genética , Linfoma de Células del Manto/metabolismo , Linfoma de Células del Manto/patología , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Proteína Quinasa de Distrofia Miotónica/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Mol Biol Cell ; 24(18): 2954-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23904267

RESUMEN

We previously reported that Asunder (ASUN) is essential for recruitment of dynein motors to the nuclear envelope (NE) and nucleus-centrosome coupling at the onset of cell division in cultured human cells and Drosophila spermatocytes, although the mechanisms underlying this regulation remain unknown. We also identified ASUN as a functional component of Integrator (INT), a multisubunit complex required for 3'-end processing of small nuclear RNAs. We now provide evidence that ASUN acts in the nucleus in concert with other INT components to mediate recruitment of dynein to the NE. Knockdown of other individual INT subunits in HeLa cells recapitulates the loss of perinuclear dynein in ASUN-small interfering RNA cells. Forced localization of ASUN to the cytoplasm via mutation of its nuclear localization sequence blocks its capacity to restore perinuclear dynein in both cultured human cells lacking ASUN and Drosophila asun spermatocytes. In addition, the levels of several INT subunits are reduced at G2/M when dynein is recruited to the NE, suggesting that INT does not directly mediate this step. Taken together, our data support a model in which a nuclear INT complex promotes recruitment of cytoplasmic dynein to the NE, possibly via a mechanism involving RNA processing.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Dineínas Citoplasmáticas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Complejos Multiproteicos/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas de Ciclo Celular/química , División Celular , Proteínas de Drosophila/química , Drosophila melanogaster/citología , Fase G2 , Células HeLa , Humanos , Masculino , Datos de Secuencia Molecular , Membrana Nuclear/metabolismo , Señales de Localización Nuclear/metabolismo , Subunidades de Proteína/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Espermatocitos/citología , Espermatocitos/metabolismo , Fracciones Subcelulares/metabolismo
9.
Biol Open ; 2(12): 1390-6, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24285713

RESUMEN

We previously reported that the small nuclear RNA processing complex, Integrator, is required for dynein recruitment to the nuclear envelope at mitotic onset in cultured human cells. We now report an additional role for INT in ciliogenesis. Depletion of INT subunits from cultured human cells results in loss of primary cilia. We provide evidence that the requirements for INT in dynein localization and ciliogenesis are uncoupled: proteins essential for ciliogenesis are not essential for dynein recruitment to the nuclear envelope, while depletion of known regulators of perinuclear dynein has minimal effects on ciliogenesis. Taken together, our data support a model in which INT ensures proper processing of distinct pools of transcripts encoding components that independently promote perinuclear dynein enrichment and ciliogenesis.

10.
Mol Cell Biol ; 32(6): 1112-23, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22252320

RESUMEN

The Integrator Complex is a group of proteins responsible for the endonucleolytic cleavage of primary small nuclear RNA (snRNA) transcripts within the nucleus. Integrator subunits 9 and 11 (IntS9/11) are thought to contain the catalytic activity based on their high sequence similarity to CPSF100 and CPSF73, which have been shown to be components of both the poly(A)(+) and histone pre-mRNA cleavage complex. Here we demonstrate that the specific heterodimeric interaction between IntS9 and IntS11 is mediated by a discrete domain present at the extreme C terminus of IntS9 and within the C terminus of IntS11, adjacent to the predicted active site of this endonuclease. This domain is highly conserved within IntS11 but conspicuously absent in CPSF73. Using a cell-based complementation assay that measures Integrator activity, we determined that the IntS9 interaction domain within IntS11 is required for its ability to restore snRNA 3' end processing after RNA interference (RNAi)-mediated depletion of IntS11. Moreover, overexpression of these interaction domains alone elicits snRNA misprocessing through a dominant-negative titration of endogenous Integrator subunits. These data collectively explain the mechanism by which the IntS11/9 and, by analogy, the CPSF73/100 heterodimeric cleavage factors distinguish themselves from each other and demonstrate that the heterodimeric interaction is functionally required for snRNA 3' end formation.


Asunto(s)
Proteínas Portadoras/metabolismo , Endorribonucleasas/metabolismo , Procesamiento de Término de ARN 3' , ARN Nuclear Pequeño/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/genética , Endorribonucleasas/química , Endorribonucleasas/genética , Expresión Génica , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA