Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 33(12): 3621-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26303183

RESUMEN

The intestinal epithelium is very peculiar for its continuous cell renewal, fuelled by multipotent stem cells localized within the crypts of Lieberkühn. Several lines of evidence have established the evolutionary conserved RNA-binding protein Musashi1 as a marker of adult stem cells, including those of the intestinal epithelium, and revealed its roles in stem cell self-renewal and cell fate determination. Previous studies from our laboratories have shown that Musashi1 controls stem cell-like features in medulloblastoma, glioblastoma, and breast cancer cells, and has pro-proliferative and pro-tumorigenic properties in intestinal epithelial progenitor cells in vitro. To undertake a detailed study of Musashi1's function in the intestinal epithelium in vivo, we have generated a mouse model, referred to as v-Msi, overexpressing Musashi1 specifically in the entire intestinal epithelium. Compared with wild type litters, v-Msi1 mice exhibited increased intestinal crypt size accompanied by enhanced proliferation. Comparative transcriptomics by RNA-seq revealed Musashi1's association with gut stem cell signature, cell cycle, DNA replication, and drug metabolism. Finally, we identified and validated three novel mRNA targets that are stabilized by Musashi1, Ccnd1 (Cyclin D1), Cdk6, and Sox4. In conclusion, the targeted expression of Musashi1 in the intestinal epithelium in vivo increases the cell proliferation rate and strongly suggests its action on stem cells activity. This is due to the modulation of a complex network of gene functions and pathways including drug metabolism, cell cycle, and DNA synthesis and repair.


Asunto(s)
Ciclo Celular , Mucosa Intestinal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Madre/metabolismo , Animales , Marcación de Gen , Mucosa Intestinal/citología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Células Madre/citología
3.
Mol Cell Biol ; 21(6): 1986-96, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11238934

RESUMEN

Maintenance of female sexual identity in Drosophila melanogaster involves an autoregulatory loop in which the protein Sex-lethal (SXL) promotes skipping of exon 3 from its own pre-mRNA. We have used transient transfection of Drosophila Schneider cells to analyze the role of exon 3 splice sites in regulation. Our results indicate that exon 3 repression requires competition between the 5' splice sites of exons 2 and 3 but is independent of their relative strength. Two 3' splice site AG's precede exon 3. We report here that, while the distal site plays a critical role in defining the exon, the proximal site is preferentially used for the actual splicing reaction, arguing for a switch in 3' splice site recognition between exon definition and splicing catalysis. Remarkably, the presence of the two 3' splice sites is important for the efficient regulation by SXL, suggesting that SXL interferes with molecular events occurring between initial splice site communication across the exon and the splice site pairing that leads to intron removal.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila , Hormonas de Insectos/genética , Sitios de Empalme de ARN , Proteínas de Unión al ARN/genética , Nucleótidos de Adenina , Animales , Secuencia de Bases , Catálisis , Células Cultivadas , Drosophila/genética , Exones , Hormonas de Insectos/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas de Unión al ARN/metabolismo
4.
Genetics ; 155(1): 129-39, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10790389

RESUMEN

The Drosophila gene female-lethal(2)d [fl(2)d] interacts genetically with the master regulatory gene for sex determination, Sex-lethal. Both genes are required for the activation of female-specific patterns of alternative splicing on transformer and Sex-lethal pre-mRNAs. We have used P-element-mediated mutagenesis to identify the fl(2)d gene. The fl(2)d transcription unit generates two alternatively spliced mRNAs that can encode two protein isoforms differing at their amino terminus. The larger isoform contains a domain rich in histidine and glutamine but has no significant homology to proteins in databases. Several lines of evidence indicate that this protein is responsible for fl(2)d function. First, the P-element insertion that inactivates fl(2)d interrupts this ORF. Second, amino acid changes within this ORF have been identified in fl(2)d mutants, and the nature of the changes correlates with the severity of the mutations. Third, all of the phenotypes associated with fl(2)d mutations can be rescued by expression of this cDNA in transgenic flies. Fl(2)d protein can be detected in extracts from Drosophila cell lines, embryos, larvae, and adult animals, without apparent differences between sexes, as well as in adult ovaries. Consistent with a possible function in posttranscriptional regulation, Fl(2)d protein has nuclear localization and is enriched in nuclear extracts.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila , Glutamina/metabolismo , Histidina/metabolismo , Proteínas de Insectos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Precursores del ARN , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , ADN Complementario , Drosophila/genética , Femenino , Genes de Insecto/genética , Glutamina/genética , Histidina/genética , Humanos , Proteínas de Insectos/genética , Datos de Secuencia Molecular , Transcripción Genética
5.
Proc Natl Acad Sci U S A ; 94(14): 7343-8, 1997 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-9207093

RESUMEN

The protein Sex-lethal (SXL) controls pre-mRNA splicing of two genes involved in Drosophila sex determination: transformer (tra) and the Sxl gene itself. Previous in vitro results indicated that SXL antagonizes the general splicing factor U2AF65 to regulate splicing of tra. In this report, we have used transgenic flies expressing chimeric proteins between SXL and the effector domain of U2AF65 to study the mechanisms of splicing regulation by SXL in vivo. Conferring U2AF activity to SXL relieves its inhibitory activity on tra splicing but not on Sxl splicing. Therefore, antagonizing U2AF65 can explain tra splicing regulation both in vitro and in vivo, but this mechanism cannot explain splicing regulation of Sxl pre-mRNA. These results are a direct proof that Sxl, the master regulatory gene in sex determination, has multiple and separable activities in the regulation of pre-mRNA splicing.


Asunto(s)
Proteínas de Drosophila , Drosophila/genética , Genes de Insecto , Empalme del ARN , Proteínas de Unión al ARN/genética , Animales , Hormonas de Insectos/genética , ARN Mensajero/genética
6.
Gene ; 193(2): 163-72, 1997 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-9256073

RESUMEN

We have mapped a region of about 33 kb which includes the transcription unit of the C-3 DNA puff gene of Rhynchosciara americana. The C-3 TU and a region extending approximately 800 bp upstream of the C-3 promoter were characterized. The TU is composed of three exons and produces a 1.1-kb mRNA whose level in salivary glands increases with the expansion of the C-3 puff. The C-3 messenger appears to undergo rapid deadenylation resulting in an RNA of about 0.95 kb which can still be observed in gland cells 15 h after the puff has regressed. The 1.1-kb mRNA codes for a 32.4-kDa, predominantly alpha-helical polypeptide with three conserved parallel coiled-coil stretches. The aa composition and structure of this polypeptide suggests that it is secreted and contributes to the formation of the cocoon in which the larvae pupate. The region upstream of the promoter contains several A-rich sequences with similarity to the ACS of yeast which might have a role in the initiation of replication/amplification.


Asunto(s)
Dípteros/genética , Genes de Insecto , Proteínas de Insectos , Proteínas y Péptidos Salivales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Transcripción Genética
7.
Genetics ; 144(4): 1653-64, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8978052

RESUMEN

The Drosophila gene Sex-lethal (Sxl) controls the processes of sex determination and dosage compensation. A Drosophila subobscura genomic fragment containing all the exons and the late and early promotors in the Sxl gene of D. melanogaster was isolated. Early Sxl expression in D. subobscura seems to be controlled at the transcriptional level, possibly by the X:A signal. In the region upstream of the early Sxl transcription initiation site are two conserved regions suggested to be involved in the early activation of Sxl. Late Sxl expression in D. subobscura produces four transcripts in adult females and males. In males, the transcripts have an additional exon which contains three translational stop codons so that a truncated, presumably nonfunctional Sxl protein is produced. The Sxl pre-mRNA of D. subobscura lacks the poly-U sequence presented at the polypirimidine tract of the 3' splice site of the male-specific exon present in D. melanogaster. Introns 2 and 3 contain the Sxl-binding poly-U stretches, whose localization in intron 2 varies but in intron 3 is conserved. The Sxl protein is fully conserved at the amino acid level in both species.


Asunto(s)
Proteínas de Drosophila , Drosophila/genética , Proteínas de Unión al ARN/genética , Análisis para Determinación del Sexo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Femenino , Dosificación de Gen , Hormonas de Insectos/genética , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia
8.
Mol Gen Genet ; 253(1-2): 26-31, 1996 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-9003283

RESUMEN

In Drosophila melanogaster, regulation of the sex determination genes throughout development occurs by sex-specific splicing of their products. The first gene is Sex-lethal(Sxl). The downstream target of Sxl is the gene transformer (tra): the Sxl protein controls the female-specific splicing of the Tra pre-mRNA. The downstream target of the gene tra is the gene double-sex (dsx): the Tra protein of females, controls the female-specific splicing of the Dsx pre-mRNA. We have identified a gene, female-lethal-2-d fl(2) d, whose function is required for the female-specific splicing of Sxl pre-mRNA. In this report we analyze whether the gene fl(2)d is also required for the sex-specific splicing of both Tra and Dsx pre-mRNAs. We found that the Sxl protein is not sufficient for the female-specific splicing of Tra pre-mRNA, the fl(2)d function also being necessary. This gene, however, is not required for the female-specific splicing of Dsx pre-mRNA.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila , Drosophila melanogaster/genética , Genes de Insecto , Hormonas de Insectos/genética , Proteínas Nucleares/genética , Empalme del ARN , Proteínas de Unión al ARN/genética , Animales , Animales Modificados Genéticamente , Femenino , Masculino , Precursores del ARN/genética , Caracteres Sexuales , Temperatura
9.
Mol Gen Genet ; 250(1): 89-96, 1996 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8569692

RESUMEN

Crosses between Drosophila melanogaster females and D. simulans males produce viable hybrid females, while males are lethal. These males are rescued if they carry the D. simulans Lhr gene. This paper reports that females of the wild-type D. melanogaster population Staket do not produce viable hybrid males when crossed with D. simulans Lhr males, a phenomenon which we designate as the Staket phenotype. The agent responsible for this phenomenon was found to be the Staket X chromosome (Xmel, Stk). Analysis of the Staket phenotype showed that it is suppressed by extra copies of D. melanogaster rDNA genes and that the Xmel, Stk chromosome manifests a weak bobbed phenotype in D. melanogaster Xmel, Stk/0 males. The numbers of functional rDNA genes in Xmel, Stk and Xmel, y w (control) chromosomes were found not to differ significantly. Thus a reduction in rDNA gene number cannot account for the weak bobbed Xmel, Stk phenotype let alone the Staket phenotype. The rRNA precursor molecules transcribed from the Xmel, Stk rDNA genes seem to be correctly processed in both intraspecific (melanogaster) and interspecific (melanogaster-simulans) conditions. It is therefore suggested that the Xmel, Stk rDNA genes are inefficiently transcribed in the melanogaster-simulans hybrids.


Asunto(s)
ADN Ribosómico , Drosophila melanogaster/genética , Drosophila/genética , Expresión Génica , Hibridación Genética , Animales , Femenino , Masculino , Fenotipo , Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA