Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Cell ; 159(4): 800-13, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25417157

RESUMEN

We sequenced the MSY (male-specific region of the Y chromosome) of the C57BL/6J strain of the laboratory mouse Mus musculus. In contrast to theories that Y chromosomes are heterochromatic and gene poor, the mouse MSY is 99.9% euchromatic and contains about 700 protein-coding genes. Only 2% of the MSY derives from the ancestral autosomes that gave rise to the mammalian sex chromosomes. Instead, all but 45 of the MSY's genes belong to three acquired, massively amplified gene families that have no homologs on primate MSYs but do have acquired, amplified homologs on the mouse X chromosome. The complete mouse MSY sequence brings to light dramatic forces in sex chromosome evolution: lineage-specific convergent acquisition and amplification of X-Y gene families, possibly fueled by antagonism between acquired X-Y homologs. The mouse MSY sequence presents opportunities for experimental studies of a sex-specific chromosome in its entirety, in a genetically tractable model organism.


Asunto(s)
Evolución Biológica , Cromosomas de los Mamíferos , Ratones Endogámicos C57BL/genética , Análisis de Secuencia de ADN , Cromosoma Y , Animales , Centrómero , Cromosomas Artificiales Bacterianos/genética , Femenino , Humanos , Masculino , Filogenia , Primates/genética , Cromosoma X
2.
PLoS Genet ; 13(4): e1006704, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380054

RESUMEN

The meiosis-specific chromosomal events of homolog pairing, synapsis, and recombination occur over an extended meiotic prophase I that is many times longer than prophase of mitosis. Here we show that, in mice, maintenance of an extended meiotic prophase I requires the gene Meioc, a germ-cell specific factor conserved in most metazoans. In mice, Meioc is expressed in male and female germ cells upon initiation of and throughout meiotic prophase I. Mouse germ cells lacking Meioc initiate meiosis: they undergo pre-meiotic DNA replication, they express proteins involved in synapsis and recombination, and a subset of cells progress as far as the zygotene stage of prophase I. However, cells in early meiotic prophase-as early as the preleptotene stage-proceed to condense their chromosomes and assemble a spindle, as if having progressed to metaphase. Meioc-deficient spermatocytes that have initiated synapsis mis-express CYCLIN A2, which is normally expressed in mitotic spermatogonia, suggesting a failure to properly transition to a meiotic cell cycle program. MEIOC interacts with YTHDC2, and the two proteins pull-down an overlapping set of mitosis-associated transcripts. We conclude that when the meiotic chromosomal program is initiated, Meioc is simultaneously induced so as to extend meiotic prophase. Specifically, MEIOC, together with YTHDC2, promotes a meiotic (as opposed to mitotic) cell cycle program via post-transcriptional control of their target transcripts.


Asunto(s)
Proteínas de Ciclo Celular/genética , Ciclina A2/biosíntesis , Meiosis/genética , Profase/genética , Proteínas de Unión al ARN/genética , Animales , Proteínas de Ciclo Celular/biosíntesis , Emparejamiento Cromosómico/genética , Ciclina A2/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Mitosis/genética , Proteínas de Unión al ARN/metabolismo , Espermatocitos , Espermatogénesis/genética , Espermatogonias/crecimiento & desarrollo , Espermatogonias/metabolismo
3.
PLoS Genet ; 11(9): e1005531, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26378784

RESUMEN

The chromosomal program of meiotic prophase, comprising events such as laying down of meiotic cohesins, synapsis between homologs, and homologous recombination, must be preceded and enabled by the regulated induction of meiotic prophase genes. This gene regulatory program is poorly understood, particularly in organisms with a segregated germline. We characterized the gene regulatory program of meiotic prophase as it occurs in the mouse fetal ovary. By profiling gene expression in the mouse fetal ovary in mutants with whole tissue and single-cell techniques, we identified 104 genes expressed specifically in pre-meiotic to pachytene germ cells. We characterized the regulation of these genes by 1) retinoic acid (RA), which induces meiosis, 2) Dazl, which is required for germ cell competence to respond to RA, and 3) Stra8, a downstream target of RA required for the chromosomal program of meiotic prophase. Initial induction of practically all identified meiotic prophase genes requires Dazl. In the presence of Dazl, RA induces at least two pathways: one Stra8-independent, and one Stra8-dependent. Genes vary in their induction by Stra8, spanning fully Stra8-independent, partially Stra8-independent, and fully Stra8-dependent. Thus, Stra8 regulates the entirety of the chromosomal program but plays a more nuanced role in governing the gene expression program. We propose that Stra8-independent gene expression enables the stockpiling of selected meiotic structural proteins prior to the commencement of the chromosomal program. Unexpectedly, we discovered that Stra8 is required for prompt down-regulation of itself and Rec8. Germ cells that have expressed and down-regulated Stra8 are refractory to further Stra8 expression. Negative feedback of Stra8, and subsequent resistance to further Stra8 expression, may ensure a single, restricted pulse of Stra8 expression. Collectively, our findings reveal a gene regulatory logic by which germ cells prepare for the chromosomal program of meiotic prophase, and ensure that it is induced only once.


Asunto(s)
Redes Reguladoras de Genes , Profase Meiótica I , Ovario/embriología , Ovinos/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Regulación hacia Abajo , Femenino , Regulación del Desarrollo de la Expresión Génica , Ovario/citología
4.
PLoS Genet ; 11(3): e1005019, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25739037

RESUMEN

In mouse embryos at mid-gestation, primordial germ cells (PGCs) undergo licensing to become gametogenesis-competent cells (GCCs), gaining the capacity for meiotic initiation and sexual differentiation. GCCs then initiate either oogenesis or spermatogenesis in response to gonadal cues. Germ cell licensing has been considered to be a cell-autonomous and gonad-independent event, based on observations that some PGCs, having migrated not to the gonad but to the adrenal gland, nonetheless enter meiosis in a time frame parallel to ovarian germ cells -- and do so regardless of the sex of the embryo. Here we test the hypothesis that germ cell licensing is cell-autonomous by examining the fate of PGCs in Gata4 conditional mutant (Gata4 cKO) mouse embryos. Gata4, which is expressed only in somatic cells, is known to be required for genital ridge initiation. PGCs in Gata4 cKO mutants migrated to the area where the genital ridge, the precursor of the gonad, would ordinarily be formed. However, these germ cells did not undergo licensing and instead retained characteristics of PGCs. Our results indicate that licensing is not purely cell-autonomous but is induced by the somatic genital ridge.


Asunto(s)
Gametogénesis , Células Germinativas/citología , Células Germinativas/metabolismo , Animales , Embrión de Mamíferos/metabolismo , Factor de Transcripción GATA4/metabolismo , Gónadas/metabolismo , Meiosis , Ratones , Proteínas de Unión al ARN/metabolismo
5.
PLoS Genet ; 10(8): e1004541, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25102060

RESUMEN

In all sexually reproducing organisms, cells of the germ line must transition from mitosis to meiosis. In mice, retinoic acid (RA), the extrinsic signal for meiotic initiation, activates transcription of Stra8, which is required for meiotic DNA replication and the subsequent processes of meiotic prophase. Here we report that RA also activates transcription of Rec8, which encodes a component of the cohesin complex that accumulates during meiotic S phase, and which is essential for chromosome synapsis and segregation. This RA induction of Rec8 occurs in parallel with the induction of Stra8, and independently of Stra8 function, and it is conserved between the sexes. Further, RA induction of Rec8, like that of Stra8, requires the germ-cell-intrinsic competence factor Dazl. Our findings strengthen the importance of RA and Dazl in the meiotic transition, provide important details about the Stra8 pathway, and open avenues to investigate early meiosis through analysis of Rec8 induction and function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Meiosis/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Replicación del ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Germinativas/crecimiento & desarrollo , Masculino , Ratones , Mitosis/genética , Proteínas Nucleares/biosíntesis , Ovario/efectos de los fármacos , Ovario/crecimiento & desarrollo , Fosfoproteínas/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Transducción de Señal/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Transcripción Genética/efectos de los fármacos , Tretinoina/administración & dosificación
6.
Viruses ; 13(7)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206520

RESUMEN

Antivirals are used not only in the current treatment of influenza but are also stockpiled as a first line of defense against novel influenza strains for which vaccines have yet to be developed. Identifying drug resistance mutations can guide the clinical deployment of the antiviral and can additionally define the mechanisms of drug action and drug resistance. Pimodivir is a first-in-class inhibitor of the polymerase basic protein 2 (PB2) subunit of the influenza A virus polymerase complex. A number of resistance mutations have previously been identified in treated patients or cell culture. Here, we generate a complete map of the effect of all single-amino-acid mutations to an avian PB2 on resistance to pimodivir. We identified both known and novel resistance mutations not only in the previously implicated cap-binding and mid-link domains, but also in the N-terminal domain. Our complete map of pimodivir resistance thus enables the evaluation of whether new viral strains contain mutations that will confer pimodivir resistance.


Asunto(s)
Antivirales/farmacología , Aves/virología , Farmacorresistencia Viral/genética , Virus de la Influenza A/genética , Mutación , Piridinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Proteínas Virales/genética , Células A549 , Animales , Variación Genética , Humanos , Virus de la Influenza A/clasificación , Gripe Aviar/virología , Proteínas de Unión a Caperuzas de ARN/antagonistas & inhibidores , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA