Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 629(8012): 652-659, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693261

RESUMEN

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Asunto(s)
Disbiosis , Padre , Microbioma Gastrointestinal , Masculino , Animales , Femenino , Ratones , Embarazo , Disbiosis/microbiología , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/microbiología , Aptitud Genética , Leptina/metabolismo , Ratones Endogámicos C57BL , Placenta/microbiología , Placenta/metabolismo
2.
Commun Biol ; 4(1): 485, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859315

RESUMEN

Female mammals achieve dosage compensation by inactivating one of their two X chromosomes during development, a process entirely dependent on Xist, an X-linked long non-coding RNA (lncRNA). At the onset of X chromosome inactivation (XCI), Xist is up-regulated and spreads along the future inactive X chromosome. Contextually, it recruits repressive histone and DNA modifiers that transcriptionally silence the X chromosome. Xist regulation is tightly coupled to differentiation and its expression is under the control of both pluripotency and epigenetic factors. Recent evidence has suggested that chromatin remodelers accumulate at the X Inactivation Center (XIC) and here we demonstrate a new role for Chd8 in Xist regulation in differentiating ES cells, linked to its control and prevention of spurious transcription factor interactions occurring within Xist regulatory regions. Our findings have a broader relevance, in the context of complex, developmentally-regulated gene expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Inactivación del Cromosoma X , Cromosoma X/genética , Animales , Proteínas de Unión al ADN/metabolismo , Compensación de Dosificación (Genética) , Femenino , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
Cell Res ; 29(3): 221-232, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30617251

RESUMEN

Several developmental stages of spermatogenesis are transcriptionally quiescent which presents major challenges associated with the regulation of gene expression. Here we identify that the zygotene to pachytene transition is not only associated with the resumption of transcription but also a wave of programmed mRNA degradation that is essential for meiotic progression. We explored whether terminal uridydyl transferase 4- (TUT4-) or TUT7-mediated 3' mRNA uridylation contributes to this wave of mRNA degradation during pachynema. Indeed, both TUT4 and TUT7 are expressed throughout most of spermatogenesis, however, loss of either TUT4 or TUT7 does not have any major impact upon spermatogenesis. Combined TUT4 and TUT7 (TUT4/7) deficiency results in embryonic growth defects, while conditional gene targeting revealed an essential role for TUT4/7 in pachytene progression. Loss of TUT4/7 results in the reduction of miRNA, piRNA and mRNA 3' uridylation. Although this reduction does not greatly alter miRNA or piRNA expression, TUT4/7-mediated uridylation is required for the clearance of many zygotene-expressed transcripts in pachytene cells. We find that TUT4/7-regulated transcripts in pachytene spermatocytes are characterized by having long 3' UTRs with length-adjusted enrichment for AU-rich elements. We also observed these features in TUT4/7-regulated maternal transcripts whose dosage was recently shown to be essential for sculpting a functional maternal transcriptome and meiosis. Therefore, mRNA 3' uridylation is a critical determinant of both male and female germline transcriptomes. In conclusion, we have identified a novel requirement for 3' uridylation-programmed zygotene mRNA clearance in pachytene spermatocytes that is essential for male meiotic progression.


Asunto(s)
Profase Meiótica I/genética , Fase Paquiteno/genética , Procesamiento Postranscripcional del ARN/fisiología , Espermatogénesis/genética , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Estabilidad del ARN/genética , ARN Mensajero/genética , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/metabolismo
4.
J Cell Sci ; 131(6)2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29437857

RESUMEN

In somatic cells, H2afx and Mdc1 are close functional partners in DNA repair and damage response. However, it is not known whether they are also involved in the maintenance of genome integrity in meiosis. By analyzing chromosome dynamics in H2afx-/- spermatocytes, we found that the synapsis of autosomes and X-Y chromosomes was impaired in a fraction of cells. Such defects correlated with an abnormal recombination profile. Conversely, Mdc1 was dispensable for the synapsis of the autosomes and played only a minor role in X-Y synapsis, compared with the action of H2afx This suggested that those genes have non-overlapping functions in chromosome synapsis. However, we observed that both genes play a similar role in the assembly of MLH3 onto chromosomes, a key step in crossover formation. Moreover, we show that H2afx and Mdc1 cooperate in promoting the activation of the recombination-dependent checkpoint, a mechanism that restrains the differentiation of cells with unrepaired DSBs. This occurs by a mechanism that involves P53. Overall, our data show that, in male germ cells, H2afx and Mdc1 promote the maintenance of genome integrity.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Espermatocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Emparejamiento Cromosómico , Inestabilidad Genómica , Genómica , Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas MutL/genética , Proteínas MutL/metabolismo , Recombinación Genética , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Espermatocitos/citología
5.
Mol Cell ; 67(6): 1059-1067.e4, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28867294

RESUMEN

YTHDF2 binds and destabilizes N6-methyladenosine (m6A)-modified mRNA. The extent to which this branch of m6A RNA-regulatory pathway functions in vivo and contributes to mammalian development remains unknown. Here we find that YTHDF2 deficiency is partially permissive in mice and results in female-specific infertility. Using conditional mutagenesis, we demonstrate that YTHDF2 is autonomously required within the germline to produce MII oocytes that are competent to sustain early zygotic development. Oocyte maturation is associated with a wave of maternal RNA degradation, and the resulting relative changes to the MII transcriptome are integral to oocyte quality. The loss of YTHDF2 results in the failure to regulate transcript dosage of a cohort of genes during oocyte maturation, with enrichment observed for the YTHDF2-binding consensus and evidence of m6A in these upregulated genes. In summary, the m6A-reader YTHDF2 is an intrinsic determinant of mammalian oocyte competence and early zygotic development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Meiosis , Oocitos/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Transcriptoma , Cigoto/metabolismo , Animales , Sitios de Unión , Femenino , Fertilidad , Genotipo , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/patología , Fenotipo , Unión Proteica , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Cigoto/patología
6.
J Biol Chem ; 291(8): 3821-36, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26694612

RESUMEN

Cumulus cells sustain the development and fertilization of the mammalian oocyte. These cells are retained around the oocyte by a hyaluronan-rich extracellular matrix synthesized before ovulation, a process called cumulus cell-oocyte complex (COC) expansion. Hyaluronan release and dispersion of the cumulus cells progressively occur after ovulation, paralleling the decline of oocyte fertilization. We show here that, in mice, postovulatory changes of matrix are temporally correlated to cumulus cell death. Cumulus cell apoptosis and matrix disassembly also occurred in ovulated COCs cultured in vitro. COCs expanded in vitro with FSH or EGF underwent the same changes, whereas those expanded with 8-bromo-adenosine-3',5'-cyclic monophosphate (8-Br-cAMP) maintained integrity for a longer time. It is noteworthy that 8-Br-cAMP treatment was also effective on ovulated COCs cultured in vitro, prolonging the vitality of the cumulus cells and the stability of the matrix from a few hours to >2 days. Stimulation of endogenous adenylate cyclase with forskolin or inhibition of phosphodiesterase with rolipram produced similar effects. The treatment with selective cAMP analogues suggests that the effects of cAMP elevation are exerted through an EPAC-independent, PKA type II-dependent signaling pathway, probably acting at the post-transcriptional level. Finally, overnight culture of ovulated COCs with 8-Br-cAMP significantly counteracted the decrease of fertilization rate, doubling the number of fertilized oocytes compared with control conditions. In conclusion, these studies suggest that cAMP-elevating agents prevent cumulus cell senescence and allow them to continue to exert beneficial effects on oocyte and sperm, thereby extending in vitro the time frame of oocyte fertilizability.


Asunto(s)
Células del Cúmulo/metabolismo , AMP Cíclico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Fertilización/efectos de los fármacos , Hormona Folículo Estimulante/farmacología , Ácido Hialurónico/metabolismo , Oocitos/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células del Cúmulo/citología , Matriz Extracelular/metabolismo , Femenino , Ratones , Oocitos/citología , Rolipram/farmacología , Transducción de Señal/efectos de los fármacos
7.
Chromosoma ; 125(2): 189-203, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26440409

RESUMEN

Meiosis is the biological process that, after a cycle of DNA replication, halves the cellular chromosome complement, leading to the formation of haploid gametes. Haploidization is achieved via two successive rounds of chromosome segregation, meiosis I and II. In mammals, during prophase of meiosis I, homologous chromosomes align and synapse through a recombination-mediated mechanism initiated by the introduction of DNA double-strand breaks (DSBs) by the SPO11 protein. In male mice, if SPO11 expression and DSB number are reduced below heterozygosity levels, chromosome synapsis is delayed, chromosome tangles form at pachynema, and defective cells are eliminated by apoptosis at epithelial stage IV at a spermatogenesis-specific endpoint. Whether DSB levels produced in Spo11 (+/-) spermatocytes represent, or approximate, the threshold level required to guarantee successful homologous chromosome pairing is unknown. Using a mouse model that expresses Spo11 from a bacterial artificial chromosome, within a Spo11 (-/-) background, we demonstrate that when SPO11 expression is reduced and DSBs at zygonema are decreased (approximately 40 % below wild-type level), meiotic chromosome pairing is normal. Conversely, DMC1 foci number is increased at pachynema, suggesting that under these experimental conditions, DSBs are likely made with delayed kinetics at zygonema. In addition, we provide evidences that when zygotene-like cells receive enough DSBs before chromosome tangles develop, chromosome synapsis can be completed in most cells, preventing their apoptotic elimination.


Asunto(s)
Emparejamiento Cromosómico , Roturas del ADN de Doble Cadena , Endodesoxirribonucleasas/metabolismo , Meiosis , Espermatocitos/citología , Animales , Cromosomas/genética , Cromosomas/metabolismo , Endodesoxirribonucleasas/genética , Femenino , Masculino , Profase Meiótica I , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espermatocitos/metabolismo , Espermatogénesis
8.
PLoS Genet ; 10(10): e1004597, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25329700

RESUMEN

Male fertility requires the continuous production of high quality motile spermatozoa in abundance. Alterations in all three metrics cause oligoasthenoteratozoospermia, the leading cause of human sub/infertility. Post-mitotic spermatogenesis inclusive of several meiotic stages and spermiogenesis (terminal spermatozoa differentiation) are transcriptionally inert, indicating the potential importance for the post-transcriptional microRNA (miRNA) gene-silencing pathway therein. We found the expression of miRNA generating enzyme Dicer within spermatogenesis peaks in meiosis with critical functions in spermatogenesis. In an expression screen we identified two miRNA loci of the miR-34 family (miR-34b/c and miR-449) that are specifically and highly expressed in post-mitotic male germ cells. A reduction in several miRNAs inclusive of miR-34b/c in spermatozoa has been causally associated with reduced fertility in humans. We found that deletion of both miR34b/c and miR-449 loci resulted in oligoasthenoteratozoospermia in mice. MiR-34bc/449-deficiency impairs both meiosis and the final stages of spermatozoa maturation. Analysis of miR-34bc-/-;449-/- pachytene spermatocytes revealed a small cohort of genes deregulated that were highly enriched for miR-34 family target genes. Our results identify the miR-34 family as the first functionally important miRNAs for spermatogenesis whose deregulation is causal to oligoasthenoteratozoospermia and infertility.


Asunto(s)
Astenozoospermia/genética , MicroARNs/genética , Oligospermia/genética , Animales , ARN Helicasas DEAD-box/genética , Regulación de la Expresión Génica , Infertilidad Masculina/genética , Masculino , Ratones Transgénicos , Mitosis , Ribonucleasa III/genética , Espermatogénesis/genética , Espermatozoides/fisiología
9.
Artículo en Inglés | MEDLINE | ID: mdl-25276231

RESUMEN

BACKGROUND: Repression of retrotransposons is essential for genome integrity and the development of germ cells. Among retrotransposons, the establishment of CpG DNA methylation and epigenetic silencing of LINE1 (L1) elements and the intracisternal A particle (IAP) endogenous retrovirus (ERV) is dependent upon the piRNA pathway during embryonic germ cell reprogramming. Furthermore, the Piwi protein Mili, guided by piRNAs, cleaves expressed L1 transcripts to post-transcriptionally enforce L1 silencing in meiotic cells. The loss of both DNA methylation and the Mili piRNA pathway does not affect L1 silencing in the mitotic spermatogonia where histone H3 lysine 9 dimethylation (H3K9me2) is postulated to co-repress these elements. RESULTS: Here we show that the histone H3 lysine 9 dimethyltransferase G9a co-suppresses L1 elements in spermatogonia. In the absence of both a functional piRNA pathway and L1 DNA methylation, G9a is both essential and sufficient to silence L1 elements. In contrast, H3K9me2 alone is insufficient to maintain IAP silencing in spermatogonia. The loss of all three repressive mechanisms has a major impact on spermatogonial populations inclusive of spermatogonial stem cells, with the loss of all germ cells observed in a high portion of seminiferous tubules. CONCLUSIONS: Our study identifies G9a-mediated H3K9me2 as a novel and important L1 repressive mechanism in the germ line. We also demonstrate fundamental differences in the requirements for the maintenance of L1 and IAP silencing during adult spermatogenesis, where H3K9me2 is sufficient to maintain L1 but not IAP silencing. Finally, we demonstrate that repression of retrotransposon activation in spermatogonia is important for the survival of this population and testicular homeostasis.

10.
Mol Cell ; 50(4): 601-8, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706823

RESUMEN

Transposons present an acute challenge to the germline, and mechanisms that repress their activity are essential for transgenerational genomic integrity. LINE1 (L1) is the most successful retrotransposon and is epigenetically repressed by CpG DNA methylation. Here, we identify two additional important mechanisms by which L1 is repressed during spermatogenesis. We demonstrate that the Piwi protein Mili and the piRNA pathway are required to posttranscriptionally silence L1 in meiotic pachytene cells even in the presence of normal L1 DNA methylation. Strikingly, in the absence of both a functional piRNA pathway and DNA methylation, L1 elements are normally repressed in mitotic stages of spermatogenesis. Accordingly, we find that the euchromatic repressive histone H3 dimethylated lysine 9 modification cosuppresses L1 expression therein. We demonstrate the existence of multiple epigenetic mechanisms that in conjunction with the piRNA pathway sequentially enforce L1 silencing and genomic stability during mitotic and meiotic stages of adult spermatogenesis.


Asunto(s)
Epigénesis Genética , Silenciador del Gen , Elementos de Nucleótido Esparcido Largo/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Espermatogénesis/genética , Factores de Edad , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Western Blotting , Metilación de ADN , Expresión Génica , Histonas/metabolismo , Lisina/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Mitosis/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatocitos/metabolismo , Testículo/citología , Testículo/metabolismo
11.
Nature ; 480(7376): 259-63, 2011 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-22020280

RESUMEN

Piwi proteins and Piwi-interacting RNAs (piRNAs) have conserved functions in transposon silencing. The murine Piwi proteins Mili and Miwi2 (also called Piwil2 and Piwil4, respectively) direct epigenetic LINE1 and intracisternal A particle transposon silencing during genome reprogramming in the embryonic male germ line. Piwi proteins are proposed to be piRNA-guided endonucleases that initiate secondary piRNA biogenesis; however, the actual contribution of their endonuclease activities to piRNA biogenesis and transposon silencing remain unknown. To investigate the role of Piwi-catalysed endonucleolytic activity, we engineered point mutations in mice that substitute the second aspartic acid to an alanine in the DDH catalytic triad of Mili and Miwi2, generating the Mili(DAH) and Miwi2(DAH) alleles, respectively. Analysis of Mili-bound piRNAs from homozygous Mili(DAH) fetal gonadocytes revealed a failure of transposon piRNA amplification, resulting in the marked reduction of piRNA bound within Miwi2 ribonuclear particles. We find that Mili-mediated piRNA amplification is selectively required for LINE1, but not intracisternal A particle, silencing. The defective piRNA pathway in Mili(DAH) mice results in spermatogenic failure and sterility. Surprisingly, homozygous Miwi2(DAH) mice are fertile, transposon silencing is established normally and no defects in secondary piRNA biogenesis are observed. In addition, the hallmarks of piRNA amplification are observed in Miwi2-deficient gonadocytes. We conclude that cycles of intra-Mili secondary piRNA biogenesis fuel piRNA amplification that is absolutely required for LINE1 silencing.


Asunto(s)
Proteínas Argonautas/metabolismo , Silenciador del Gen , Elementos de Nucleótido Esparcido Largo/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Alelos , Animales , Proteínas Argonautas/genética , Elementos Transponibles de ADN/genética , Masculino , Ratones , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Espermatogénesis/genética
12.
J Exp Med ; 207(7): 1351-8, 2010 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-20513743

RESUMEN

The process of erythropoiesis must be efficient and robust to supply the organism with red bloods cells both under condition of homeostasis and stress. The microRNA (miRNA) pathway was recently shown to regulate erythroid development. Here, we show that expression of the locus encoding miR-144 and miR-451 is strictly dependent on Argonaute 2 and is required for erythroid homeostasis. Mice deficient for the miR-144/451 cluster display a cell autonomous impairment of late erythroblast maturation, resulting in erythroid hyperplasia, splenomegaly, and a mild anemia. Analysis of gene expression profiles from wild-type and miR-144/451-deficient erythroblasts revealed that the miR-144/451 cluster acts as a "tuner" of gene expression, influencing the expression of many genes. MiR-451 imparts a greater impact on target gene expression than miR-144. Accordingly, mice deficient in miR-451 alone exhibited a phenotype indistinguishable from miR-144/451-deficient mice. Thus, the miR-144/451 cluster tunes gene expression to impart a robustness to erythropoiesis that is critical under conditions of stress.


Asunto(s)
Células Eritroides/metabolismo , Sitios Genéticos/genética , Homeostasis/genética , MicroARNs/genética , Anemia/genética , Anemia/patología , Animales , Proteínas Argonautas , Diferenciación Celular/genética , Linaje de la Célula/genética , Eritroblastos/metabolismo , Eritroblastos/patología , Células Eritroides/patología , Eritropoyesis/genética , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hiperplasia , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo
13.
PLoS Genet ; 4(5): e1000076, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18497861

RESUMEN

During meiosis in most sexually reproducing organisms, recombination forms crossovers between homologous maternal and paternal chromosomes and thereby promotes proper chromosome segregation at the first meiotic division. The number and distribution of crossovers are tightly controlled, but the factors that contribute to this control are poorly understood in most organisms, including mammals. Here we provide evidence that the ATM kinase or protein is essential for proper crossover formation in mouse spermatocytes. ATM deficiency causes multiple phenotypes in humans and mice, including gonadal atrophy. Mouse Atm-/- spermatocytes undergo apoptosis at mid-prophase of meiosis I, but Atm(-/-) meiotic phenotypes are partially rescued by Spo11 heterozygosity, such that ATM-deficient spermatocytes progress to meiotic metaphase I. Strikingly, Spo11+/-Atm-/- spermatocytes are defective in forming the obligate crossover on the sex chromosomes, even though the XY pair is usually incorporated in a sex body and is transcriptionally inactivated as in normal spermatocytes. The XY crossover defect correlates with the appearance of lagging chromosomes at metaphase I, which may trigger the extensive metaphase apoptosis that is observed in these cells. In addition, control of the number and distribution of crossovers on autosomes appears to be defective in the absence of ATM because there is an increase in the total number of MLH1 foci, which mark the sites of eventual crossover formation, and because interference between MLH1 foci is perturbed. The axes of autosomes exhibit structural defects that correlate with the positions of ongoing recombination. Together, these findings indicate that ATM plays a role in both crossover control and chromosome axis integrity and further suggests that ATM is important for coordinating these features of meiotic chromosome dynamics.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Cromosomas Sexuales/genética , Espermatocitos/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Aberraciones Cromosómicas , Emparejamiento Cromosómico , Intercambio Genético , Proteínas de Unión al ADN/genética , Endodesoxirribonucleasas , Esterasas/genética , Femenino , Heterocigoto , Masculino , Meiosis , Metafase , Ratones , Ratones Noqueados , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Espermatocitos/citología , Proteínas Supresoras de Tumor/genética
14.
Mol Cell Biol ; 25(16): 7203-15, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16055729

RESUMEN

Fundamentally different recombination defects cause apoptosis of mouse spermatocytes at the same stage in development, stage IV of the seminiferous epithelium cycle, equivalent to mid-pachynema in normal males. To understand the cellular response(s) that triggers apoptosis, we examined markers of spermatocyte development in mice with different recombination defects. In Spo11(-)(/)(-) mutants, which lack the double-strand breaks (DSBs) that initiate recombination, spermatocytes express markers of early to mid-pachynema, forming chromatin domains that contain sex body-associated proteins but that rarely encompass the sex chromosomes. Dmc1(-)(/)(-) spermatocytes, impaired in DSB repair, appear to arrest at or about late zygonema. Epistasis analysis reveals that this earlier arrest is a response to unrepaired DSBs, and cytological analysis implicates the BRCT-containing checkpoint protein TOPBP1. Atm(-)(/)(-) spermatocytes show similarities to Dmc1(-)(/)(-) spermatocytes, suggesting that ATM promotes meiotic DSB repair. Msh5(-)(/)(-) mutants display a set of characteristics distinct from these other mutants. Thus, despite equivalent stages of spermatocyte elimination, different recombination-defective mutants manifest distinct responses, providing insight into surveillance mechanisms in male meiosis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Recombinación Genética , Espermatocitos/citología , Animales , Apoptosis , Cromatina/metabolismo , Reparación del ADN , Epistasis Genética , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Modelos Genéticos , Mutación , Espermatocitos/metabolismo , Testículo/metabolismo , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 102(3): 737-42, 2005 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-15640358

RESUMEN

Defects in meiotic recombination in many organisms result in arrest because of activation of a meiotic checkpoint(s). The proximal defect that triggers this checkpoint in mammalian germ cells is not understood, but it has been suggested to involve either the presence of DNA damage in the form of unrepaired recombination intermediates or defects in homologous chromosome pairing and synapsis independent of DNA damage per se. To distinguish between these possibilities in the female germ line, we compared mouse oocyte development in a mutant that fails to form the double-strand breaks (DSBs) that initiate meiotic recombination (Spo11-/-) to mutants with defects in processing DSBs when they are formed (Dmc1-/- and Msh5-/-), and we examined the epistasis relationships between these mutations. Absence of DSB formation caused a partial defect in follicle formation, whereas defects in DSB repair caused earlier and more severe meiotic arrest, which could be suppressed by eliminating DSB formation. Therefore, our analysis reveals that there are both DNA-damage-dependent and -independent responses to recombination errors in mammalian oocytes. By using these findings as a paradigm, we also examined oocyte loss in mutants lacking the DNA-damage checkpoint kinase ATM. The absence of ATM caused defects in folliculogenesis that were similar to those in Dmc1 mutants and that could be suppressed by Spo11 mutation, implying that oocyte death in Atm-deficient animals is a response to defective DSB repair.


Asunto(s)
Daño del ADN/fisiología , Oocitos , Recombinación Genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endodesoxirribonucleasas , Epistasis Genética , Esterasas/deficiencia , Esterasas/genética , Esterasas/fisiología , Femenino , Meiosis , Ratones , Ratones Noqueados , Proteínas Nucleares , Folículo Ovárico , Proteínas de Unión a Fosfato , Proteínas/genética , Proteínas/fisiología , Complejo Sinaptonémico
16.
Proc Natl Acad Sci U S A ; 99(18): 11712-7, 2002 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-12181493

RESUMEN

Overexpression of the TCL1 oncogene has been shown to play a causative role in T cell leukemias of humans and mice. The characterization of Tcl1-deficient mice in these studies indicates an important developmental role for Tcl1 in early embryogenesis. In wild-type embryos, Tcl1 is abundant in the first three mitotic cycles, during which it shuttles between nuclei and the embryo cortical regions in a cell-cycle-dependent fashion. The absence of this protein in early embryogenesis results in reduced fertility of female mice. The present studies elucidate the mechanism responsible for the reduced female fertility through analysis of the oogenesis stages and early embryo development in Tcl1-deficient mice. Even though Tcl1(-/-) females display normal oogenesis and rates of oocyte maturation/ovulation and fertilization, the lack of maternally derived Tcl1 impairs the embryo's ability to undergo normal cleavage and develop to the morula stage, especially under in vitro culture conditions. Beyond this crisis point, differentiative traits of zygotic genome activation and embryo compaction can take place normally. In contrast with this unanticipated role in early embryogenesis, we observed an overexpression of TCL1 in human seminomas. This finding suggests that TCL1 dysregulation could contribute to the development of this germinal cell cancer as well as lymphoid malignancies.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Desarrollo Embrionario y Fetal/fisiología , Proteínas Proto-Oncogénicas , Seminoma/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Blastómeros , Cartilla de ADN , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Infertilidad Femenina/genética , Ratones , Ratones Noqueados , Embarazo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...