Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cell ; 139(5): 999-1011, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19945382

RESUMEN

In somatic cells of female placental mammals, one X chromosome is inactivated to minimize sex-related dosage differences of X-encoded genes. Random X chromosome inactivation (XCI) in the embryo is a stochastic process, in which each X has an independent probability to initiate XCI, triggered by the nuclear concentration of one or more X-encoded XCI-activators. Here, we identify the E3 ubiquitin ligase RNF12 as an important XCI-activator. Additional copies of mouse Rnf12 or human RNF12 result in initiation of XCI in male mouse ES cells and on both X chromosomes in a substantial percentage of female mouse ES cells. This activity is dependent on an intact open reading frame of Rnf12 and correlates with the transgenic expression level of RNF12. Initiation of XCI is markedly reduced in differentiating female heterozygous Rnf12(+/-) ES cells. These findings provide evidence for a dose-dependent role of RNF12 in the XCI counting and initiation process.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras/metabolismo , Inactivación del Cromosoma X , Animales , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Masculino , Ratones , Secuencias Reguladoras de Ácidos Nucleicos , Ubiquitina-Proteína Ligasas
2.
Mol Cell ; 53(6): 965-78, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24613346

RESUMEN

X chromosome inactivation (XCI) in female placental mammals is a vital mechanism for dosage compensation between X-linked and autosomal genes. XCI starts with activation of Xist and silencing of the negative regulator Tsix, followed by cis spreading of Xist RNA over the future inactive X chromosome (Xi). Here, we show that XCI does not require physical contact between the two X chromosomes (X-pairing) but is regulated by trans-acting diffusible factors. We found that the X-encoded trans-acting and dose-dependent XCI-activator RNF12 acts in concert with the cis-regulatory region containing Jpx, Ftx, and Xpr to activate Xist and to overcome repression by Tsix. RNF12 acts at two subsequent steps; two active copies of Rnf12 drive initiation of XCI, and one copy needs to remain active to maintain XCI toward establishment of the Xi. This two-step mechanism ensures that XCI is very robust and fine-tuned, preventing XCI of both X chromosomes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , ARN Largo no Codificante/genética , Ubiquitina-Proteína Ligasas/genética , Inactivación del Cromosoma X , Cromosoma X , Animales , Transporte Biológico , Línea Celular , Emparejamiento Cromosómico , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Ratones Noqueados , ARN Largo no Codificante/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
3.
Genome Res ; 26(9): 1202-10, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27510564

RESUMEN

The X and Y sex chromosomes of placental mammals show hallmarks of a tumultuous evolutionary past. The X Chromosome has a rich and conserved gene content, while the Y Chromosome has lost most of its genes. In the Transcaucasian mole vole Ellobius lutescens, the Y Chromosome including Sry has been lost, and both females and males have a 17,X diploid karyotype. Similarly, the closely related Ellobius talpinus, has a 54,XX karyotype in both females and males. Here, we report the sequencing and assembly of the E. lutescens and E. talpinus genomes. The results indicate that the loss of the Y Chromosome in E. lutescens and E. talpinus occurred in two independent events. Four functional homologs of mouse Y-Chromosomal genes were detected in both female and male E. lutescens, of which three were also detected in the E. talpinus genome. One of these is Eif2s3y, known as the only Y-derived gene that is crucial for successful male meiosis. Female and male E. lutescens can carry one and the same X Chromosome with a largely conserved gene content, including all genes known to function in X Chromosome inactivation. The availability of the genomes of these mole vole species provides unique models to study the dynamics of sex chromosome evolution.


Asunto(s)
Cromosomas Sexuales/genética , Procesos de Determinación del Sexo/genética , Cromosoma X/genética , Cromosoma Y/genética , Animales , Arvicolinae/genética , Cromosomas de los Mamíferos/genética , Femenino , Cariotipificación , Masculino , Mamíferos/genética , Ratones
4.
PLoS Genet ; 12(10): e1006358, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27716834

RESUMEN

In mouse female preimplantation embryos, the paternal X chromosome (Xp) is silenced by imprinted X chromosome inactivation (iXCI). This requires production of the noncoding Xist RNA in cis, from the Xp. The Xist locus on the maternally inherited X chromosome (Xm) is refractory to activation due to the presence of an imprint. Paternal inheritance of an Xist deletion (XpΔXist) is embryonic lethal to female embryos, due to iXCI abolishment. Here, we circumvented the histone-to-protamine and protamine-to-histone transitions of the paternal genome, by fertilization of oocytes via injection of round spermatids (ROSI). This did not affect initiation of XCI in wild type female embryos. Surprisingly, ROSI using ΔXist round spermatids allowed survival of female embryos. This was accompanied by activation of the intact maternal Xist gene, initiated with delayed kinetics, around the morula stage, resulting in Xm silencing. Maternal Xist gene activation was not observed in ROSI-derived males. In addition, no Xist expression was detected in male and female morulas that developed from oocytes fertilized with mature ΔXist sperm. Finally, the expression of the X-encoded XCI-activator RNF12 was enhanced in both male (wild type) and female (wild type as well as XpΔXist) ROSI derived embryos, compared to in vivo fertilized embryos. Thus, high RNF12 levels may contribute to the specific activation of maternal Xist in XpΔXist female ROSI embryos, but upregulation of additional Xp derived factors and/or the specific epigenetic constitution of the round spermatid-derived Xp are expected to be more critical. These results illustrate the profound impact of a dysregulated paternal epigenome on embryo development, and we propose that mouse ROSI can be used as a model to study the effects of intergenerational inheritance of epigenetic marks.


Asunto(s)
Desarrollo Embrionario/genética , Herencia Paterna/genética , ARN Largo no Codificante/genética , Inactivación del Cromosoma X/genética , Animales , Blastocisto , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Oocitos/crecimiento & desarrollo , Eliminación de Secuencia/genética , Espermátides/metabolismo , Cromosoma X/genética
5.
Nature ; 485(7398): 386-90, 2012 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-22596162

RESUMEN

Evolution of the mammalian sex chromosomes has resulted in a heterologous X and Y pair, where the Y chromosome has lost most of its genes. Hence, there is a need for X-linked gene dosage compensation between XY males and XX females. In placental mammals, this is achieved by random inactivation of one X chromosome in all female somatic cells. Upregulation of Xist transcription on the future inactive X chromosome acts against Tsix antisense transcription, and spreading of Xist RNA in cis triggers epigenetic changes leading to X-chromosome inactivation. Previously, we have shown that the X-encoded E3 ubiquitin ligase RNF12 is upregulated in differentiating mouse embryonic stem cells and activates Xist transcription and X-chromosome inactivation. Here we identify the pluripotency factor REX1 as a key target of RNF12 in the mechanism of X-chromosome inactivation. RNF12 causes ubiquitination and proteasomal degradation of REX1, and Rnf12 knockout embryonic stem cells show an increased level of REX1. Using chromatin immunoprecipitation sequencing, REX1 binding sites were detected in Xist and Tsix regulatory regions. Overexpression of REX1 in female embryonic stem cells was found to inhibit Xist transcription and X-chromosome inactivation, whereas male Rex1(+/-) embryonic stem cells showed ectopic X-chromosome inactivation. From this, we propose that RNF12 causes REX1 breakdown through dose-dependent catalysis, thereby representing an important pathway to initiate X-chromosome inactivation. Rex1 and Xist are present only in placental mammals, which points to co-evolution of these two genes and X-chromosome inactivation.


Asunto(s)
Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Inactivación del Cromosoma X , Cromosoma X/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células Madre Embrionarias/metabolismo , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , ARN Largo no Codificante , ARN no Traducido/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
6.
PLoS Genet ; 9(6): e1003538, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23754961

RESUMEN

In mammalian meiotic prophase, the initial steps in repair of SPO11-induced DNA double-strand breaks (DSBs) are required to obtain stable homologous chromosome pairing and synapsis. The X and Y chromosomes pair and synapse only in the short pseudo-autosomal regions. The rest of the chromatin of the sex chromosomes remain unsynapsed, contains persistent meiotic DSBs, and the whole so-called XY body undergoes meiotic sex chromosome inactivation (MSCI). A more general mechanism, named meiotic silencing of unsynapsed chromatin (MSUC), is activated when autosomes fail to synapse. In the absence of SPO11, many chromosomal regions remain unsynapsed, but MSUC takes place only on part of the unsynapsed chromatin. We asked if spontaneous DSBs occur in meiocytes that lack a functional SPO11 protein, and if these might be involved in targeting the MSUC response to part of the unsynapsed chromatin. We generated mice carrying a point mutation that disrupts the predicted catalytic site of SPO11 (Spo11(YF/YF)), and blocks its DSB-inducing activity. Interestingly, we observed foci of proteins involved in the processing of DNA damage, such as RAD51, DMC1, and RPA, both in Spo11(YF/YF) and Spo11 knockout meiocytes. These foci preferentially localized to the areas that undergo MSUC and form the so-called pseudo XY body. In SPO11-deficient oocytes, the number of repair foci increased during oocyte development, indicating the induction of S phase-independent, de novo DNA damage. In wild type pachytene oocytes we observed meiotic silencing in two types of pseudo XY bodies, one type containing DMC1 and RAD51 foci on unsynapsed axes, and another type containing only RAD51 foci, mainly on synapsed axes. Taken together, our results indicate that in addition to asynapsis, persistent SPO11-induced DSBs are important for the initiation of MSCI and MSUC, and that SPO11-independent DNA repair foci contribute to the MSUC response in oocytes.


Asunto(s)
Emparejamiento Cromosómico/genética , Reparación del ADN/genética , Endodesoxirribonucleasas/genética , Meiosis/genética , Inactivación del Cromosoma X/genética , Animales , Roturas del ADN de Doble Cadena , Endodesoxirribonucleasas/metabolismo , Femenino , Masculino , Ratones , Oogénesis/genética , Espermatocitos/citología , Espermatocitos/metabolismo , Cromosoma X/genética , Cromosoma Y/genética
7.
BMC Genomics ; 16: 291, 2015 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-25884295

RESUMEN

BACKGROUND: In mammalian meiotic prophase, homologous chromosome recognition is aided by formation and repair of programmed DNA double-strand breaks (DSBs). Subsequently, stable associations form through homologous chromosome synapsis. In male mouse meiosis, the largely heterologous X and Y chromosomes synapse only in their short pseudoautosomal regions (PARs), and DSBs persist along the unsynapsed non-homologous arms of these sex chromosomes. Asynapsis of these arms and the persistent DSBs then trigger transcriptional silencing through meiotic sex chromosome inactivation (MSCI), resulting in formation of the XY body. This inactive state is partially maintained in post-meiotic haploid spermatids (postmeiotic sex chromatin repression, PSCR). For the human, establishment of MSCI and PSCR have also been reported, but X-linked gene silencing appears to be more variable compared to mouse. To gain more insight into the regulation and significance of MSCI and PSCR among different eutherian species, we have performed a global analysis of XY pairing dynamics, DSB repair, MSCI and PSCR in the domestic dog (Canis lupus familiaris), for which the complete genome sequence has recently become available, allowing a thorough comparative analyses. RESULTS: In addition to PAR synapsis between X and Y, we observed extensive self-synapsis of part of the dog X chromosome, and rapid loss of known markers of DSB repair from that part of the X. Sequencing of RNA from purified spermatocytes and spermatids revealed establishment of MSCI. However, the self-synapsing region of the X displayed higher X-linked gene expression compared to the unsynapsed area in spermatocytes, and was post-meiotically reactivated in spermatids. In contrast, genes in the PAR, which are expected to escape MSCI, were expressed at very low levels in both spermatocytes and spermatids. Our comparative analysis was then used to identify two X-linked genes that may escape MSCI in spermatocytes, and 21 that are specifically re-activated in spermatids of human, mouse and dog. CONCLUSIONS: Our data indicate that MSCI is incomplete in the dog. This may be partially explained by extensive, but transient, self-synapsis of the X chromosome, in association with rapid completion of meiotic DSB repair. In addition, our comparative analysis identifies novel candidate male fertility genes.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Perros/genética , Meiosis , Cromosomas Sexuales/metabolismo , Espermatogénesis , Inactivación del Cromosoma X , Animales , Animales Domésticos , Roturas del ADN de Doble Cadena , Reparación del ADN , Perros/metabolismo , Humanos , Masculino , Ratones , Espermatocitos/citología , Espermatocitos/metabolismo , Testículo
8.
PLoS Genet ; 7(1): e1002001, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21298085

RESUMEN

In somatic cells of female placental mammals, one of the two X chromosomes is transcriptionally silenced to accomplish an equal dose of X-encoded gene products in males and females. Initiation of random X chromosome inactivation (XCI) is thought to be regulated by X-encoded activators and autosomally encoded suppressors controlling Xist. Spreading of Xist RNA leads to silencing of the X chromosome in cis. Here, we demonstrate that the dose dependent X-encoded XCI activator RNF12/RLIM acts in trans and activates Xist. We did not find evidence for RNF12-mediated regulation of XCI through Tsix or the Xist intron 1 region, which are both known to be involved in inhibition of Xist. In addition, we found that Xist intron 1, which contains a pluripotency factor binding site, is not required for suppression of Xist in undifferentiated ES cells. Analysis of female Rnf12⁻/⁻ knockout ES cells showed that RNF12 is essential for initiation of XCI and is mainly involved in the regulation of Xist. We conclude that RNF12 is an indispensable factor in up-regulation of Xist transcription, thereby leading to initiation of random XCI.


Asunto(s)
Silenciador del Gen , ARN no Traducido/genética , Proteínas Represoras/fisiología , Inactivación del Cromosoma X/genética , Animales , Células Madre Embrionarias/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hibridación Fluorescente in Situ , Intrones/genética , Masculino , Ratones , Proteína Homeótica Nanog , ARN Largo no Codificante , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas
9.
J Cell Sci ; 124(Pt 16): 2837-50, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21807948

RESUMEN

RAD18 is an ubiquitin ligase that is involved in replication damage bypass and DNA double-strand break (DSB) repair processes in mitotic cells. Here, we investigated the testicular phenotype of Rad18-knockdown mice to determine the function of RAD18 in meiosis, and in particular, in the repair of meiotic DSBs induced by the meiosis-specific topoisomerase-like enzyme SPO11. We found that RAD18 is recruited to a specific subfraction of persistent meiotic DSBs. In addition, RAD18 is recruited to the chromatin of the XY chromosome pair, which forms the transcriptionally silent XY body. At the XY body, RAD18 mediates the chromatin association of its interaction partners, the ubiquitin-conjugating enzymes HR6A and HR6B. Moreover, RAD18 was found to regulate the level of dimethylation of histone H3 at Lys4 and maintain meiotic sex chromosome inactivation, in a manner similar to that previously observed for HR6B. Finally, we show that RAD18 and HR6B have a role in the efficient repair of a small subset of meiotic DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Meiosis , Testículo/metabolismo , Animales , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Histonas/genética , Histonas/metabolismo , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño/genética , Testículo/patología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Inactivación del Cromosoma X/genética
10.
Nucleic Acids Res ; 39(18): e121, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21737430

RESUMEN

The use of bacterial artificial chromosomes (BACs) provides a consistent and high targeting efficiency of homologous recombination in embryonic stem (ES) cells, facilitated by long stretches of sequence homology. Here, we introduce a BAC targeting method which employs restriction fragment length polymorphisms (RFLPs) in targeted polymorphic C57BL/6/Cast/Ei F1 mouse ES cell lines to identify properly targeted ES cell clones. We demonstrate that knockout alleles can be generated either by targeting of an RFLP located in the open reading frame thereby disrupting the RFLP and ablating gene function, or by introduction of a transcription stop cassette that prematurely stops transcription of an RFLP located downstream of the stop cassette. With both methods we have generated Rnf12 heterozygous knockout ES cells, which were identified by allele specific PCR using genomic DNA or cDNA as a template. Our results indicate that this novel strategy is efficient and precise, by combining a high targeting efficiency with a convenient PCR based readout and reliable detection of correct targeting events.


Asunto(s)
Cromosomas Artificiales Bacterianos , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Polimorfismo de Longitud del Fragmento de Restricción , Animales , Línea Celular , Células Madre Embrionarias/citología , Ratones , Ratones Endogámicos C57BL , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética
11.
J Cell Sci ; 123(Pt 3): 331-9, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20053632

RESUMEN

The cytoplasmic chromatoid body (CB) organizes mRNA metabolism and small regulatory RNA pathways, in relation to haploid gene expression, in mammalian round spermatids. However, little is known about functions and fate of the CB at later steps of spermatogenesis, when elongating spermatids undergo chromatin compaction and transcriptional silencing. In mouse elongating spermatids, we detected accumulation of the testis-specific serine/threonine kinases TSSK1 and TSSK2, and the substrate TSKS, in a ring-shaped structure around the base of the flagellum and in a cytoplasmic satellite, both corresponding to structures described to originate from the CB. At later steps of spermatid differentiation, the ring is found at the caudal end of the newly formed mitochondrial sheath. Targeted deletion of the tandemly arranged genes Tssk1 and Tssk2 in mouse resulted in male infertility, with loss of the CB-derived ring structure, and with elongating spermatids possessing a collapsed mitochondrial sheath. These results reveal TSSK1- and TSSK2-dependent functions of a transformed CB in post-meiotic cytodifferentiation of spermatids.


Asunto(s)
Gránulos Citoplasmáticos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Espermátides/enzimología , Espermátides/metabolismo , Testículo/enzimología , Animales , Western Blotting , Proteínas del Citoesqueleto , Electroforesis en Gel de Poliacrilamida , Femenino , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Fosfoproteínas , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Espermátides/ultraestructura , Espermatogénesis/genética , Espermatogénesis/fisiología , Testículo/ultraestructura
12.
PLoS Genet ; 5(5): e1000466, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19461881

RESUMEN

During meiotic prophase in male mammals, the heterologous X and Y chromosomes remain largely unsynapsed, and meiotic sex chromosome inactivation (MSCI) leads to formation of the transcriptionally silenced XY body. In birds, the heterogametic sex is female, carrying Z and W chromosomes (ZW), whereas males have the homogametic ZZ constitution. During chicken oogenesis, the heterologous ZW pair reaches a state of complete heterologous synapsis, and this might enable maintenance of transcription of Z- and W chromosomal genes during meiotic prophase. Herein, we show that the ZW pair is transiently silenced, from early pachytene to early diplotene using immunocytochemistry and gene expression analyses. We propose that ZW inactivation is most likely achieved via spreading of heterochromatin from the W on the Z chromosome. Also, persistent meiotic DNA double-strand breaks (DSBs) may contribute to silencing of Z. Surprisingly, gammaH2AX, a marker of DSBs, and also the earliest histone modification that is associated with XY body formation in mammalian and marsupial spermatocytes, does not cover the ZW during the synapsed stage. However, when the ZW pair starts to desynapse, a second wave of gammaH2AX accumulates on the unsynapsed regions of Z, which also show a reappearance of the DSB repair protein RAD51. This indicates that repair of meiotic DSBs on the heterologous part of Z is postponed until late pachytene/diplotene, possibly to avoid recombination with regions on the heterologously synapsed W chromosome. Two days after entering diplotene, the Z looses gammaH2AX and shows reactivation. This is the first report of meiotic sex chromosome inactivation in a species with female heterogamety, providing evidence that this mechanism is not specific to spermatogenesis. It also indicates the presence of an evolutionary force that drives meiotic sex chromosome inactivation independent of the final achievement of synapsis.


Asunto(s)
Pollos/genética , Meiosis/genética , Cromosomas Sexuales/genética , Acetilación , Animales , Secuencia de Bases , Pollos/metabolismo , Roturas del ADN de Doble Cadena , Metilación de ADN , Cartilla de ADN/genética , Reparación del ADN , Compensación de Dosificación (Genética) , Femenino , Perfilación de la Expresión Génica , Silenciador del Gen , Histonas/química , Histonas/metabolismo , Hibridación Fluorescente in Situ , Lisina/química , Masculino , Microscopía Fluorescente , Oocitos/citología , Oocitos/metabolismo , Oogénesis/genética , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cromosomas Sexuales/metabolismo , Espermatogénesis/genética , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo
13.
Br J Sports Med ; 46(8): 614-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21540190

RESUMEN

Based on DNA analysis of a historical case, the authors describe how a female athlete can be unknowingly confronted with the consequences of a disorder of sex development resulting in hyperandrogenism emerging early in her sports career. In such a situation, it is harmful and confusing to question sex and gender. Exposure to either a low or high level of endogenous testosterone from puberty is a decisive factor with respect to sexual dimorphism of physical performance. Yet, measurement of testosterone is not the means by which questions of an athlete's eligibility to compete with either women or men are resolved. The authors discuss that it might be justifiable to use the circulating testosterone level as an endocrinological parameter, to try to arrive at an objective criterion in evaluating what separates women and men in sports competitions, which could prevent the initiation of complicated, lengthy and damaging sex and gender verification procedures.


Asunto(s)
Rendimiento Atlético/fisiología , Trastornos del Desarrollo Sexual/diagnóstico , Análisis para Determinación del Sexo/métodos , Rendimiento Atlético/historia , Cromosomas Humanos X/fisiología , Cromosomas Humanos Y/fisiología , Trastornos del Desarrollo Sexual/sangre , Trastornos del Desarrollo Sexual/genética , Trastornos del Desarrollo Sexual/historia , Femenino , Identidad de Género , Historia del Siglo XX , Humanos , Masculino , Mosaicismo , Países Bajos , Caracteres Sexuales , Análisis para Determinación del Sexo/historia , Desarrollo Sexual/fisiología , Testosterona/sangre
14.
Chromosoma ; 119(3): 311-24, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20162291

RESUMEN

During male meiotic prophase in mammals, X and Y are in a largely unsynapsed configuration, which is thought to trigger meiotic sex chromosome inactivation (MSCI). In avian species, females are ZW, and males ZZ. Although Z and W in chicken oocytes show complete, largely heterologous synapsis, they too undergo MSCI, albeit only transiently. The W chromosome is already inactive in early meiotic prophase, and inactive chromatin marks may spread on to the Z upon synapsis. Mammalian MSCI is considered as a specialised form of the general meiotic silencing mechanism, named meiotic silencing of unsynapsed chromatin (MSUC). Herein, we studied the avian form of MSUC, by analysing the behaviour of the peculiar germline restricted chromosome (GRC) that is present as a single copy in zebra finch spermatocytes. In the female germline, this chromosome is present in two copies, which normally synapse and recombine. In contrast, during male meiosis, the single GRC is always eliminated. We found that the GRC in the male germline is silenced from early leptotene onwards, similar to the W chromosome in avian oocytes. The GRC remains largely unsynapsed throughout meiotic prophase I, although patches of SYCP1 staining indicate that part of the GRC may self-synapse. In addition, the GRC is largely devoid of meiotic double strand breaks. We observed a lack of the inner centromere protein INCENP on the GRC and elimination of the GRC following metaphase I. Subsequently, the GRC forms a micronucleus in which the DNA is fragmented. We conclude that in contrast to MSUC in mammals, meiotic silencing of this single chromosome in the avian germline occurs prior to, and independent of DNA double strand breaks and chromosome pairing, hence we have named this phenomenon meiotic silencing prior to synapsis (MSPS).


Asunto(s)
Fragmentación del ADN , Meiosis , Passeriformes/genética , Cromosomas Sexuales/genética , Espermatocitos/citología , Animales , Femenino , Masculino , Especificidad de la Especie
15.
Biochim Biophys Acta ; 1794(2): 193-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19059367

RESUMEN

An oligonucleotide-based assay (OBA) was used to identify novel co-factors that can be recruited by the deoxyribonucleic acid (DNA)-bound androgen receptor (AR). Nuclear extracts obtained from LNCaP cells, after incubation with R1881, were incubated with biotinylated oligonucleotides bound to streptavidin coated beads. The oligonucleotides contain 3 copies in tandem of the androgen responsive element ARE1 from the prostate specific antigen (PSA) gene promoter. As control incubation, a scrambled version of the tandem ARE1 was used. Immunoblots of the eluents revealed that the AR was bound to the ARE1 oligonucleotide and to a much lesser extent to the scrambled oligonucleotide. Proteins eluted from the oligonucleotides, were separated on a 5-15% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gradient gel, followed by identification using mass spectrometry. Identified proteins were scored for having one or more of the following known properties: nuclear localization, involved in transcription regulation, involvement in steroid hormone receptor (SHR) function, or specifical involvement in AR function. A total number of 85 nuclear proteins were found in two separate OBAs. Based on peptide counting, we found enrichment of 7 proteins eluted from the ARE1 oligonucleotide, compared to the scrambled oligonucleotide. Taken together with the obtained scores, these proteins are considered putative AR co-factors. One of these proteins, DDX17, is known to be a co-factor for estrogen receptor alpha (ERalpha), but has never been associated with AR function. The results indicate that the ARE oligonucleotide-based assay may allow enrichment of new candidate DNA-bound AR interacting proteins.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ADN/metabolismo , Receptores Androgénicos/fisiología , Biotina , Línea Celular Tumoral , Humanos , Masculino , Metribolona/farmacología , Oligonucleótidos , Regiones Promotoras Genéticas , Antígeno Prostático Específico/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores Androgénicos/genética , Elementos de Respuesta , Estreptavidina
16.
BMC Genomics ; 11: 367, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20537150

RESUMEN

BACKGROUND: The ubiquitin-conjugating enzyme HR6B is required for spermatogenesis in mouse. Loss of HR6B results in aberrant histone modification patterns on the trancriptionally silenced X and Y chromosomes (XY body) and on centromeric chromatin in meiotic prophase. We studied the relationship between these chromatin modifications and their effects on global gene expression patterns, in spermatocytes and spermatids. RESULTS: HR6B is enriched on the XY body and on centromeric regions in pachytene spermatocytes. Global gene expression analyses revealed that spermatid-specific single- and multicopy X-linked genes are prematurely expressed in Hr6b knockout spermatocytes. Very few other differences in gene expression were observed in these cells, except for upregulation of major satellite repeat transcription. In contrast, in Hr6b knockout spermatids, 7298 genes were differentially expressed; 65% of these genes was downregulated, but we observed a global upregulation of gene transcription from the X chromosome. In wild type spermatids, approximately 20% of the single-copy X-linked genes reach an average expression level that is similar to the average expression from autosomes. CONCLUSIONS: Spermatids maintain an enrichment of repressive chromatin marks on the X chromosome, originating from meiotic prophase, but this does not interfere with transcription of the single-copy X-linked genes that are reactivated or specifically activated in spermatids. HR6B represses major satellite repeat transcription in spermatocytes, and functions in the maintenance of X chromosome silencing in spermatocytes and spermatids. It is discussed that these functions involve modification of chromatin structure, possibly including H2B ubiquitylation.


Asunto(s)
Espermátides/metabolismo , Espermatocitos/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Inactivación del Cromosoma X , Cromosoma X/genética , Animales , Proteínas de Ciclo Celular/genética , Centrómero/genética , Centrómero/metabolismo , Cromatina/genética , Cromatina/metabolismo , Dosificación de Gen/genética , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Ligados a X/genética , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Especificidad de Órganos , Fosfoproteínas/genética , Testículo/metabolismo , Transcripción Genética , Activación Transcripcional , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Regulación hacia Arriba , Cromosoma X/metabolismo , Cromosoma Y/genética
17.
DNA Repair (Amst) ; 8(2): 190-201, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19013543

RESUMEN

The ubiquitin ligase RAD18 is involved in different DNA repair processes. Here, we show that in G1 phase, human RAD18 accumulates in a few relatively large spontaneous foci that contain proteins involved in double-strand break (DSB) repair. These foci persist until cells enter S phase, when numerous small foci appear. At these sites, only 20% of RAD18 colocalizes with PCNA, a known RAD18 substrate. In late G2 phase, RAD18 relocates to nucleoli. After UVC irradiation, PCNA accumulates at the damaged site, followed by RAD18, independent of the cell cycle phase. After induction of DSBs, using low-power multi-photon laser, RAD18 accumulated at the DSB sites, but no PCNA accumulation was observed. Our data show that RAD18 accumulates on DSBs independent of the cell cycle phase. DSBs marked by RAD18 and RAD51 are also positive for RPA in G1 phase, and these DSBs persist until S phase. In addition, we show that DSBs generated in G2 phase are not all repaired, and are observed again in the next G1 phase. We conclude that repair of induced and spontaneous DSBs that accumulate RAD18 and RAD51 in G1 phase cells is delayed until S phase.


Asunto(s)
Ciclo Celular , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Ciclo Celular/efectos de la radiación , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Recuperación de Fluorescencia tras Fotoblanqueo , Fase G1/efectos de la radiación , Células HeLa , Humanos , Cinética , Rayos Láser , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transporte de Proteínas/efectos de la radiación , Radiación Ionizante , Proteínas Recombinantes de Fusión/metabolismo , Fase S/efectos de la radiación , Ubiquitina-Proteína Ligasas , Rayos Ultravioleta
18.
Dev Biol ; 317(1): 270-81, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18384767

RESUMEN

In meiotic prophase of male placental mammals, the heterologous X and Y chromosomes remain largely unsynapsed, which activates meiotic sex chromosome inactivation (MSCI), leading to formation of the transcriptionally silenced XY body. MSCI is most likely related to meiotic silencing of unsynapsed chromatin (MSUC), a mechanism that can silence autosomal unsynapsed chromatin. However, heterologous synapsis and escape from silencing also occur. In mammalian species, formation of DNA double strand breaks (DSBs) during leptotene precedes meiotic chromosome pairing. These DSBs are essential to achieve full synapsis of homologous chromosomes. We generated 25% extra meiotic DSBs by whole body irradiation of mice. This leads to a significant increase in meiotic recombination frequency. In mice carrying translocation chromosomes with synaptic problems, we observed an approximately 35% increase in asynapsis and MSUC of the nonhomologous region in the smallest chromosome pair following irradiation. However, the same nonhomologous region in the largest chromosome pair, shows complete synapsis and escape from MSUC in almost 100% of the nuclei, irrespective of exposure to irradiation. We propose that prevention of synapsis and associated activation of MSUC is linked to the presence of unrepaired meiotic DSBs in the nonhomologous region. Also, spreading of synaptonemal complex formation from regions of homology may act as an opposing force, and drive heterologous synapsis.


Asunto(s)
Cromatina/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Rayos gamma , Profase Meiótica I , Animales , Emparejamiento Cromosómico , Intercambio Genético , Masculino , Ratones , Recombinasa Rad51/metabolismo
19.
Mol Cell Biol ; 26(3): 976-89, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428451

RESUMEN

Homologous recombination is a versatile DNA damage repair pathway requiring Rad51 and Rad54. Here we show that a mammalian Rad54 paralog, Rad54B, displays physical and functional interactions with Rad51 and DNA that are similar to those of Rad54. While ablation of Rad54 in mouse embryonic stem (ES) cells leads to a mild reduction in homologous recombination efficiency, the absence of Rad54B has little effect. However, the absence of both Rad54 and Rad54B dramatically reduces homologous recombination efficiency. Furthermore, we show that Rad54B protects ES cells from ionizing radiation and the interstrand DNA cross-linking agent mitomycin C. Interestingly, at the ES cell level the paralogs do not display an additive or synergic interaction with respect to mitomycin C sensitivity, yet animals lacking both Rad54 and Rad54B are dramatically sensitized to mitomycin C compared to either single mutant. This suggests that the paralogs possibly function in a tissue-specific manner. Finally, we show that Rad54, but not Rad54B, is needed for a normal distribution of Rad51 on meiotic chromosomes. Thus, even though the paralogs have similar biochemical properties, genetic analysis in mice uncovered their nonoverlapping roles.


Asunto(s)
Daño del ADN , ADN Helicasas/fisiología , Reparación del ADN , Proteínas Nucleares/fisiología , Recombinación Genética , Animales , Antibióticos Antineoplásicos/farmacología , Aberraciones Cromosómicas , Cromosomas/química , ADN Helicasas/genética , Proteínas de Unión al ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Meiosis , Ratones , Ratones Mutantes , Mitomicina/farmacología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Recombinasa Rad51/análisis , Recombinasa Rad51/metabolismo , Tolerancia a Radiación/genética , Células Madre/efectos de los fármacos , Células Madre/enzimología , Células Madre/efectos de la radiación
20.
Mol Cell Endocrinol ; 292(1-2): 69-78, 2008 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-18656523

RESUMEN

A novel mutation F826L located within the ligand binding domain (LBD) of the human androgen receptor (AR) was investigated. This mutation was found in a boy with severe penoscrotal hypospadias (classified as 46,XY DSD). The AR mutant F826L appeared to be indistinguishable from the wild-type AR, with respect to ligand binding affinity, transcriptional activation of MMTV-luciferase and ARE2-TATA-luciferase reporter genes, protein level in genital skin fibroblasts (GSFs), and sub-cellular distribution in transfected cells. However, an at least two-fold higher NH2-/COOH-terminal domain interaction was found in luciferase and GST pull-down assays. A two-fold increase was also observed for TIF2 (transcription intermediary factor 2) co-activation of the AR F826L COOH-terminal domain. This increase could not be explained by a higher stability of the mutant protein, which was within wild-type range. Repression of transactivation by the nuclear receptor co-repressor (N-CoR) was not affected by the AR F826L mutation. The observed properties of AR F826L would be in agreement with an increased activity rather than with a partial defective AR transcriptional activation. It is concluded that the penoscrotal hypospadias in the present case is caused by an as yet unknown mechanism, which still may involve the mutant AR.


Asunto(s)
Sustitución de Aminoácidos , Síndrome de Resistencia Androgénica/genética , Mutación/genética , Coactivador 2 del Receptor Nuclear/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética , Línea Celular , Preescolar , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Prepucio/citología , Humanos , Inmunoprecipitación , Lactante , Ligandos , Masculino , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Fracciones Subcelulares/metabolismo , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA