Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 15(7): e1008290, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31329581

RESUMEN

[This corrects the article DOI: 10.1371/journal.pgen.1002900.].

2.
Reproduction ; 159(4): X1, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32065737

RESUMEN

The journal and the authors apologise for an error in the above titled article published in this journal (vol 144, pp 433­445). The authors inadvertently presented duplicate sperm images for XY and XESxrbO mouse testes of Fig. 6 (bottom panels). This error does not change the findings of the paper, as this figure does not give a quantitative breakdown of the proportions of different shapes.

3.
Hum Mol Genet ; 25(24): 5300-5310, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27742779

RESUMEN

During spermatogenesis, germ cells that fail to synapse their chromosomes or fail to undergo meiotic sex chromosome inactivation (MSCI) are eliminated via apoptosis during mid-pachytene. Previous work showed that Y-linked genes Zfy1 and Zfy2 act as 'executioners' for this checkpoint, and that wrongful expression of either gene during pachytene triggers germ cell death. Here, we show that in mice, Zfy genes are also necessary for efficient MSCI and the sex chromosomes are not correctly silenced in Zfy-deficient spermatocytes. This unexpectedly reveals a triple role for Zfy at the mid-pachytene checkpoint in which Zfy genes first promote MSCI, then monitor its progress (since if MSCI is achieved, Zfy genes will be silenced), and finally execute cells with MSCI failure. This potentially constitutes a negative feedback loop governing this critical checkpoint mechanism.


Asunto(s)
Proteínas de Unión al ADN/genética , Espermatocitos/metabolismo , Factores de Transcripción/genética , Inactivación del Cromosoma X/genética , Animales , Masculino , Meiosis/genética , Ratones , Espermatocitos/crecimiento & desarrollo , Espermatogénesis/genética , Cromosoma X/genética
4.
Chromosoma ; 125(2): 177-88, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26596988

RESUMEN

In a male mouse, meiosis markers of processed DNA double strand breaks (DSBs) such as DMC1 and RAD51 are regularly seen in the non-PAR region of the X chromosome; these disappear late in prophase prior to entry into the first meiotic metaphase. Marker evidence for DSBs occurring in the non-PAR region of the Y chromosome is limited. Nevertheless, historically it has been documented that recombination can occur within the mouse Y short arm (Yp) when an additional Yp segment is attached distal to the X and/or the Y pseudoautosomal region (PAR). A number of recombinants identified among offsprings involved unequal exchanges involving repeated DNA segments; however, equal exchanges will have frequently been missed because of the paucity of markers to differentiate between the two Yp segments. Here, we discuss this historical data and present extensive additional data obtained for two mouse models with Yp additions to the X PAR. PCR genotyping enabled identification of a wider range of potential recombinants; the proportions of Yp exchanges identified among the recombinants were 9.7 and 22.4 %. The frequency of these exchanges suggests that the Yp segment attached to the X PAR is subject to the elevated level of recombinational DSBs that characterizes the PAR.


Asunto(s)
Ratones/genética , Regiones Pseudoautosómicas/genética , Recombinación Genética , Cromosoma X/genética , Cromosoma Y/genética , Animales , Animales no Consanguíneos , Femenino , Masculino , Meiosis
6.
Development ; 141(4): 855-66, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24496622

RESUMEN

Outbred XY(Sry-) female mice that lack Sry due to the 11 kb deletion Sry(dl1Rlb) have very limited fertility. However, five lines of outbred XY(d) females with Y chromosome deletions Y(Del(Y)1Ct)-Y(Del(Y)5Ct) that deplete the Rbmy gene cluster and repress Sry transcription were found to be of good fertility. Here we tested our expectation that the difference in fertility between XO, XY(d-1) and XY(Sry-) females would be reflected in different degrees of oocyte depletion, but this was not the case. Transgenic addition of Yp genes to XO females implicated Zfy2 as being responsible for the deleterious Y chromosomal effect on fertility. Zfy2 transcript levels were reduced in ovaries of XY(d-1) compared with XY(Sry-) females in keeping with their differing fertility. In seeking the biological basis of the impaired fertility we found that XY(Sry-), XY(d-1) and XO,Zfy2 females produce equivalent numbers of 2-cell embryos. However, in XY(Sry-) and XO,Zfy2 females the majority of embryos arrested with 2-4 cells and almost no blastocysts were produced; by contrast, XY(d-1) females produced substantially more blastocysts but fewer than XO controls. As previously documented for C57BL/6 inbred XY females, outbred XY(Sry-) and XO,Zfy2 females showed frequent failure of the second meiotic division, although this did not prevent the first cleavage. Oocyte transcriptome analysis revealed major transcriptional changes resulting from the Zfy2 transgene addition. We conclude that Zfy2-induced transcriptional changes in oocytes are sufficient to explain the more severe fertility impairment of XY as compared with XO females.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Infertilidad Femenina/genética , Meiosis/genética , Oocitos/metabolismo , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/genética , Proteína de la Región Y Determinante del Sexo/deficiencia , Factores de Transcripción/metabolismo , Cromosoma Y/genética , Animales , Western Blotting , Cruzamiento , Fase de Segmentación del Huevo/patología , Fase de Segmentación del Huevo/fisiología , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Genotipo , Modelos Lineales , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Factores de Transcripción/genética
7.
PLoS Genet ; 10(6): e1004444, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24967676

RESUMEN

Mouse Zfy1 and Zfy2 encode zinc finger transcription factors that map to the short arm of the Y chromosome (Yp). They have previously been shown to promote meiotic quality control during pachytene (Zfy1 and Zfy2) and at the first meiotic metaphase (Zfy2). However, from these previous studies additional roles for genes encoded on Yp during meiotic progression were inferred. In order to identify these genes and investigate their function in later stages of meiosis, we created three models with diminishing Yp and Zfy gene complements (but lacking the Y-long-arm). Since the Y-long-arm mediates pairing and exchange with the X via their pseudoautosomal regions (PARs) we added a minute PAR-bearing X chromosome derivative to enable formation of a sex bivalent, thus avoiding Zfy2-mediated meiotic metaphase I (MI) checkpoint responses to the unpaired (univalent) X chromosome. Using these models we obtained definitive evidence that genetic information on Yp promotes meiosis II, and by transgene addition identified Zfy1 and Zfy2 as the genes responsible. Zfy2 was substantially more effective and proved to have a much more potent transactivation domain than Zfy1. We previously established that only Zfy2 is required for the robust apoptotic elimination of MI spermatocytes in response to a univalent X; the finding that both genes potentiate meiosis II led us to ask whether there was de novo Zfy1 and Zfy2 transcription in the interphase between meiosis I and meiosis II, and this proved to be the case. X-encoded Zfx was also expressed at this stage and Zfx over-expression also potentiated meiosis II. An interphase between the meiotic divisions is male-specific and we previously hypothesised that this allows meiosis II critical X and Y gene reactivation following sex chromosome silencing in meiotic prophase. The interphase transcription and meiosis II function of Zfx, Zfy1 and Zfy2 validate this hypothesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Interfase/genética , Meiosis/genética , Espermatogénesis/genética , Factores de Transcripción/genética , Animales , Apoptosis/fisiología , Proteínas de Unión al ADN/biosíntesis , Femenino , Genes Ligados a Y , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Espermatocitos/fisiología , Factores de Transcripción/biosíntesis , Activación Transcripcional/genética , Cromosoma Y/genética
8.
Nat Rev Genet ; 10(3): 207-16, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19188923

RESUMEN

During mammalian meiosis, synapsis of paternal and maternal chromosomes and the generation of DNA breaks are needed to allow reshuffling of parental genes. In mammals errors in synapsis are associated with a male-biased meiotic impairment, which has been attributed to a response to persisting DNA double-stranded breaks in the asynapsed chromosome segments. Recently it was discovered that the chromatin of asynapsed chromosome segments is transcriptionally silenced, providing new insights into the connection between asynapsis and meiotic impairment.


Asunto(s)
Emparejamiento Cromosómico , Cromosomas/metabolismo , Meiosis , Animales , Femenino , Humanos , Masculino
9.
PLoS Genet ; 8(9): e1002900, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028340

RESUMEN

Intragenomic conflicts arise when a genetic element favours its own transmission to the detriment of others. Conflicts over sex chromosome transmission are expected to have influenced genome structure, gene regulation, and speciation. In the mouse, the existence of an intragenomic conflict between X- and Y-linked multicopy genes has long been suggested but never demonstrated. The Y-encoded multicopy gene Sly has been shown to have a predominant role in the epigenetic repression of post meiotic sex chromatin (PMSC) and, as such, represses X and Y genes, among which are its X-linked homologs Slx and Slxl1. Here, we produced mice that are deficient for both Sly and Slx/Slxl1 and observed that Slx/Slxl1 has an opposite role to that of Sly, in that it stimulates XY gene expression in spermatids. Slx/Slxl1 deficiency rescues the sperm differentiation defects and near sterility caused by Sly deficiency and vice versa. Slx/Slxl1 deficiency also causes a sex ratio distortion towards the production of male offspring that is corrected by Sly deficiency. All in all, our data show that Slx/Slxl1 and Sly have antagonistic effects during sperm differentiation and are involved in a postmeiotic intragenomic conflict that causes segregation distortion and male sterility. This is undoubtedly what drove the massive gene amplification on the mouse X and Y chromosomes. It may also be at the basis of cases of F1 male hybrid sterility where the balance between Slx/Slxl1 and Sly copy number, and therefore expression, is disrupted. To the best of our knowledge, our work is the first demonstration of a competition occurring between X and Y related genes in mammals. It also provides a biological basis for the concept that intragenomic conflict is an important evolutionary force which impacts on gene expression, genome structure, and speciation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Nucleares/genética , Proteínas/genética , Cromosoma X/genética , Cromosoma Y/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras del Transporte Vesicular , Animales , Epigénesis Genética , Femenino , Dosificación de Gen , Regulación de la Expresión Génica , Especiación Genética , Infertilidad Masculina , Masculino , Meiosis/genética , Ratones , Ratones Transgénicos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Proteínas/antagonistas & inhibidores , Cromatina Sexual/genética , Cromatina Sexual/metabolismo , Razón de Masculinidad , Espermátides/metabolismo , Espermatozoides/crecimiento & desarrollo , Espermatozoides/metabolismo
10.
Hum Mol Genet ; 21(12): 2631-45, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22407129

RESUMEN

Mammalian ZFY genes are located on the Y chromosome, and code putative transcription factors with 12-13 zinc fingers preceded by a large acidic (activating) domain. In mice, there are two genes, Zfy1 and Zfy2, which are expressed mainly in the testis. Their transcription increases in germ cells as they enter meiosis, both are silenced by meiotic sex chromosome inactivation (MSCI) during pachytene, and Zfy2 is strongly reactivated later in spermatids. Recently, we have shown that mouse Zfy2, but not Zfy1, is involved in triggering the apoptotic elimination of specific types of sex chromosomally aberrant spermatocytes. In humans, there is a single widely transcribed ZFY gene, and there is no evidence for a specific role in the testis. Here, we characterize ZFY transcription during spermatogenesis in mice and humans. In mice, we define a variety of Zfy transcripts, among which is a Zfy2 transcript that predominates in spermatids, and a Zfy1 transcript, lacking an exon encoding approximately half of the acidic domain, which predominates prior to MSCI. In humans, we have identified a major testis-specific ZFY transcript that encodes a protein with the same short acidic domain. This represents the first evidence that ZFY has a conserved function during human spermatogenesis. We further show that, in contrast to the full acidic domain, the short domain does not activate transcription in yeast, and we hypothesize that this explains the functional difference observed between Zfy1 and Zfy2 during mouse meiosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores de Transcripción de Tipo Kruppel/genética , Testículo/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Activación Transcripcional , Empalme Alternativo , Animales , Secuencia de Bases , Sitios de Unión/genética , Secuencia Conservada/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Espermatocitos/metabolismo , Espermatogénesis/genética , Testículo/citología , Testículo/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Dedos de Zinc/genética
11.
Nat Genet ; 37(1): 41-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15580272

RESUMEN

In Neurospora, DNA unpaired in meiosis both is silenced and induces silencing of all DNA homologous to it. This process, called meiotic silencing by unpaired DNA, is thought to protect the host genome from invasion by transposable elements. We now show that silencing of unpaired (unsynapsed) chromosome regions also takes place in the mouse during both male and female meiosis. The tumor suppressor protein BRCA1 is implicated in this silencing, mirroring its role in the meiotic silencing of the X and Y chromosomes in normal male meiosis. These findings impact on the interpretation of the relationship between synaptic errors and sterility in mammals and extend our understanding of the biology of Brca1.


Asunto(s)
Emparejamiento Cromosómico , Silenciador del Gen , Meiosis , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN , Femenino , Genes BRCA1/fisiología , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Oocitos/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Espermatocitos/fisiología , Translocación Genética , Cromosoma X , Cromosoma Y
12.
Reproduction ; 144(4): 433-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22869781

RESUMEN

We recently used three XO male mouse models with varying Y short-arm (Yp) gene complements, analysed at 30 days post partum, to demonstrate a Yp gene requirement for the apoptotic elimination of spermatocytes with a univalent X chromosome at the first meiotic metaphase. The three mouse models were i) XSxr(a)O in which the Yp-derived Tp(Y)1Ct(Sxr-a) sex reversal factor provides an almost complete Yp gene complement, ii) XSxr(b)O,Eif2s3y males in which Tp(Y)1Ct(Sxr-b) has a deletion completely or partially removing eight Yp genes - the Yp gene Eif2s3y has been added as a transgene to support spermatogonial proliferation, and iii) XOSry,Eif2s3y males in which the Sry transgene directs gonad development along the male pathway. In this study, we have used the same mouse models analysed at 6 weeks of age to investigate potential Yp gene involvement in spermiogenesis. We found that all three mouse models produce haploid and diploid spermatids and that the diploid spermatids showed frequent duplication of the developing acrosomal cap during the early stages. However, only in XSxr(a)O males did spermiogenesis continue to completion. Most strikingly, in XOSry,Eif2s3y males, spermatid development arrested at round spermatid step 7 so that no sperm head restructuring or tail development was observed. In contrast, in XSxr(b)O,Eif2s3y males, spermatids with substantial sperm head and tail morphogenesis could be easily found, although this was delayed compared with XSxr(a)O. We conclude that Sxr(a) (and therefore Yp) includes genetic information essential for sperm morphogenesis and that this is partially retained in Sxr(b).


Asunto(s)
Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/metabolismo , Genes Ligados a Y , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/metabolismo , Proteína de la Región Y Determinante del Sexo/metabolismo , Espermátides/metabolismo , Espermatogénesis , Acrosoma/metabolismo , Acrosoma/patología , Animales , Deleción Cromosómica , Cromosomas Humanos Y/metabolismo , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Eliminación de Gen , Infertilidad Masculina , Masculino , Meiosis , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Recombinantes de Fusión/metabolismo , Aberraciones Cromosómicas Sexuales , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual/patología , Proteína de la Región Y Determinante del Sexo/genética , Cola del Espermatozoide/metabolismo , Cola del Espermatozoide/patología , Espermátides/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
PLoS Biol ; 7(11): e1000244, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19918361

RESUMEN

Studies of mice with Y chromosome long arm deficiencies suggest that the male-specific region (MSYq) encodes information required for sperm differentiation and postmeiotic sex chromatin repression (PSCR). Several genes have been identified on MSYq, but because they are present in more than 40 copies each, their functions cannot be investigated using traditional gene targeting. Here, we generate transgenic mice producing small interfering RNAs that specifically target the transcripts of the MSYq-encoded multicopy gene Sly (Sycp3-like Y-linked). Microarray analyses performed on these Sly-deficient males and on MSYq-deficient males show a remarkable up-regulation of sex chromosome genes in spermatids. SLY protein colocalizes with the X and Y chromatin in spermatids of normal males, and Sly deficiency leads to defective repressive marks on the sex chromatin, such as reduced levels of the heterochromatin protein CBX1 and of histone H3 methylated at lysine 9. Sly-deficient mice, just like MSYq-deficient mice, have severe impairment of sperm differentiation and are near sterile. We propose that their spermiogenesis phenotype is a consequence of the change in spermatid gene expression following Sly deficiency. To our knowledge, this is the first successful targeted disruption of the function of a multicopy gene (or of any Y gene). It shows that SLY has a predominant role in PSCR, either via direct interaction with the spermatid sex chromatin or via interaction with sex chromatin protein partners. Sly deficiency is the major underlying cause of the spectrum of anomalies identified 17 y ago in MSYq-deficient males. Our results also suggest that the expansion of sex-linked spermatid-expressed genes in mouse is a consequence of the enhancement of PSCR that accompanies Sly amplification.


Asunto(s)
Dosificación de Gen , Células Germinativas/citología , Meiosis , Cromosoma Y , Animales , Cromosomas de los Mamíferos , Regulación de la Expresión Génica , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Transgénicos , Cromosomas Sexuales , Espermátides
14.
Dev Cell ; 10(4): 521-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16580996

RESUMEN

Transcriptional silencing of the sex chromosomes during male meiosis (MSCI) is conserved among organisms with limited sex chromosome synapsis, including mammals. Since the 1990s the prevailing view has been that MSCI in mammals is transient, with sex chromosome reactivation occurring as cells exit meiosis. Recently, we found that any chromosome region unsynapsed during pachytene of male and female mouse meiosis is subject to transcriptional silencing (MSUC), and we hypothesized that MSCI is an inevitable consequence of this more general meiotic silencing mechanism. Here, we provide direct evidence that asynapsis does indeed drive MSCI. We also show that a substantial degree of transcriptional repression of the sex chromosomes is retained postmeiotically, and we provide evidence that this postmeiotic repression is a downstream consequence of MSCI/MSUC. While this postmeiotic repression occurs after the loss of MSUC-related proteins at the end of prophase, other histone modifications associated with transcriptional repression have by then become established.


Asunto(s)
Meiosis/genética , Fase Paquiteno/genética , Cromosomas Sexuales/genética , Espermátides/fisiología , Animales , Femenino , Silenciador del Gen , Hibridación Fluorescente in Situ/métodos , Masculino , Ratones , Cromosomas Sexuales/metabolismo , Espermátides/citología , Transcripción Genética , Cromosoma X/genética , Cromosoma X/metabolismo , Inactivación del Cromosoma X/genética , Cariotipo XYY/genética , Cromosoma Y/genética , Cromosoma Y/metabolismo
15.
BMC Genomics ; 11: 82, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20122165

RESUMEN

BACKGROUND: X monosomic mice (39,XO) have a remarkably mild phenotype when compared to women with Turner syndrome (45,XO). The generally accepted hypothesis to explain this discrepancy is that the number of genes on the mouse X chromosome which escape X inactivation, and thus are expressed at higher levels in females, is very small. However this hypothesis has never been tested and only a small number of genes have been assayed for their X-inactivation status in the mouse. We performed a global expression analysis in four somatic tissues (brain, liver, kidney and muscle) of adult 40,XX and 39,XO mice using the Illumina Mouse WG-6 v1_1 Expression BeadChip and an extensive validation by quantitative real time PCR, in order to identify which genes are expressed from both X chromosomes. RESULTS: We identified several genes on the X chromosome which are overexpressed in XX females, including those previously reported as escaping X inactivation, as well as new candidates. However, the results obtained by microarray and qPCR were not fully concordant, illustrating the difficulty in ascertaining modest fold changes, such as those expected for genes escaping X inactivation. Remarkably, considerable variation was observed between tissues, suggesting that inactivation patterns may be tissue-dependent. Our analysis also exposed several autosomal genes involved in mitochondrial metabolism and in protein translation which are differentially expressed between XX and XO mice, revealing secondary transcriptional changes to the alteration in X chromosome dosage. CONCLUSIONS: Our results support the prediction that the mouse inactive X chromosome is largely silent, while providing a list of the genes potentially escaping X inactivation in rodents. Although the lower expression of X-linked genes in XO mice may not be relevant in the particular tissues/systems which are affected in human X chromosome monosomy, genes deregulated in XO mice are good candidates for further study in an involvement in Turner Syndrome phenotype.


Asunto(s)
Perfilación de la Expresión Génica , Síndrome de Turner/genética , Inactivación del Cromosoma X , Cromosoma X/genética , Alelos , Animales , Femenino , Redes Reguladoras de Genes , Genes Ligados a X , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
16.
Biol Reprod ; 81(2): 250-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19176879

RESUMEN

Deletion analysis has established that genes on the Y chromosome are essential for normal sperm production in humans, mice, and Drosophila. In mice, long-arm deletions have an impact on spermiogenesis, with the most extensive deletions resulting in severe sperm head malformations and infertility. Intriguingly, smaller deletions are compatible with fertility but result in a distorted sex ratio in favor of females, and recently it was found that Y long-arm deletions are also associated with a marked upregulation of several X-encoded and Y-encoded spermatid-expressed genes. The mouse Y long arm encodes a number of distinct transcripts, each of which derives from multiple gene copies. Of these multicopy genes, the recently described Sly has been favored as the gene underlying the spermiogenic defects associated with Y long-arm deletions. To assess the candidacy of Sly, the expression of this gene was examined in the testis at the transcript and protein levels. Sly is transcribed after the first meiotic division in secondary spermatocytes and round spermatids and encodes two transcript variants, Sly_v1 and Sly_v2 (proteins referred to as SLY1 and SLY2). We raised an antibody against SLY1 which detected the protein in round and early elongating spermatids, where it is predominantly cytoplasmic. Yeast two-hybrid and coimmunoprecipitation studies demonstrated that SLY1 interacts with the acrosomal protein DKKL1, the histone acetyltransferase KAT5 (also known as TIP60), and the microtubule-associated protein APPBP2. Together, these data suggest SLY1 may be involved in multiple processes during spermiogenesis, including the control of gene expression and the development or function of the acrosome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Genes Ligados a Y , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Deleción Cromosómica , Epidídimo/metabolismo , Femenino , Expresión Génica , Biblioteca de Genes , Histona Acetiltransferasas/genética , Inmunohistoquímica , Inmunoprecipitación , Lisina Acetiltransferasa 5 , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Unión Proteica , Isoformas de Proteínas , Transporte de Proteínas , Proteínas de Unión al ARN/genética , Túbulos Seminíferos/citología , Túbulos Seminíferos/metabolismo , Alineación de Secuencia , Espermatozoides/citología , Simportadores/genética , Simportadores/metabolismo , Testículo/citología , Testículo/metabolismo , Transactivadores , Técnicas del Sistema de Dos Híbridos
17.
Biol Reprod ; 81(2): 353-61, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19420387

RESUMEN

The mouse Y chromosome long arm (Yq) comprises approximately 70 Mb of repetitive, male-specific DNA together with a short (0.7-Mb) pseudoautosomal region (PAR). The repetitive non-PAR region (NPYq) encodes genes whose deficiency leads to subfertility and infertility, resulting from impaired spermiogenesis. In XSxr(a)Y*(X) mice, the only Y-specific material is provided by the Y chromosome short arm-derived sex reversal factor Sxr(a), which is attached to the X chromosome PAR; these males (NPYq- males) produce sperm with severely malformed heads and are infertile. In the present study, we investigated sperm function in these mice in the context of intracytoplasmic sperm injection (ICSI). Of 261 oocytes injected, 103 reached the 2-cell stage, and 46 developed to liveborn offspring. Using Xist RT-PCR genotyping as well as gamete and somatic cell karyotyping, all six predicted genotypes were identified among ICSI-derived progeny. The sex chromosome constitution of NPYq- males does not allow production of offspring with the same genotype, but one of the expected offspring genotypes is XY*(X)Sxr(a) (NPYq-(2)), which has the same Y gene complement as NPYq-. Analysis of NPYq-(2) males revealed they had normal-sized testes with ongoing spermatogenesis. Like NPYq- males, these males were infertile, and their sperm had malformed heads that nevertheless fertilized eggs via ICSI. In vitro fertilization (IVF), however, was unsuccessful. Overall, we demonstrated that a lack of NPYq-encoded genes does not interfere with the ability of sperm to fertilize oocytes via ICSI but does prevent fertilization via IVF. Thus, NPYq-encoded gene functions are not required after the sperm have entered the oocyte. The present work also led to development of a new mouse model lacking NPYq gene complement that will facilitate future studies of Y-encoded gene function.


Asunto(s)
Genes Ligados a Y/genética , Infertilidad Masculina/genética , Nacimiento Vivo/genética , Aberraciones Cromosómicas Sexuales , Inyecciones de Esperma Intracitoplasmáticas , Espermatogénesis/genética , Cromosoma Y/genética , Análisis de Varianza , Animales , Células de la Médula Ósea , Epidídimo/citología , Femenino , Fertilidad , Fertilización In Vitro , Cariotipificación , Funciones de Verosimilitud , Modelos Lineales , Masculino , Ratones , Ratones Endogámicos C57BL , Oocitos , Tamaño de los Órganos , Embarazo , Capacitación Espermática , Recuento de Espermatozoides , Cabeza del Espermatozoide/ultraestructura , Motilidad Espermática , Testículo/citología , Testículo/patología
18.
Genome Biol ; 20(1): 160, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399122

RESUMEN

Following publication of the original article [1], the following error was reported: The actin control panel in Fig. 3 of this paper is reproduced from Fig. 7 of Touré et al, 2004 [2] by kind permission of the Genetics Society of America. Touré et al, 2004 used Northern blotting to show that the Y-linked genes Ssty1 and Ssty2 have reduced expression in a range of mouse genotypes with deletions on the Y chromosome long arm. This paper shows that two novel genes, Sly and Asty are also present on mouse Yq and have reduced expression in these deleted genotypes. A further companion paper was published in Human Molecular Genetics (Ellis et al, 2005 [3]) showing that X-linked genes are upregulated in the various deleted genotypes. Since two of the genotypes concerned are sterile and very hard to generate, all the Northern blot experiments in these papers were performed on a single membrane that was stripped and re-probed with a range of different X- and Y-linked genes. The same beta-actin loading control image thus necessarily applies to all the data presented, and was shown in all three papers. We regret that this was not mentioned appropriately in the Methods and figure legends at the time of publication.

19.
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
20.
J Neurosci ; 26(8): 2335-42, 2006 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-16495461

RESUMEN

Across human cultures and mammalian species, sex differences can be found in the expression of aggression and parental nurturing behaviors: males are typically more aggressive and less parental than females. These sex differences are primarily attributed to steroid hormone differences during development and/or adulthood, especially the higher levels of androgens experienced by males, which are caused ultimately by the presence of the testis-determining gene Sry on the Y chromosome. The potential for sex differences arising from the different complements of sex-linked genes in male and female cells has received little research attention. To directly test the hypothesis that social behaviors are influenced by differences in sex chromosome complement other than Sry, we used a transgenic mouse model in which gonadal sex and sex chromosome complement are uncoupled. We find that latency to exhibit aggression and one form of parental behavior, pup retrieval, can be influenced by both gonadal sex and sex chromosome complement. For both behaviors, females but not males with XX sex chromosomes differ from XY. We also measured vasopressin immunoreactivity in the lateral septum, which was higher in gonadal males than females, but also differed according to sex chromosome complement. These results imply that a gene(s) on the sex chromosomes (other than Sry) affects sex differences in brain and behavior. Identifying the specific X and/or Y genes involved will increase our understanding of normal and abnormal aggression and parental behavior, including behavioral abnormalities associated with mental illness.


Asunto(s)
Agresión/psicología , Conducta Animal/fisiología , Trastornos del Desarrollo Sexual , Genes sry/fisiología , Padres , Cromosomas Sexuales/fisiología , Procesos de Determinación del Sexo , Animales , Conducta Exploratoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Caracteres Sexuales , Conducta Social
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA