Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Reproduction ; 159(4): X1, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32065737

RESUMO

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.
Genome Biol ; 20(1): 160, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399122

RESUMO

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.

4.
PLoS Genet ; 15(7): e1008290, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31329581

RESUMO

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

5.
Biol Sex Differ ; 7: 68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27999654

RESUMO

In animals with heteromorphic sex chromosomes, all sex differences originate from the sex chromosomes, which are the only factors that are consistently different in male and female zygotes. In mammals, the imbalance in Y gene expression, specifically the presence vs. absence of Sry, initiates the differentiation of testes in males, setting up lifelong sex differences in the level of gonadal hormones, which in turn cause many sex differences in the phenotype of non-gonadal tissues. The inherent imbalance in the expression of X and Y genes, or in the epigenetic impact of X and Y chromosomes, also has the potential to contribute directly to the sexual differentiation of non-gonadal cells. Here, we review the research strategies to identify the X and Y genes or chromosomal regions that cause direct, sexually differentiating effects on non-gonadal cells. Some mouse models are useful for separating the effects of sex chromosomes from those of gonadal hormones. Once direct "sex chromosome effects" are detected in these models, further studies are required to narrow down the list of candidate X and/or Y genes and then to identify the sexually differentiating genes themselves. Logical approaches to the search for these genes are reviewed here.

6.
Hum Mol Genet ; 25(24): 5300-5310, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27742779

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Espermatócitos/metabolismo , Fatores de Transcrição/genética , Inativação do Cromossomo X/genética , Animais , Masculino , Meiose/genética , Camundongos , Espermatócitos/crescimento & desenvolvimento , Espermatogênese/genética , Cromossomo X/genética
7.
PLoS One ; 11(1): e0145398, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26765744

RESUMO

A previous study indicated that genetic information encoded on the mouse Y chromosome short arm (Yp) is required for efficient completion of the second meiotic division (that generates haploid round spermatids), restructuring of the sperm head, and development of the sperm tail. Using mouse models lacking a Y chromosome but with varying Yp gene complements provided by Yp chromosomal derivatives or transgenes, we recently identified the Y-encoded zinc finger transcription factors Zfy1 and Zfy2 as the Yp genes promoting the second meiotic division. Using the same mouse models we here show that Zfy2 (but not Zfy1) contributes to the restructuring of the sperm head and is required for the development of the sperm tail. The preferential involvement of Zfy2 is consistent with the presence of an additional strong spermatid-specific promotor that has been acquired by this gene. This is further supported by the fact that promotion of sperm morphogenesis is also seen in one of the two markedly Yp gene deficient models in which a Yp deletion has created a Zfy2/1 fusion gene that is driven by the strong Zfy2 spermatid-specific promotor, but encodes a protein almost identical to that encoded by Zfy1. Our results point to there being further genetic information on Yp that also has a role in restructuring the sperm head.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Cabeça do Espermatozoide/metabolismo , Cauda do Espermatozoide/metabolismo , Espermatogênese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cromossomo Y/genética , Animais , Masculino , Camundongos , Modelos Animais , Morfogênese/genética , Mapeamento Físico do Cromossomo , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Cauda do Espermatozoide/ultraestrutura
8.
Chromosoma ; 125(2): 177-88, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26596988

RESUMO

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.


Assuntos
Camundongos/genética , Regiões Pseudoautossômicas/genética , Recombinação Genética , Cromossomo X/genética , Cromossomo Y/genética , Animais , Animais não Endogâmicos , Feminino , Masculino , Meiose
9.
PLoS Genet ; 10(6): e1004444, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967676

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Interfase/genética , Meiose/genética , Espermatogênese/genética , Fatores de Transcrição/genética , Animais , Apoptose/fisiologia , Proteínas de Ligação a DNA/biossíntese , Feminino , Genes Ligados ao Cromossomo Y , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Espermatócitos/fisiologia , Fatores de Transcrição/biossíntese , Ativação Transcricional/genética , Cromossomo Y/genética
10.
Development ; 141(4): 855-66, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24496622

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Infertilidade Feminina/genética , Meiose/genética , Oócitos/metabolismo , Transtornos do Cromossomo Sexual no Desenvolvimento Sexual/genética , Proteína da Região Y Determinante do Sexo/deficiência , Fatores de Transcrição/metabolismo , Cromossomo Y/genética , Animais , Western Blotting , Cruzamento , Fase de Clivagem do Zigoto/patologia , Fase de Clivagem do Zigoto/fisiologia , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Genótipo , Modelos Lineares , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Fatores de Transcrição/genética
11.
PLoS Genet ; 8(9): e1002900, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23028340

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Nucleares/genética , Proteínas/genética , Cromossomo X/genética , Cromossomo Y/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transporte Vesicular , Animais , Epigênese Genética , Feminino , Dosagem de Genes , Regulação da Expressão Gênica , Especiação Genética , Infertilidade Masculina , Masculino , Meiose/genética , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/deficiência , Proteínas/antagonistas & inibidores , Cromatina Sexual/genética , Cromatina Sexual/metabolismo , Razão de Masculinidade , Espermátides/metabolismo , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/metabolismo
12.
Reproduction ; 144(4): 433-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22869781

RESUMO

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).


Assuntos
Modelos Animais de Doenças , Fator de Iniciação 2 em Eucariotos/metabolismo , Genes Ligados ao Cromossomo Y , Transtornos do Cromossomo Sexual no Desenvolvimento Sexual/metabolismo , Proteína da Região Y Determinante do Sexo/metabolismo , Espermátides/metabolismo , Espermatogênese , Acrossomo/metabolismo , Acrossomo/patologia , Animais , Deleção Cromossômica , Cromossomos Humanos Y/metabolismo , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Deleção de Genes , Infertilidade Masculina , Masculino , Meiose , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/metabolismo , Aberrações dos Cromossomos Sexuais , Transtornos do Cromossomo Sexual no Desenvolvimento Sexual/patologia , Proteína da Região Y Determinante do Sexo/genética , Cauda do Espermatozoide/metabolismo , Cauda do Espermatozoide/patologia , Espermátides/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Hum Mol Genet ; 21(12): 2631-45, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407129

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Fatores de Transcrição Kruppel-Like/genética , Testículo/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional , Processamento Alternativo , Animais , Sequência de Bases , Sítios de Ligação/genética , Sequência Conservada/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização in Situ Fluorescente , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Espermatócitos/metabolismo , Espermatogênese/genética , Testículo/citologia , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética
14.
Psychoneuroendocrinology ; 37(2): 221-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21723668

RESUMO

Attention Deficit Hyperactivity Disorder (ADHD) is a common neurodevelopmental condition characterised by inattention, impulsivity and hyperactivity; it is frequently co-morbid with anxiety and conduct disorders, sleep perturbation and abnormal consummatory behaviours. Recent studies have implicated the neurosteroid-modulating enzyme steroid sulfatase (STS) as a modulator of ADHD-related endophenotypes. The effects of steroid sulfatase deficiency on homecage activity, feeding/drinking behaviours, anxiety-related behaviours (assayed in light-dark box and open field paradigms), social dominance and serum steroid hormone levels were determined by comparing 40,XY and 39,X(Y*)O mice. Subsequently, mice administered the steroid sulfatase inhibitor COUMATE acutely were compared to vehicle-treated mice on behavioural tasks sensitive to enzyme deficiency to dissociate between its developmental and ongoing effects. 39,X(Y*)O mice exhibited heightened reactivity to a novel environment, hyperactivity in the active phase, and increased water (but not food) consumption relative to 40,XY mice during a 24h period; the former group also demonstrated evidence for heightened emotional reactivity. There was no difference in social dominance between the 40,XY and 39,X(Y*)O mice. COUMATE administration had no effect on homecage activity, water consumption or anxiety measures in the open field. 39,X(Y*)O mice exhibited significantly lower dehydroepiandrosterone (DHEA) serum levels than 40,XY mice, but equivalent corticosterone levels. Together with previous data, the present results suggest that steroid sulfatase may influence core and associated ADHD behavioural endophenotypes via both developmental and ongoing mechanisms, and that the 39,X(Y*)O model may represent a useful tool for elucidating the neurobiological basis of these endophenotypes.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/enzimologia , Comportamento Animal/efeitos dos fármacos , Cumarínicos/farmacologia , Ictiose Ligada ao Cromossomo X/psicologia , Sulfonamidas/farmacologia , Animais , Transtorno do Deficit de Atenção com Hiperatividade/genética , Endofenótipos , Comportamento Exploratório/efeitos dos fármacos , Ictiose Ligada ao Cromossomo X/genética , Camundongos , Camundongos Mutantes
15.
Curr Biol ; 21(9): 787-93, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21530259

RESUMO

During male but not female mammalian meiosis, there is efficient apoptotic elimination of cells with unpaired (univalent) chromosomes at the first meiotic metaphase (MI) [1]. Apoptotic elimination of MI spermatocytes is seen in response to the univalent X chromosome of XSxr(a)O male mice [2], in which the X chromosome carries Sxr(a) [3, 4], the Y-chromosome-derived sex-reversal factor that includes the testis determinant Sry. Sxr(b) is an Sxr(a)-derived variant in which a deletion has removed six Y short-arm genes and created a Zfy2/Zfy1 fusion gene spanning the deletion breakpoint [4, 5]. XSxr(b)O males have spermatogonial arrest that can be overcome by the re-addition of Eif2s3y from the deletion as a transgene; however, XSxr(b)OEif2s3y transgenic males do not show the expected elimination of MI spermatocytes in response to the univalent [6]. Here we show that these XSxr(b)OEif2s3y males have an impaired apoptotic response with completion of the first meiotic division, but there is no second meiotic division. We then show that Zfy2 (but not the closely related Zfy1) is sufficient to reinstate the apoptotic response to the X univalent. These findings provide further insight into the basis for the much lower transmission of chromosomal errors originating at the first meiotic division in men than in women [7].


Assuntos
Apoptose/fisiologia , Pareamento Cromossômico/fisiologia , Proteínas de Ligação a DNA/metabolismo , Meiose/fisiologia , Metáfase/fisiologia , Espermatócitos/fisiologia , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Feminino , Imunofluorescência , Técnicas Histológicas , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Ovário/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Cromossomos Sexuais/genética , Proteína da Região Y Determinante do Sexo/genética , Fatores de Transcrição/genética , Transgenes/genética
16.
Curr Biol ; 20(23): 2117-23, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21093264

RESUMO

The mammalian X and Y chromosomes share little homology and are largely unsynapsed during normal meiosis. This asynapsis triggers inactivation of X- and Y-linked genes, or meiotic sex chromosome inactivation (MSCI). Whether MSCI is essential for male meiosis is unclear. Pachytene arrest and apoptosis is observed in mouse mutants in which MSCI fails, e.g., Brca1(-/-), H2afx(-/-), Sycp1(-/-), and Msh5(-/-). However, these also harbor defects in synapsis and/or recombination and as such may activate a putative pachytene checkpoint. Here we present evidence that MSCI failure is sufficient to cause pachytene arrest. XYY males exhibit Y-Y synapsis and Y chromosomal escape from MSCI without accompanying synapsis/recombination defects. We find that XYY males, like synapsis/recombination mutants, display pachytene arrest and that this can be circumvented by preventing Y-Y synapsis and associated Y gene expression. Pachytene expression of individual Y genes inserted as transgenes on autosomes shows that expression of the Zfy 1/2 paralogs in XY males is sufficient to phenocopy the pachytene arrest phenotype; insertion of Zfy 1/2 on the X chromosome where they are subject to MSCI prevents this response. Our findings show that MSCI is essential for male meiosis and, as such, provide insight into the differential severity of meiotic mutations' effects on male and female meiosis.


Assuntos
Fertilidade/genética , Inativação Gênica , Meiose/genética , Cromossomo X/genética , Cromossomo Y/genética , Animais , Animais Geneticamente Modificados , Apoptose , Pareamento Cromossômico , Feminino , Células Germinativas/citologia , Células Germinativas/fisiologia , Masculino , Camundongos
17.
Dev Cell ; 19(3): 477-84, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20833369

RESUMO

Differences between males and females are normally attributed to developmental and hormonal differences between the sexes. Here, we demonstrate differences between males and females in gene silencing using a heterochromatin-sensitive reporter gene. Using "sex-reversal" mouse models with varying sex chromosome complements, we found that this differential gene silencing was determined by X chromosome complement, rather than sex. Genome-wide transcription profiling showed that the expression of hundreds of autosomal genes was also sensitive to sex chromosome complement. These genome-wide analyses also uncovered a role for Sry in modulating autosomal gene expression in a sex chromosome complement-specific manner. The identification of this additional layer in the establishment of sexual dimorphisms has implications for understanding sexual dimorphisms in physiology and disease.


Assuntos
Proteínas do Sistema Complemento/genética , Transtornos do Desenvolvimento Sexual , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Cromossomos Sexuais/genética , Proteína da Região Y Determinante do Sexo/genética , Animais , Biomarcadores/metabolismo , Western Blotting , Proteínas do Sistema Complemento/metabolismo , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína da Região Y Determinante do Sexo/metabolismo
18.
Mol Biol Cell ; 21(20): 3497-505, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20739462

RESUMO

The human and mouse sex chromosomes are enriched in multicopy genes required for postmeiotic differentiation of round spermatids into sperm. The gene Sly is present in multiple copies on the mouse Y chromosome and encodes a protein that is required for the epigenetic regulation of postmeiotic sex chromosome expression. The X chromosome carries two multicopy genes related to Sly: Slx and Slxl1. Here we investigate the role of Slx/Slxl1 using transgenically-delivered small interfering RNAs to disrupt their function. We show that Slx and Slxl1 are important for normal sperm differentiation and male fertility. Slx/Slxl1 deficiency leads to delay in spermatid elongation and sperm release. A high proportion of delayed spermatids are eliminated via apoptosis, with a consequent reduced sperm count. The remaining spermatozoa are abnormal with impaired motility and fertilizing abilities. Microarray analyses reveal that Slx/Slxl1 deficiency affects the metabolic processes occurring in the spermatid cytoplasm but does not lead to a global perturbation of sex chromosome expression; this is in contrast with the effect of Sly deficiency which leads to an up-regulation of X and Y chromosome genes. This difference may be due to the fact that SLX/SLXL1 are cytoplasmic while SLY is found in the nucleus and cytoplasm of spermatids.


Assuntos
Dosagem de Genes/genética , Proteínas Nucleares/deficiência , Espermátides/patologia , Espermatogênese/genética , Animais , Apoptose , Fertilidade/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Cromossomos Sexuais/genética , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermátides/metabolismo , Espermátides/ultraestrutura , Testículo/metabolismo , Testículo/patologia
19.
Genome Biol ; 11(6): R66, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20573212

RESUMO

BACKGROUND: Mice with severe non-PAR Y chromosome long arm (NPYq) deficiencies are infertile in vivo and in vitro. We have previously shown that sperm from these males, although having grossly malformed heads, were able to fertilize oocytes via intracytoplasmic sperm injection (ICSI) and yield live offspring. However, in continuing ICSI trials we noted a reduced efficiency when cryopreserved sperm were used and with epididymal sperm as compared to testicular sperm. In the present study we tested if NPYq deficiency is associated with sperm DNA damage - a known cause of poor ICSI success. RESULTS: We observed that epididymal sperm from mice with severe NPYq deficiency (that is, deletion of nine-tenths or the entire NPYq gene complement) are impaired in oocyte activation ability following ICSI and there is an increased incidence of oocyte arrest and paternal chromosome breaks. Comet assays revealed increased DNA damage in both epididymal and testicular sperm from these mice, with epididymal sperm more severely affected. In all mice the level of DNA damage was increased by freezing. Epididymal sperm from mice with severe NPYq deficiencies also suffered from impaired membrane integrity and abnormal chromatin condensation and suboptimal chromatin protamination. It is therefore likely that the increased DNA damage associated with NPYq deficiency is a consequence of disturbed chromatin remodeling. CONCLUSIONS: This study provides the first evidence of DNA damage in sperm from mice with NPYq deficiencies and indicates that NPYq-encoded gene/s may play a role in processes regulating chromatin remodeling and thus in maintaining DNA integrity in sperm.


Assuntos
Aberrações Cromossômicas , Cromossomos de Mamíferos/genética , Dano ao DNA , Genes Ligados ao Cromossomo Y/genética , Espermatozoides/metabolismo , Cromossomo Y/genética , Análise de Variância , Animais , Western Blotting , Membrana Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestrutura , Quebra Cromossômica , Cromossomos de Mamíferos/metabolismo , Ensaio Cometa , Criopreservação , Reparo do DNA/genética , Epididimo/metabolismo , Feminino , Congelamento , Cariotipagem , Masculino , Camundongos , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Protaminas/metabolismo , Injeções de Esperma Intracitoplásmicas , Espermatozoides/citologia , Espermatozoides/ultraestrutura , Testículo/citologia , Testículo/metabolismo
20.
BMC Genomics ; 11: 82, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20122165

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica , Síndrome de Turner/genética , Inativação do Cromossomo X , Cromossomo X/genética , Alelos , Animais , Feminino , Redes Reguladoras de Genes , Genes Ligados ao Cromossomo X , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA