Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Biol Reprod ; 110(5): 985-999, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38376238

RESUMEN

Sry on the Y-chromosome upregulates Sox9, which in turn upregulates a set of genes such as Fgf9 to initiate testicular differentiation in the XY gonad. In the absence of Sry expression, genes such as Rspo1, Foxl2, and Runx1 support ovarian differentiation in the XX gonad. These two pathways antagonize each other to ensure the development of only one gonadal sex in normal development. In the B6.YTIR mouse, carrying the YTIR-chromosome on the B6 genetic background, Sry is expressed in a comparable manner with that in the B6.XY mouse, yet, only ovaries or ovotestes develop. We asked how testicular and ovarian differentiation pathways interact to determine the gonadal sex in the B6.YTIR mouse. Our results showed that (1) transcript levels of Sox9 were much lower than in B6.XY gonads while those of Rspo1 and Runx1 were as high as B6.XX gonads at 11.5 and 12.5 days postcoitum. (2) FOXL2-positive cells appeared in mosaic with SOX9-positive cells at 12.5 days postcoitum. (3) SOX9-positive cells formed testis cords in the central area while those disappeared to leave only FOXL2-positive cells in the poles or the entire area at 13.5 days postcoitum. (4) No difference was found at transcript levels of all genes between the left and right gonads up to 12.5 days postcoitum, although ovotestes developed much more frequently on the left than the right at 13.5 days postcoitum. These results suggest that inefficient Sox9 upregulation and the absence of Rspo1 repression prevent testicular differentiation in the B6.YTIR gonad.


Asunto(s)
Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Testículo , Trombospondinas , Regulación hacia Arriba , Animales , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Masculino , Femenino , Ratones , Trombospondinas/genética , Trombospondinas/metabolismo , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología , Testículo/metabolismo , Gónadas/metabolismo , Ovario/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Diferenciación Sexual/genética , Ratones Endogámicos C57BL
2.
Mamm Genome ; 34(1): 44-55, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36454369

RESUMEN

Several lines of evidence suggest that the presence of the Y chromosome influences DNA methylation of autosomal loci. To better understand the impact of the Y chromosome on autosomal DNA methylation patterns and its contribution to sex bias in methylation, we identified Y chromosome dependent differentially methylated regions (yDMRs) using whole-genome bisulfite sequencing methylation data from livers of mice with different combinations of sex-chromosome complement and gonadal sex. Nearly 90% of the autosomal yDMRs mapped to transposable elements (TEs) and most of them had lower methylation in XY compared to XX or XO mice. Follow-up analyses of four reporter autosomal yDMRs showed that Y-dependent methylation levels were consistent across most somatic tissues but varied in strains with different origins of the Y chromosome, suggesting that genetic variation in the Y chromosome influenced methylation levels of autosomal regions. Mice lacking the q-arm of the Y chromosome (B6.NPYq-2) as well as mice with a loss-of-function mutation in Kdm5d showed no differences in methylation levels compared to wild type mice. In conclusion, the Y-linked modifier of TE methylation is likely to reside on the short arm of Y chromosome and further studies are required to identify this gene.


Asunto(s)
Metilación de ADN , Sexismo , Ratones , Animales , Cromosoma Y , Variación Genética
3.
Chromosoma ; 130(1): 41-52, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33492414

RESUMEN

In mammalian oocytes, proper chromosome segregation at the first meiotic division is dictated by the presence and site of homologous chromosome recombination, which takes place in fetal life. Our current understanding of how homologous chromosomes find each other and initiate synapsis, which is prerequisite for homologous recombination, is limited. It is known that chromosome telomeres are anchored into the nuclear envelope (NE) at the early meiotic prophase I (MPI) and move along NE to facilitate homologous chromosome search and pairing. However, the mouse (Mus musculus) carries all acrocentric chromosomes with one telomeric end close to the centromere (subcentromeric telomere; C-telomere) and the other far away from the centromere (distal telomere; D-telomere), and how C- and D-telomeres participate in chromosome pairing and synapsis during the MPI progression is not well understood. Here, we found in the mouse oocyte that C- and D-telomeres transiently clustered in one area, but D-telomeres soon separated together from C-telomeres and then dispersed to preferentially initiate synapsis, while C-telomeres remained in clusters and synapsed at the last. In the Spo11 null oocyte, which is deficient in SPO11-dependent DSBs formation and homologous synapsis, the pattern of C- and D-telomere clustering and resolution was not affected, but synapsis was more frequently initiated at C-telomeres. These results suggest that SPO11 suppresses the early synapsis between C-telomeres in clusters.


Asunto(s)
Emparejamiento Cromosómico , Segregación Cromosómica , Cromosomas/genética , Recombinación Homóloga , Profase Meiótica I , Oocitos/fisiología , Telómero , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oocitos/citología
4.
Am J Hum Genet ; 103(5): 740-751, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388401

RESUMEN

Androgenetic complete hydatidiform moles are human pregnancies with no embryos and affect 1 in every 1,400 pregnancies. They have mostly androgenetic monospermic genomes with all the chromosomes originating from a haploid sperm and no maternal chromosomes. Androgenetic complete hydatidiform moles were described in 1977, but how they occur has remained an open question. We identified bi-allelic deleterious mutations in MEI1, TOP6BL/C11orf80, and REC114, with roles in meiotic double-strand breaks formation in women with recurrent androgenetic complete hydatidiform moles. We investigated the occurrence of androgenesis in Mei1-deficient female mice and discovered that 8% of their oocytes lose all their chromosomes by extruding them with the spindles into the first polar body. We demonstrate that Mei1-/- oocytes are capable of fertilization and 5% produce androgenetic zygotes. Thus, we uncover a meiotic abnormality in mammals and a mechanism for the genesis of androgenetic zygotes that is the extrusion of all maternal chromosomes and their spindles into the first polar body.


Asunto(s)
Andrógenos/genética , Mola Hidatiforme/genética , Mutación/genética , Alelos , Animales , Cromosomas/genética , Femenino , Humanos , Masculino , Mamíferos/genética , Ratones , Ratones Endogámicos C57BL , Oocitos/patología , Embarazo , Cigoto/patología
5.
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
6.
Cryobiology ; 76: 98-103, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28412286

RESUMEN

Oocyte cryopreservation is imperative for assisted reproductive technologies (ART). Although cryopreservation of oocytes at the Metaphase II has been widely used, immature oocytes at the germinal vesicle stage (GV-oocytes) need to be cryopreserved in certain situations such as cancer patients; however, the success rate of embryonic development from the GV-oocytes remains low largely due to the requirement for in vitro maturation (IVM). Our aim was to investigate the effects of glutathione (GSH) supplementation during vitrification and warming of mouse GV-oocytes on the preservation of developmental competence. GV-oocytes within cumulus oocyte complexes (COCs) were collected from C57BL/6J (B6) and (B6.DBA)F1 mouse strains and subjected to vitrification and warming, followed by IVM. The vitrification, warming or IVM medium was supplemented with GSH at 0-4.0 mM. In vitro matured oocytes were then fertilized in vitro and cultured in KSOMaa up to 4 days. The first cleavage and blastocyst development were evaluated morphologically, and their rates were statistically analysed by one-way ANOVA followed by Tukey's multiple comparisons test. The difference was considered significant at P < 0.05. The results showed that GSH supplementation in the IVM medium exhibited no or rather inhibitory effects on the first cleavage or blastocyst development in both mouse strains except that 1.0 mM GSH increased the blastocyst development rate in B6. By contrast, 1 mM GSH supplementation during vitrification and warming increased the blastocyst development rate in both mouse strains, more efficiently in B6 than (B6.DBA)F1. In conclusion, GSH supplementation during vitrification and warming of GV-oocytes protects the oocytes from freezing-inflicted loss of developmental competence.


Asunto(s)
Crioprotectores/farmacología , Glutatión/farmacología , Oocitos , Vitrificación/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Criopreservación/métodos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilización In Vitro/métodos , Congelación , Técnicas de Maduración In Vitro de los Oocitos/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Embarazo
8.
Dev Biol ; 385(2): 242-52, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24247007

RESUMEN

The oocyte becomes competent for embryonic development by involving mutual communication with cumulus cells (CCs) during folliculogenesis. How this communication takes place under physiological conditions is not fully understood. Current study examined oocyte-CCs communication in the XY sex-revered female mouse. We have previously found that the XY oocyte is defective in its cytoplasm, causing abnormal MII-spindle assembly and a failure in embryonic development. Our present study showed that transcript levels of Pfkp, Pkm2 and Ldh1 involved in glycolysis were lower in the CCs surrounding XY oocytes than in those surrounding XX oocytes. ATP contents in XY oocytes were also lower than those in XX oocytes, suggesting that lower glycolytic gene expression in CCs resulted in lower ATP contents in the enclosed oocyte. Co-culture of oocytectomized CC-oocyte complexes (COCs) with denuded oocytes showed that XY oocytes were less efficient than XX oocytes in promoting glycolytic gene expression in CCs. Furthermore, both glycolytic gene expression levels in CCs and ATP contents in oocytes of XY COCs increased to similar levels to those of XX COCs after culture for 20h in the presence of milrinone (=preincubation), which prevented spontaneous oocyte maturation. By increasing ATP levels in XY oocytes by either COC preincubation or ATP microinjection into oocytes prior to in vitro maturation, an improvement in MII-spindle assembly was observed. We conclude that the XY oocyte produces lesser amounts of paracrine factors that affect its companion CCs, which in turn make the ooplasm deficient in its components, including ATP, essential for MII-spindle assembly.


Asunto(s)
Células del Cúmulo/citología , Meiosis , Oocitos/citología , Huso Acromático/fisiología , Cromosoma X , Cromosoma Y , Animales , Secuencia de Bases , Medios de Cultivo , Cartilla de ADN , Expresión Génica , Glucólisis/genética , Ratones , Microscopía Confocal , Milrinona/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Dev Biol ; 377(1): 213-23, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23384561

RESUMEN

In many mammalian species, more than half of the initial oocyte population is eliminated by neonatal life, thus limiting the oocyte reserve for reproduction. The cause or mechanism of this major oocyte loss remains poorly understood. We examined the apoptotic pathway involved in oocyte elimination in wild-type mouse ovaries as well as in Msh5 -/- ovaries, in which all oocytes were eliminated due to a lack of double strand break repair. Immunoblot and immunofluorescence staining showed that an initiator caspase 9 and an effector caspase 7 were constitutively activated in almost all oocytes in fetal ovaries regardless of their genotypes. In caspase 9 -/- ovaries, the total number of oocytes remained high while that in wild-type ovaries steadily declined during ovarian development. Therefore, the activation of caspase 9 was required for but did not immediately lead to oocyte demise. We found that XIAP, an endogenous inhibitor of apoptosis, was also abundant in oocytes during meiotic prophase progression. On the other hand, a cleaved form of PARP1, a target of effector caspases, was localized to the nuclei of a limited number of oocytes, and the frequency of cleaved PARP1-positive oocyte nuclei increased significantly higher before all oocytes disappeared in Msh5 -/- ovaries. We conclude that the mitochondrial apoptotic pathway mediated by caspase 9 is constitutively activated in oocytes and renders the elimination of oocytes with meiotic errors, which can be captured by the cleavage of PARP1.


Asunto(s)
Caspasa 9/metabolismo , Profase Meiótica I , Oocitos/citología , Oocitos/enzimología , Animales , Animales Recién Nacidos , Caspasa 7/metabolismo , Caspasa 9/deficiencia , Recuento de Células , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Femenino , Feto/citología , Técnica del Anticuerpo Fluorescente , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ovario/citología , Ovario/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte de Proteínas , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
11.
Chromosoma ; 122(5): 337-49, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23760560

RESUMEN

In the XY pachytene spermatocyte, the sex chromosomes do not synapse except for the pseudoautosomal region and become transcriptionally silenced. It has been suggested that the meiotic silencing of unsynapsed chromatin (MSUC) also occurs in oocytes. In the XY sex-reversed female mouse, the sex chromosomes fail to pair in the majority of oocytes and a greater number of oocytes are eliminated during the meiotic prophase compared to the XX female. Yet, many XY oocytes survive to reach the second meiotic metaphase. The goal of our current study was to determine whether the single X chromosome shows the characteristics of asynapsis and meiotic silencing in a proportion of XY oocytes, which can explain the survival of the remaining oocytes. We first examined the accumulation of markers associated with asynapsis or transcriptional silencing, i.e., BRCA1, γH2AX, H3K9me3, and H3K27me3, at the single X chromosome in the XY oocyte. We found that γH2AX and BRCA1 were enriched on the single X chromosome whereas H3K9me3 was not, and H3K27me3 was enriched at all chromosomes in the majority of XY oocytes. We next examined the meiotic silencing of the single X chromosome using enrichment of the X-encoded ATRX protein. On average, ATRX enrichment was lower in XY oocytes than in XX oocytes as expected from its half gene dosage. However, the intensity of ATRX staining in XY oocytes harboring γH2AX domains showed a remarkable heterogeneity. We conclude that MSUC occurs with varying consequences, resulting in a heterogeneous population of oocytes with respect to protein enrichment in the XY female mouse.


Asunto(s)
Disgenesia Gonadal 46 XY/genética , Meiosis/genética , Oocitos , Cromosoma X/genética , Animales , Proteína BRCA1/genética , Cromatina/genética , Emparejamiento Cromosómico/genética , ADN Helicasas/genética , Femenino , Dosificación de Gen , Silenciador del Gen , Heterogeneidad Genética , Histonas/genética , Ratones , Proteínas Nucleares/genética , Proteína Nuclear Ligada al Cromosoma X
12.
Hum Reprod ; 29(10): 2256-68, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25113843

RESUMEN

STUDY QUESTION: How does l-carnitine (LC) supplementation during vitrification and in vitro maturation (IVM) of germinal vesicle stage (GV)-oocytes improve the developmental competence of the resultant metaphase II (MII) oocytes? SUMMARY ANSWER: LC supplementation during both vitrification of GV-oocytes and their subsequent IVM improved nuclear maturation as well as meiotic spindle assembly and mitochondrial distribution in MII oocytes. WHAT IS KNOWN ALREADY: Vitrification of GV-oocytes results in a lower success rate of blastocyst development compared with non-vitrified oocytes. LC supplementation during both vitrification and IVM of mouse GV-oocytes significantly improves embryonic development after IVF. STUDY DESIGN, SIZE, DURATION: GV-oocytes were collected from (B6.DBA)F1 and B6 mouse strains and subjected to vitrification and warming with or without 3.72 mM LC supplementation. After IVM with or without LC supplementation, the rate of nuclear maturation and the quality of MII oocytes were evaluated. At least 20 oocytes/group were examined, and each experiment was repeated at least three times. All experiments were conducted during 2013-2014. PARTICIPANTS/MATERIALS, SETTING, METHODS: Extrusion of the first polar body in IVM oocytes was observed as an indication of nuclear maturation. Spindle assembly and chromosomal alignment were examined by immunostaining of α-tubulin and nuclear staining with 4,6-diamidino-2-phenylindole (DAPI). Mitochondrial distribution and oxidative activity were measured by staining with Mitotracker Green Fluorescence Mitochondria (Mitotracker Green FM) and chloromethyltetramethylrosamine (Mitotracker Orange CMTMRos), respectively. ATP levels were determined by using the Bioluminescent Somatic Cell Assay Kit. MAIN RESULTS AND THE ROLE OF CHANCE: LC supplementation during both vitrification and IVM of GV-oocytes significantly increased the proportions of oocytes with normal MII spindles to the levels comparable with those of non-vitrified oocytes in both mouse strains. While vitrification of GV-oocytes lowered the proportions of MII oocytes with peripherally concentrated mitochondrial distribution compared with non-vitrified oocytes, LC supplementation significantly increased the proportion of such oocytes in the (B6.DBA)F1 strain. LC supplementation decreased the proportion of oocytes with mitochondrial aggregates in both vitrified and non-vitrified oocytes in the B6 strain. The oxidative activity of mitochondria was mildly decreased by vitrification and drastically increased by LC supplementation irrespective of vitrification in both mouse strains. No change was found in ATP levels irrespective of vitrification or LC supplementation. Results were considered to be statistically significant at P < 0.05 by either χ(2)- or t-test. LIMITATIONS, REASONS FOR CAUTION: It remains to be tested whether beneficial effect of LC supplementation during vitrification and IVM of GV-oocytes leads to fetal development and birth of healthy offspring after embryo transfer to surrogate females. WIDER IMPLICATIONS OF THE FINDINGS: This protocol has the potential to improve the quality of vitrified human oocytes and embryos during assisted reproduction treatment. STUDY FUNDING/COMPETING INTEREST: Partially supported by the Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery Grant and Mitacs Elevate Postdoctoral Fellowship, Canada.


Asunto(s)
Carnitina/farmacología , Técnicas de Maduración In Vitro de los Oocitos , Metafase/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Vitrificación , Adenosina Trifosfato/metabolismo , Animales , Técnicas de Cultivo de Célula , Femenino , Masculino , Ratones , Ratones Endogámicos DBA , Mitocondrias/ultraestructura , Oocitos/crecimiento & desarrollo , Huso Acromático/ultraestructura
13.
Biol Reprod ; 88(4): 104, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23446455

RESUMEN

Oocyte cryopreservation is important for assisted reproductive technologies (ART). Although cryopreservation of metaphase II (MII) oocytes has been successfully used, MII oocytes are vulnerable to the damage inflicted by the freezing procedure. Cryopreservation of germinal vesicle stage oocytes (GV-oocytes) is an alternative choice; however, blastocyst development from GV-oocytes is limited largely due to the need for in vitro maturation (IVM). Herein, we evaluated the effects of l-carnitine (LC) supplementation during vitrification and thawing of mouse GV-oocytes, IVM, and embryo culture on preimplantation development after in vitro fertilization (IVF). We first compared the rate of embryonic development from the oocytes that had been collected at the GV stage from three mouse strains, (B6.DBA)F1, (B6.C3H)F1, and B6, and processed for IVM and IVF, as well as that from the oocytes matured in vivo, i.e. ovulated (IVO). Our results demonstrated that the rate of blastocyst development was the highest in the (B6.DBA)F1 strain and the lowest in the B6 strain. We then supplemented the IVM medium with 0.6 mg/ml LC. The rate of blastocyst development improved in the B6 but not in the (B6.DBA)F1 strain. Vitrification of GV-oocytes in the basic medium alone reduced the rate of blastocyst development in both of those mouse strains. LC supplementation to the IVM medium alone did not change the percentage of blastocyst development. However, LC supplementation to both vitrification and IVM media significantly improved blastocyst development to the levels comparable with those obtained from vitrified/thawed IVO oocytes in both of the (B6.DBA)F1 and B6 strains. We conclude that LC supplementation during vitrification is particularly efficient in improving the preimplantation development from the GV-oocytes that otherwise have lower developmental competence in culture.


Asunto(s)
Carnitina/administración & dosificación , Fase de Segmentación del Huevo/fisiología , Criopreservación/métodos , Desarrollo Embrionario/efectos de los fármacos , Fertilización In Vitro , Técnicas de Maduración In Vitro de los Oocitos , Oocitos , Vitrificación , Animales , Blastocisto , Células Cultivadas , Fase de Segmentación del Huevo/efectos de los fármacos , Femenino , Fertilización In Vitro/métodos , Técnicas de Maduración In Vitro de los Oocitos/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Vitrificación/efectos de los fármacos
14.
Differentiation ; 82(1): 18-27, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21592645

RESUMEN

SRY on the Y-chromosome acts as a transcription factor to initiate testicular differentiation in mammals. Sox9 is a SRY target gene, upregulated immediately after Sry expression, and plays a key role in testicular differentiation. In the present study, we examined the expression of SRY and SOX9 proteins in the B6.Y(TIR) gonad, which undergoes partial or complete sex reversal. The results show that the ontogeny of SRY expression in the B6.Y(TIR) gonad was comparable with that in the B6.XY gonad. On the other hand, while SOX9 expression immediately followed SRY expression in the B6.XY gonad, it was considerably delayed compared to SRY expression in the B6.Y(TIR) gonad or SOX9 expression in the B6.XY gonad. Although SOX9 expression reached the entire gonad at a time point, it was downregulated and became restricted to the central area in which testis cords were organized. MIS, a marker of Sertoli cells, appeared only in well-organized testis cords. We speculate that the SRY protein from the Y(TIR)-chromosome is inefficient in upregulating the Sox9 gene on the B6 background, allowing the initiation of ovarian differentiation.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Disgenesia Gonadal 46 XY , Ovario/citología , Ovario/embriología , Factor de Transcripción SOX9/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Animales , Femenino , Disgenesia Gonadal 46 XY/genética , Disgenesia Gonadal 46 XY/metabolismo , Gónadas/metabolismo , Masculino , Ratones , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Transcripción SOX9/metabolismo , Testículo/citología , Testículo/embriología , Testículo/metabolismo , Regulación hacia Arriba
15.
Sci Rep ; 12(1): 12028, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35835815

RESUMEN

Meiotic homologous recombination during fetal development dictates proper chromosome segregation in adult mammalian oocytes. Successful homologous synapsis and recombination during Meiotic Prophase I (MPI) depends on telomere-led chromosome movement along the nuclear envelope. In mice, all chromosomes are acrocentric, while other mammalian species carry a mixture of acrocentric and metacentric chromosomes. Such differences in telomeric structures may explain the exceptionally low aneuploidy rates in mice. Here, we tested whether the presence of metacentric chromosomes carrying Robertsonian translocations (RbT) affects the rate of homologous recombination or aneuploidy. We found a delay in MPI progression in RbT-carrier vs. wild-type (WT) fetal ovaries. Furthermore, resolution of distal telomere clusters, associated with synapsis initiation, was delayed and centromeric telomere clusters persisted until later MPI substages in RbT-carrier oocytes compared to WT oocytes. When chromosomes fully synapsed, higher percentages of RbT-carrier oocytes harbored at least one chromosome pair lacking MLH1 foci, which indicate crossover sites, compared to WT oocytes. Aneuploidy rates in ovulated eggs were also higher in RbT-carrier females than in WT females. In conclusion, the presence of metacentric chromosomes among acrocentric chromosomes in mouse oocytes delays MPI progression and reduces the efficiency of homologous crossover, resulting in a higher frequency of aneuploidy.


Asunto(s)
Meiosis , Oocitos , Aneuploidia , Animales , Cromosomas , Femenino , Mamíferos , Meiosis/genética , Profase Meiótica I/genética , Ratones , Telómero/genética , Translocación Genética
16.
Sex Dev ; 16(5-6): 355-364, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35235936

RESUMEN

BACKGROUND: In eutherian mammals, the sex chromosome complement, XX and XY, determines sexual differentiation of gonadal primordia into testes and ovaries, which in turn direct differentiation of germ cells into haploid sperm and oocytes, respectively. When gonadal sex is reversed, however, the germ cell sex becomes discordant with the chromosomal sex. XY females in humans are infertile, while XY females in the mouse (Mus musculus) are subfertile or infertile dependent on the cause of sex reversal and the genetic background. This article reviews publications to understand how the sex chromosome complement affects the fertility of XY oocytes by comparing with XX and monosomy X (XO) oocytes. SUMMARY: The results highlight 2 folds disadvantage of XY oocytes over XX oocytes: (1) the X and Y chromosomes fail to pair during the meiotic prophase I, resulting in sex chromosome aneuploidy at the first meiotic division and (2) expression of the Y-linked genes during oocyte growth affects the transcriptome landscape and renders the ooplasmic component incompetent for embryonic development. KEY MESSAGE: The XX chromosome complement gives the oocyte the highest competence for embryonic development.

17.
Proc Natl Acad Sci U S A ; 105(37): 13918-23, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18772381

RESUMEN

The B6.Y(TIR) sex-reversed female mouse is anatomically normal at young ages but fails to produce offspring. We have previously shown that its oocytes go through the meiotic cell cycle up to the second metaphase; however, the meiotic spindle is not properly organized, the second meiotic division goes awry after activation or fertilization, and none of the oocytes initiate embryonic development. In the present study, we transferred the nuclei of GV-stage oocytes from XY females into the enucleated GV-stage oocytes from (B6.DBA)F1.XX females. The resultant reconstructed oocytes properly assembled second meiotic spindles after in vitro maturation and produced healthy offspring after in vitro fertilization. Some male pups inherited maternal Y chromosomes. We conclude that the cytoplasm of the XY oocyte is insufficient to support spindle formation at the second metaphase whereas its replacement with the cytoplasmic material from an XX oocyte allows normal development.


Asunto(s)
Diferenciación Celular , Citoplasma/metabolismo , Oocitos/citología , Oocitos/metabolismo , Cromosoma X/genética , Cromosoma Y/genética , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Femenino , Cariotipificación , Masculino , Meiosis , Ratones , Polimorfismo Genético/genética , Huso Acromático/genética , Factores de Transcripción/genética
18.
Front Genet ; 12: 792604, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34987552

RESUMEN

The sex chromosome complement, XX or XY, determines sexual differentiation of the gonadal primordium into a testis or an ovary, which in turn directs differentiation of the germ cells into sperm and oocytes, respectively, in eutherian mammals. When the X monosomy or XY sex reversal occurs, XO and XY females exhibit subfertility and infertility in the mouse on the C57BL/6J genetic background, suggesting that functional germ cell differentiation requires the proper sex chromosome complement. Using these mouse models, we asked how the sex chromosome complement affects gene transcription in the oocytes during follicular growth. An oocyte accumulates cytoplasmic components such as mRNAs and proteins during follicular growth to support subsequent meiotic progression, fertilization, and early embryonic development without de novo transcription. However, how gene transcription is regulated during oocyte growth is not well understood. Our results revealed that XY oocytes became abnormal in chromatin configuration, mitochondria distribution, and de novo transcription compared to XX or XO oocytes near the end of growth phase. Therefore, we compared transcriptomes by RNA-sequencing among the XX, XO, and XY oocytes of 50-60 µm in diameter, which were still morphologically comparable. The results showed that the X chromosome dosage limited the X-linked and autosomal gene transcript levels in XO oocytes whereas many genes were transcribed from the Y chromosome and made the transcriptome in XY oocytes closer to that in XX oocytes. We then compared the transcript levels of 3 X-linked, 3 Y-linked and 2 autosomal genes in the XX, XO, and XY oocytes during the entire growth phase as well as at the end of growth phase using quantitative RT-PCR. The results indicated that the transcript levels of most genes increased with oocyte growth while largely maintaining the X chromosome dosage dependence. Near the end of growth phase, however, transcript levels of some X-linked genes did not increase in XY oocytes as much as XX or XO oocytes, rendering their levels much lower than those in XX oocytes. Thus, XY oocytes established a distinct transcriptome at the end of growth phase, which may be associated with abnormal chromatin configuration and mitochondria distribution.

19.
Sci Rep ; 11(1): 13766, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215813

RESUMEN

Sexual dimorphism in gene regulation, including DNA methylation, is the main driver of sexual dimorphism in phenotypes. However, the questions of how and when sex shapes DNA methylation remain unresolved. Recently, using mice with different combinations of genetic and phenotypic sex, we identified sex-associated differentially methylated regions (sDMRs) that depended on the sex phenotype. Focusing on a panel of validated sex-phenotype dependent male- and female-biased sDMRs, we tested the developmental dynamics of sex bias in liver methylation and the impacts of mutations in the androgen receptor, estrogen receptor alpha, or the transcriptional repressor Bcl6 gene. True hermaphrodites that carry both unilateral ovaries and contralateral testes were also tested. Our data show that sex bias in methylation either coincides with or follows sex bias in the expression of sDMR-proximal genes, suggesting that sex bias in gene expression may be required for demethylation at certain sDMRs. Global ablation of AR, ESR1, or a liver-specific loss of BCL6, all alter sDMR methylation, whereas presence of both an ovary and a testis delays the establishment of male-type methylation levels in hermaphrodites. Moreover, the Bcl6-LKO shows dissociation between expression and methylation, suggesting a distinct role of BCL6 in demethylation of intragenic sDMRs.


Asunto(s)
Metilación de ADN/genética , Receptor alfa de Estrógeno/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores Androgénicos/genética , Animales , Trastornos del Desarrollo Sexual/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Ratones , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Caracteres Sexuales , Sexismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
20.
Mamm Genome ; 21(7-8): 377-87, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20577743

RESUMEN

Meiotic silencing of unsynapsed chromatin (MSUC) occurs in the germ cells of translocation carriers and may cause meiotic arrest and infertility. We hypothesized that if bypassing meiotic checkpoints MSUC may cause epigenetic defects in sperm. We investigated the meiotic behavior of the Robertsonian translocation Rb (8.12) in mice. The unsynapsed 8 and 12 trivalent was associated with the XY body during early and mid-pachynema in heterozygous Rb (8.12) carriers, suggesting possible silencing of pericentromeric genes, such as the Dnmt3a gene. In wild-type mice, DNMT3A protein showed a dramatic accumulation in the nucleus during the mid-pachytene stage and distinct association with the XY body. In translocation carriers, DNMT3A was less abundant in a proportion of pachytene spermatocytes that also had unsynapsed pericentromeric regions of chromosomes 8 and 12. The same mice had incomplete methylation of the imprinted H19 differentially methylated region (DMR) in sperm. We propose that impaired H19 imprint establishment results from lack of synapsis in chromosomes 8 and 12 probably through transient silencing of a chromosome 8 or 12 gene during pachynema. Furthermore, our findings support the notion that imprint establishment at the H19 locus extends into pachynema.


Asunto(s)
Impresión Genómica/fisiología , Translocación Genética/genética , Animales , Núcleo Celular/metabolismo , Emparejamiento Cromosómico/genética , Emparejamiento Cromosómico/fisiología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Femenino , Impresión Genómica/genética , Heterocigoto , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Espermatocitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA