Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biol Reprod ; 95(6): 123, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27760750

RESUMEN

The first lineage specification during mammalian embryo development can be visually distinguished at the blastocyst stage. Two cell lineages are observed on the embryonic-abembryonic axis of the blastocyst: the inner cell mass and the trophectoderm. The timing and mechanisms driving this process are still not fully understood. In mouse embryos, cells seem prepatterned to become certain cell lineage because the first cleavage plane has been related with further embryonic-abembryonic axis at the blastocyst stage. Nevertheless, this possibility has been very debatable. Our objective was to determine whether this would be the case in another mammalian species, the bovine. To achieve this, cells of in vitro produced bovine embryos were traced from the 2-cell stage to the blastocyst stage. Blastocysts were then classified according to the allocation of the labeled cells in the embryonic and/or abembryonic part of the blastocyst. Surprisingly, we found that there is a significant percentage of the embryos (∼60%) with labeled and nonlabeled cells randomly distributed and intermingled. Using time-lapse microscopy, we have identified the emergence of this random pattern at the third to fourth cell cycle, when cells started to intermingle. Even though no differences were found on morphokinetics among different embryos, these random blastocysts and those with labeled cells separated by the embryonic-abembryonic axis (deviant pattern) are significantly bigger; moreover deviant embryos have a significantly higher number of cells. Interestingly, we observed that daughter cells allocation at the blastocyst stage is not affected by biopsies performed at an earlier stage.


Asunto(s)
Blastocisto/citología , Blastómeros/citología , Linaje de la Célula/fisiología , Desarrollo Embrionario/fisiología , Animales , Blastocisto/metabolismo , Blastómeros/metabolismo , Bovinos , Metilación de ADN , Histonas/metabolismo
2.
Curr Biol ; 24(4): 404-8, 2014 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-24485832

RESUMEN

The origin of sex reversal in XX goats homozygous for the polled intersex syndrome (PIS) mutation was unclear because of the complexity of the mutation that affects the transcription of both FOXL2 and several long noncoding RNAs (lncRNAs). Accumulating evidence suggested that FOXL2 could be the sole gene of the PIS locus responsible for XX sex reversal, the lncRNAs being involved in transcriptional regulation of FOXL2. In this study, using zinc-finger nuclease-directed mutagenesis, we generated several fetuses, of which one XX individual bears biallelic mutations of FOXL2. Our analysis demonstrates that FOXL2 loss of function dissociated from loss of lncRNA expression is sufficient to cause an XX female-to-male sex reversal in the goat model and, as in the mouse model, an agenesis of eyelids. Both developmental defects were reproduced in two newborn animals cloned from the XX FOXL2(-/-) fibroblasts. These results therefore identify FOXL2 as a bona fide female sex-determining gene in the goat. They also highlight a stage-dependent role of FOXL2 in the ovary, different between goats and mice, being important for fetal development in the former but for postnatal maintenance in the latter.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Cabras/metabolismo , Procesos de Determinación del Sexo , Animales , Femenino , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Cabras/embriología , Cabras/genética , Masculino , Ovario/embriología , Ovario/metabolismo , Testículo/embriología , Testículo/metabolismo , Cromosoma X , Cromosoma Y
3.
Cell Reprogram ; 14(6): 497-504, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23194453

RESUMEN

The somatic cell nuclear transfer (SCNT) procedure requires nuclear remodeling to return differentiated somatic nuclei to the totipotent undifferentiated stage. We hypothesize that mechanical constraints might occur upon SCNT and thereby affect nuclear remodeling. Therefore, we analyzed the nuclear structures upon SCNT using as donors either wild-type fibroblasts with a dense vimentin network or vimentin-deprived cells [embryonic stem cells (ESCs) and fibroblasts invalidated for vimetin]. We demonstrated that following nuclear transfer of wild-type fibroblasts, vimentin intermediate filaments (IFs) persisted around the transplanted nuclei and 88% of them presented severe distortions. We also showed that the presence of vimentin filaments in the reconstructed embryos was correlated with DNA damage, as evidenced by γH2A.X foci. On the other hand, when ESCs or vimentin-null (Vim(-/-)) fibroblasts devoid of IFs were used as nuclear donors, no nuclear distortion and less DNA damage were observed. Altogether we believe that the introduction of vimentin into recipient oocytes during SCNT induces a mechanical constraint on the transplanted nucleus that is responsible for nuclear distortions and DNA damage. This could lead to incomplete reprogramming that would be detrimental to further embryonic development.


Asunto(s)
Núcleo Celular/metabolismo , Células Madre Embrionarias/metabolismo , Fibroblastos/metabolismo , Filamentos Intermedios/metabolismo , Técnicas de Transferencia Nuclear , Vimentina/metabolismo , Animales , Núcleo Celular/genética , Células Cultivadas , Desarrollo Embrionario , Células Madre Embrionarias/citología , Fibroblastos/citología , Ratones , Ratones Mutantes , Vimentina/genética
4.
Epigenetics ; 7(7): 747-57, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22647320

RESUMEN

Early embryonic development is characterized by dramatic changes in cell potency and chromatin organization. The role of histone variants in the context of chromatin remodeling during embryogenesis remains under investigated. In particular, the nuclear distribution of the histone variant H2A.Z and its modifications have not been examined. Here we investigated the dynamics of acetylation of H2A.Z and two other active chromatin marks, H3K9ac and H3K36me3, throughout murine and bovine pre-implantation development. We show that H2A.Z distribution is dynamic during the earliest stages of mouse development, with protein levels significantly varying across stages and lowest at the 2-cell stage. When present, H2A.Z localizes preferentially to euchromatin at all stages analyzed. H2A.Z is acetylated in pre-implantation blastomeres and is preferentially localized to euchromatin, in line with the known role of H2A.Zac in transcriptional activation. Interestingly, however, H2A.Zac is undetectable in mouse embryos at the 2-cell stage, the time of major embryonic genome activation (EGA). Similarly, H3K36me3 is present exclusively in the maternal chromatin immediately after fertilization but becomes undetectable in interphase nuclei at the 2-cell stage, suggesting uncoupling of these active marks with global embryonic transcription activation. In bovine embryos, which undergo EGA at the 8-cell stage, H2A.Zac can be detected in zygotes, 4-, 8- and 16-cell stage embryos as well as in blastocysts, indicating that the dynamics of H2A.Zac is not conserved in mammals. In contrast, H3K36me3 displays mostly undetectable and heterogeneous localization pattern throughout bovine pre-implantation development. Thus, our results suggest that 'canonical' active chromatin marks exhibit a dynamic behavior in embryonic nuclei, which is both stage- and species-specific. We hypothesize that chromatin of early embryonic nuclei is subject to fine-tuning through differential acquisition of histone marks, allowing for proper chromatin remodeling and developmental progression in a species-specific fashion.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Epigénesis Genética , Histonas/metabolismo , Acetilación , Animales , Blastocisto/metabolismo , Bovinos , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Metilación , Ratones , Activación Transcripcional
5.
Biol Reprod ; 87(2): 32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623620

RESUMEN

FOXL2, a winged-helix/forkhead domain transcription factor, is a key gene involved in the differentiation and biological functions of the ovary. In a recent transcriptomic analysis, we found that FOXL2 expression in bovine caruncular endometrium was different from that in intercaruncular endometrium. In order to gain new insights into FOXL2 in this tissue, we determined the expression of this transcription factor during the estrous cycle and the establishment of pregnancy in cattle. The endometrial expression of FOXL2 did not vary during maternal recognition of pregnancy (Days 16-20). Using an in vivo bovine model and primary cell cultures, we showed that FOXL2 was not an interferon-tau target gene. Both FOXL2 transcript and protein were expressed from Day 5 to Day 20 of the estrous cycle, and their levels showed a significant increase during the luteolytic phase. A 2-day progesterone supplementation in heifers led to a clear down-regulation of FOXL2 protein levels, suggesting the negative impact of progesterone on FOXL2 expression. Immunohistochemistry data revealed the localization of FOXL2 in endometrial stromal and glandular cells. FOXL2 subcellular distribution was shown to be nuclear in endometrial samples collected during the luteolytic period, while it was not detected in nuclei during the luteal phase and at implantation. Collectively, our findings provide the first evidence that FOXL2 is involved in the regulation of endometrial tissue physiology.


Asunto(s)
Bovinos/fisiología , Endometrio/metabolismo , Ciclo Estral , Factores de Transcripción Forkhead/metabolismo , Animales , Implantación del Embrión , Femenino , Interferón Tipo I/fisiología , Embarazo , Proteínas Gestacionales/fisiología , Progesterona/farmacología
6.
J Reprod Dev ; 58(4): 467-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22572731

RESUMEN

Phosphorylation of histone H3 at Ser10 (H3S10P) has been linked to a variety of cellular processes, such as chromosome condensation and gene activation/silencing. Remarkably, in mammalian somatic cells, H3S10P initiates in the pericentromeric heterochromatin during the late G2 phase, and phosphorylation spreads throughout the chromosomes arms in prophase, being maintained until the onset of anaphase when it gets dephosphorylated. Considerable studies have been carried out about H3S10P in different organisms; however, there is little information about this histone modification in mammalian embryos. We hypothesized that this epigenetic modification could also be a marker of pericentromeric heterochromatin in preimplantation embryos. We therefore followed the H3S10P distribution pattern in the G1/S and G2 phases through the entire preimplantation development in in vivo mouse embryos. We paid special attention to its localization relative to another pericentromeric heterochromatin marker, HP1ß and performed immunoFISH using specific pericentromeric heterochromatin probes. Our results indicate that H3S10P presents a remarkable distribution pattern in preimplantation mouse embryos until the 4-cell stage and is a better marker of pericentromeric heterochromatin than HP1ß. After the 8-cell stage, H3S10P kinetic is more similar to the somatic one, initiating during G2 in chromocenters and disappearing upon telophase. Based on these findings, we believe that H3S10P is a good marker of pericentromeric heterochromatin, especially in the late 1- and 2-cell stages as it labels both parental genomes and that it can be used to further investigate epigenetic regulation and heterochromatin mechanisms in early preimplantation embryos.


Asunto(s)
Blastocisto/metabolismo , Desarrollo Embrionario , Epigénesis Genética , Heterocromatina/metabolismo , Histonas/metabolismo , Interfase , Serina/metabolismo , Animales , Biomarcadores/metabolismo , Blastocisto/citología , Femenino , Hibridación Fluorescente in Situ , Metafase , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Fosforilación , Embarazo , Profase , Procesamiento Proteico-Postraduccional , Telofase
7.
PLoS One ; 7(3): e34110, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479535

RESUMEN

Pluripotency genes are implicated in mouse embryonic genome activation (EGA) and pluripotent lineage specification. Moreover, their expression levels have been correlated with embryonic term development. In bovine, however, little information is available about dynamics of pluripotency genes during these processes. In this study, we charted quantitative and/or qualitative spatio-temporal expression patterns of transcripts and proteins of pluripotency genes (OCT4, SOX2 and NANOG) and mRNA levels of some of their downstream targets in bovine oocytes and early embryos. Furthermore, to correlate expression patterns of these genes with term developmental potential, we used cloned embryos, having similar in vitro but different full term development rates. Our findings affirm: firstly, the core triad of pluripotency genes is probably not implicated in bovine EGA since their proteins were not detected during pre-EGA phase, despite the transcripts for OCT4 and SOX2 were present. Secondly, an earlier ICM specification of transcripts and proteins of SOX2 and NANOG makes them pertinent candidates of bovine pluripotent lineage specification than OCT4. Thirdly, embryos with low term development potential have higher transcription rates; nevertheless, precarious balance between pluripotency genes is maintained. This balance presages normal in vitro development but, probably higher transcription rate disturbs it at later stage that abrogates term development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genoma , Células Madre Pluripotentes/citología , Animales , Bovinos , Linaje de la Célula , Clonación de Organismos , Fertilización In Vitro , Fibroblastos/citología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Ratones , Proteína Homeótica Nanog , Técnicas de Transferencia Nuclear , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Oocitos/citología , Factores de Transcripción SOXB1/biosíntesis , Factores de Tiempo
8.
Biol Reprod ; 84(6): 1248-57, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21293029

RESUMEN

During the periovulatory period, the induction of prostaglandin G/H synthase-2 (PTGS2) expression in cumulus cells and associated prostaglandin E2 (PGE2) production are implicated in the terminal differentiation of the cumulus-oocyte complex. During the present study, the effects of the PTGS2/PGE2 pathway on the developmental competence of bovine oocytes were investigated using an in vitro model of maturation, fertilization, and early embryonic development. The specific inhibition of PTGS2 activity with NS-398 during in vitro maturation (IVM) significantly restricted mitogen-activated protein kinase (MAPK) activation in oocytes at the germinal vesicle breakdown stage and reduced both cumulus expansion and the maturation rate after 22 h of culture. In addition, significantly higher rates of abnormal meiotic spindle organization were observed after 26 h of culture. Periconceptional PTGS2 inhibition did not affect fertilization but significantly reduced the speed of embryo development. Embryo output rates were significantly decreased on Day 6 postfertilization but not on Day 7. However, total blastomere number was significantly lower in embryos obtained after PTGS2 inhibition. The addition of PGE2 to IVM and in vitro fertilization cultures containing NS-398 overrode oocyte maturation and early embryonic developmental defects. Protein and mRNA expression for the prostaglandin E receptor PTGER2 were found in oocytes, whereas the PTGER2, PTGER3, and PTGER4 subtypes were expressed in cumulus cells. This study is the first to report the involvement of PGE2 in oocyte MAPK activation during the maturation process. Taken together, these results indicate that PGE2-mediated interactions between somatic and germ cells during the periconceptional period promote both in vitro oocyte maturation and preimplantation embryonic development in cattle.


Asunto(s)
Bovinos/embriología , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Meiosis/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Oocitos/metabolismo , Animales , Células Cultivadas , Células del Cúmulo/metabolismo , Ciclooxigenasa 2/genética , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Oocitos/citología , Fosforilación
9.
Cell Reprogram ; 12(6): 729-38, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108537

RESUMEN

The early events in the nuclear reprogramming process during somatic cell nuclear transfer (SCNT) consist of morphological remodeling of the donor nucleus including premature chromosome condensation (PCC). In the present study, the objective was to increase oocyte M-Phase Promoting Factor (MPF) kinase activity and to examine the fate of the donor nucleus and the development of SCNT embryos thereafter. Indeed, in controls, recipient oocytes activated upon nuclear transfer, undergo a decrease in MPF activity, responsible for the inability to promote PCC in 77.8% of reconstituted embryos. Here we showed that exposure of the recipient oocyte to the proteasome inhibitor MG132 prior to fusion inhibited the degradation of cyclin B, which normally occurred immediately after activation by electro stimulation, and therefore sustained a high level of MPF. Treatment with MG132 also significantly increased the percentage of SCNT embryos with PCC when compared to the nontreated SCNT control embryos (94.1 vs. 22.2%, respectively, p < 0.01). The frequency of development to the blastocyst stage did not differ between MG132-treated or untreated recipient oocytes. However, we observed a significant increase of the total cells number in embryos produced after MG132 treatment. Investigation of the global nuclear organization by immunodetection of heterochromatin protein 1 (CBX1) showed that SCNT embryos derived from MG132-treated recipient oocytes displayed organization patterns similar to the ones observed in IVF embryos in contrast to the nontreated SCNT controls. Taken together, these results suggest that the PCC induced by MG132 treatment allows reorganization of the chromatin at an appropriate time potentially, leading to better reprogramming.


Asunto(s)
Núcleo Celular/metabolismo , Reprogramación Celular , Inhibidores de Cisteína Proteinasa/farmacología , Embrión de Mamíferos/fisiología , Leupeptinas/farmacología , Técnicas de Transferencia Nuclear , Oocitos/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , Cromatina/metabolismo , Clonación de Organismos/métodos , Embrión de Mamíferos/citología , Desarrollo Embrionario/fisiología , Fibroblastos/citología , Factor Promotor de Maduración/metabolismo , Oocitos/citología , Oocitos/fisiología
10.
Reproduction ; 135(4): 431-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18296513

RESUMEN

It is clear from a wide range of studies that the nuclear/cytoplasmic distribution of Cdc25C has important functional consequences for cell cycle control. It is now admitted that in somatic cells, the localization of Cdc25C in the cytoplasm is required to maintain the cell in an interphasic state and that Cdc25C has to translocate to the nucleus just before M-phase to induce mitotic events. We characterized the expression and localization of Cdc25C during oocyte maturation, the first embryo mitosis, and the first steps of somatic cell nuclear transfer (SCNT) in cattle. We demonstrated that Cdc25C was expressed throughout the maturation process and the early development. We clearly showed that Cdc25C was localized in the nucleus at the germinal vesicle stage and during the early development until the blastocyst stage. However, the signal change in blastocyst and Cdc25C became cytoplasmic as is the case in somatic cells. Thus, oocytes and early embryonic cells presented a specific nuclear Cdc25C localization different from the one observed in somatic cells, suggesting that Cdc25C could have a particular localization/regulation in undifferentiated cells. Following SCNT, Cdc25C became nuclear as soon as the nucleus swelled, and this localization persisted until the blastocyst stage, as is the case in in vitro fertilized embryos. The Cdc25C nuclear localization appeared to constitute a major change, which could be associated with the reorganization of the somatic nucleus upon nuclear transfer.


Asunto(s)
Blastocisto/enzimología , Núcleo Celular/enzimología , Técnicas de Transferencia Nuclear , Oocitos/enzimología , Fosfatasas cdc25/análisis , Animales , Western Blotting , Bovinos , Citoplasma/enzimología , Femenino , Fertilización In Vitro , Inmunohistoquímica , Microscopía Fluorescente , Embarazo
11.
Mol Reprod Dev ; 71(4): 489-94, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15858794

RESUMEN

EGF has been shown to influence meiotic maturation and development competence of oocyte in various mammalian species. We previously reported, in goat, that the EGF receptor (EGF-R) was present both on cumulus cells and oocytes. Here, EGF-induced signaling was investigated during the in vitro maturation process in goat cumulus-oocyte complexes (COCs). Cumulus cells and oocytes were subjected to Western immunoblotting analysis using anti-MAP kinase, anti-phosphotyrosine, anti-phospho MAP kinase, and anti-phospho EGF-R antibodies. We demonstrated that treatment with EGF during the in vitro maturation process induced rapid tyrosine phosphorylation of EGF-R in a time and concentration dependent manner in cumulus cells. A similar pattern of activation by phosphorylation was observed for MAP kinase upon EGF stimulation. AG 1478, an inhibitor of the EGF kinase, suppressed EGF-stimulated phosphorylation of EGF-R and also affected the MAP kinase activation. Treatment with the MEK inhibitor PD 98059 abolished EGF-induced MAP kinase activation. We did not observe oocyte EGF-R phosphorylation in our experiments during the in vitro maturation process. Our data indicate, in goat cumulus cells, that activation of EGF-R by EGF triggers signaling through the MAP kinase pathway during in vitro maturation. This supports the hypothesis that the major site of action for EGF, that regulates oocyte maturation, is the cumulus cell.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Receptores ErbB/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Folículo Ovárico/fisiología , Animales , Receptores ErbB/antagonistas & inhibidores , Femenino , Flavonoides/farmacología , Cabras , Inmunoensayo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Fosforilación , Quinazolinas , Factores de Tiempo , Tirfostinos/farmacología
12.
Mol Reprod Dev ; 67(4): 439-45, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14991735

RESUMEN

It has been previously reported that epidermal growth factor (EGF) influences meiotic maturation and development competence of oocytes in various mammalian species. The present study was undertaken to analyze the expression of the gene encoding the EGF-receptor (EGF-R) in the goat cumulus-oocyte complex during meiotic competence acquisition. Expression of EGF-R mRNA was evaluated by PCR on reverse transcribed mRNA from follicular cells and oocytes, using EGF-R specific primers designed from human cDNA. The presence of the EGF-R transcript was evidenced in follicular cells as well as in meiotically competent and incompetent oocytes. Western blot analysis performed with specific anti EGF-R antibody revealed in meiotically competent and incompetent oocytes and in follicular cells a 170 kD polypeptide corresponding to the goat EGF-R protein. In oocytes the amount of EGF-R increased with meiotic competence acquisition. EGF-R distribution was examined by indirect immunofluorescence on frozen sections of cumulus-oocyte complexes (COCs). EGF-R immunoreactivity was observed in cumulus cells and in oocytes. Staining appeared to be confined to the periphery of the cells for both oocytes and cumulus cells. In this study, we identified the main component required for signaling via EGF-R in the goat oocyte and in follicular cells. These results suggest a possible involvement of EGF in the regulation of follicular growth and oocyte maturation in goat.


Asunto(s)
Receptores ErbB/genética , Expresión Génica , Cabras/anatomía & histología , Oocitos/citología , Folículo Ovárico/citología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Receptores ErbB/metabolismo , Femenino , Cabras/metabolismo , Datos de Secuencia Molecular , Oocitos/metabolismo , Folículo Ovárico/metabolismo
13.
Mol Reprod Dev ; 62(1): 4-12, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11933156

RESUMEN

Change in Cdc25C expression and localization during maturation and meiotic competence acquisition was investigated in goat oocytes. Western blot analysis revealed that Cdc25C is constitutively expressed throughout meiosis in competent goat oocytes, with changes in its phosphorylation level. Cdc25C was detected at 55 and 70 kDa, representing the nonphosphorylated form and the hyperphosphorylated active form, respectively. During the G2-M transition at meiosis resumption, Cdc25C was hyperphosphorylated as evidenced by a clear shift from 55 to 70 kDa. Okadaic acid which induced premature meiosis resumption associated with MPF activation also involved a premature shift from 55 to 70 kDa in goat competent oocytes. After artificial activation of goat oocytes, Cdc25C returned to its 55 kDa form. By indirect immunofluorescence, Cdc25C was found essentially localized in the nucleus at the germinal vesicle stage, suggesting that Cdc25C functions within the nucleus to regulate MPF activation. Concomitantly with germinal vesicle breakdown, Cdc25C was redistributed throughout the cytoplasm. The amount of Cdc25C, very low in incompetent oocytes, increased with meiosis competence acquisition. On the other hand, during oocyte growth while the expression of Cdc25C increased, its phosphorylation level increased concomitantly as well as its nuclear translocation. These results suggest that meiosis resumption needs a sufficient amount of Cdc25C which must be completely phosphorylated and nuclear and that the amount of Cdc25C may be a limiting factor for meiotic competence acquisition. We could consider that Cdc25C nuclear translocation and phosphorylation, during oocyte growth, prepare the oocytes in advance for the G2-M phase transition occurring during meiosis resumption.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Meiosis/fisiología , Oocitos/crecimiento & desarrollo , Fosfatasas cdc25/biosíntesis , Animales , Núcleo Celular/metabolismo , Femenino , Cabras , Oocitos/química , Oocitos/metabolismo
14.
Dev Growth Differ ; 38(5): 517-525, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37281669

RESUMEN

When in vitro-matured oocytes were enucleated, aged and kept at 10°C before reconstitution, the in vitro development of nuclear transfer embryos to the blastocyst stage did not differ from that obtained with in vitro fertilization. This suggests that these recipient cytoplasts constitute a suitable environment for the development of the nuclear transplant. The aim of the present study was to investigate, at the biochemical level, the result of the preparation of recipient oocytes, including enucleation, ageing and cooling. For this purpose the phosphorylation profiles of four groups of in vitro-matured bovine oocytes (aged oocytes, aged-cooled oocytes, enucleated-aged oocytes and enucleated-aged-cooled oocytes (recipient cytoplasts)) were analyzed. These recipient cytoplasts exhibited a phosphorylation profile similar to that of activated oocytes. Maturation promoting factor (MPF) activity, which was high in young metaphase II oocytes, in aged oocytes, in enucleated-aged oocytes and in aged-cooled oocytes, dropped to the basal level in enucleated-aged-cooled oocytes (recipient cytoplasts), while mitogen-activated protein kinase (MAPK) activity remained elevated. The combination of enucleation, ageing and cooling following oocyte in vitro maturation resulted in an interphase-like stage cytoplasm having a phosphorylation profile and low MPF activity similar to activated oocytes, but exhibiting high MAPK activity.

15.
Dev Growth Differ ; 34(5): 579-587, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37281151

RESUMEN

The association between germ cells and somatic granulosa cells persists throughout the growth of the oocyte by means of foot processes of the cumulus corona cells that cross the zona pellucida. During meiotic maturation important nuclear and cytoplasmic events occur in cumulus-oocyte complex suggesting implication of cytoskeletal elements. Immunoblotting analysis of cytoskeletal proteins of the cumulus cells revealed the presence of vimentin polypeptide and of at least two cytokeratin polypeptides. Using immunofluorescence techniques on cryostat sections through frozen tissue, we provided evidence for the presence of cytokeratins of the simple epithelial type in addition to vimentin in sheep cumulus cells. These two types of intermediate filaments were localized throughout the cytoplasm and especially in the foot processes which cross the zona pellucida. The contact area between the two cell types was also labelled with the antibodies. Acrylamide treatment of cumulus-oocyte complexes involved a drastic disorganization of the intermediate filament network and triggered the isolation of the oocyte from its cumulus cells. This isolation resulted in resumption of meiosis. From these results it appears that intermediate filaments could participate in the process of gap junction loss and indirectly in the control of meiosis resumption.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA