Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 3(10): e3516, 2008.
Article in English | MEDLINE | ID: mdl-18949044

ABSTRACT

BACKGROUND: Homologous chromosomes separate in meiosis I and sister chromatids separate in meiosis II, generating haploid gametes. To address the question why sister chromatids do not separate in meiosis I, we explored the roles of Shogoshin1 (Sgo1) in chromosome separation during oocyte meiosis. METHODOLOGY/PRINCIPAL FINDINGS: Sgo1 function was evaluated by exogenous overexpression to enhance its roles and RNAi to suppress its roles during two meioses of mouse oocytes. Immunocytochemistry and chromosome spread were used to evaluate phenotypes. The exogenous Sgo1 overexpression kept homologous chromosomes and sister chromatids not to separate in meiosis I and meiosis II, respectively, while the Sgo1 RNAi promoted premature separation of sister chromatids. CONCLUSIONS: Our results reveal that prevention of premature separation of sister chromatids in meiosis I requires the retention of centromeric Sgo1, while normal separation of sister chromatids in meiosis II requires loss of centromeric Sgo1.


Subject(s)
Cell Cycle Proteins/physiology , Chromatids/physiology , Chromosome Segregation/genetics , Meiosis/genetics , Oocytes/physiology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centromere/drug effects , Centromere/metabolism , Chromatids/drug effects , Chromatids/genetics , Chromosome Segregation/drug effects , Female , Meiosis/drug effects , Mice , Mice, Inbred ICR , Oocytes/drug effects , Oocytes/metabolism , RNA, Small Interfering/pharmacology , Sequence Homology , Tissue Distribution , Transfection
2.
Biol Reprod ; 79(5): 897-905, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18667751

ABSTRACT

The role of androgen and androgen receptors (ARs) in males has been well established. This steroid and its receptor also exist in follicles, but their functions are still unclear. In this study, using a culture system containing a low dose of hypoxanthine, we revealed the positive contribution of testosterone to oocyte meiotic resumption. By performing ultracentrifugation to allow clear visualization of porcine germinal vesicles, our results provide evidence that mitogen-activated protein kinase (MAPK) in the oocyte itself but not in cumulus cells was activated before germinal vesicle breakdown (GVBD) after testosterone treatment. We further explored the signal cascade of testosterone-triggered GVBD and showed significant contributions of AR to testosterone-induced MAPK activation and GVBD. By using a potent and selective inhibitor of SRC and detecting activation of the kinase, we found that testosterone activated SRC in oocytes but not in cumulus cells and that SRC (as an essential upstream molecule of MAPK) mediated this testosterone- and AR-promoted reinitiation of meiosis. The present findings propose an undefined signaling pathway and suggest the potential competence of testosterone for meiotic resumption in mammalian oocytes.


Subject(s)
Meiosis , Oocytes/physiology , Receptors, Androgen/metabolism , Testosterone/physiology , src Homology Domains , Animals , Butadienes , Cumulus Cells/metabolism , Enzyme Activation , Epidermal Growth Factor/metabolism , Female , Hypoxanthine/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitriles , Phosphatidylinositol 3-Kinases/metabolism , Pyrimidines , Swine
3.
Biol Reprod ; 79(4): 718-26, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18596218

ABSTRACT

BRCA1 as a tumor suppressor has been widely investigated in mitosis, but its functions in meiosis are unclear. In the present study, we examined the expression, localization, and function of BRCA1 during mouse oocyte meiotic maturation. We found that expression level of BRCA1 was increased progressively from germinal vesicle to metaphase I stage, and then remained stable until metaphase II stage. Immunofluorescent analysis showed that BRCA1 was localized to the spindle poles at metaphase I and metaphase II stages, colocalizing with centrosomal protein gamma-tubulin. Taxol treatment resulted in the presence of BRCA1 onto the spindle microtubule fibers, whereas nocodazole treatment induced the localization of BRCA1 onto the chromosomes. Depletion of BRCA1 by both antibody injection and siRNA injection caused severely impaired spindles and misaligned chromosomes. Furthermore, BRCA1-depleted oocytes could not arrest at the metaphase I in the presence of low-dose nocodazole, suggesting that the spindle checkpoint is defective. Also, in BRCA1-depleted oocytes, gamma-tubulin dissociated from spindle poles and MAD2L1 failed to rebind to the kinetochores when exposed to nocodazole at metaphase I stage. Collectively, these data indicate that BRCA1 regulates not only meiotic spindle assembly, but also spindle assembly checkpoint, implying a link between BRCA1 deficiency and aneuploid embryos.


Subject(s)
BRCA1 Protein/physiology , Cell Cycle Proteins/metabolism , Oocytes/metabolism , Spindle Apparatus/metabolism , Aneuploidy , Animals , BRCA1 Protein/metabolism , Cells, Cultured , Embryo, Mammalian , Female , Mad2 Proteins , Meiosis/drug effects , Meiosis/physiology , Mice , Nocodazole/pharmacology , Nuclear Proteins/metabolism , Oocytes/drug effects , Oocytes/physiology , Oogenesis/drug effects , Oogenesis/physiology , Paclitaxel/pharmacology , Spindle Apparatus/drug effects , Tissue Distribution/drug effects , Tubulin Modulators/pharmacology
4.
J Reprod Dev ; 54(5): 391-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18594125

ABSTRACT

Spindle movements, including spindle migration from the center to the cortex of oocytes during first meiosis and spindle rotation during second meiosis, are required for asymmetric meiotic divisions in many species. However, little is currently known in relation to the rat oocyte. To explore how spindles move and the mechanism controlling spindle movements in rat oocytes, we observed the spindle dynamics during the two meiotic divisions in the rat oocyte by confocal microscopy. Drugs that depolymerize microtubules or microfilaments were employed to further determine the roles of these two cytoskeletons in spindle movements. The results showed that peripheral spindle migration took place during first meiosis and spindle rotation took place during second meiosis in the rat oocytes. Microfilament inhibitor inhibited both spindle migration and spindle rotation, and depolymerization of microtubules inhibited spindle rotation. Severe depolymerization of microtubules inhibited spindle migration, while migration was achieved by partial but not complete depolymerization of microtubules. We thus conclude that microfilaments are important for both spindle migration and spindle rotation and that spindle microtubules are essential for spindle movements in rat oocytes.


Subject(s)
Actin Cytoskeleton/physiology , Meiosis/physiology , Microtubules/physiology , Oocytes/physiology , Spindle Apparatus/physiology , Actin Cytoskeleton/drug effects , Animals , Cytochalasin B/pharmacology , Female , Meiosis/drug effects , Microtubules/drug effects , Movement/physiology , Nocodazole/pharmacology , Oocytes/drug effects , Rats , Rats, Sprague-Dawley , Rotation , Spindle Apparatus/drug effects , Tubulin Modulators/pharmacology
5.
Microsc Microanal ; 14(4): 349-56, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18598570

ABSTRACT

Spindle movement, including spindle migration during first meiosis and spindle rotation during second meiosis, is essential for asymmetric divisions in mouse oocytes. Previous studies by others and us have shown that microfilaments are required for both spindle migration and rotation. In the present study, we aimed to further investigate the mechanism controlling spindle movement during mouse oocyte meiosis. By employing drug treatment and immunofluorescence microscopy, we showed that dynamic microtubule assembly was involved in both spindle migration and rotation. Furthermore, we found that the calcium/CaM/CaMKII pathway was important for regulating spindle rotation.


Subject(s)
Meiosis , Oocytes/physiology , Spindle Apparatus , Animals , Calcium/metabolism , Mice , Microscopy, Fluorescence , Microtubules/metabolism , Movement , Rotation
7.
Cell Cycle ; 7(4): 513-20, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18239470

ABSTRACT

Heterochromatin protein 1 (HP1) is closely associated with diverse chromatin organization and function in mitosis. However, we almost know nothing about HP1 in mammalian oocyte. Here, we investigated the subcellular distribution of HP1alpha and its spatial relationship to histone modifications during mouse oocyte maturation. Dynamic migration of HP1alpha was observed in germinal vesicle with non-surrounded nucleolus (NSN) to surrounded nucleolus (SN) oocytes, which may be essential for the transition of chromatin conformation during the development of antral oocytes. In meiosis, HP1alpha was clearly detectable at the periphery of chromosomes from pre-metaphase I stage to anaphase-telophase I stage. Spatial correlation between HP1alpha and histone modifications is highly variable around the time of meiotic resumption. In germinal vesicle oocytes, HP1alpha almost colocalized with all histone modifications examined in this study except for phosphorylation of serine 28 on histone H3. However, with the breakdown of germinal vesicle, HP1alpha was detected mostly in the chromosomal domains with strong phosphorylation of serine 10 and 28 on histone H3, and they also partially associated with methylated histones. These results presented the functional implication of histone modifications in the regulation of HP1alpha during oocyte maturation. In addition, we also showed that blocking the function of HP1alpha by microinjecting anti-HP1alpha antibody caused the delay of GVBD, however, this effect may not be achieved through modifying histones.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Histones/metabolism , Meiosis/physiology , Oocytes/cytology , Analysis of Variance , Animals , Chromobox Protein Homolog 5 , Hydroxamic Acids , Mice , Microscopy, Fluorescence
8.
Mol Reprod Dev ; 75(1): 89-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17486610

ABSTRACT

So far, standard follicle culture systems can produce blastocyst from less than 40% of the in vitro matured oocytes compared to over 70% in the in vivo counterpart. Because the capacity for embryonic development is strictly associated with the terminal stage of oocyte growth, the nuclear maturity status of the in vitro grown oocyte was the subject of this study. Mouse early preantral follicles (100-130 microm) and early antral follicles (170-200 microm) isolated enzymatically were cultured for 12 and 4 days, respectively, in a collagen-free dish. The serum-based media were supplemented with either 100 mIU/ml FSH (FSH only); 100 mIU/ml FSH + 10 mIU/ml LH (FSH-LH); 100 mIU/ml FSH + 1 mIU/ml GH (FSH-GH) or 100 mIU/ml FSH + 100 ng/ml activin A (FSH-AA). Follicle survival was highest in follicle stimulating hormone (FSH)-AA group in both cultured preantral (91.8%) and antral follicles (82.7%). Survival rates in the other groups ranged between 48% (FSH only, preantral follicle culture) and 78.7% (FSH only, antral follicle culture). Estradiol and progesterone were undetectable in medium lacking gonadotrophins while AA supplementation in synergy with FSH caused increased estradiol secretion and a simultaneously lowered progesterone secretion. Chromatin configuration of oocytes from surviving follicles at the end of culture revealed that there were twice more developmentally incompetent non-surrounded nucleolus (NSN) oocytes (>65%) than the competent surrounded nucleolus (SN) oocytes (<34%). We conclude that the present standard follicle culture system does not produce optimum proportion of developmentally competent oocytes.


Subject(s)
Cell Culture Techniques , Gonadal Steroid Hormones/metabolism , Gonadotropins/pharmacology , Oocytes/drug effects , Ovarian Follicle/drug effects , Activins/pharmacology , Animals , Cells, Cultured , Chromatin/ultrastructure , Culture Media/metabolism , Culture Media/pharmacology , Enzymes/chemistry , Female , Follicle Stimulating Hormone/pharmacology , Growth Hormone/pharmacology , Mice , Oocytes/growth & development , Oocytes/ultrastructure , Ovarian Follicle/growth & development , Ovarian Follicle/ultrastructure
10.
Cell Cycle ; 6(12): 1521-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17507801

ABSTRACT

We recently reported that MEK1/2 plays an important role in microtubule organization and spindle pole tethering in mouse oocytes, but how the intracellular transport of this protein is regulated remains unknown. In the present study, we investigated the mechanisms of poleward MEK1/2 transport during the prometaphase I/metaphase I transition and MEK1/2 release from the spindle poles during the metaphase I/anaphase I transition in mouse oocytes. Firstly, we found that p-MEK1/2 was colocalized with dynactin at the spindle poles. Inhibition of the cytoplasmic dynein/dynactin complex by antibody microinjection blocked polar accumulation of p-MEK1/2 and caused obvious spindle abnormalities. Moreover, coimmunoprecipitation of p-MEK1/2 and dynein or dynactin from mouse oocyte extracts confirmed their association at metaphase I. Secondly, disruption of microtubules by nocodazole resulted in the failure of poleward p-MEK1/2 transport. Whereas, when the nocodazole-treated oocytes were recovered in fresh culture medium, the spindle reformed and p-MEK1/2 relocalized to the spindle poles. Finally, we examined the mechanism of p-MEK1/2 release from the spindle poles. In control oocytes, polar p-MEK1/2 was gradually released during metaphase I/anaphase I transition. By contrast, in the presence of nondegradable cyclin B (Delta90), p-MEK1/2 still remained at the spindle poles at anaphase I. Our results indicate that poleward MEK1/2 transport is a cytoplasmic dynein/dynactin-mediated and spindle microtubule-dependent intracellular movement, and that its subsequent anaphase release from spindle poles is dependent on cyclin B degradation.


Subject(s)
Cyclin B/metabolism , Cytoplasm/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Metaphase/physiology , Spindle Apparatus/metabolism , Animals , Dynactin Complex , Dyneins/metabolism , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Nocodazole/toxicity , Oocytes/cytology , Oocytes/physiology , Protein Transport/drug effects , Protein Transport/physiology , Spindle Apparatus/drug effects , Tubulin Modulators/toxicity
11.
Mol Reprod Dev ; 74(5): 591-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17034048

ABSTRACT

Chromatin transformation from a diffused or NSN configuration to a compacted or SN shape that forms a ring around the nucleolus is regarded as one of the modifications necessary for successful embryonic development. But the process of the transformation is poorly understood. In this study we cultured mouse antral oocytes under meiotic arrest with IBMX for periods between 3 and 24 hr. We observed the chromatin status of the oocytes before and after culture under UV illumination. We reported here that the NSN configured oocytes transformed temporally through an intermediate form into the SN configuration while under meiotic arrest in vitro. Meiotic rate was improved in the NSN oocytes after the meiotic arrest but decreased in the SN oocytes. We also reported that chromatin of both the NSN and SN oocytes was acetylated and the two groups underwent the same pattern of H4/K5 deacetylation during meiotic maturation. We hypothesized that the transformation of mouse oocyte from the NSN to SN type may be time rather than oocyte size specific and the abrupt deacetylation of NSN oocyte during spontaneous maturation may explain its poor meiotic and developmental competence.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Meiosis/physiology , Oocytes/physiology , Ultraviolet Rays , Acetylation , Animals , Cell Size/drug effects , Cells, Cultured , Chromatin/physiology , Female , Histones/metabolism , Meiosis/drug effects , Mice , Microscopy, Fluorescence , Oocytes/cytology , Oocytes/drug effects , Pregnancy
12.
Cell Cycle ; 5(17): 1974-82, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16969090

ABSTRACT

Epigenetic regulation of pericentromeric heterochromatin is crucial for proper interactions between kinetochores and spindle microtubules governing accurate chromosome segregation. Here, we first examined the dynamic distribution of phosphorylated serine 10 and 28 on H3 during mouse oocyte maturation and early embryo development using immunofluorescent staining and confocal microscopy. Our results revealed strong signals of phosphorylated H3/ser10 and 28 in the pericentromeric heterochromatin area and continuous persistent staining of the chromosome periphery, respectively. A panel of specific antibodies against various acetylated lysine, dimethylated lysine or phosphorylated serine residues on histone H3 or H4 were used to investigate the effects of Trichostatin A (TSA), a general inhibitor of histone deacetylases (HDACs), on histone modifications of pericentromeric heterochromatin. Unexpectedly, TSA treatment was unable to alter the acetylation and methylation status of pericentromeric heterochromatin, however, it resulted in significant dephosphorylation of H3/ser10 at this site during mouse oocyte meiosis, which is likely to play a role in the TSA-induced defective chromosome segregation. Furthermore, by using ZM447439, an inhibitor of Aurora kinases, we revealed that Aurora kinases may participate in the regulation of histone phosphorylation during mouse oocyte maturation.


Subject(s)
Heterochromatin/enzymology , Histones/metabolism , Meiosis , Oocytes/growth & development , Acetylation , Animals , Aurora Kinases , Blastocyst/enzymology , Centromere , Chromosome Segregation , Embryonic Development , Enzyme Inhibitors/pharmacology , Heterochromatin/drug effects , Histone Deacetylase Inhibitors , Histones/chemistry , Hydroxamic Acids/pharmacology , Methylation , Mice , Microscopy, Confocal , Oocytes/drug effects , Oocytes/enzymology , Phosphorylation/drug effects , Phosphoserine/analysis , Protein Serine-Threonine Kinases/metabolism
13.
Cell Cycle ; 5(18): 2130-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16969117

ABSTRACT

In mitosis the checkpoint proteins ensure faithful chromosome segregation by delaying onset of anaphase until all sister chromatids align at the metaphase plate of the bipolar spindle correctly. In the present study we blocked the function of Bub1 during meiosis by microinjecting anti-Bub1 specific antibody into cytoplasm of mouse oocytes, and found that depletion of Bub1 induced evident cyclin B degradation and precocious anaphase onset. Bub1 suppression also overrode the checkpoint-dependent cell cycle arrest provoked by a low dosage of nocodazole. Furthermore, Bub1 depletion induced a significantly higher percentage of oocytes with misaligned chromosomes. In addition, we depicted the localization dynamics of Bub1 in response to spindle damage and its relationship with microtubules and chromosomes, providing further evidence for Bub1's role as a spindle checkpoint protein. Our data suggest that Bub1 is a critical spindle checkpoint protein that regulates accurate chromosome alignment and homolog disjunction in mammalian oocyte meiosis.


Subject(s)
Anaphase/genetics , Chromosome Segregation/genetics , Chromosomes/metabolism , Meiosis/genetics , Oocytes/metabolism , Protein Kinases/metabolism , Animals , Antibodies/pharmacology , Cell Cycle/genetics , Chromosomes/genetics , Cyclin B/metabolism , Cyclin B1 , Female , Genes, cdc/physiology , Mice , Oocytes/cytology , Oogenesis/genetics , Protein Kinases/genetics , Protein Kinases/immunology , Protein Serine-Threonine Kinases , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
14.
Front Biosci ; 11: 2193-202, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16720305

ABSTRACT

Although securin/separase/cohesion pathway was reported to regulate chromosome segregation during meiotic metaphase-to-anaphase transition, little biochemical evidence was provided. We recently found that oocytes could not progress beyond meiotic metaphase when ubiquitin-proteasome pathway was inhibited, but the mechanisms remain unclear. In the present study, we investigated the quantity of securin and Rec8 protein and the localization of securin, a cohesion subunit, during oocyte meiosis providing data in support of the hypothesis that the effect of ubiquitin-proteasome pathway on metaphase-to-anaphase transition was mediated by regulating securin and Rec8 degradation in mouse and pig oocytes. In germinal vesicle-stage oocytes, immunostaining of securin was mainly localized in the germinal vesicle. Shortly after germinal vesicle breakdown, immunoreactive securin accumulated around the condensed chromosomes at prometaphase I. At metaphase I and metaphase II, when chromosomes were organized at the equatorial plate, immunoreactive securin was concentrated around the aligned chromosomes, putatively associated with the position of the metaphase spindle. The accumulation of securin could not be detected at anaphase I and anaphase II. In both mouse and pig oocytes, Western blot analysis showed that securin protein was low at germinal vesicle stage, reached the highest level at metaphase I, while decreased at anaphase I. Securin was increased again at metaphase II, while it was decreased at anaphase II. Rec8 protein was present in germinal vesicle-stage oocytes and remained until metaphase I, while it was decreased at anaphase I. Like securin, Rec8 was increased at metaphase II, while it was decreased again at anaphase II. The inhibition of the ubiquitin-proteasome pathway inhibited the decrease in securin and Rec8 at metaphase-to-anaphase transitions in both mouse and pig oocytes. Microinjection of securin antibody into MII-arrested oocytes leads to the degradation of Rec8. In conclusion, these results suggest that the proteolysis of securin is dependent on ubiquitin-proteasome pathway and is necessary for the degradation of Rec8 during meiotic metaphase-to-anaphase transitions in mouse and pig oocytes.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Oocytes/metabolism , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/physiology , Ubiquitin/physiology , Anaphase/physiology , Animals , Antibodies , Blotting, Western , Carrier Proteins/analysis , Cell Culture Techniques , Cell Cycle Proteins , Female , Meiosis , Metaphase/physiology , Mice , Securin , Swine
15.
Cell Cycle ; 5(7): 766-74, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16582639

ABSTRACT

Histone acetylation is associated with a diversity of chromatin-related processes in mitosis. However, its roles in mammalian oocyte meiosis are largely unknown. In the present study, we first investigated in detail the acetylation changes during porcine oocyte maturation using a panel of antibodies specific for the critical acetylated forms of histone H3 and H4, and showed meiosis stage-dependent and lysine residue-specific patterns of histone acetylation. By using trichostatin A (TSA), a general inhibitor of histone deacetylases (HDACs), we further determined that selective inhibition of histone deacetylation (thereby maintaining hyperacetylation) delayed the onset of germinal vesicle breakdown and produced a high frequency of lagging chromosomes or chromatin bridges at anaphase and telophase I (AT-I), suggesting that histone deacetylation is required for orderly meiotic resumption and accurate chromosome segregation in porcine oocytes. In addition, we examined the localization and expression of HDAC1 by performing immunofluorescence and immunoblotting analysis. The results showed that subcellular translocation, expression level and phosphorylated modification of HDAC1 were temporally regulated and likely to coparticipate in the establishment of histone acetylation profiles in oocyte meiosis.


Subject(s)
Histones/metabolism , Meiosis/physiology , Acetylation/drug effects , Anaphase/drug effects , Animals , Cell Nucleus/metabolism , Cells, Cultured , Chromosome Segregation/drug effects , Gene Expression , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Lysine/metabolism , Oocytes/cytology , Oocytes/drug effects , Protein Transport , Swine , Telophase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...