Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Development ; 151(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38785133

RESUMO

The RNA-binding protein cytoplasmic polyadenylation element binding 1 (CPEB1) plays a fundamental role in regulating mRNA translation in oocytes. However, the specifics of how and which protein kinase cascades modulate CPEB1 activity are still controversial. Using genetic and pharmacological tools, and detailed time courses, we have re-evaluated the relationship between CPEB1 phosphorylation and translation activation during mouse oocyte maturation. We show that both the CDK1/MAPK and AURKA/PLK1 pathways converge on CPEB1 phosphorylation during prometaphase of meiosis I. Only inactivation of the CDK1/MAPK pathway disrupts translation, whereas inactivation of either pathway alone leads to CPEB1 stabilization. However, CPEB1 stabilization induced by inactivation of the AURKA/PLK1 pathway does not affect translation, indicating that destabilization and/or degradation is not linked to translational activation. The accumulation of endogenous CCNB1 protein closely recapitulates the translation data that use an exogenous template. These findings support the overarching hypothesis that the activation of translation during prometaphase in mouse oocytes relies on a CDK1/MAPK-dependent CPEB1 phosphorylation, and that translational activation precedes CPEB1 destabilization.


Assuntos
Meiose , Oócitos , Biossíntese de Proteínas , Fatores de Transcrição , Fatores de Poliadenilação e Clivagem de mRNA , Animais , Feminino , Camundongos , Aurora Quinase A/metabolismo , Aurora Quinase A/genética , Proteína Quinase CDC2/metabolismo , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ciclina B1/metabolismo , Ciclina B1/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/genética , Oócitos/metabolismo , Oócitos/citologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
2.
J Vis Exp ; (187)2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36190266

RESUMO

Aneuploidy is the leading genetic abnormality causing early miscarriage and pregnancy failure in humans. Most errors in chromosome segregation that give rise to aneuploidy occur during meiosis in oocytes, but why oocyte meiosis is error-prone is still not fully understood. During cell division, cells prevent errors in chromosome segregation by activating the spindle assembly checkpoint (SAC). This control mechanism relies on detecting kinetochore (KT)-microtubule (MT) attachments and sensing tension generated by spindle fibers. When KTs are unattached, the SAC is activated and prevents cell-cycle progression. The SAC is activated first by MPS1 kinase, which triggers the recruitment and formation of the mitotic checkpoint complex (MCC), composed of MAD1, MAD2, BUB3, and BUBR1. Then, the MCC diffuses into the cytoplasm and sequesters CDC20, an anaphase-promoting complex/cyclosome (APC/C) activator. Once KTs become attached to microtubules and chromosomes are aligned at the metaphase plate, the SAC is silenced, CDC20 is released, and the APC/C is activated, triggering the degradation of Cyclin B and Securin, thereby allowing anaphase onset. Compared to somatic cells, the SAC in oocytes is not as effective because cells can undergo anaphase despite having unattached KTs. Understanding why the SAC is more permissive and if this permissiveness is one of the causes of chromosome segregation errors in oocytes still needs further investigation. The present protocol describes the three techniques to comprehensively evaluate SAC integrity in mouse oocytes. These techniques include using nocodazole to depolymerize MTs to evaluate the SAC response, tracking SAC silencing by following the kinetics of Securin destruction, and evaluating the recruitment of MAD2 to KTs by immunofluorescence. Together these techniques probe mechanisms needed to produce healthy eggs by providing a complete evaluation of SAC integrity.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Fuso Acromático , Ciclossomo-Complexo Promotor de Anáfase/genética , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Aneuploidia , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Camundongos , Nocodazol , Oócitos , Securina/genética , Securina/metabolismo , Fuso Acromático/metabolismo
3.
Reproduction ; 164(4): V5-V7, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36125382

RESUMO

In brief: The Aurora protein kinases have critical functions in controlling oocyte meiotic maturation. In this study, we describe an assay for examining their activation state in oocytes and establish the best working doses of three commonly used inhibitors. Abstract: Several small molecule inhibitors exist for targeting Aurora kinase proteins in somatic cells. From this point of view, we evaluate the specificity of these inhibitors in mouse oocytes, and we demonstrate that MLN 8237 and AZD 1152 are specific for Aurora kinase A and Aurora kinase C, respectively, only when used at low concentrations.


Assuntos
Aurora Quinase A , Meiose , Animais , Aurora Quinase A/metabolismo , Aurora Quinase C/metabolismo , Camundongos , Oócitos/metabolismo , Proteínas Quinases/metabolismo
4.
Development ; 148(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34636397

RESUMO

Mammalian oocytes are transcriptionally quiescent, and meiosis and early embryonic divisions rely on translation of stored maternal mRNAs. Activation of these mRNAs is mediated by polyadenylation. Cytoplasmic polyadenylation binding element 1 (CPEB1) regulates mRNA polyadenylation. One message is aurora kinase C (Aurkc), encoding a protein that regulates chromosome segregation. We previously demonstrated that AURKC levels are upregulated in oocytes lacking aurora kinase B (AURKB), and this upregulation caused increased aneuploidy rates, a role we investigate here. Using genetic and pharmacologic approaches, we found that AURKB negatively regulates CPEB1-dependent translation of many messages. To determine why translation is increased, we evaluated aurora kinase A (AURKA), a kinase that activates CPEB1 in other organisms. We find that AURKA activity is increased in Aurkb knockout mouse oocytes and demonstrate that this increase drives the excess translation. Importantly, removal of one copy of Aurka from the Aurkb knockout strain background reduces aneuploidy rates. This study demonstrates that AURKA is required for CPEB1-dependent translation, and it describes a new AURKB requirement to maintain translation levels through AURKA, a function crucial to generating euploid eggs.


Assuntos
Aurora Quinase A/metabolismo , Aurora Quinase B/metabolismo , Oócitos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro Estocado/metabolismo , Animais , Aurora Quinase A/genética , Aurora Quinase B/genética , Meiose , Camundongos , Camundongos Knockout , Fatores de Transcrição/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
5.
Nat Commun ; 12(1): 5005, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34408140

RESUMO

Embryonic aneuploidy from mis-segregation of chromosomes during meiosis causes pregnancy loss. Proper disjunction of homologous chromosomes requires the mismatch repair (MMR) genes MLH1 and MLH3, essential in mice for fertility. Variants in these genes can increase colorectal cancer risk, yet the reproductive impacts are unclear. To determine if MLH1/3 single nucleotide polymorphisms (SNPs) in human populations could cause reproductive abnormalities, we use computational predictions, yeast two-hybrid assays, and MMR and recombination assays in yeast, selecting nine MLH1 and MLH3 variants to model in mice via genome editing. We identify seven alleles causing reproductive defects in mice including female subfertility and male infertility. Remarkably, in females these alleles cause age-dependent decreases in litter size and increased embryo resorption, likely a consequence of fewer chiasmata that increase univalents at meiotic metaphase I. Our data suggest that hypomorphic alleles of meiotic recombination genes can predispose females to increased incidence of pregnancy loss from gamete aneuploidy.


Assuntos
Aborto Espontâneo/genética , Aneuploidia , Perda do Embrião/genética , Proteína 1 Homóloga a MutL/genética , Proteínas MutL/genética , Aborto Espontâneo/metabolismo , Aborto Espontâneo/fisiopatologia , Alelos , Animais , Troca Genética , Reparo de Erro de Pareamento de DNA , Perda do Embrião/fisiopatologia , Feminino , Recombinação Homóloga , Humanos , Tamanho da Ninhada de Vivíparos , Masculino , Meiose , Camundongos , Proteína 1 Homóloga a MutL/metabolismo , Proteínas MutL/metabolismo , Gravidez , Reprodução , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
6.
PLoS Genet ; 17(4): e1009327, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33901174

RESUMO

The Aurora protein kinases are well-established regulators of spindle building and chromosome segregation in mitotic and meiotic cells. In mouse oocytes, there is significant Aurora kinase A (AURKA) compensatory abilities when the other Aurora kinase homologs are deleted. Whether the other homologs, AURKB or AURKC can compensate for loss of AURKA is not known. Using a conditional mouse oocyte knockout model, we demonstrate that this compensation is not reciprocal because female oocyte-specific knockout mice are sterile, and their oocytes fail to complete meiosis I. In determining AURKA-specific functions, we demonstrate that its first meiotic requirement is to activate Polo-like kinase 1 at acentriolar microtubule organizing centers (aMTOCs; meiotic spindle poles). This activation induces fragmentation of the aMTOCs, a step essential for building a bipolar spindle. We also show that AURKA is required for regulating localization of TACC3, another protein required for spindle building. We conclude that AURKA has multiple functions essential to completing MI that are distinct from AURKB and AURKC.


Assuntos
Aurora Quinase A/genética , Proteínas de Ciclo Celular/genética , Proteínas Fetais/genética , Meiose/genética , Proteínas Associadas aos Microtúbulos/genética , Oócitos/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Aurora Quinase B/genética , Aurora Quinase C/genética , Divisão do Núcleo Celular/genética , Segregação de Cromossomos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Oócitos/metabolismo , Fuso Acromático/genética , Polos do Fuso/genética , Quinase 1 Polo-Like
7.
Chromosoma ; 128(3): 369-383, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31256246

RESUMO

Sirtuins are NAD+-dependent protein deacylases and ADP-ribosyltransferases that are involved in a wide range of cellular processes including genome homeostasis and metabolism. Sirtuins are expressed in human and mouse oocytes yet their role during female gamete development are not fully understood. Here, we investigated the role of a mammalian sirtuin member, SIRT7, in oocytes using a mouse knockout (KO) model. Sirt7 KO females have compromised fecundity characterized by a rapid fertility decline with age, suggesting the existence of a diminished oocyte pool. Accordingly, Sirt7 KO females produced fewer oocytes and ovulated fewer eggs. Because of the documented role of SIRT7 in DNA repair, we investigated whether SIRT7 regulates prophase I when meiotic recombination occurs. Sirt7 KO pachynema-like staged oocytes had approximately twofold increased γH2AX signals associated with regions with unsynapsed chromosomes. Consistent with the presence of asynaptic chromosome regions, Sirt7 KO oocytes had fewer MLH1 foci (~one less), a mark of crossover-mediated repair, than WT oocytes. Moreover, this reduced level of crossing over is consistent with an observed twofold increased incidence of aneuploidy in Metaphase II eggs. In addition, we found that acetylated lysine 18 of histone H3 (H3K18ac), an established SIRT7 substrate, was increased at asynaptic chromosome regions suggesting a functional relationship between this epigenetic mark and chromosome synapsis. Taken together, our findings demonstrate a pivotal role for SIRT7 in oocyte meiosis by promoting chromosome synapsis and have unveiled the importance of SIRT7 as novel regulator of the reproductive lifespan.


Assuntos
Pareamento Cromossômico , Prófase Meiótica I , Sirtuínas/metabolismo , Acetilação , Aneuploidia , Animais , Troca Genética , Feminino , Fertilidade/genética , Imunofluorescência , Histonas/metabolismo , Homozigoto , Camundongos , Camundongos Knockout , Oócitos/metabolismo , Ovário/metabolismo , Ovário/patologia , Sirtuínas/genética
8.
Curr Biol ; 28(21): 3458-3468.e5, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30415701

RESUMO

Errors in chromosome segregation during female meiosis I occur frequently, and aneuploid embryos account for 1/3 of all miscarriages in humans [1]. Unlike mitotic cells that require two Aurora kinase (AURK) homologs to help prevent aneuploidy (AURKA and AURKB), mammalian germ cells also require a third (AURKC) [2, 3]. AURKA is the spindle-pole-associated homolog, and AURKB/C are the chromosome-localized homologs. In mitosis, AURKB has essential roles as the catalytic subunit of the chromosomal passenger complex (CPC), regulating chromosome alignment, kinetochore-microtubule attachments, cohesion, the spindle assembly checkpoint, and cytokinesis [4, 5]. In mouse oocyte meiosis, AURKC takes over as the predominant CPC kinase [6], although the requirement for AURKB remains elusive [7]. In the absence of AURKC, AURKB compensates, making defining potential non-overlapping functions difficult [6, 8]. To investigate the role(s) of AURKB and AURKC in oocytes, we analyzed oocyte-specific Aurkb and Aurkc single- and double-knockout (KO) mice. Surprisingly, we find that double KO female mice are fertile. We demonstrate that, in the absence of AURKC, AURKA localizes to chromosomes in a CPC-dependent manner. These data suggest that AURKC prevents AURKA from localizing to chromosomes by competing for CPC binding. This competition is important for adequate spindle length to support meiosis I. We also describe a unique requirement for AURKB to negatively regulate AURKC to prevent aneuploidy. Together, our work reveals oocyte-specific roles for the AURKs in regulating each other's localization and activity. This inter-kinase regulation is critical to support wild-type levels of fecundity in female mice.


Assuntos
Aurora Quinase A/genética , Aurora Quinase B/genética , Aurora Quinase C/genética , Meiose , Oócitos/metabolismo , Aneuploidia , Animais , Aurora Quinase A/metabolismo , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Segregação de Cromossomos/genética , Feminino , Fertilidade/genética , Camundongos
9.
Methods Mol Biol ; 1818: 67-76, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29961256

RESUMO

Immunofluorescence is a useful technique for analysis of protein expression and localization, thereby providing information regarding protein function, regulation, and protein-protein interactions. It is a standard approach to determine the temporal and spatial location of gene products that function in oocyte meiotic maturation. Fixation is one of the critical steps in the immunofluorescence protocol. Here, we describe the use of antibodies that are widely utilized in oocytes studies: anti-centromeric antigen (ACA), anti-Aurora kinase A (AURKA) and anti-alpha and gamma-tubulin antibodies that require different technical approaches for successful visualization, and we provide protocols for these conditions that are amenable to mouse oocyte studies. Detection of these proteins provides phenotypic information about spindle morphology, chromosome alignment, and microtubule attachments to kinetochores critical to assessing oocyte quality.


Assuntos
Imunofluorescência/métodos , Técnicas de Maturação in Vitro de Oócitos/métodos , Oócitos/citologia , Frações Subcelulares/metabolismo , Animais , Aurora Quinase A/metabolismo , Células Cultivadas , Centrômero/metabolismo , Segregação de Cromossomos , Feminino , Meiose , Camundongos , Microtúbulos/metabolismo , Oócitos/fisiologia , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
10.
Trends Genet ; 33(5): 349-363, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28359584

RESUMO

The aurora kinases (AURKs) comprise an evolutionarily conserved family of serine/threonine kinases involved in mitosis and meiosis. While most mitotic cells express two AURK isoforms (AURKA and AURKB), mammalian germ cells also express a third, AURKC. Although much is known about the functions of the kinases in mitosis, less is known about how the three isoforms function to coordinate meiosis. This review is aimed at describing what is known about the three isoforms in female meiosis, the similarities and differences between kinase functions, and speculates as to why mammalian germ cells require expression of three AURKs instead of two.


Assuntos
Aurora Quinase A/genética , Aurora Quinase B/genética , Aurora Quinase C/genética , Oócitos/crescimento & desenvolvimento , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Humanos , Meiose/genética , Camundongos , Oócitos/metabolismo , Oogênese/genética
11.
Hum Mol Genet ; 25(13): 2698-2711, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27106102

RESUMO

Aneuploidy is the leading genetic abnormality that leads to miscarriage, and it is caused by a failure of accurate chromosome segregation during gametogenesis or early embryonic divisions. Aurora kinase C (AURKC) is essential for formation of euploid sperm in humans because mutations in AURKC are correlated with macrozoospermia and these sperm are tetraploid. These mutations are currently the most frequent mutations that cause macrozoospermia and result from an inability to complete meiosis I (MI). Three of these mutations AURKC c.144delC (AURKC p.L49Wfs22), AURKC c.686G > A (AURKC p.C229Y) and AURKC c.744C > G (AURKC p.Y248*) occur in the coding region of the gene and are the focus of this study. By expressing these alleles in oocytes isolated from Aurkc-/- mice, we show that the mutations have different effects on AURKC function during MI. AURKC p.L49Wfs22 is a loss-of-function mutant that perturbs localization of the chromosomal passenger complex (CPC), AURKC p.C229Y is a hypomorph that cannot fully support cell-cycle progression, and AURKC p.Y248* fails to localize and function with the CPC to support chromosome segregation yet retains catalytic activity in the cytoplasm. Finally, we show that these variants of AURKC cause meiotic failure and polyploidy due to a failure in AURKC-CPC function that results in metaphase chromosome misalignment. This study is the first to assess the function of mutant alleles of AURKC that affect human fertility in a mammalian meiotic system.


Assuntos
Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Alelos , Aneuploidia , Animais , Segregação de Cromossomos , Cromossomos , Humanos , Infertilidade Masculina/genética , Masculino , Mamíferos , Meiose , Camundongos , Mutação , Espermatozoides
12.
Front Cell Dev Biol ; 3: 50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26347867

RESUMO

The mammalian genome encodes three Aurora kinase protein family members: A, B, and C. While Aurora kinase A (AURKA) and B (AURKB) are found in cells throughout the body, significant protein levels of Aurora kinase C (AURKC) are limited to cells that undergo meiosis (sperm and oocyte). Despite its discovery nearly 20 years ago, we know little about the function of AURKC compared to that of the other 2 Aurora kinases. This lack of understanding can be attributed to the high sequence homology between AURKB and AURKC preventing the use of standard approaches to understand non-overlapping and meiosis I (MI)-specific functions of the two kinases. Recent evidence has revealed distinct functions of AURKC in meiosis and may aid in our understanding of why chromosome segregation during MI often goes awry in oocytes. Many cancers aberrantly express AURKC, but because we do not fully understand AURKC function in its normal cellular context, it is difficult to predict the biological significance of this expression on the disease. Here, we consolidate and update what is known about AURKC signaling in meiotic cells to better understand why it has oncogenic potential.

13.
Biol Reprod ; 92(4): 105, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25788661

RESUMO

During meiosis I (MI) in oocytes, the maturation-associated decrease of histone acetylation is critical for normal meiotic progression and accurate chromosome segregation. RBBP4 is a component of several different histone deacetylase containing chromatin-remodeling complexes, but RBBP4's role in regulating MI is not known. Depleting RBBP4 in mouse oocytes resulted in multipolar spindles at metaphase (Met) I with subsequent perturbed meiotic progression and increased incidence of abnormal spindles, chromosome misalignment, and aneuploidy at Met II. We attribute these defects to improper deacetylation of histones because histones H3K4, H4K8, H4K12, and H4K16 were hyperacetylated in RBBP4-depleted oocytes. Importantly, we show that RBBP4-mediated histone deacetylation is essential for regulating bipolar spindle assembly, at least partially, through promoting Aurora kinase (AURK) C function. To our knowledge, these results are the first to identify RBBP4 as a regulator of histone deacetylation during oocyte maturation, and they provide evidence that deacetylation is required for bipolar spindle assembly through AURKC.


Assuntos
Histona Desacetilases/metabolismo , Oócitos/fisiologia , Proteína 4 de Ligação ao Retinoblastoma/fisiologia , Fuso Acromático/fisiologia , Animais , Aurora Quinase C/metabolismo , Cromossomos/genética , Desenvolvimento Embrionário , Feminino , Técnicas de Silenciamento de Genes , Meiose/genética , Meiose/fisiologia , Camundongos , Gravidez , Proteína 4 de Ligação ao Retinoblastoma/genética , Proteína 7 de Ligação ao Retinoblastoma/genética , Proteína 7 de Ligação ao Retinoblastoma/fisiologia
14.
J Cell Sci ; 127(Pt 23): 5066-78, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25315835

RESUMO

Meiosis I (MI), the division that generates haploids, is prone to errors that lead to aneuploidy in females. Haspin is a kinase that phosphorylates histone H3 on threonine 3, thereby recruiting Aurora kinase B (AURKB) and the chromosomal passenger complex (CPC) to kinetochores to regulate mitosis. Haspin and AURKC, an AURKB homolog, are enriched in germ cells, yet their significance in regulating MI is not fully understood. Using inhibitors and overexpression approaches, we show a role for haspin during MI in mouse oocytes. Haspin-perturbed oocytes display abnormalities in chromosome morphology and alignment, improper kinetochore-microtubule attachments at metaphase I and aneuploidy at metaphase II. Unlike in mitosis, kinetochore localization remained intact, whereas the distribution of the CPC along chromosomes was absent. The meiotic defects following haspin inhibition were similar to those observed in oocytes where AURKC was inhibited, suggesting that the correction of microtubule attachments during MI requires AURKC along chromosome arms rather than at kinetochores. Our data implicate haspin as a regulator of the CPC and chromosome segregation during MI, while highlighting important differences in how chromosome segregation is regulated between MI and mitosis.


Assuntos
Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Prófase Meiótica I , Oócitos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Adenosina Trifosfatases/metabolismo , Aneuploidia , Animais , Aurora Quinase C/antagonistas & inibidores , Aurora Quinase C/metabolismo , Células Cultivadas , Segregação de Cromossomos , Proteínas de Ligação a DNA/metabolismo , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinetocoros/enzimologia , Prófase Meiótica I/efeitos dos fármacos , Camundongos , Microtúbulos/enzimologia , Complexos Multiproteicos/metabolismo , Oócitos/efeitos dos fármacos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Transdução de Sinais , Treonina , Fatores de Tempo , Transfecção
15.
Cell Cycle ; 13(4): 600-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24317350

RESUMO

During mouse oocyte maturation histones are deacetylated, and inhibiting this deacetylation leads to abnormal chromosome segregation and aneuploidy. RBBP7 is a component of several different complexes that contain histone deacetylases, and therefore could be implicated in histone deacetylation. We find that Rbbp7 is a dormant maternal mRNA that is recruited for translation during oocyte maturation to regulate the histone deacetylation. Importantly, we show that the maturation-associated decrease of histone acetylation is required for localization and function of the chromosomal passenger complex (CPC) during oocyte meiotic maturation. This finding can explain the phenotypes of oocytes where Rbbp7 is depleted by an siRNA/morpholino cocktail including severe chromosome misalignment, improper kinetochore-microtubule attachments, impaired SAC function, cytokinesis defects, and increased incidence of aneuploidy at metaphase II (Met II). These results implicate RBBP7 as a novel regulator of histone deacetylation during oocyte maturation and provide evidence that such deacetylation is required for proper chromosome segregation by regulating localized CPC function.


Assuntos
Aurora Quinase B/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Oócitos/metabolismo , Proteínas Repressoras/metabolismo , Proteína 7 de Ligação ao Retinoblastoma/genética , Acetilação , Animais , Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Segregação de Cromossomos , Feminino , Técnicas de Silenciamento de Genes , Meiose , Camundongos , Complexos Multiproteicos/metabolismo , Oócitos/citologia , RNA Interferente Pequeno/genética , Proteína 7 de Ligação ao Retinoblastoma/metabolismo , Survivina
16.
J Cell Sci ; 126(Pt 5): 1081-5, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23345398

RESUMO

Mammalian oocytes are arrested in metaphase of second meiosis (MII) until fertilization. This arrest is enforced by the cytostatic factor (CSF), which maintains the M-phase promoting factor (MPF) in a highly active state. Although the continuous synthesis and degradation of cyclin B to maintain the CSF-mediated MII arrest is well established, it is unknown whether cyclin-dependent kinase 1 (Cdk1) phosphorylations are involved in this arrest in mouse oocytes. Here, we show that a dynamic equilibrium of Cdk1 phosphorylation is required to maintain MII arrest. When the Cdc25A phosphatase is downregulated, mouse oocytes are released from MII arrest and MPF becomes inactivated. This inactivation occurs in the absence of cyclin B degradation and is dependent on Wee1B-mediated phosphorylation of Cdk1. Thus, our data demonstrate that Cdk1 activity is maintained during MII arrest not only by cyclin turnover but also by steady state phosphorylation.


Assuntos
Metáfase/fisiologia , Oócitos/citologia , Fosfatases cdc25/metabolismo , Células Cultivadas , Humanos , Immunoblotting , Fator Promotor de Maturação/metabolismo , Meiose/genética , Meiose/fisiologia , Mesotelina , Metáfase/genética , Oócitos/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fosfatases cdc25/genética
17.
Proc Natl Acad Sci U S A ; 109(33): E2215-22, 2012 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-22778418

RESUMO

Aurora kinases are highly conserved, essential regulators of cell division. Two Aurora kinase isoforms, A and B (AURKA and AURKB), are expressed ubiquitously in mammals, whereas a third isoform, Aurora C (AURKC), is largely restricted to germ cells. Because AURKC is very similar to AURKB, based on sequence and functional analyses, why germ cells express AURKC is unclear. We report that Aurkc(-/-) females are subfertile, and that AURKB function declines as development progresses based on increasing severity of cytokinesis failure and arrested embryonic development. Furthermore, we find that neither Aurkb nor Aurkc is expressed after the one-cell stage, and that AURKC is more stable during maturation than AURKB using fluorescently tagged reporter proteins. In addition, Aurkc mRNA is recruited during maturation. Because maturation occurs in the absence of transcription, posttranscriptional regulation of Aurkc mRNA, coupled with the greater stability of AURKC protein, provides a means to ensure sufficient Aurora kinase activity, despite loss of AURKB, to support both meiotic and early embryonic cell divisions. These findings suggest a model for the presence of AURKC in oocytes: that AURKC compensates for loss of AURKB through differences in both message recruitment and protein stability.


Assuntos
Desenvolvimento Embrionário , Oócitos/citologia , Oócitos/enzimologia , Oogênese , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aurora Quinase A , Aurora Quinase B , Aurora Quinase C , Aurora Quinases , Sequência de Bases , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Estabilidade Enzimática , Feminino , Meiose , Camundongos , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteólise , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo
18.
J Vis Exp ; (53)2011 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-21808228

RESUMO

Mistakes in chromosome segregation lead to aneuploid cells. In somatic cells, aneuploidy is associated with cancer but in gametes, aneuploidy leads to infertility, miscarriages or developmental disorders like Down syndrome. Haploid gametes form through species-specific developmental programs that are coupled to meiosis. The first meiotic division (MI) is unique to meiosis because sister chromatids remain attached while homologous chromosomes are segregated. For reasons not fully understood, this reductional division is prone to errors and is more commonly the source of aneuploidy than errors in meiosis II (MII) or than errors in male meiosis. In mammals, oocytes arrest at prophase of MI with a large, intact germinal vesicle (GV; nucleus) and only resume meiosis when they receive ovulatory cues. Once meiosis resumes, oocytes complete MI and undergo an asymmetric cell division, arresting again at metaphase of MII. Eggs will not complete MII until they are fertilized by sperm. Oocytes also can undergo meiotic maturation using established in vitro culture conditions. Because generation of transgenic and gene-targeted mouse mutants is costly and can take long periods of time, manipulation of female gametes in vitro is a more economical and time-saving strategy. Here, we describe methods to isolate prophase-arrested oocytes from mice and for microinjection. Any material of choice may be introduced into the oocyte, but because meiotically-competent oocytes are transcriptionally silent cRNA, and not DNA, must be injected for ectopic expression studies. To assess ploidy, we describe our conditions for in vitro maturation of oocytes to MII eggs. Historically, chromosome-spreading techniques are used for counting chromosome number. This method is technically challenging and is limited to only identifying hyperploidies. Here, we describe a method to determine hypo-and hyperploidies using intact eggs. This method uses monastrol, a kinesin-5 inhibitor, that collapses the bipolar spindle into a monopolar spindle thus separating chromosomes such that individual kinetochores can readily be detected and counted by using an anti-CREST autoimmune serum. Because this method is performed in intact eggs, chromosomes are not lost due to operator error.


Assuntos
Microinjeções/métodos , Oócitos/fisiologia , Ploidias , Animais , DNA/administração & dosagem , Feminino , Camundongos , Oócitos/ultraestrutura , Prófase , RNA Complementar/administração & dosagem
19.
Mol Reprod Dev ; 76(11): 1094-105, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19565641

RESUMO

The elevated incidence of aneuploidy in human oocytes warrants study of the molecular mechanisms regulating proper chromosome segregation. The Aurora kinases are a well-conserved family of serine/threonine kinases that are involved in proper chromosome segregation during mitosis and meiosis. Here we report the expression and localization of all three Aurora kinase homologs, AURKA, AURKB, and AURKC, during meiotic maturation of mouse oocytes. AURKA, the most abundantly expressed homolog, localizes to the spindle poles during meiosis I (MI) and meiosis II (MII), whereas AURKB is concentrated at kinetochores, specifically at metaphase of MI (Met I). The germ cell-specific homolog, AURKC, is found along the entire length of chromosomes during both meiotic divisions. Maturing oocytes in the presence of the small molecule pan-Aurora kinase inhibitor, ZM447439 results in defects in meiotic progression and chromosome alignment at both Met I and Met II. Over-expression of AURKB, but not AURKA or AURKC, rescues the chromosome alignment defect suggesting that AURKB is the primary Aurora kinase responsible for regulating chromosome dynamics during meiosis in mouse oocytes.


Assuntos
Segregação de Cromossomos/fisiologia , Oócitos/enzimologia , Oócitos/ultraestrutura , Proteínas Serina-Treonina Quinases/metabolismo , Análise de Variância , Animais , Aurora Quinase A , Aurora Quinase B , Aurora Quinase C , Aurora Quinases , Benzamidas/farmacologia , Segregação de Cromossomos/efeitos dos fármacos , Cromossomos/metabolismo , Imuno-Histoquímica , Cinetocoros/metabolismo , Meiose , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Quinazolinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA