Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 42(23): e113955, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37850882

RESUMO

Recent studies have reported the differentiation of pluripotent cells into oocytes in vitro. However, the developmental competence of in vitro-generated oocytes remains low. Here, we perform a comprehensive comparison of mouse germ cell development in vitro over all culture steps versus in vivo with the goal to understand mechanisms underlying poor oocyte quality. We show that the in vitro differentiation of primordial germ cells to growing oocytes and subsequent follicle growth is critical for competence for preimplantation development. Systematic transcriptome analysis of single oocytes that were subjected to different culture steps identifies genes that are normally upregulated during oocyte growth to be susceptible for misregulation during in vitro oogenesis. Many misregulated genes are Polycomb targets. Deregulation of Polycomb repression is therefore a key cause and the earliest defect known in in vitro oocyte differentiation. Conversely, structurally normal in vitro-derived oocytes fail at zygotic genome activation and show abnormal acquisition of 5-hydroxymethylcytosine on maternal chromosomes. Our data identify epigenetic regulation at an early stage of oogenesis limiting developmental competence and suggest opportunities for future improvements.


Assuntos
Epigênese Genética , Oócitos , Feminino , Animais , Camundongos , Folículo Ovariano , Oogênese/genética , Células Germinativas
2.
Nature ; 615(7954): 900-906, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36922585

RESUMO

Sex chromosome disorders severely compromise gametogenesis in both males and females. In oogenesis, the presence of an additional Y chromosome or the loss of an X chromosome disturbs the robust production of oocytes1-5. Here we efficiently converted the XY chromosome set to XX without an additional Y chromosome in mouse pluripotent stem (PS) cells. In addition, this chromosomal alteration successfully eradicated trisomy 16, a model of Down's syndrome, in PS cells. Artificially produced euploid XX PS cells differentiated into mature oocytes in culture with similar efficiency to native XX PS cells. Using this method, we differentiated induced pluripotent stem cells from the tail of a sexually mature male mouse into fully potent oocytes, which gave rise to offspring after fertilization. This study provides insights that could ameliorate infertility caused by sex chromosome or autosomal disorders, and opens the possibility of bipaternal reproduction.


Assuntos
Engenharia Genética , Técnicas In Vitro , Oócitos , Cromossomo X , Animais , Feminino , Masculino , Camundongos , Oócitos/metabolismo , Oócitos/fisiologia , Cromossomo X/genética , Cromossomo Y/genética , Células-Tronco Pluripotentes/metabolismo , Síndrome de Down/genética , Síndrome de Down/terapia , Fertilização , Infertilidade/terapia , Homossexualidade Masculina , Transtornos dos Cromossomos Sexuais/complicações , Transtornos dos Cromossomos Sexuais/genética , Transtornos dos Cromossomos Sexuais/terapia , Engenharia Genética/métodos
3.
J Reprod Dev ; 69(1): 1-9, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36436912

RESUMO

The study of the size of cells and organelles has a long history, dating back to the 1600s when cells were defined. In particular, various methods have elucidated the size of the nucleus and the mitotic spindle in several species. However, little research has been conducted on oocyte size and organelles in mammals, and many questions remain to be answered. The appropriate size is essential to cell function properly. Oocytes have a very large cytoplasm, which is more than 100 times larger than that of general somatic cells in mammals. In this review, we discuss how oocytes acquire an enormous cytoplasmic size and the adverse effects of a large cytoplasmic size on cellular functions.


Assuntos
Meiose , Oócitos , Animais , Citoplasma , Fuso Acromático/fisiologia , Mamíferos
4.
EMBO Rep ; 23(7): e54992, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35587095

RESUMO

Microinjection of spermatozoa or spermatids into oocytes is a major choice for infertility treatment. However, the use of premeiotic spermatocytes has never been considered because of its technical problems. Here, we show that the efficiency of spermatocyte injection in mice can be improved greatly by reducing the size of the recipient oocytes. Live imaging showed that the underlying mechanism involves reduced premature separation of the spermatocyte's meiotic chromosomes, which produced much greater (19% vs. 1%) birth rates in smaller oocytes. Application of this technique to spermatocyte arrest caused by STX2 deficiency, an azoospermia factor also found in humans, resulted in the production of live offspring. Thus, the microinjection of primary spermatocytes into oocytes may be a potential treatment for overcoming a form of nonobstructive azoospermia caused by meiotic failure.


Assuntos
Azoospermia , Espermatócitos , Animais , Humanos , Masculino , Meiose , Camundongos , Oócitos , Espermátides
5.
J Cell Biol ; 220(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34424312

RESUMO

Zygotes require two accurate sets of parental chromosomes, one each from the mother and the father, to undergo normal embryogenesis. However, upon egg-sperm fusion in vertebrates, the zygote has three sets of chromosomes, one from the sperm and two from the egg. The zygote therefore eliminates one set of maternal chromosomes (but not the paternal chromosomes) into the polar body through meiosis, but how the paternal chromosomes are protected from maternal meiosis has been unclear. Here we report that RanGTP and F-actin dynamics prevent egg-sperm fusion in proximity to maternal chromosomes. RanGTP prevents the localization of Juno and CD9, egg membrane proteins that mediate sperm fusion, at the cell surface in proximity to maternal chromosomes. Following egg-sperm fusion, F-actin keeps paternal chromosomes away from maternal chromosomes. Disruption of these mechanisms causes the elimination of paternal chromosomes during maternal meiosis. This study reveals a novel critical mechanism that prevents aneuploidy in zygotes.


Assuntos
Citoesqueleto de Actina/metabolismo , Cromossomos/metabolismo , Fertilização , Proteína ran de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos
6.
Aging Cell ; 20(8): e13428, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34245092

RESUMO

Chromosome segregation errors in oocytes lead to the production of aneuploid eggs, which are the leading cause of pregnancy loss and of several congenital diseases such as Down syndrome. The frequency of chromosome segregation errors in oocytes increases with maternal age, especially at a late stage of reproductive life. How aging at various life stages affects oocytes differently remains poorly understood. In this study, we describe aging-associated changes in the transcriptome profile of mouse oocytes throughout reproductive life. Our single-oocyte comprehensive RNA sequencing using RamDA-seq revealed that oocytes undergo transcriptome changes at a late reproductive stage, whereas their surrounding cumulus cells exhibit transcriptome changes at an earlier stage. Calorie restriction, a paradigm that reportedly prevents aging-associated egg aneuploidy, promotes a transcriptome shift in oocytes with the up-regulation of genes involved in chromosome segregation. This shift is accompanied by the improved maintenance of chromosomal cohesin, the loss of which is a hallmark of oocyte aging and causes chromosome segregation errors. These findings have implications for understanding how oocytes undergo aging-associated functional decline throughout their reproductive life in a context-dependent manner.


Assuntos
Envelhecimento/genética , Restrição Calórica/métodos , Perfilação da Expressão Gênica/métodos , Oócitos/metabolismo , Animais , Feminino , Humanos , Camundongos
7.
EMBO Rep ; 22(4): e51400, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33655692

RESUMO

In mouse oocytes, acentriolar MTOCs functionally replace centrosomes and act as microtubule nucleation sites. Microtubules nucleated from MTOCs initially assemble into an unorganized ball-like structure, which then transforms into a bipolar spindle carrying MTOCs at its poles, a process called spindle bipolarization. In mouse oocytes, spindle bipolarization is promoted by kinetochores but the mechanism by which kinetochore-microtubule attachments contribute to spindle bipolarity remains unclear. This study demonstrates that the stability of kinetochore-microtubule attachment is essential for confining MTOC positions at the spindle poles and for limiting spindle elongation. MTOC sorting is gradual and continues even in the metaphase spindle. When stable kinetochore-microtubule attachments are disrupted, the spindle is unable to restrict MTOCs at its poles and fails to terminate its elongation. Stable kinetochore fibers are directly connected to MTOCs and to the spindle poles. These findings suggest a role for stable kinetochore-microtubule attachments in fine-tuning acentrosomal spindle bipolarity.


Assuntos
Cinetocoros , Fuso Acromático , Animais , Camundongos , Centro Organizador dos Microtúbulos , Microtúbulos , Oócitos
8.
Nature ; 589(7841): 264-269, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33328630

RESUMO

During female germline development, oocytes become a highly specialized cell type and form a maternal cytoplasmic store of crucial factors. Oocyte growth is triggered at the transition from primordial to primary follicle and is accompanied by dynamic changes in gene expression1, but the gene regulatory network that controls oocyte growth remains unknown. Here we identify a set of transcription factors that are sufficient to trigger oocyte growth. By investigation of the changes in gene expression and functional screening using an in vitro mouse oocyte development system, we identified eight transcription factors, each of which was essential for the transition from primordial to primary follicle. Notably, enforced expression of these transcription factors swiftly converted pluripotent stem cells into oocyte-like cells that were competent for fertilization and subsequent cleavage. These transcription-factor-induced oocyte-like cells were formed without specification of primordial germ cells, epigenetic reprogramming or meiosis, and demonstrate that oocyte growth and lineage-specific de novo DNA methylation are separable from the preceding epigenetic reprogramming in primordial germ cells. This study identifies a core set of transcription factors for orchestrating oocyte growth, and provides an alternative source of ooplasm, which is a unique material for reproductive biology and medicine.


Assuntos
Oócitos/metabolismo , Oogênese/genética , Fatores de Transcrição/metabolismo , Animais , Linhagem da Célula , Epigênese Genética , Feminino , Fertilização , Meiose , Metilação , Camundongos , Oócitos/citologia , Folículo Ovariano/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
9.
Genes Cells ; 25(10): 685-694, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32865279

RESUMO

Chromosome segregation requires the formation of a bipolar spindle. The timely bipolarization of the acentrosomal spindle during meiosis I in mouse oocytes depends on the antiparallel microtubule crosslinker Prc1. How Prc1 is regulated in oocytes remains poorly understood. In this study, we show that the kinase Cdk1 negatively regulates the spindle localization of Prc1 in mouse oocytes. The acute inhibition of Cdk1 activity led to excessive localization of Prc1 at the spindle and kinetochores, whereas the overactivation of Cdk1 had opposite effects. The overexpression of Prc1 carrying mutations at Cdk1-mediated phosphorylation sites increased its localization to the spindle, accelerated spindle bipolarization and caused spindle-checkpoint-dependent arrest at metaphase I. Overactivation of Cdk1 delayed spindle bipolarization, which was reversed by the overexpression of a phospho-mutant form but not the wild-type form of Prc1. These results suggest that Cdk1-mediated phosphorylation negatively regulates Prc1 localization to ensure the timely bipolarization of the acentrosomal spindle during meiosis I in mammalian oocytes.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Animais , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos , Feminino , Cinetocoros/metabolismo , Masculino , Metáfase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microtúbulos/metabolismo , Oócitos/metabolismo , Fuso Acromático/genética
10.
Nat Commun ; 11(1): 2652, 2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32461611

RESUMO

Acentrosomal meiosis in oocytes represents a gametogenic challenge, requiring spindle bipolarization without predefined bipolar cues. While much is known about the structures that promote acentrosomal microtubule nucleation, less is known about the structures that mediate spindle bipolarization in mammalian oocytes. Here, we show that in mouse oocytes, kinetochores are required for spindle bipolarization in meiosis I. This process is promoted by oocyte-specific, microtubule-independent enrichment of the antiparallel microtubule crosslinker Prc1 at kinetochores via the Ndc80 complex. In contrast, in meiosis II, cytoplasm that contains upregulated factors including Prc1 supports kinetochore-independent pathways for spindle bipolarization. The kinetochore-dependent mode of spindle bipolarization is required for meiosis I to prevent chromosome segregation errors. Human oocytes, where spindle bipolarization is reportedly error prone, exhibit no detectable kinetochore enrichment of Prc1. This study reveals an oocyte-specific function of kinetochores in acentrosomal spindle bipolarization in mice, and provides insights into the error-prone nature of human oocytes.


Assuntos
Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Oócitos/metabolismo , Fuso Acromático/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos , Proteínas do Citoesqueleto/metabolismo , Feminino , Gametogênese/fisiologia , Meiose/fisiologia , Camundongos , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo
11.
EMBO Rep ; 20(8): e47905, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31290587

RESUMO

The accuracy of the two sequential meiotic divisions in oocytes is essential for creating a haploid gamete with a normal chromosomal content. Here, we have analysed the 3D dynamics of chromosomes during the second meiotic division in live mouse oocytes. We find that chromosomes form stable kinetochore-microtubule attachments at the end of prometaphase II stage that are retained until anaphase II onset. Remarkably, we observe that more than 20% of the kinetochore-microtubule attachments at the metaphase II stage are merotelic or lateral. However, < 1% of all chromosomes at onset of anaphase II are found to lag at the spindle equator and < 10% of the laggards missegregate and give rise to aneuploid gametes. Our results demonstrate that aberrant kinetochore-microtubule attachments are not corrected at the metaphase stage of the second meiotic division. Thus, the accuracy of the chromosome segregation process in mouse oocytes during meiosis II is ensured by an efficient correction process acting at the anaphase stage.


Assuntos
Anáfase , Cinetocoros/ultraestrutura , Metáfase , Microtúbulos/ultraestrutura , Oócitos/ultraestrutura , Sequência de Aminoácidos , Animais , Cromátides/metabolismo , Cromátides/ultraestrutura , Segregação de Cromossomos , Feminino , Humanos , Cinetocoros/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Oócitos/metabolismo , Espermatócitos/metabolismo , Espermatócitos/ultraestrutura , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura , Imagem com Lapso de Tempo
12.
Nat Commun ; 10(1): 847, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30783089

RESUMO

Autophagy, an evolutionarily conserved cytoplasmic degradation system, has been implicated as a convergent mechanism in various longevity pathways. Autophagic activity decreases with age in several organisms, but the underlying mechanism is unclear. Here, we show that the expression of Rubicon, a negative regulator of autophagy, increases in aged worm, fly and mouse tissues at transcript and/or protein levels, suggesting that an age-dependent increase in Rubicon impairs autophagy over time, and thereby curtails animal healthspan. Consistent with this idea, knockdown of Rubicon extends worm and fly lifespan and ameliorates several age-associated phenotypes. Tissue-specific experiments reveal that Rubicon knockdown in neurons has the greatest effect on lifespan. Rubicon knockout mice exhibits reductions in interstitial fibrosis in kidney and reduced α-synuclein accumulation in the brain. Rubicon is suppressed in several long-lived worms and calorie restricted mice. Taken together, our results suggest that suppression of autophagic activity by Rubicon is one of signatures of aging.


Assuntos
Envelhecimento/fisiologia , Proteínas Relacionadas à Autofagia/metabolismo , Autofagia/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas de Drosophila/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Envelhecimento/genética , Animais , Animais Geneticamente Modificados , Autofagia/genética , Proteínas Relacionadas à Autofagia/genética , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Drosophila/genética , Drosophila/fisiologia , Proteínas de Drosophila/genética , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intracelular/genética , Longevidade , Masculino , Camundongos Endogâmicos C57BL
14.
PLoS One ; 13(8): e0202663, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30125305

RESUMO

Mammalian oocytes and zygotes have nucleoli that are transcriptionally inactive and structurally distinct from nucleoli in somatic cells. These nucleoli have been termed nucleolus precursor bodies (NPBs). Recent research has shown that NPBs are important for embryonic development, but they are only required during pronuclear formation. After fertilization, multiple small NPBs are transiently formed in male and female pronuclei and then fuse into a single large NPB in zygotes. In cloned embryos produced by somatic cell nuclear transfer (SCNT), multiple NPBs are formed and maintained in the pseudo-pronucleus, and this is considered an abnormality of the cloned embryos. Despite this difference between SCNT and normal embryos, it is unclear how the size and number of NPBs in pronuclei is determined. Here, we show that in mouse embryos, the volume of NPB materials plays a major role in the NPB scaling through a limiting component mechanism and determines whether a single or multiple NPBs will form in the pronucleus. Extra NPB- and extra MII spindle-injection experiments demonstrated that the total volume of NPBs was maintained regardless of the pronucleus number and the ratio of pronucleus/NPB is important for fusion into a single NPB. Based on these results, we examined whether extra-NPB injection rescued multiple NPB maintenance in SCNT embryos. When extra-NPBs were injected into enucleated-MII oocytes before SCNT, the number of NPBs in pseudo-pronuclei of SCNT embryos was reduced. These results indicate that multiple NPB maintenance in SCNT embryos is caused by insufficient volume of NPB.


Assuntos
Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Oócitos/crescimento & desenvolvimento , Zigoto/crescimento & desenvolvimento , Animais , Transferência Embrionária , Desenvolvimento Embrionário , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Técnicas de Transferência Nuclear , Oócitos/metabolismo , Gravidez , Zigoto/metabolismo
15.
Methods Mol Biol ; 1818: 89-97, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29961258

RESUMO

Meiotic division is a dynamic process that exhibits active interactive behaviors amongst different intracellular structures and components for spindle assembly and chromosome segregation. Understanding the mechanisms of meiotic spindle assembly and chromosome segregation therefore requires a quantitative analysis of spatiotemporal relationships among different structures and components. In this chapter, we describe a method for triple-color live imaging of meiotic division in mouse oocytes. This approach combines the microinjection of RNAs encoding proteins tagged with green and red fluorescent proteins and the visualization of microtubules with the fluorogenic far-red probe SiR-Tubulin. This method enables the simultaneous spatiotemporal mapping of three different components of the spindle and chromosomes, which opens the way to quantitative analysis of their interactive behaviors.


Assuntos
Meiose , Microscopia de Fluorescência/métodos , Oócitos/citologia , Animais , Células Cultivadas , Segregação de Cromossomos , Feminino , Proteínas de Fluorescência Verde/metabolismo , Processamento de Imagem Assistida por Computador , Proteínas Luminescentes/metabolismo , Camundongos , Microtúbulos/metabolismo , Oócitos/fisiologia , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
16.
Curr Biol ; 28(10): 1661-1669.e4, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29754905

RESUMO

The production of haploid gametes requires the maintenance of centromeric cohesion between sister chromatids through the transition between two successive meiotic divisions, meiosis I and meiosis II. One mechanism for the cohesion maintenance is shugoshin-dependent protection of centromeric cohesin at anaphase I onset [1-3]. However, how centromeric cohesion is maintained during late anaphase I and telophase I, when centromeric shugoshin is undetectable [1-3], remains largely unexplored. Here we show that the centromeric small ubiquitin-related modifier (SUMO) pathway is critical for the maintenance of centromeric cohesion during post-anaphase-I periods in mouse oocytes. SUMO2/3 and E3 ligase PIAS are enriched near centromeres during late anaphase I and telophase I. Specific perturbation of the centromeric SUMO pathway results in precocious loss of centromeric cohesin at telophase I, although shugoshin-dependent centromeric protection at anaphase I onset remains largely intact. Prevention of the SUMO perturbation during post-anaphase-I periods restores the maintenance of centromeric cohesion through the meiosis I-II transition. Thus, the post-anaphase-I centromeric SUMO pathway ensures continuous maintenance of centromeric cohesion through the meiosis I-II transition.


Assuntos
Centrômero/fisiologia , Meiose/fisiologia , Oócitos/fisiologia , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Animais , Feminino , Camundongos
17.
Methods Cell Biol ; 144: 459-474, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29804683

RESUMO

Meiotic divisions in females occur in fully grown oocytes that have a large cytoplasmic volume. The intracellular processes that are needed to accomplish meiotic divisions, such as spindle formation, chromosome segregation, and polar body extrusion, are controlled by the concerted actions of nuclear and cytoplasmic factors, which exhibit dynamic quantitative and spatiotemporal changes during meiotic maturation. Thus, distinguishing between meiotic controls that are mediated by cytoplasmic factors and those mediated by nuclear factors helps in the understanding of the mechanisms underlying meiotic divisions. Here, we describe a method to artificially modify the number of nuclei and the volume of the cytoplasm of mouse oocytes through cytoplasmic removal, enucleation, and cell fusion. The oocytes generated by this method are viable and undergo reproducible meiotic divisions exhibiting the effects of altered amounts of cytoplasmic and nuclear factors, which can be analyzed by various assays, such as live imaging microscopy.


Assuntos
Fusão Celular/métodos , Núcleo Celular/metabolismo , Oócitos/citologia , Animais , Separação Celular , Feminino , Camundongos , Fuso Acromático/metabolismo
18.
Dev Growth Differ ; 60(1): 33-43, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29318599

RESUMO

Proper kinetochore-microtubule attachment is essential for correct chromosome segregation. Therefore, cells normally possess multiple mechanisms for the prevention of errors in kinetochore-microtubule attachments and for selective stabilization of correct attachments. However, the oocyte, a cell that produces an egg through meiosis, exhibits a high frequency of errors in kinetochore-microtubule attachments. These attachment errors predispose oocytes to chromosome segregation errors, resulting in aneuploidy in eggs. This review aims to provide possible explanations for the error-prone nature of oocytes by examining key differences among other cell types in the mechanisms for the establishment of kinetochore-microtubule attachments.


Assuntos
Aneuploidia , Cinetocoros/fisiologia , Meiose , Microtúbulos/fisiologia , Oócitos/citologia , Oócitos/fisiologia , Animais , Feminino , Humanos , Mamíferos
19.
Dev Cell ; 41(3): 287-298.e4, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28486131

RESUMO

Chromosome segregation during meiosis in oocytes is error prone. The uniquely large cytoplasmic size of oocytes, which provides support for embryogenesis after fertilization, might be a predisposing factor for meiotic errors. However, this hypothesis remains unproven. Here, we show that cytoplasmic size affects the functionality of the acentrosomal spindle. Artificially decreasing the cytoplasmic size in mouse oocytes allows the acentrosomal spindle poles to have a better-focused distribution of microtubule-organizing centers and to biorient chromosomes more efficiently, whereas enlargement of the cytoplasmic size has the opposite effects. Moreover, we found that the cytoplasmic size-dependent dilution of nuclear factors, including anaphase inhibitors that are preformed at the nuclear membrane, limits the spindle's capacity to prevent anaphase entry with misaligned chromosomes. The present study defines a large cytoplasmic volume as a cell-intrinsic feature linked to the error-prone nature of oocytes. This may represent a trade-off between meiotic fidelity and post-fertilization developmental competence.


Assuntos
Segregação de Cromossomos/fisiologia , Meiose/fisiologia , Microtúbulos/metabolismo , Oócitos/citologia , Fuso Acromático/metabolismo , Anáfase/fisiologia , Animais , Citoplasma/metabolismo , Fertilização/fisiologia , Camundongos
20.
Methods Mol Biol ; 1457: 241-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27557586

RESUMO

Fluorescence live imaging is a powerful approach to study intracellular dynamics during cellular events such as cell division. By applying automated confocal live imaging to mouse oocytes, in which meiotic maturation can be induced in vitro after the introduction of fluorescent proteins through microinjection, the meiotic dynamics of intracellular structures, such as chromosomes, can be monitored at high resolution. A combination of this method with approaches for the perturbation of specific proteins opens up opportunities for understanding the molecular and intracellular basis of mammalian meiosis.


Assuntos
Meiose , Imagem Molecular/métodos , Oócitos/metabolismo , Animais , Biomarcadores , Separação Celular/métodos , Feminino , Espaço Intracelular/metabolismo , Meiose/genética , Camundongos , Microinjeções/métodos , Oócitos/citologia , RNA/genética , RNA/isolamento & purificação , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...