Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Adv Sci (Weinh) ; 10(27): e2301940, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37493331

RESUMO

Sperm-induced Ca2+ rise is critical for driving oocyte activation and subsequent embryonic development, but little is known about how lasting Ca2+ oscillations are regulated. Here it is shown that NLRP14, a maternal effect factor, is essential for keeping Ca2+ oscillations and early embryonic development. Few embryos lacking maternal NLRP14 can develop beyond the 2-cell stage. The impaired developmental potential of Nlrp14-deficient oocytes is mainly caused by disrupted cytoplasmic function and calcium homeostasis due to altered mitochondrial distribution, morphology, and activity since the calcium oscillations and development of Nlrp14-deficient oocytes can be rescued by substitution of whole cytoplasm by spindle transfer. Proteomics analysis reveal that cytoplasmic UHRF1 (ubiquitin-like, containing PHD and RING finger domains 1) is significantly decreased in Nlrp14-deficient oocytes, and Uhrf1-deficient oocytes also show disrupted calcium homeostasis and developmental arrest. Strikingly, it is found that the mitochondrial Na+ /Ca2+ exchanger (NCLX) encoded by Slc8b1 is significantly decreased in the Nlrp14mNull oocyte. Mechanistically, NLRP14 interacts with the NCLX intrinsically disordered regions (IDRs) domain and maintain its stability by regulating the K27-linked ubiquitination. Thus, the study reveals NLRP14 as a crucial player in calcium homeostasis that is important for early embryonic development.


Assuntos
Cálcio , Nucleosídeo-Trifosfatase , Sêmen , Humanos , Masculino , Cálcio/metabolismo , Homeostase/fisiologia , Oócitos/metabolismo , Sêmen/metabolismo , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo , Ubiquitinação , Animais , Camundongos , Nucleosídeo-Trifosfatase/metabolismo
2.
Cell Prolif ; 56(3): e13377, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36479743

RESUMO

Pre-replication complex (pre-RC) is critical for DNA replication initiation. CDT1 and MCM2 are the subunits of pre-RC, and proper regulation of CDT1 and MCM2 are necessary for DNA replication and cell proliferation. The present study aimed to explore the role of CDT1 and MCM2 in oocyte meiotic maturation and early embryonic development. The depletion and overexpression of Cdt1 and Mcm2 in oocyte and zygote were achieved by microinjecting specific siRNA and mRNA to explored their functions in oocyte meiotic maturation and embryonic development. Then, we examined the effect of CDT1 and MCM2 on other signal pathways by immunostaining the expression of related maker genes. We showed that neither depletion nor overexpression of Cdt1 affected oocyte meiotic progressions. The CDT1 was degraded in S phase and remained at a low level in G2 phase of zygote. Exogenous expression of Cdt1 in G2 phase led to embryo attest at zygote stage. Mechanistically, CDT1 overexpression induced DNA re-replication and thus DNA damage check-point activation. Protein abundance of MCM2 was stable throughout the cell cycle, and embryos with overexpressed MCM2 could develop to blastocysts normally. Overexpression or depletion of Mcm2 also had no effect on oocyte meiotic maturation. Our results indicate that pre-RC subunits CDT1 and MCM2 are not involved in oocyte meiotic maturation. In zygote, CDT1 but not MCM2 is the major regulator of DNA replication in a cell cycle dependent manner. Furthermore, its' degradation is essential for zygotes to prevent from DNA re-replication in G2 stage.


Assuntos
Proteínas de Ciclo Celular , Zigoto , Zigoto/metabolismo , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Ciclo Celular , DNA
3.
Int J Biol Sci ; 18(11): 4513-4531, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35864958

RESUMO

During oocyte growth, various epigenetic modifications are gradually established, accompanied by accumulation of large amounts of mRNAs and proteins. However, little is known about the relationship between epigenetic modifications and meiotic progression. Here, by using Gdf9-Cre to achieve oocyte-specific ablation of Ehmt2 (Euchromatic-Histone-Lysine-Methyltransferase 2) from the primordial follicle stage, we found that female mutant mice were infertile. Oocyte-specific knockout of Ehmt2 caused failure of homologous chromosome separation independent of persistently activated SAC during the first meiosis. Further studies revealed that lacking maternal Ehmt2 affected the transcriptional level of Ccnb3, while microinjection of exogenous Ccnb3 mRNA could partly rescue the failure of homologous chromosome segregation. Of particular importance was that EHMT2 regulated ccnb3 transcriptions by regulating CTCF binding near ccnb3 gene body in genome in oocytes. In addition, the mRNA level of Ccnb3 significantly decreased in the follicles microinjected with Ctcf siRNA. Therefore, our findings highlight the novel function of maternal EHMT2 on the metaphase I-to-anaphase I transition in mouse oocytes: regulating the transcription of Ccnb3.


Assuntos
Segregação de Cromossomos , Meiose , Anáfase , Animais , Feminino , Meiose/genética , Camundongos , Oócitos/metabolismo , RNA Mensageiro/metabolismo
4.
Reprod Biol Endocrinol ; 20(1): 90, 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35710416

RESUMO

BACKGROUND: Nonobstructive azoospermia (NOA) is one of the most difficult forms of male infertility to treat, and its pathogenesis is still unclear. miRNAs can regulate autophagy by affecting their target gene expression. Our previous study found that miR-188-3p expression in NOA patients was low. There are potential binding sites between the autophagy gene ATG7 and miR-188-3p. This study aimed to verify the binding site between miR-188-3p and ATG7 and whether miR-188-3p affects autophagy and participates in NOA by regulating ATG7 to influence the autophagy marker genes LC3 and Beclin-1. METHODS: Testicular tissue from 16 NOA patients and 16 patients with normal spermatogenesis and 5 cases in each group of pathological sections were collected. High-throughput sequencing was performed to detect mRNA expression differences. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, immunohistochemical staining and immunofluorescence were used to detect protein localization and expression. Autophagosome changes were detected by electron microscopy. The targeting relationship between miR-188-3p and ATG7 was confirmed by a luciferase assay. RESULTS: ATG7 protein was localized in the cytoplasm of spermatogenic cells at all levels, and the ATG7 gene (p = 0.019) and protein (p = 0.000) were more highly expressed in the NOA group. ATG7 expression after overexpression/inhibition of miR-188-3p was significantly lower (p = 0.029)/higher (p = 0.021) than in the control group. After overexpression of miR-188-3p, the ATG7 3'UTR-WT luciferase activity was impeded (p = 0.004), while the ATG7 3'UTR-MUT luciferase activity showed no significant difference (p = 0.46). LC3 (p = 0.023) and Beclin-1 (p = 0.041) expression in the NOA group was significantly higher. LC3 and Beclin-1 gene expression after miR-188-3p overexpression/inhibition was significantly lower (p = 0.010 and 0.024, respectively) and higher (p = 0.024 and 0.049, respectively). LC3 punctate aggregation in the cytoplasm decreased after overexpression of miR-188-3p, while the LC3 punctate aggregation in the miR-188-3p inhibitor group was higher. The number of autophagosomes in the miR-188-3p mimic group was lower than the number of autophagosomes in the mimic NC group. CONCLUSIONS: LC3 and Beclin-1 were more highly expressed in NOA testes and negatively correlated with the expression of miR-188-3p, suggesting that miR-188-3p may be involved in the process of autophagy in NOA. miR-188-3p may regulate its target gene ATG7 to participate in autophagy anDual luciferase experiment d affect the development of NOA.


Assuntos
Azoospermia , MicroRNAs , Regiões 3' não Traduzidas , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Azoospermia/genética , Proteína Beclina-1/genética , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo
5.
Development ; 149(10)2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35546066

RESUMO

Mammalian early embryo cells have complex DNA repair mechanisms to maintain genomic integrity, and homologous recombination (HR) plays the main role in response to double-strand DNA breaks (DSBs) in these cells. Polo-like kinase 1 (PLK1) participates in the HR process and its overexpression has been shown to occur in a variety of human cancers. Nevertheless, the regulatory mechanism of PLK1 remains poorly understood, especially during the S and G2 phase. Here, we show that protein phosphatase 4 catalytic subunit (PPP4C) deletion causes severe female subfertility due to accumulation of DNA damage in oocytes and early embryos. PPP4C dephosphorylated PLK1 at the S137 site, negatively regulating its activity in the DSB response in early embryonic cells. Depletion of PPP4C induced sustained activity of PLK1 when cells exhibited DNA lesions that inhibited CHK2 and upregulated the activation of CDK1, resulting in inefficient loading of the essential HR factor RAD51. On the other hand, when inhibiting PLK1 in the S phase, DNA end resection was restricted. These results demonstrate that PPP4C orchestrates the switch between high-PLK1 and low-PLK1 periods, which couple the checkpoint to HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo de DNA por Recombinação , Animais , Proteínas de Ciclo Celular , Linhagem Celular , DNA/genética , Reparo do DNA por Junção de Extremidades , Reparo do DNA/genética , Desenvolvimento Embrionário/genética , Feminino , Recombinação Homóloga , Mamíferos/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , Quinase 1 Polo-Like
6.
Front Endocrinol (Lausanne) ; 12: 692082, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335469

RESUMO

Calcium is a crucial factor in regulating the biological behavior of cells. The imbalance of calcium homeostasis in cytoplasm will cause abnormal behavior of cells and the occurrence of diseases. In intracytoplasmic sperm injection (ICSI) cycle, the dysfunction of oocyte activation caused by insufficient release of Ca2+ from endoplasmic reticulum is one of the main reasons for repeated fertilization failure. Calcium ionophore (A23187) is a highly selective calcium ionophore, which can form stable complex with Ca2+ and pass through the cell membrane at will, effectively increasing intracellular Ca2+ levels. It has been reported that calcium ionophore (A23187) can activate oocytes and obtain normal embryos. However, there are few studies on unfertilized oocytes after calcium ionophore (A23187) rescue activation in ICSI cycle. The purpose of this study was to analyze the effects of calcium ionophore (A23187) rescue activation on the activation of unfertilized oocytes, embryonic development potential, embryonic development timing and chromosomal aneuploidy, and to compare and analyze the clinical data of patients with calcium ionophore (A23187) activation in clinical application. The results showed that a certain proportion of high-quality blastocysts with normal karyotype could be obtained after calcium ionophore (A23187) rescue activation of unfertilized oocytes, and it did not have a significant effect on the timing of embryo development. In clinical practice, direct activation with calcium ionophore (A23187) after ICSI was better than rescue activation the next day. In conclusions, the studies on the effectiveness and safety of calcium ionophore (A23187) rescue activation for oocytes with ICSI fertilization failure can enable some patients to obtain usable, high-quality embryos during the first ICSI cycle.


Assuntos
Calcimicina/farmacologia , Ionóforos de Cálcio/farmacologia , Oócitos/efeitos dos fármacos , Aneuploidia , Blastocisto/efeitos dos fármacos , Cromossomos , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Humanos , Injeções de Esperma Intracitoplásmicas
7.
Front Endocrinol (Lausanne) ; 12: 619437, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34367060

RESUMO

Background: The definition of recurrent implantation failure (RIF) differs clinically, one of the most controversial diagnostic criteria is the number of failed treatment cycles. We tried to investigate whether the two implantation failure could be included in the diagnostic criteria of RIF. Methods: A retrospective analysis of the clinical data of patients (N=1518) aged under 40 years with two or more implantation failure, recruited from the Center for Reproductive Medicine of the First Affiliated Hospital of Zhengzhou University from January 2016 to June 2019. Results: After adjusting for confounding factors by using binary logistic regression, the results showed that partial general information and: distribution of associated factors were significant differences such as maternal age (aOR=1.054, P=0.001), type of cycle (aOR=2.040, P<0.001), stage of embryos development (aOR=0.287, P<0.001), number of embryos transferred (aOR=0.184, P<0.001), female factor (tubal pathology) (aOR=0.432, P=0.031) and male factor (aOR=1.734, P=0.002) between the groups with two and three or more unexplained implantation failure. And further explored whether these differential factors had a significant negative impact on pregnancy outcome, the results showed that: for patients who had three unexplained implantation failure, in the fourth cycle of ET, the live birth rate decreased significantly with age (aOR=0.921, P<0.001), and the live birth rate of blastocyst transfer was significantly higher than that of cleavage embryo transfer (aOR=1.826, P=0.007). At their first assisted pregnancy treatment after the diagnosis of RIF according to these two different definitions, there were no significant difference in the biochemical pregnancy rate, clinical pregnancy rate, ectopic pregnancy rate and abortion rate (P>0.05), but the live birth rate (35.64% vs 42.95%, P=0.004) was significantly different. According to the definition of 'two or more failed treatment cycles', the live birth rate of the first ET treatment after RIF diagnosis was significantly lower than that of patients according to the definition of 'three or more failed treatment cycles'. Conclusion: For patients with unexplained recurrent implantation failure, two implantation failure cannot be included in the diagnostic criteria of RIF. This study supports the generally accepted definition of three or more failed treatment cycles for RIF.


Assuntos
Implantação do Embrião , Adulto , Feminino , Humanos , Modelos Logísticos , Gravidez , Resultado da Gravidez , Recidiva , Estudos Retrospectivos , Falha de Tratamento
8.
Genome Res ; 31(9): 1519-1530, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34330789

RESUMO

Uniparental embryos derived from only the mother (gynogenetic [GG]) or the father (androgenetic [AG]) are unique models for studying genomic imprinting and parental contributions to embryonic development. Human parthenogenetic embryos can be obtained following artificial activation of unfertilized oocytes, but the production of AG embryos by injection of two sperm into one denucleated oocyte leads to an extra centriole, resulting in multipolar spindles, abnormal cell division, and developmental defects. Here, we improved androgenote production by transferring the male pronucleus from one zygote into another haploid androgenote to prevent extra centrioles and successfully generated human diploid AG embryos capable of developing into blastocysts with an identifiable inner cell mass (ICM) and trophectoderm (TE). The GG embryos were also generated. The zygotic genome was successfully activated in both the AG and GG embryos. DNA methylome analysis showed that the GG blastocysts partially retain the oocyte transcription-dependent methylation pattern, whereas the AG blastocyst methylome showed more extensive demethylation. The methylation states of most known imprinted differentially methylated regions (DMRs) were recapitulated in the AG and GG blastocysts. Novel candidate imprinted DMRs were also identified. The production of uniparental human embryos followed by transcriptome and methylome analysis is valuable for identifying parental contributions and epigenome memory transitions during early human development.


Assuntos
Blastocisto , Epigenoma , Blastocisto/metabolismo , Metilação de DNA , Feminino , Impressão Genômica , Humanos , Masculino , Oócitos/metabolismo , Pais , Gravidez
9.
Fertil Steril ; 115(2): 463-473, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33579525

RESUMO

OBJECTIVE: To investigate the expression of aquaporin 7 (AQP7) and aquaporin 9 (AQP9) in the granulosa cells of patients with polycystic ovary syndrome (PCOS) and healthy women and detect their localization in oocytes at the germinal vesicle (GV), metaphase I (MI), MII, embryo, and blastocyst stages and the in vitro response to insulin stimulation. DESIGN: Randomized, assessor-blinded study. SETTING: Reproductive medical center. PATIENT(S): A total of 40 women (aged 20-38 years) comprising 29 cases of primary infertility and 11 cases of secondary infertility, of whom 17 had an initial diagnosis of PCOS and three received a PCOS diagnosis after an infertility examination. INTERVENTION(S): Controlling different concentrations of insulin and different treatment times in cultures of normal human granulosa cells in vitro. MAIN OUTCOME MEASURE(S): Expression of AQP7 and AQP9 genes and proteins in granulosa cells detected by real-time quantitative polymerase chain reaction, and localization in oocytes at the GV, MI, MII, embryo, and blastocyst stages by Western blot, immunohistochemical, and immunofluorescence assays, and concentrations of insulin in follicular fluid by enzyme-linked immunosorbent assay. RESULT(S): The expression levels of the AQP7 mRNA and protein in the granulosa cells of patients with PCOS were higher than found in healthy controls. We found AQP7 protein expressed in human oocytes at GV, MI, MII, embryo, and blastocyst stages; it was mainly located in the nucleoplasm. In the PCOS group, the expression level of AQP9 mRNA and protein in granulosa cells was lower, and AQP9 protein was expressed in oocytes at the GV, MI, MII, embryo, and blastocyst stages; it was localized on the nuclear membrane. Compared with healthy women, the insulin expression in patients with PCOS was higher. In cultures of normal human granulosa cells in vitro, the expression of AQP7 and AQP9 mRNA and protein decreased with the increase in insulin concentration; expression statistically significantly decreased when the insulin concentration was 100 nmol/L, and after 6 to 24 hours of exposure the lowest expression levels were found at 12 hours. CONCLUSION(S): The different localization and expression of AQP7 and AQP9 between the two groups suggests that they might be involved in oocyte maturation and embryonic development through different regulatory pathways. The expression levels of AQP7 and AQP9 were negatively correlated with insulin regulation, suggesting that insulin might affect the maturation of PCOS follicles by changing AQP7 and AQP9 expression.


Assuntos
Aquaporinas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/metabolismo , Insulina/metabolismo , Oócitos/metabolismo , Síndrome do Ovário Policístico/metabolismo , Adulto , Aquaporinas/genética , Feminino , Humanos , Infertilidade Feminina/epidemiologia , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Insulina/genética , Síndrome do Ovário Policístico/epidemiologia , Síndrome do Ovário Policístico/genética , Método Simples-Cego , Adulto Jovem
10.
Nat Commun ; 11(1): 6354, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311485

RESUMO

The formation of zygote is the beginning of mammalian life, and dynamic epigenetic modifications are essential for mammalian normal development. H3K27 di-methylation (H3K27me2) and H3K27 tri-methylation (H3K27me3) are marks of facultative heterochromatin which maintains transcriptional repression established during early development in many eukaryotes. However, the mechanism underlying establishment and regulation of epigenetic asymmetry in the zygote remains obscure. Here we show that maternal EZH2 is required for the establishment of H3K27me3 in mouse zygotes. However, combined immunostaining with ULI-NChIP-seq (ultra-low-input micrococcal nuclease-based native ChIP-seq) shows that EZH1 could partially safeguard the role of EZH2 in the formation of H3K27me2. Meanwhile, we identify that EHMT1 is involved in the establishment of H3K27me2, and that H3K27me2 might be an essential prerequisite for the following de novo H3K27me3 modification on the male pronucleus. In this work, we clarify the establishment and regulatory mechanisms of H3K27me2 and H3K27me3 in mouse zygotes.


Assuntos
Genoma , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Zigoto/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigenômica , Heterocromatina , Histona-Lisina N-Metiltransferase/genética , Masculino , Metilação , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Nuclease do Micrococo , Oogênese/fisiologia , Complexo Repressor Polycomb 2/genética , Processamento de Proteína Pós-Traducional
11.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(11): 1296-1300, 2020 Nov 10.
Artigo em Chinês | MEDLINE | ID: mdl-33179244

RESUMO

With the in-depth exploration of all stages in early-stage embryos, in particular zygotic genome activation and first cell lineage differentiation, researchers have found that early embryonic epigenetics follows a strict pattern of temporal and spatial modification. Previous studies have determined the inhibitory effect of H3K9me3 and H3K27me3 on genomic expression, and found that they are involved in many core biological events in the genome such as chromatin reprogramming, genomic imprinting, maintenance of embryonic stem cell pluripotency and somatic cell nuclear transfer, though the detailed molecular mechanism has remained elusive. From the point of developmental biology and epigenetics, this article has expounded the research progress on the methylation of H3K9 and H3K27 histones in early-stage embryos, which may provide a clue for the complex mechanism of embryonic development and improvement of culture method for embryos in vitro.


Assuntos
Desenvolvimento Embrionário , Histonas , Cromatina , Epigênese Genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Histonas/metabolismo , Humanos , Metilação , Gravidez
12.
J Cell Physiol ; 235(7-8): 5541-5554, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31984513

RESUMO

Cell division cycle protein, CDC6, is essential for the initiation of DNA replication. CDC6 was recently shown to inhibit the microtubule-organizing activity of the centrosome. Here, we show that CDC6 is localized to the spindle from pro-metaphase I (MI) to MII stages of oocytes, and it plays important roles at two critical steps of oocyte meiotic maturation. CDC6 depletion facilitated the G2/M transition (germinal vesicle breakdown [GVBD]) through regulation of Cdh1 and cyclin B1 expression and CDK1 (CDC2) phosphorylation in a GVBD-inhibiting culture system containing milrinone. Furthermore, GVBD was significantly decreased after knockdown of cyclin B1 in CDC6-depleted oocytes, indicating that the effect of CDC6 loss on GVBD stimulation was mediated, at least in part, by raising cyclin B1. Knockdown of CDC6 also caused abnormal localization of γ-tubulin, resulting in defective spindles, misaligned chromosomes, cyclin B1 accumulation, and spindle assembly checkpoint (SAC) activation, leading to significant pro-MI/MI arrest and PB1 extrusion failure. These phenotypes were also confirmed by time-lapse live cell imaging analysis. The results indicate that CDC6 is indispensable for maintaining G2 arrest of meiosis and functions in G2/M checkpoint regulation in mouse oocytes. Moreover, CDC6 is also a key player regulating meiotic spindle assembly and metaphase-to-anaphase transition in meiotic oocytes.


Assuntos
Proteínas de Ciclo Celular/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Meiose/genética , Proteínas Nucleares/genética , Oócitos/crescimento & desenvolvimento , Anáfase/genética , Animais , Centrossomo , Feminino , Pontos de Checagem da Fase M do Ciclo Celular/genética , Metáfase/genética , Camundongos , Oócitos/metabolismo , Fuso Acromático/genética
13.
Science ; 365(6451): 353-360, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31273069

RESUMO

Histone modifications regulate gene expression and development. To address how they are reprogrammed in human early development, we investigated key histone marks in human oocytes and early embryos. Unlike that in mouse oocytes, the permissive mark trimethylated histone H3 lysine 4 (H3K4me3) largely exhibits canonical patterns at promoters in human oocytes. After fertilization, prezygotic genome activation (pre-ZGA) embryos acquire permissive chromatin and widespread H3K4me3 in CpG-rich regulatory regions. By contrast, the repressive mark H3K27me3 undergoes global depletion. CpG-rich regulatory regions then resolve to either active or repressed states upon ZGA, followed by subsequent restoration of H3K27me3 at developmental genes. Finally, by combining chromatin and transcriptome maps, we revealed transcription circuitry and asymmetric H3K27me3 patterning during early lineage specification. Collectively, our data unveil a priming phase connecting human parental-to-zygotic epigenetic transition.


Assuntos
Blastocisto/metabolismo , Epigênese Genética , Código das Histonas , Histonas/metabolismo , Zigoto/metabolismo , Animais , Cromatina/metabolismo , Ilhas de CpG , Humanos , Camundongos , Oócitos/metabolismo , Transcriptoma
14.
FASEB J ; 33(1): 1179-1187, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30110177

RESUMO

N6-methyladenosine (m6A) is the most prevalent and reversible internal modification of mammalian messenger and noncoding RNAs mediated by specific m6A writer, reader, and eraser proteins. As an m6A writer, the methyltransferase-like 3-methyltransferase-like 14 (METTL14)-Wilms tumor 1-associated protein complex dynamically regulates m6A modification and plays important roles in diverse biologic processes. However, our knowledge about the complete functions of this RNA methyltransferase complex, the contributions of each component to the methylation, and their effects on different biologic pathways are still limited. By using both in vivo and in vitro models, we here report that METTL14 is indispensable for postimplantation embryonic development by facilitating the conversion from naive to primed state of the epiblast. Depletion of Mettl14 leads to conspicuous embryonic growth retardation from embryonic d 6.5, mainly as a result of resistance to differentiation, which further leads to embryonic lethality early in gestation. Our data highlight the critical function of METTL14 as an m6A modification regulator in orchestrating early mouse embryogenesis.-Meng, T.-G., Lu, X., Guo, L., Hou, G.-M., Ma, X.-S., Li, Q.-N., Huang, L., Fan, L.-H., Zhao, Z.-H., Ou, X.-H., OuYang, Y.-C., Schatten, H., Li, L., Wang, Z.-B., Sun, Q.-Y. Mettl14 is required for mouse postimplantation development by facilitating epiblast maturation.


Assuntos
Desenvolvimento Embrionário/genética , Camadas Germinativas/citologia , Metiltransferases/fisiologia , Adenosina/análogos & derivados , Adenosina/genética , Animais , Sistemas CRISPR-Cas , Feminino , Perfilação da Expressão Gênica , Genes Letais , Metiltransferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , RNA Mensageiro/genética
15.
Cell Death Differ ; 26(5): 969-980, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30154445

RESUMO

Zygotic chromatin undergoes extensive reprogramming immediately after fertilization. It is generally accepted that maternal factors control this process. However, little is known about the underlying mechanisms. Here we report that maternal RAD9A, a key protein in DNA damage response pathway, is involved in post-zygotic embryo development, via a mouse model with conditional depletion of Rad9a alleles in oocytes of primordial follicles. Post-zygotic losses originate from delayed zygotic chromatin decondensation after depletion of maternal RAD9A. Pronucleus formation and DNA replication of most mutant zygotes are therefore deferred, which subsequently trigger the G2/M checkpoint and arrest development of most mutant zygotes. Delayed zygotic chromatin decondensation could also lead to increased reabsorption of post-implantation mutant embryos. In addition, our data indicate that delayed zygotic chromatin decondensation may be attributed to deferred epigenetic modification of histone in paternal chromatin after fertilization, as fertilization and resumption of secondary meiosis in mutant oocytes were both normal. More interestingly, most mutant oocytes could not support development beyond one-cell stage after parthenogenetic activation. Therefore, RAD9A may also play an important role in maternal chromatin reprogramming. In summary, our data reveal an important role of RAD9A in zygotic chromatin reprogramming and female fertility.


Assuntos
Proteínas de Ciclo Celular/genética , Cromatina/genética , Desenvolvimento Embrionário/genética , Epigênese Genética , Animais , Núcleo Celular/genética , Replicação do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Histonas/genética , Masculino , Meiose/genética , Camundongos , Oócitos/crescimento & desenvolvimento , Espermatozoides/crescimento & desenvolvimento , Zigoto/crescimento & desenvolvimento
16.
J Reprod Dev ; 63(5): 481-488, 2017 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-28690291

RESUMO

Geminin plays a critical role in cell cycle regulation by regulating DNA replication and serves as a transcriptional molecular switch that directs cell fate decisions. Spermatogonia lacking Geminin disappear during the initial wave of mitotic proliferation, while geminin is not required for meiotic progression of spermatocytes. It is unclear whether geminin plays a role in pre-meiotic DNA replication in later-stage spermatogonia and their subsequent differentiation. Here, we selectively disrupted Geminin in the male germline using the Stra8-Cre/loxP conditional knockout system. Geminin-deficient mice showed atrophic testes and infertility, concomitant with impaired spermatogenesis and reduced sperm motility. The number of undifferentiated spermatogonia and spermatocytes was significantly reduced; the pachytene stage was impaired most severely. Expression of cell proliferation-associated genes was reduced in Gmnnfl/Δ; Stra8-Cre testes compared to in controls. Increased DNA damage, decreased Cdt1, and increased phosphorylation of Chk1/Chk2 were observed in Geminin-deficient germ cells. These results suggest that geminin plays important roles in pre-meiotic DNA replication and subsequent spermatogenesis.


Assuntos
Geminina/genética , Infertilidade Masculina/genética , Meiose/genética , Espermatogênese/genética , Animais , Replicação do DNA/genética , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espermatócitos/fisiologia
17.
Cell Death Dis ; 8(6): e2846, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28569793

RESUMO

The process of follicular development involves communications between oocyte and surrounding granulosa cells. FURIN is a member of the family of proprotein convertases that is involved in the activation of a large number of zymogens and proproteins by cleavage at its recognition motif. To investigate the functions of FURIN in female fertility, furinflox/flox (furfl/fl) mice were crossed with Zp3-Cre mice and Gdf9-Cre, respectively, to achieve oocyte-specific disruption of FURIN. Here we report for the first time that FURIN is dispensable for primordial follicle maintenance and activation but important for early secondary follicular development, as ablation of FURIN in oocytes caused failure of follicle development beyond the type 4 and/or 5a follicles in mutant mice, resulting in increased number of early secondary follicles and the severely decreased number of mature follicles, thus anovulation and infertility. We also found that the developmental arrest of early secondary follicles might be rooted in the loss of the mature form of ADAMTS1 (85-kDa prodomain truncated) and compromised proliferation of granulosa cells in mutant mice. Taken together, our data highlight the importance of FURIN in follicle development beyond the early secondary follicle stage and indicate that compromised FURIN function leads to follicular dysplasia and female infertility in mice.


Assuntos
Proteína ADAMTS1/genética , Furina/genética , Células da Granulosa/enzimologia , Infertilidade Feminina/genética , Oócitos/enzimologia , Proteína ADAMTS1/deficiência , Animais , Comunicação Celular , Pontos de Checagem do Ciclo Celular/genética , Proliferação de Células , Cruzamentos Genéticos , Feminino , Furina/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/patologia , Humanos , Infertilidade Feminina/enzimologia , Infertilidade Feminina/patologia , Masculino , Camundongos , Camundongos Knockout , Oócitos/patologia
18.
J Reprod Dev ; 63(4): 383-388, 2017 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-28458300

RESUMO

LSM family member 14 (LSM14) belongs to the RNA-associated protein (RAP) family that is widely expressed in different species, and whose functions include associating and storing mRNAs. In the present study, we found that LSM14b was essential for oocyte meiotic maturation. Lack of LSM14b caused oocyte meiotic arrest at metaphase, and misalignment of chromosomes, as well as abnormal spindle assembly checkpoint (SAC) and maturation promoting factor (MPF) activation. Cyclin B1 and Cdc20 mRNAs, whose contents changed with LSM14b expression, were likely direct targets of LSM14b. We conclude that LSM14b, by functioning as a container of mRNAs, controls protein expression, and thus regulates the oocyte meiotic maturation process.


Assuntos
Meiose/fisiologia , Oócitos/metabolismo , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Animais , Proteínas Cdc20/genética , Proteínas Cdc20/metabolismo , Ciclina B1/genética , Ciclina B1/metabolismo , Fator Promotor de Maturação/genética , Fator Promotor de Maturação/metabolismo , Mesotelina , Camundongos , Proteínas/genética , RNA Mensageiro/genética , Fuso Acromático/metabolismo
19.
J Reprod Dev ; 63(3): 319-324, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28367932

RESUMO

Casein kinase 2 (CK2) is a highly conserved, ubiquitously expressed serine/threonine protein kinase with hundreds of substrates. The role of CK2 in the G2/M transition of oocytes, zygotes, and 2-cell embryos was studied in mouse by enzyme activity inhibition using the specific inhibitor 4, 5, 6, 7-tetrabromobenzotriazole (TBB). Zygotes and 2-cell embryos were arrested at G2 phase by TBB treatment, and DNA damage was increased in the female pronucleus of arrested zygotes. Further developmental ability of arrested zygotes was reduced, but that of arrested 2-cell embryos was not affected after releasing from inhibition. By contrast, the G2/M transition in oocytes was not affected by TBB. These results indicate that CK2 activity is essential for mitotic G2/M transition in early embryos but not for meiotic G2/M transition in oocytes.


Assuntos
Caseína Quinase II/metabolismo , Embrião de Mamíferos/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular , Oócitos/fisiologia , Zigoto/enzimologia , Animais , Caseína Quinase II/antagonistas & inibidores , Feminino , Camundongos Endogâmicos ICR , Triazóis
20.
Sci Rep ; 6: 38574, 2016 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-27991495

RESUMO

Kif2a is a member of the Kinesin-13 microtubule depolymerases. Here, we report the expression, subcellular localization and functions of Kif2a during mouse oocyte meiotic maturation. Immunoblotting analysis showed that Kif2a was gradually increased form GV to the M I stages, and then decreased slightly at the M II stage. Confocal microscopy identified that Kif2a localized to the meiotic spindle, especially concentrated at the spindle poles and inner centromeres in metaphase and translocated to the midbody at telophase. Kif2a depletion by siRNA microinjection generated severely defective spindles and misaligned chromosomes, reduced microtubule depolymerization, which led to significant pro-M I/M Iarrest and failure of first polar body (PB1) extrusion. Kif2a-depleted oocytes were also defective in spindle pole localization of γ-tubulin and showed spindle assembly checkpoint (SAC) protein Bub3 at the kinetochores even after 10 hr extended culture. These results demonstrate that Kif2a may act as a microtubule depolymerase, regulating microtubule dynamics, spindle assembly and chromosome congression, and thus cell cycle progression during mouse oocyte meiotic maturation.


Assuntos
Cinesinas/metabolismo , Meiose , Oócitos/citologia , Oócitos/metabolismo , Proteínas Repressoras/metabolismo , Fuso Acromático/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Segregação de Cromossomos/efeitos dos fármacos , Cromossomos de Mamíferos/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Meiose/efeitos dos fármacos , Camundongos Endogâmicos ICR , Nocodazol/farmacologia , Oócitos/efeitos dos fármacos , Paclitaxel/farmacologia , Corpos Polares/metabolismo , Fuso Acromático/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Tubulina (Proteína)/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA