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.
Int J Gen Med ; 15: 179-189, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35023956

RESUMO

OBJECTIVE: The present study aims to explore the association between phthalate exposure and the risk of gestational diabetes mellitus (GDM). MATERIALS AND METHODS: A total of 11 plasticizer metabolites were measured in patient morning urine using high-performance liquid chromatography. Furthermore, fasting blood glucose and fasting insulin were detected in first-trimester blood samples. The chemical concentration was described using the median, the metabolite concentration difference between the GDM and control groups was compared using the bootstrap method, and the correlations of the fasting blood glucose, fasting insulin, insulin resistance index, and phthalic acid ester (PAE) metabolites were analyzed using Spearman correlation analysis. The multivariate logistic regression model and predictive probability map were performed to help assess the linearity and nature of any dose-response relationship. RESULTS: Of the 224 women recruited for the present study, 200 met the inclusion criteria. Their measured outcomes and biomonitoring data were examined for the presence of chemicals. The results showed that the patients in the GDM group had higher mono-(2-ethylhexyl) phthalate (MEHP) and methylerythritol cyclophosphane concentrations in their bodies than the patients in the control group. Statistically significant MEHP-GDM associations were also observed (P < 0.001). The GDM and MEHP dose-response relationships were different among pregnant women aged <35 years and those aged >35 years (P < 0.001). Furthermore, gestational age >28 weeks exhibited similar changes to those aged ≤28 weeks (P = 0.059). CONCLUSION: The findings of the present study add to the growing body of evidence supporting phthalate exposure as a GDM risk factor.

3.
Cell Death Differ ; 27(6): 1952-1964, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31819157

RESUMO

Protein phosphatase 6 (PP6) is a member of the PP2A-like subfamily, which plays a critical role in many fundamental cellular processes. We recently reported that PP6 is essential for female fertility. Here, we report that PP6 is involved in meiotic recombination and that germ cell-specific deletion of PP6 by Stra8-Cre causes defective spermatogenesis. The PP6-deficient spermatocytes were arrested at the pachytene stage and defects in DSB repair and crossover formation were observed, indicating that PP6 facilitated meiotic double-stranded breaks (DSB) repair. Further investigations revealed that depletion of PP6 in the germ cells affected chromatin relaxation, which was dependent on MAPK pathway activity, consequently preventing programmed DSB repair factors from being recruited to proper positions on the chromatin. Taken together, our results demonstrate that PP6 has an important role in meiotic recombination and male fertility.


Assuntos
Fosfoproteínas Fosfatases/fisiologia , Espermatócitos , Espermatogênese , Animais , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estágio Paquíteno , Espermatócitos/citologia , Espermatócitos/metabolismo
4.
Cell Prolif ; 53(1): e12726, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31755150

RESUMO

OBJECTIVES: In humans, non-obstructive azoospermia (NOA) is a major cause of male infertility. However, the aetiology of NOA is largely unknown. Previous studies reported that protein CK2ß was abundantly and broadly expressed in spermatogenic cells. Here, we investigate whether protein CK2ß participates in spermatogenesis. MATERIALS AND METHODS: In this study, we separated spermatogenic cells using STA-PUT velocity sedimentation, analysed the expression pattern of protein CK2ß by immunoblotting, specifically deleted Ck2ß gene in early-stage spermatogenic cells by crossing Ck2ßfl mice with Stra8-Cre+ mice and validated the knockout efficiency by quantitative RT-PCR and immunoblotting. The phenotypes of Ck2ßfl/Δ ;SCre+ mice were studied by immunohistochemistry and immunofluorescence. The molecular mechanisms of male germ cell development arrest were elucidated by immunoblotting and TUNEL assay. RESULTS: Ablation of Ck2ß gene triggered excessive germ cell apoptosis, germ cell development arrest, azoospermia and male infertility. Inactivation of Ck2ß gene caused distinctly reduced expression of Ck2α' gene and CK2α' protein. CONCLUSIONS: Ck2ß is a vital gene for germ cell survival and male fertility in mice.


Assuntos
Apoptose/genética , Azoospermia , Caseína Quinase II/deficiência , Células Germinativas , Animais , Azoospermia/enzimologia , Azoospermia/genética , Azoospermia/patologia , Caseína Quinase II/metabolismo , Deleção de Genes , Células Germinativas/enzimologia , Células Germinativas/patologia , Masculino , Camundongos , Camundongos Knockout
5.
Cell Cycle ; 18(4): 395-412, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30730241

RESUMO

PKCßI, a member of the classical protein kinase C family, plays key roles in regulating cell cycle transition. Here, we report the expression, localization and functions of PKCßI in mouse oocyte meiotic maturation. PKCßI and p-PKCßI (phosphor-PKCßI) were expressed from germinal vesicle (GV) stage to metaphase II (MII) stage. Confocal microscopy revealed that PKCßI was localized in the GV and evenly distributed in the cytoplasm after GV breakdown (GVBD), and it was concentrated at the midbody at telophase in meiotic oocytes. While, p-PKCßI was concentrated at the spindle poles at the metaphase stages and associated with midbody at telophase. Depletion of PKCßI by specific siRNA injection resulted in defective spindles, accompanied with spindle assembly checkpoint activation, metaphase I arrest and failure of first polar body (PB1) extrusion. Live cell imaging analysis also revealed that knockdown of PKCßI resulted in abnormal spindles, misaligned chromosomes, and meiotic arrest of oocytes arrest at the Pro-MI/MI stage. PKCßI depletion did not affect the G2/M transition, but its overexpression delayed the G2/M transition through regulating Cyclin B1 level and Cdc2 activity. Our findings reveal that PKCßI is a critical regulator of meiotic cell cycle progression in oocytes. Abbreviations: PKC, protein kinase C; COC, cumulus-oocyte complexes; GV, germinal vesicle; GVBD, germinal vesicle breakdown; Pro-MI, first pro-metaphase; MI, first metaphase; Tel I, telophase I; MII, second metaphase; PB1, first polar body; SAC, spindle assembly checkpoint.


Assuntos
Metáfase/genética , Corpos Polares/metabolismo , Proteína Quinase C beta/genética , Proteína Quinase C beta/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Cromossomos/metabolismo , Ciclina B1/metabolismo , Citoplasma/metabolismo , Feminino , Pontos de Checagem da Fase M do Ciclo Celular/genética , Camundongos , Camundongos Endogâmicos ICR , Microinjeções , Plasmídeos/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Fuso Acromático/metabolismo , Telófase/genética
6.
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
7.
Aging (Albany NY) ; 10(7): 1586-1596, 2018 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-30036861

RESUMO

After ovulation, metaphase II oocytes undergo a time-dependent deterioration in vivo or in vitro, which is referred to as postovulatory oocyte aging, a process during which a series of deleterious molecular and cellular changes occur. In this study, we found that short-term injection of resveratrol (3,5,4'-trihydroxystilbene) effectively ameliorated oxidative stress-induced damage in postovulatory oocyte aging of middle-aged mice in vivo. Resveratrol induced changes that delayed the aging-induced oocyte deterioration including the elevated expression of the anti-aging molecule Sirtuin 1 (SIRT1); it reduced intracellular reactive oxygen species (ROS) level, and improved mitochondria function. In addition, these beneficial changes may also help to prevent apoptosis. Taken together, our data suggest that resveratrol can effectively protect against postovulatory oocyte aging in vivo primarily by preventing ROS production.


Assuntos
Senescência Celular/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Ovulação/fisiologia , Resveratrol/farmacologia , Envelhecimento , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio , Sirtuína 1/metabolismo
8.
Cell Cycle ; 17(12): 1471-1486, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29965788

RESUMO

Oocyte is arrested at metaphase of the second meiosis until fertilization switching on [Ca2+]i oscillations. Oocyte activation inefficiency is the most challenging problem for failed fertilization and embryonic development. Mitochondrial function and intracellular [Ca2+]i oscillations are two critical factors for the oocyte's developmental potential. We aimed to understand the possible correlation between mitochondrial function and [Ca2+]i oscillations in oocytes. To this end, mitochondrial uncoupler CCCP which damages mitochondrial function and two small molecule mitochondrial agonists, L-carnitine (LC) and BGP-15, were used to examine the regulation of [Ca2+]i by mitochondrial functions. With increasing CCCP concentrations, [Ca2+]i oscillations were gradually diminished and high concentrations of CCCP led to oocyte death. LC enhanced mitochondrial membrane potential and [Ca2+]i oscillations and even improved the damage induced by CCCP, however, BGP-15 had no beneficial effect on oocyte activation. We have found that mitochondrial function plays a vital role in the generation of [Ca2+]i oscillations in oocytes, and thus mitochondria may interact with the ER to generate [Ca2+]i oscillations during oocyte activation. Improvement of mitochondrial functions with small molecules can be expected to improve oocyte activation and embryonic development in infertile patients without invasive micromanipulation.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Meiose/fisiologia , Mitocôndrias/metabolismo , Oócitos/metabolismo , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/metabolismo , Carnitina/metabolismo , Ciclo Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos ICR , Oogênese/fisiologia , Oximas/metabolismo , Piperidinas/metabolismo , Gravidez
9.
Cell Death Dis ; 9(5): 508, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29725001

RESUMO

Premature ovarian failure (POF), a major cause of female infertility, is a complex disorder, but the molecular mechanisms underlying the disorder are only poorly understood. Here we report that protein kinase CK2 contributes to maintaining follicular survival through PI3K/AKT pathway and DNA damage response pathway. Targeted deletion of CK2ß in mouse oocytes from the primordial follicle stage resulted in female infertility, which was attributed to POF incurring by massive follicle atresia. Downregulated PI3K/AKT signaling was found after CK2ß deletion, indicated by reduced level of phosphorylated AKT (S473, T308, and S129) and altered AKT targets related to cell survival. Further studies discovered that CK2ß-deficient oocytes showed enhanced γH2AX signals, indicative of accumulative unrepaired DSBs, which activated CHK2-dependant p53 and p63 signaling. The suppressed PI3K/AKT signaling and failed DNA damage response signaling probably contribute to large-scale oocyte loss and eventually POF. Our findings provide important new clues for elucidating the mechanisms underlying follicle atresia and POF.


Assuntos
Caseína Quinase II/genética , Atresia Folicular/genética , Infertilidade Feminina/genética , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Insuficiência Ovariana Primária/genética , Animais , Caseína Quinase II/deficiência , Sobrevivência Celular , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Quebras de DNA de Cadeia Dupla , Modelos Animais de Doenças , Feminino , Atresia Folicular/metabolismo , Regulação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Camundongos , Camundongos Knockout , Oócitos/patologia , Folículo Ovariano/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
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
11.
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
12.
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
13.
Oncotarget ; 7(52): 86350-86358, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27861152

RESUMO

Spermatogenesis in testes requires precise spermatogonia differentiation. Spermatocytes lacking the Rad9a gene are arrested in pachytene prophase, implying a possible role for RAD9A in spermatogonia differentiation. However, numerous RAD9A-positive pachytene spermatocytes are still observed in mouse testes following Rad9a excision using the Stra8-Cre system, and it is unclear whether Rad9a deletion in spermatogonia interrupts differentiation. Here, we generated a mouse model in which Rad9a was specifically deleted in spermatogonial stem cells (SSCs) using Cre recombinase expression driven by the germ cell-specific Vasa promoter. Adult Rad9a-null male mice were infertile as a result of completely blocked spermatogonia differentiation. No early spermatocytes were detected in mutant testicular cords of 9-day-old mice. Mutant spermatogonia were prone to apoptosis, although proliferation rates were unaffected. Rad9a deletion also resulted in malformation of seminiferous tubules, in which cells assembled irregularly into clusters, and malformation led to testicular cord disruption. Our findings suggest that Rad9a is indispensable for spermatogonia differentiation and testicular development in mice.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Espermatogônias/citologia , Animais , Apoptose , Diferenciação Celular , RNA Helicases DEAD-box/análise , Proteínas de Ligação a DNA/análise , Infertilidade Masculina/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Fatores de Transcrição/análise
14.
Biol Reprod ; 92(4): 97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25761595

RESUMO

The mammalian oocyte undergoes two rounds of asymmetric cell divisions during meiotic maturation and fertilization. Acentric spindle positioning and cortical polarity are two major factors involved in asymmetric cell division, both of which are thought to depend on the dynamic interaction between myosin II and actin filaments. Myosin light chain kinase (MLCK), encoded by the Mylk1 gene, could directly phosphorylate and activate myosin II. To determine whether MLCK was required for oocyte asymmetric division, we specifically disrupted the Mylk1 gene in oocytes by Cre-loxP conditional knockout system. We found that Mylk1 mutant female mice showed severe subfertility. Unexpectedly, contrary to previously reported in vitro findings, our data showed that oocyte meiotic maturation including spindle organization, polarity establishment, homologous chromosomes separation, and polar body extrusion were not affected in Mylk1(fl/fl);GCre(+) females. Follicular development, ovulation, and early embryonic development up to compact morula occurred normally in Mylk1(fl/fl);GCre(+) females, but deletion of MLCK caused delayed morula-to-blastocyst transition. More than a third of embryos were at morula stage at 3.5 Days Postcoitum in vivo. The delayed embryos could develop further to early blastocyst stage in vitro on Day 4 when most control embryos reached expanded blastocysts. Our findings provide evidence that MLCK is linked to timely blastocyst formation, though it is dispensable for oocyte meiotic maturation.


Assuntos
Blastocisto/fisiologia , Fertilidade/genética , Mórula/fisiologia , Quinase de Cadeia Leve de Miosina/genética , Quinase de Cadeia Leve de Miosina/fisiologia , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Animais , Cromossomos de Mamíferos/genética , Feminino , Fertilidade/fisiologia , Fertilização/genética , Deleção de Genes , Infertilidade/genética , Infertilidade/fisiopatologia , Meiose/genética , Camundongos , Camundongos Endogâmicos C57BL , Corpos Polares/fisiologia , Gravidez , Fuso Acromático/genética , Fuso Acromático/fisiologia
15.
Biol Reprod ; 91(1): 19, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24899574

RESUMO

Ppp2r1a encodes the scaffold subunit Aalpha of protein phosphatase 2A (PP2A), which is an important and ubiquitously expressed serine threonine phosphatase family and plays a critical role in many fundamental cellular processes. To identify the physiological role of PP2A in female germ cell meiosis, we selectively disrupted Ppp2r1a expression in oocytes by using the Cre-Loxp conditional knockout system. Here we report for the first time that oocyte-specific deletion of Ppp2r1a led to severe female subfertility without affecting follicle survival, growth, and ovulation. PP2A-Aalpha was essential for regulating oocyte meiotic maturation because depletion of PP2A-Aalpha facilitated germinal vesicle breakdown, causing elongation of the MII spindle and precocious separation of sister chromatids. The resulting eggs had high risk of aneuploidy, though they could be fertilized, leading to defective embryonic development and thus subfertility. Our findings provide strong evidence that PP2A-Aalpha within the oocyte plays an indispensable role in oocyte meiotic maturation, though it is dispensable for folliculogenesis in the mouse ovary.


Assuntos
Fertilidade/fisiologia , Meiose/fisiologia , Oócitos/metabolismo , Proteína Fosfatase 2/metabolismo , Subunidades Proteicas/metabolismo , Animais , Feminino , Camundongos , Camundongos Knockout , Oogênese/fisiologia , Ovulação/genética , Ovulação/metabolismo , Proteína Fosfatase 2/genética
16.
Reprod Biol Endocrinol ; 12: 29, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24721882

RESUMO

BACKGROUND: The adverse effects on offspring of diabetic and/or obese mothers can be passed to the next generation. However, the mechanisms behind this are still unclear. Epigenetics may play a key role during this process. METHODS: To confirm the hypothesis, we investigated the DNA methylation of several imprinted genes in spermatozoa of offspring from diabetic and/or obese mothers utilizing streptozotocin (STZ)- and high-fat-diet (HFD)-induced mouse models. RESULTS: We found that the DNA methylation of Peg3 was significantly increased in spermatozoa of offspring of obese mothers compared to that in spermatozoa of offspring of normal mothers. The DNA methylation of H19 was significantly higher in spermatozoa of offspring of diabetic mothers than that in spermatozoa of offspring of non-diabetic mothers. CONCLUSIONS: These results indicate that pre-gestational diabetes and/or obesity can alter DNA methylation in offspring spermatozoa.


Assuntos
Metilação de DNA/fisiologia , Diabetes Mellitus Experimental/metabolismo , Obesidade/metabolismo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Espermatozoides/metabolismo , Animais , Diabetes Mellitus Experimental/complicações , Feminino , Masculino , Bem-Estar Materno , Camundongos , Obesidade/complicações , Gravidez , Distribuição Aleatória
17.
Environ Health Perspect ; 122(2): 159-64, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24316659

RESUMO

BACKGROUND: Maternal obesity has adverse effects on oocyte quality, embryo development, and the health of the offspring. OBJECTIVES: To understand the underlying mechanisms responsible for the negative effects of maternal obesity, we investigated the DNA methylation status of several imprinted genes and metabolism-related genes. METHODS: Using a high-fat-diet (HFD)-induced mouse model of obesity, we analyzed the DNA methylation of several imprinted genes and metabolism-related genes in oocytes from control and obese dams and in oocytes and liver from their offspring. Analysis was performed using combined bisulfite restriction analysis (COBRA) and bisulfite sequencing. RESULTS: DNA methylation of imprinted genes in oocytes was not altered in either obese dams or their offspring; however, DNA methylation of metabolism-related genes was changed. In oocytes of obese mice, the DNA methylation level of the leptin (Lep) promoter was significantly increased and that of the Ppar-α promoter was reduced. Increased methylation of Lep and decreased methylation of Ppar-α was also observed in the liver of female offspring from dams fed the high-fat diet (OHFD). mRNA expression of Lep and Ppar-α was also significantly altered in the liver of these OHFD. In OHFD oocytes, the DNA methylation level of Ppar-α promoter was increased. CONCLUSIONS: Our results indicate that DNA methylation patterns of several metabolism-related genes are changed not only in oocytes of obese mice but also in oocytes and liver of their offspring. These data may contribute to the understanding of adverse effects of maternal obesity on reproduction and health of the offspring.


Assuntos
Metilação de DNA/fisiologia , Dieta Hiperlipídica/efeitos adversos , Padrões de Herança/genética , Fígado/fisiologia , Obesidade/complicações , Obesidade/etiologia , Oócitos/fisiologia , Animais , Sequência de Bases , Metilação de DNA/genética , Feminino , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA
18.
Biol Reprod ; 88(5): 117, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23515675

RESUMO

Maternal diabetes has adverse effects not only on oocyte quality but also on embryo development. However, it is still unknown whether the DNA imprinting in oocytes is altered by diabetes. By using streptozotocin (STZ)-induced and nonobese diabetic (NOD) mouse models we investigated the effect of maternal diabetes on DNA methylation of imprinted genes in oocytes. Mice which were judged as being diabetic 4 days after STZ injection were used for experiments. In superovulated oocytes of diabetic mice, the methylation pattern of Peg3 differential methylation regions (DMR) was affected in a time-dependent manner, and evident demethylation was observed on Day 35 after STZ injection. The expression level of DNA methyltransferases (DNMTs) was also decreased in a time-dependent manner in diabetic oocytes. However, the methylation patterns of H19 and Snrpn DMRs were not significantly altered by maternal diabetes, although there were some changes in Snrpn. In NOD mice, the methylation pattern of Peg3 was similar to that of STZ-induced mice. Embryo development was adversely affected by maternal diabetes; however, no evident imprinting abnormality was observed in oocytes from female offspring derived from a diabetic mother. These results indicate that maternal diabetes has adverse effects on DNA methylation of maternally imprinted gene Peg3 in oocytes of a diabetic female in a time-dependent manner, but methylation in offspring's oocytes is normal.


Assuntos
Metilação de DNA , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Expressão Gênica , Oócitos/metabolismo , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Desenvolvimento Embrionário/genética , Feminino , Impressão Genômica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos NOD , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Centrais de snRNP
19.
Reprod Biol Endocrinol ; 11: 119, 2013 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-24378208

RESUMO

BACKGROUND: Maternal diabetes mellitus not only has severe deleterious effects on fetal development, but also it affects transmission to the next generation. However, the underlying mechanisms for these effects are still not clear. METHODS: We investigated the methylation patterns and expressions of the imprinted genes Peg3, Snrpn, and H19 in mid-gestational placental tissues and on the whole fetus utilizing the streptozotocin (STZ)-induced hyperglycemic mouse model for quantitative analysis of methylation by PCR and quantitative real-time PCR. The protein expression of Peg3 was evaluated by Western blot. RESULTS: We found that the expression of H19 was significantly increased, while the expression of Peg3 was significantly decreased in dpc10.5 placentas of diabetic mice. We further found that the methylation level of Peg3 was increased and that of H19 was reduced in dpc10.5 placentas of diabetic mice. When pronuclear embryos of normal females were transferred to normal/diabetic (NN/ND) pseudopregnant females, the methylation and expression of Peg3 in placentas was also clearly altered in the ND group compared to the NN group. However, when the pronuclear embryos of diabetic female were transferred to normal pesudopregnant female mice (DN), the methylation and expression of Peg3 and H19 in dpc10.5 placentas was similar between the two groups. CONCLUSIONS: We suggest that the effects of maternal diabetes on imprinted genes may primarily be caused by the adverse uterus environment.


Assuntos
Diabetes Mellitus/genética , Desenvolvimento Fetal/genética , Impressão Genômica , Gravidez em Diabéticas/genética , Útero/metabolismo , Animais , Western Blotting , Metilação de DNA , Feminino , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Placenta/metabolismo , Gravidez , Gravidez em Diabéticas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...