Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38426493

RESUMO

Early gestational loss occurs in approximately 20% of all clinically recognized human pregnancies and is an important cause of morbidity. Either embryonic or maternal defects can cause loss, but a functioning and receptive uterine endometrium is crucial for embryo implantation. We report that the switch/sucrose nonfermentable (SWI/SNF) remodeling complex containing polybromo-1 (PBRM1) and Brahma-related gene 1 (BRG1) is essential for implantation of the embryonic blastocyst on the wall of the uterus in mice. Although preimplantation development is unaffected, conditional ablation of Pbrm1 in uterine stromal cells disrupts progesterone pathways and uterine receptivity. Heart and neural crest derivatives expressed 2 (Hand2) encodes a basic helix-loop-helix (bHLH) transcription factor required for embryo implantation. We identify an enhancer of the Hand2 gene in stromal cells that requires PBRM1 for epigenetic histone modifications/coactivator recruitment and looping with the promoter. In Pbrm1cKO mice, perturbation of chromatin assembly at the promoter and enhancer sites compromises Hand2 transcription, adversely affects fibroblast growth factor signaling pathways, prevents normal stromal-epithelial crosstalk, and disrupts embryo implantation. The mutant female mice are infertile and provide insight into potential causes of early pregnancy loss in humans.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina , Animais , Feminino , Humanos , Camundongos , Gravidez , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cromatina/metabolismo , Implantação do Embrião/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Útero
2.
Trends Genet ; 40(3): 238-249, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38262796

RESUMO

Maternal mRNAs accumulate during egg growth and must be judiciously degraded or translated to ensure successful development of mammalian embryos. In this review we integrate recent investigations into pathways controlling rapid degradation of maternal mRNAs during the maternal-to-zygotic transition. Degradation is not indiscriminate, and some mRNAs are selectively protected and rapidly translated after fertilization for reprogramming the zygotic genome during early embryogenesis. Oocyte specific cofactors and pathways have been illustrated to control different futures of maternal mRNAs. We discuss mechanisms that control the fate of maternal mRNAs during late oogenesis and after fertilization. Issues to be resolved in current maternal mRNA research are described, and future research directions are proposed.


Assuntos
Desenvolvimento Embrionário , RNA Mensageiro Estocado , Animais , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo , Desenvolvimento Embrionário/genética , Oócitos , Oogênese/genética , Zigoto , Regulação da Expressão Gênica no Desenvolvimento/genética , Mamíferos/genética
3.
Dev Cell ; 58(18): 1716-1732.e8, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37714160

RESUMO

Early mammalian development occurs during embryo transit of the female reproductive tract. Transport is orchestrated by secreted oviduct fluid, unidirectional beating of epithelial cilia, and smooth muscle contractions. Using gene-edited mice, we document that conditional disruption of a component of the SWI/SNF chromatin remodeling complex in smooth muscle cells prevents transport through the oviduct without perturbing embryogenesis. Analysis with RNA sequencing (RNA-seq), transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunocleavage sequencing (ChIC-seq), and pharmacologic rescue experiments implicated prostaglandin signaling pathways. In comparison with controls, gene-edited mice had compromised chromatin accessibility at enhancer/promoters of Ptgs2, Pla2g16, Pla2r1, and Ptger3 (EP3) as well as decreased enhancer-promoter interactive looping critical for Ptgs2 (aka Cox-2) expression in a SWI/SNF complex-dependent manner. Treatment of wild-type mice with prostaglandin inhibitors phenocopied the genetically induced defect.


Assuntos
Montagem e Desmontagem da Cromatina , Prostaglandinas , Feminino , Animais , Camundongos , Ciclo-Oxigenase 2/genética , Músculo Liso , Cromatina , Mamíferos
4.
Genes Dev ; 37(9-10): 418-431, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37257918

RESUMO

Translation of maternal mRNAs is detected before transcription of zygotic genes and is essential for mammalian embryo development. How certain maternal mRNAs are selected for translation instead of degradation and how this burst of translation affects zygotic genome activation remain unknown. Using gene-edited mice, we document that the oocyte-specific eukaryotic translation initiation factor 4E family member 1b (eIF4E1b) is the regulator of maternal mRNA expression that ensures subsequent reprogramming of the zygotic genome. In oocytes, eIF4E1b binds to transcripts encoding translation machinery proteins, chromatin remodelers, and reprogramming factors to promote their translation in zygotes and protect them from degradation. The protein products are thought to establish an open chromatin landscape in one-cell zygotes to enable transcription of genes required for cleavage stage development. Our results define a program for rapid resetting of the zygotic epigenome that is regulated by maternal mRNA expression and provide new insights into the mammalian maternal-to-zygotic transition.


Assuntos
RNA Mensageiro Estocado , Zigoto , Animais , Camundongos , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Oócitos , Biossíntese de Proteínas , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo , Zigoto/metabolismo
5.
Aging Cell ; 19(3): e13102, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32074399

RESUMO

In female mammals, the size of the initially established primordial follicle (PF) pool within the ovaries determines the reproductive lifespan of females. Interestingly, the establishment of the PF pool is accompanied by a remarkable programmed oocyte loss for unclear reasons. Although apoptosis and autophagy are involved in the process of oocyte loss, the underlying mechanisms require substantial study. Here, we identify a new role of lysine-specific demethylase 1 (LSD1) in controlling the fate of oocytes in perinatal mice through regulating the level of autophagy. Our results show that the relatively higher level of LSD1 in fetal ovaries sharply reduces from 18.5 postcoitus (dpc). Meanwhile, the level of autophagy increases while oocytes are initiating programmed death. Specific disruption of LSD1 resulted in significantly increased autophagy and obviously decreased oocyte number compared with the control. Conversely, the oocyte number is remarkably increased by the overexpression of Lsd1 in ovaries. We further demonstrated that LSD1 exerts its role by regulating the transcription of p62 and affecting autophagy level through its H3K4me2 demethylase activity. Finally, in physiological conditions, a decrease in LSD1 level leads to an increased level of autophagy in the oocyte when a large number of oocytes are being lost. Collectively, LSD1 may be one of indispensible epigenetic molecules who protects oocytes against preterm death through repressing the autophagy level in a time-specific manner. And epigenetic modulation contributes to programmed oocyte death by regulating autophagy in mice.


Assuntos
Apoptose/genética , Autofagia/genética , Histona Desmetilases/metabolismo , Oócitos/metabolismo , Proteína Sequestossoma-1/metabolismo , Transcrição Gênica/genética , Animais , Células Cultivadas , Feminino , Desenvolvimento Fetal/genética , Histona Desmetilases/genética , Histonas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Folículo Ovariano/metabolismo , Gravidez , Proteína Sequestossoma-1/genética , Transdução de Sinais/genética , Transfecção
6.
J Mol Cell Biol ; 12(3): 230-244, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-31282930

RESUMO

Establishment of the primordial follicle (PF) pool is pivotal for the female reproductive lifespan; however, the mechanism of primordial folliculogenesis is poorly understood. Here, the transcription factor SP1 was shown to be essential for PF formation in mice. Our results showed that SP1 is present in both oocytes and somatic cells during PF formation in the ovary. Knockdown of Sp1 expression, especially in pregranulosa cells, significantly suppressed nest breakdown, oocyte apoptosis, and PF formation, suggesting that SP1 expressed by somatic cells functions in the process of primordial folliculogenesis. We further demonstrated that SP1 governs the recruitment and maintenance of Forkhead box L2-positive (FOXL2+) pregranulosa cells using an Lgr5-EGFP-IRES-CreERT2 (Lgr5-KI) reporter mouse model and a FOXL2+ cell-specific knockdown model. At the molecular level, SP1 functioned mainly through manipulation of NOTCH2 expression by binding directly to the promoter of the Notch2 gene. Finally, consistent with the critical role of granulosa cells in follicle survival in vitro, massive loss of oocytes in Sp1 knockdown ovaries was evidenced before puberty after the ovaries were transplanted under the renal capsules. Conclusively, our results reveal that SP1 controls the establishment of the ovarian reserve by regulating pregranulosa cell development in the mammalian ovary.


Assuntos
Células da Granulosa/citologia , Células da Granulosa/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Folículo Ovariano/metabolismo , Animais , Apoptose/genética , Biomarcadores , Suscetibilidade a Doenças , Feminino , Imunofluorescência , Proteína Forkhead Box L2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Oócitos/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Insuficiência Ovariana Primária/etiologia , Insuficiência Ovariana Primária/metabolismo , Regiões Promotoras Genéticas , Receptor Notch2/genética , Receptor Notch2/metabolismo , Maturidade Sexual/genética , Transdução de Sinais
7.
Cell Death Dis ; 10(6): 438, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31165749

RESUMO

The placenta, responsible for the nutrient and gas exchange between the mother and fetus, is pivotal for successful pregnancy. It has been shown that Rbpj, the core transcriptional mediator of Notch signaling pathway, is required for normal placentation in mice. However, it remains largely unclear how Rbpj signaling in different placental compartments coordinates with other important regulators to ensure normal placental morphogenesis. In this study, we found that systemic deletion of Rbpj led to abnormal chorioallantoic morphogenesis and defective trophoblast differentiation in the ectoplacental cone (EPC). Employing mouse models with selective deletion of Rbpj in the allantois versus trophoblast, combining tetraploid aggregation assay, we demonstrated that allantois-expressed Rbpj is essential for chorioallantoic attachment and subsequent invagination of allantoic blood vessels into the chorionic ectoderm. Further studies uncovered that allantoic Rbpj regulates chorioallantoic fusion and morphogenesis via targeting Vcam1 in a Notch-dependent manner. Meanwhile, we also revealed that trophoblast-expressed Rbpj in EPC facilitates Mash2's transcriptional activity, promoting the specification of Tpbpα-positive trophoblasts, which differentiate into trophoblast subtypes responsible for interstitial and endovascular invasion at the later stage of placental development. Collectively, our study further shed light on the molecular network governing placental development and functions, highlighting the necessity of a spatiotemporal coordination of Rbpj signaling for normal placental morphogenesis.


Assuntos
Alantoide/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Morfogênese/genética , Placenta/metabolismo , Placentação/genética , Trofoblastos/metabolismo , Alantoide/crescimento & desenvolvimento , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Córion/crescimento & desenvolvimento , Córion/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Células HEK293 , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/genética , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
8.
J Clin Invest ; 128(1): 175-189, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202468

RESUMO

Natural and synthetic progestogens have been commonly used to prevent recurrent pregnancy loss in women with inadequate progesterone secretion or reduced progesterone sensitivity. However, the clinical efficacy of progesterone and its analogs for maintaining pregnancy is variable. Additionally, the underlying cause of impaired endometrial progesterone responsiveness during early pregnancy remains unknown. Here, we demonstrated that uterine-selective depletion of BMI1, a key component of the polycomb repressive complex-1 (PRC1), hampers uterine progesterone responsiveness and derails normal uterine receptivity, resulting in implantation failure in mice. We further uncovered genetic and biochemical evidence that BMI1 interacts with the progesterone receptor (PR) and the E3 ligase E6AP in a polycomb complex-independent manner and regulates the PR ubiquitination that is essential for normal progesterone responsiveness. A close association of aberrantly low endometrial BMI1 expression with restrained PR responsiveness in women who had previously had a miscarriage indicated that the role of BMI1 in endometrial PR function is conserved in mice and in humans. In addition to uncovering a potential regulatory mechanism of BMI1 that ensures normal endometrial progesterone responsiveness during early pregnancy, our findings have the potential to help clarify the underlying causes of spontaneous pregnancy loss in women.


Assuntos
Implantação do Embrião , Endométrio/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Progesterona/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Progesterona/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Aborto Espontâneo/patologia , Animais , Endométrio/patologia , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Gravidez , Proteínas Proto-Oncogênicas/genética , Receptores de Progesterona/genética , Ubiquitina-Proteína Ligases/genética
9.
Theriogenology ; 104: 55-61, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28822241

RESUMO

Uterine receptivity is defined as a state when the uterine milieu is favorable for blastocyst implantation and it can only last for a limited time period. In this study, by utilizing the embryo transfer model, it was observed that a portion of blastocysts could initiate implantation even when transferred beyond the timing of normal uterine receptivity, while their mid-gestational embryo development exhibited severe retardation, suggesting that the uterine status beyond the normal implantation window is unconducive for normal implantation. We further performed microarray analysis to explore the molecular basis that distinguishes the normal and defective uterine receptivity. A total of 229 genes was found to be differentially expressed, and a large amount of them were epithelium-expressing genes and responsive to progesterone signaling.


Assuntos
Implantação do Embrião/fisiologia , Regulação da Expressão Gênica/fisiologia , Transcriptoma , Útero/metabolismo , Animais , Transferência Embrionária , Feminino , Masculino , Camundongos , Análise Serial de Proteínas
10.
Cell Death Differ ; 24(12): 2013-2021, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28731465

RESUMO

Formation of secretary endometrial glands in the uterus known as adenogenesis is a typical process of branching morphogenesis involving dynamic epithelial growth and differentiation. Unsuccessful adenogenesis often leads to female infertility. However, it remains largely unexplored so far regarding the epigenetic machinery governing normal endometrial gland formation. Here, we demonstrated that PR-Set7, an epigenetic regulator for H4K20me1 modification, was extensively expressed in the postnatal uteri, and its conditional deletion resulted in a complete lack of endometrial glands and infertility in mice. Subsequent analysis revealed that uterine PR-Set7 deficiency abolishes the dynamic endometrial epithelial population growth during the short span of gland formation from postnatal days 3 to 9. This markedly reduced epithelial population growth in PR-Set7-null mutant uteri is well associated with DNA damage accumulation and massive apoptotic death in the epithelium, due to blockade of 53BP1 recruitment to DNA damage sites upon reduced levels of H4K20me1/2. Using PgrCre/+/Rosa26DTA/+ mouse line and postnatal progesterone injection mouse model, we further confirmed that an impaired epithelial cell population growth either by inducing epithelial death in the diphtheria toxin-A (DTA)-mouse model or attenuating epithelial growth upon postnatal progesterone treatment similarly hampers uterine adenogenesis. Collectively, we establish here a novel 'epithelial population growth threshold' model for successful gland development. Besides further shedding light on the regulatory machinery governing uterine gland formation, our findings raise a safety concern on progesterone supplementation to prevent preterm birth in women bearing a female fetus, as exogenous progesterone may hamper uterine adenogenesis via attenuating epithelial population growth.


Assuntos
Endométrio/crescimento & desenvolvimento , Histona-Lisina N-Metiltransferase/deficiência , Animais , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Reparo do DNA , Endométrio/metabolismo , Feminino , Fertilidade/fisiologia , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Camundongos , Morfogênese , Transfecção , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
11.
Proc Natl Acad Sci U S A ; 114(18): 4816-4821, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28424251

RESUMO

Estrogen and progesterone coupled with locally produced signaling molecules are essential for embryo implantation. However, the hierarchical landscape of the molecular pathways that governs this process remains largely unexplored. Here we show that the protein tyrosine phosphatase Shp2, a positive transducer of RTK signaling, is predominately localized in the nuclei in the periimplantation mouse uterus. Uterine-specific deletion of Shp2 exhibits reduced progesterone receptor (PR) expression and progesterone resistance, which derails normal uterine receptivity, leading to complete implantation failure in mice. Notably, the PR expression defects are attributed to the limited estrogen receptor α (ERα) activation in uterine stroma. Further analysis reveals that nuclear Shp2, rather than cytosolic Shp2, promotes the ERα transcription activity. This function is achieved by enhancing the Src kinase-mediated ERα tyrosine phosphorylation, which facilitates ERα binding to Pgr promoter in an ERK-independent manner in periimplantation uteri. Besides uncovering a regulatory mechanism, this study could be clinically relevant to dysfunctional ERα-caused endometrial disorders in women.


Assuntos
Núcleo Celular/enzimologia , Implantação do Embrião/fisiologia , Receptor alfa de Estrogênio/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Útero/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Núcleo Celular/genética , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Fosforilação/fisiologia , Gravidez , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Quinases da Família src/genética
12.
Drug Deliv ; 24(1): 651-659, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28283003

RESUMO

Nano-scaled materials have been proved to be ideal DNA carriers for transgene. Bacterial magnetic particles (BMPs) help to reduce the toxicity of polyethylenimine (PEI), an efficient gene-transferring agent, and assist tissue transgene ex vivo. Here, the effectiveness of the BMP-PEI complex-conjugated foreign DNAs (BPDs) in promoting testes-mediated gene transfer (TMGT) in mouse was compared with that of liposome-conjugated foreign DNAs. The results proved that through testes injection, the clusters of BPDs successfully reached the cytoplasm and the nuclear of spermatogenesis cell, and expressed in testes of transgene founder mice. Additionally, the ratio of founder mice obtained from BPDs (88%) is about 3 times higher than the control (25%) (p < 0.05). Interestingly, the motility of sperms recovered from epididymis of the founder mice from BPD group were significantly improved, as compared with the control (p < 0.01). Based on classic breeding, the ratio of transgene mice within the first filial was significantly higher in BPDs compared with the control (73.8% versus 11.6%, p < 0.05). TMGT in this study did not produce visible histological changes in the testis. In conclusion, nano-scaled BPDs could be an alternative strategy for efficiently producing transgene mice in vivo.


Assuntos
Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Magnetossomos/genética , Magnetospirillum/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Transgenes , Animais , Efeito Fundador , Regulação da Expressão Gênica , Genótipo , Iminas/química , Iminas/metabolismo , Lipossomos , Magnetossomos/metabolismo , Magnetospirillum/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Polietilenos/química , Polietilenos/metabolismo , Motilidade dos Espermatozoides , Espermatogênese , Testículo/citologia , Fatores de Tempo
13.
Cell Death Dis ; 8(3): e2662, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28277543

RESUMO

In mammals, oocytes are arrested at the diplotene stage of meiosis I until the pre-ovulatory luteinizing hormone (LH) surge triggers meiotic resumption through the signals in follicular granulosa cells. In this study, we show that the estradiol (E2)-estrogen receptors (ERs) system in follicular granulosa cells has a dominant role in controlling oocyte meiotic resumption in mammals. We found that the expression of ERs was controlled by gonadotropins under physiological conditions. E2-ERs system was functional in maintaining oocyte meiotic arrest by regulating the expression of natriuretic peptide C and natriuretic peptide receptor 2 (NPPC/NPR2), which was achieved through binding to the promoter regions of Nppc and Npr2 genes directly. In ER knockout mice, meiotic arrest was not sustained by E2 in most cumulus-oocyte complexes in vitro and meiosis resumed precociously in pre-ovulatory follicles in vivo. In human granulosa cells, similar conclusions are reached that ER levels were controlled by gonadotropins and E2-ERs regulated the expression of NPPC/NPR2 levels. In addition, our results revealed that the different regulating patterns of follicle-stimulating hormone and LH on ER levels in vivo versus in vitro determined their distinct actions on oocyte maturation. Taken together, these findings suggest a critical role of E2-ERs system during oocyte meiotic progression and may propose a novel approach for oocyte in vitro maturation treatment in clinical practice.


Assuntos
Meiose/genética , Peptídeo Natriurético Tipo C/genética , Receptores do Fator Natriurético Atrial/genética , Receptores de Estradiol/genética , Receptores de Estrogênio/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/metabolismo , Humanos , Técnicas de Maturação in Vitro de Oócitos , Hormônio Luteinizante/genética , Camundongos Knockout , Oócitos/crescimento & desenvolvimento , Oogênese/genética , Folículo Ovariano/crescimento & desenvolvimento
14.
Sheng Li Xue Bao ; 68(4): 435-54, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27546504

RESUMO

Estrogen receptor (ER) and progesterone receptor (PR) are two important members of steroid receptors family, an evolutionarily conserved family of transcription factors. Upon binding to their ligands, ER and PR enter cell nucleus to interact with specific DNA element in the context of chromatin to initiate the transcription of diverse target genes, which largely depends on the timely recruitment of a wide range of cofactors. Moreover, the interactions between steroid hormones and their respective receptors also trigger post-translational modifications on these receptors to fine-tune their transcriptional activities. Besides the well-known phosphorylation modifications on tyrosine and serine/threonine residues, recent studies have identified several other covalent modifications, such as ubiquitylation and sumoylation. These post-translational modifications of steroid receptors affect its stability, subcellular localization, and/or cofactor recruitment; eventually influence the duration and extent of transcriptional activation. This review is to focus on the recent research progress on the transcriptional activation of nuclear ER and PR as well as their physiological functions in early pregnancy, which may help us to better understand related female reproductive diseases.


Assuntos
Ativação Transcricional , Ligantes , Fosforilação , Receptores de Estrogênio , Receptores de Progesterona , Sumoilação
15.
J Cell Sci ; 129(11): 2202-12, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27084580

RESUMO

Ovarian follicles are the basic functional units of female reproduction in the mammalian ovary. We show here that the protein a disintegrin and metalloproteinase domain 10 (ADAM10), a cell surface sheddase, plays an indispensable role in controlling primordial follicle formation by regulating the recruitment of follicle supporting cells in mice. We demonstrate that suppressing ADAM10 in vitro or deletion of Adam10 in vivo disrupts germline cyst breakdown and primordial follicle formation. Using a cell lineage tracing approach, we show that ADAM10 governs the recruitment of ovarian follicle cells by regulating the differentiation and proliferation of LGR5-positive follicle supporting progenitor cells. By detecting the development of FOXL2-positive pregranulosa cells, we found that inhibiting ADAM10 reduced the number of FOXL2-positive cells in perinatal ovaries. Furthermore, inhibiting ADAM10 suppressed the activation of Notch signaling, and blocking Notch signaling also disrupted the recruitment of follicle progenitor cells. Taken together, these results show that ADAM10-Notch signaling in ovarian somatic cells governs the primordial follicle formation by controlling the development of ovarian pregranulosa cells. The proper recruitment of ovarian follicle supporting cells is essential for establishment of the ovarian reserve in mice.


Assuntos
Proteína ADAM10/metabolismo , Células da Granulosa/metabolismo , Organogênese , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/metabolismo , Células Germinativas/metabolismo , Células da Granulosa/citologia , Camundongos , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
16.
Development ; 143(10): 1778-87, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27013242

RESUMO

Physiologically, the size of the primordial follicle pool determines the reproductive lifespan of female mammals, while its establishment largely depends on a process of germline cyst breakdown during the perinatal period. The mechanisms regulating this process are poorly understood. Here we demonstrate that c-Jun amino-terminal kinase (JNK) signaling is crucial for germline cyst breakdown and primordial follicle formation. JNK was specifically localized in oocytes and its activity increased as germline cyst breakdown progressed. Importantly, disruption of JNK signaling with a specific inhibitor (SP600125) or knockdown technology (Lenti-JNK-shRNAs) resulted in significantly suppressed cyst breakdown and primordial follicle formation in cultured mouse ovaries. Our results show that E-cadherin is intensely expressed in germline cysts, and that its decline is necessary for oocyte release from the cyst. However, inhibition of JNK signaling leads to aberrantly enhanced localization of E-cadherin at oocyte-oocyte contact sites. WNT4 expression is upregulated after SP600125 treatment. Additionally, similar to the effect of SP600125 treatment, WNT4 overexpression delays cyst breakdown and is accompanied by abnormal E-cadherin expression patterns. In conclusion, our results suggest that JNK signaling, which is inversely correlated with WNT4, plays an important role in perinatal germline cyst breakdown and primordial follicle formation by regulating E-cadherin junctions between oocytes in mouse ovaries.


Assuntos
Caderinas/metabolismo , Células Germinativas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Organogênese , Folículo Ovariano/metabolismo , Animais , Feminino , Técnicas de Silenciamento de Genes , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Proteólise , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Wnt4/metabolismo
17.
Sci Rep ; 5: 13735, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26334131

RESUMO

Human endometrium decidualization, which involves endometrial stromal proliferation and differentiation, is a prerequisite for embryo implantation, thus successful pregnancy. The Forkhead Box M1 (FoxM1), previously known as HNF-3, HFH-11, MPP2, Win, and Trident, is a transcriptional factor that plays crucial roles in cell proliferation and cell cycle progression. However, the molecular mechanism of FoxM1 during human endometrial decidualization remains unexplored. In this study, we first found FoxM1 is dynamically expressed in human endometrium during menstrual cycle. Employing a human endometrial stromal cell (HESC) line, we then demonstrated that FoxM1 inhibition downregulates cyclin B1 expression, delaying G2/M phase transition during HESC proliferation. Additionally, loss of FoxM1 expression blocks the differentiation of HESCs in response to estrogen, progesterone, and dbcAMP. Applying chromatin immunoprecipitation (ChIP) technique and luciferase assay, we further approved that FoxM1 can transcriptionally active signal transducer and activator of transcription 3 (STAT3), ensuring normal HESC differentiation. Besides enriching our knowledge on molecular basis underlying stromal decidualization, these findings help to shed light on the potential molecular causes for the endometrial disorders in humans.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/citologia , Endométrio/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator de Transcrição STAT3/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Proteína Forkhead Box M1 , Humanos , Ciclo Menstrual/fisiologia , Células Estromais/citologia , Células Estromais/fisiologia
18.
Toxicol Ind Health ; 27(4): 341-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21078691

RESUMO

Alcoholic liver disease (ALD) is a major medical complication of drinking alcohol, and commonly accompanied with hepatic iron overload and liver injuries. Oxidative stress plays an important role in pathogenesis of ALD and also leads to iron-metabolic disorders. In this study, the effects of vitamin C (Vc) on iron metabolism-related genes expression and liver protection from drinking in mice were investigated. Twenty-four male kunming mice were divided into four groups (six mice per group): control (water drinking); alcohol group (20% alcohol drinking), alcohol + low Vc group (adding 50 mg/kg Vc daily) and alcohol + high Vc group (adding 100 mg/kg Vc daily). All these mice were sacrificed after 7 days. Vc can ameliorate the increase of sera alanine aminotransferase (ALT) activity and hepatic iron overload of drinking alcohol in mice. Vc increases the expression of the iron-regulated hormone hepcidin and decreases transferrin receptor 1 (TfR1) expression in liver. Vc also down-regulates the expression of ferroportin 1 (Fpn1) in the intestine and decreases the iron release to blood. In conclusion, Vc ameliorated the alcoholic liver injuries through regulating the iron metabolism-related genes expression.


Assuntos
Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Etanol/toxicidade , Sobrecarga de Ferro/tratamento farmacológico , Ferro/metabolismo , Hepatopatias Alcoólicas/tratamento farmacológico , Alanina Transaminase/sangue , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Western Blotting , Modelos Animais de Doenças , Etanol/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hepcidinas , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/metabolismo , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...