Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Materials (Basel) ; 16(13)2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37445010

RESUMEN

In this paper, we examined the parameters of the Mooney-Rivlin model based on the effects of alternative aging and sea corrosion tests for natural rubber bearings and rubber materials in seawater. The model parameters for rubber material used in natural rubber bearings were determined using the least-squares method. Meanwhile, the time-varying law formula of the Mooney-Rivlin model parameters of rubber were fitted, and the fitting and calculated values were compared. Both fitting values and calculated values coincide with each other well. Then, the rubber material parameters were predicted based on the calculated results and combined with nonlinear auto-regressive (NAR). The predicted values were compared with both the fitting and calculated values. The average deviations between predicted and fitting values for C10 and C01 were 2.6% and 5.1%, respectively, and average deviations between predicted and calculated values for C10 and C01 were 5.2% and 4.1%. Compared results show that the predicted values are in good agreement with both the fitting and calculated values; meanwhile, the proposed time-varying law formula of the Mooney-Rivlin model parameters of rubber material have been well verified.

2.
Elife ; 112022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36445738

RESUMEN

Recent studies show that pre-follicular mouse oogenesis takes place in germline cysts, highly conserved groups of oogonial cells connected by intercellular bridges that develop as nurse cells as well as an oocyte. Long studied in Drosophila and insect gametogenesis, female germline cysts acquire cytoskeletal polarity and traffic centrosomes and organelles between nurse cells and the oocyte to form the Balbiani body, a conserved marker of polarity. Mouse oocyte development and nurse cell dumping are supported by dynamic, cell-specific programs of germline gene expression. High levels of perinatal germ cell death in this species primarily result from programmed nurse cell turnover after transfer rather than defective oocyte production. The striking evolutionary conservation of early oogenesis mechanisms between distant animal groups strongly suggests that gametogenesis and early embryonic development in vertebrates and invertebrates share even more in common than currently believed.


Asunto(s)
Drosophila , Oogénesis , Femenino , Embarazo , Ratones , Animales , Células Germinativas , Oocitos , Gametogénesis
3.
Cell ; 185(14): 2576-2590.e12, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35623357

RESUMEN

Mouse germline cysts, on average, develop into six oocytes supported by 24 nurse cells that transfer cytoplasm and organelles to generate a Balbiani body. We showed that between E14.5 and P5, cysts periodically activate some nurse cells to begin cytoplasmic transfer, which causes them to shrink and turnover within 2 days. Nurse cells die by a programmed cell death (PCD) pathway involving acidification, similar to Drosophila nurse cells, and only infrequently by apoptosis. Prior to initiating transfer, nurse cells co-cluster by scRNA-seq with their pro-oocyte sisters, but during their final 2 days, they cluster separately. The genes promoting oocyte development and nurse cell PCD are upregulated, whereas the genes that repress transfer, such as Tex14, and oocyte factors, such as Nobox and Lhx8, are under-expressed. The transferred nurse cell centrosomes build a cytocentrum that establishes a large microtubule aster in the primordial oocyte that organizes the Balbiani body, defining the earliest oocyte polarity.


Asunto(s)
Linaje de la Célula , Quistes , Oocitos , Animales , Apoptosis , Aumento de la Célula , Quistes/genética , Quistes/metabolismo , Citoplasma/metabolismo , Drosophila melanogaster , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Oocitos/citología , Oocitos/metabolismo , Ovario/citología , Ovario/embriología , Ovario/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(33): 20015-20026, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32759216

RESUMEN

We sequenced more than 52,500 single cells from embryonic day 11.5 (E11.5) postembryonic day 5 (P5) gonads and performed lineage tracing to analyze primordial follicles and wave 1 medullar follicles during mouse fetal and perinatal oogenesis. Germ cells clustered into six meiotic substages, as well as dying/nurse cells. Wnt-expressing bipotential precursors already present at E11.5 are followed at each developmental stage by two groups of ovarian pregranulosa (PG) cells. One PG group, bipotential pregranulosa (BPG) cells, derives directly from bipotential precursors, expresses Foxl2 early, and associates with cysts throughout the ovary by E12.5. A second PG group, epithelial pregranulosa (EPG) cells, arises in the ovarian surface epithelium, ingresses cortically by E12.5 or earlier, expresses Lgr5, but delays robust Foxl2 expression until after birth. By E19.5, EPG cells predominate in the cortex and differentiate into granulosa cells of quiescent primordial follicles. In contrast, medullar BPG cells differentiate along a distinct pathway to become wave 1 granulosa cells. Reflecting their separate somatic cellular lineages, second wave follicles were ablated by diptheria toxin treatment of Lgr5-DTR-EGFP mice at E16.5 while first wave follicles developed normally and supported fertility. These studies provide insights into ovarian somatic cells and a resource to study the development, physiology, and evolutionary conservation of mammalian ovarian follicles.


Asunto(s)
Células de la Granulosa/citología , Ratones/embriología , Folículo Ovárico/embriología , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Células de la Granulosa/metabolismo , Ratones/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Embarazo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
BMC Biol ; 16(1): 73, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976179

RESUMEN

BACKGROUND: In mammalian females, progressive activation of dormant primordial follicles in adulthood is crucial for the maintenance of the reproductive lifespan. Misregulated activation of primordial follicles leads to various ovarian diseases, such as premature ovarian insufficiency (POI). Although recent studies have revealed that several functional genes and pathways, such as phosphoinositide 3-kinase (PI3K) signaling, play roles in controlling the activation of primordial follicles, our understanding of the molecular networks regulating the activation progress is still incomplete. RESULTS: Here, we identify a new role for cell division cycle 42 (CDC42) in regulating the activation of primordial follicles in mice. Our results show that CDC42 expression increases in oocytes during the activation of primordial follicles in the ovary. Disruption of CDC42 activity with specific inhibitors or knockdown of Cdc42 expression significantly suppresses primordial follicle activation in cultured mouse ovaries. Conversely, the follicle activation ratio is remarkably increased by overexpression of CDC42 in ovaries. We further demonstrate that CDC42 governs the process of primordial follicle activation by binding to phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta (p110ß) and regulating the expression levels of PTEN in oocytes. Finally, we extend our study to potential clinical applications and show that a short-term in vitro treatment with CDC42 activators could significantly increase the activation rates of primordial follicles in both neonatal and adult mouse ovaries. CONCLUSION: Our results reveal that CDC42 controls the activation of primordial follicles in the mammalian ovary and that increasing the activity of CDC42 with specific activators might improve the efficiency of in vitro activation approaches, opening avenues for infertility treatments.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Oocitos/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Ovario/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteína de Unión al GTP cdc42/fisiología , Animales , Femenino , Ratones , Proteína de Unión al GTP cdc42/genética
6.
Sci Rep ; 6: 23972, 2016 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-27050391

RESUMEN

The size of the primordial follicle pool determines the reproductive potential of mammalian females, and establishment of the pool is highly dependent on specific genes expression. However, the molecular mechanisms by which the essential genes are regulated coordinately to ensure primordial follicle assembly remain a mystery. Here, we show that the small GTPase Rac1 plays an indispensable role in controlling the formation of primordial follicles in mouse ovary. Employing fetal mouse ovary organ culture system, we demonstrate that disruption of Rac1 retarded the breakdown of germline cell cysts while Rac1 overexpression accelerated the formation of primordial follicles. In addition, in vivo inhibitor injection resulted in the formation of multi-oocyte follicles. Subsequent investigation showed that Rac1 induced nuclear import of STAT3 by physical binding. In turn, nuclear STAT3 directly activated the transcription of essential oocyte-specific genes, including Jagged1, GDF9, BMP15 and Nobox. Further, GDF9 and BMP15 regulated the translation of Notch2 via mTORC1 activation in pregranulosa cells. Overexression or addition of Jagged1, GDF9 and BMP15 not only reversed the effect of Rac1 disruption, but also accelerated primordial follicle formation via Notch2 signaling activation. Collectively, these results indicate that Rac1 plays important roles as a key regulator in follicular assembly.


Asunto(s)
Proteína Morfogenética Ósea 15/metabolismo , Factor 9 de Diferenciación de Crecimiento/metabolismo , Proteína Jagged-1/metabolismo , Folículo Ovárico/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Transporte Activo de Núcleo Celular/genética , Aminoquinolinas/farmacología , Animales , Western Blotting , Proteína Morfogenética Ósea 15/genética , Núcleo Celular/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor 9 de Diferenciación de Crecimiento/genética , Inmunohistoquímica , Proteína Jagged-1/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Modelos Genéticos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Técnicas de Cultivo de Órganos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/embriología , Unión Proteica , Pirimidinas/farmacología , Interferencia de ARN , Receptor Notch2/genética , Receptor Notch2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
7.
J Cell Sci ; 129(11): 2202-12, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27084580

RESUMEN

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.


Asunto(s)
Proteína ADAM10/metabolismo , Células de la Granulosa/metabolismo , Organogénesis , Receptores Notch/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Diferenciación Celular , Proliferación Celular , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Células Germinativas/metabolismo , Células de la Granulosa/citología , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Células Madre/metabolismo
8.
Development ; 143(10): 1778-87, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27013242

RESUMEN

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.


Asunto(s)
Cadherinas/metabolismo , Células Germinativas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Organogénesis , Folículo Ovárico/metabolismo , Animales , Femenino , Técnicas de Silenciamiento del Gen , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína Wnt4/metabolismo
9.
Reproduction ; 151(2): 105-15, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26554027

RESUMEN

The reserve of primordial follicles determines the reproductive ability of the female mammal over its reproductive life. The primordial follicle is composed of two types of cells: oocytes and surrounding pre-granulosa cells. However, the underlying mechanism regulating primordial follicle assembly is largely undefined. In this study, we found that gap junction communication (GJC) established between the ovarian cells in the perinatal mouse ovary may be involved in the process. First, gap junction structures between the oocyte and surrounding pre-granulosa cells appear at about 19.0 dpc (days post coitum). As many as 12 gap junction-related genes are upregulated at birth, implying that a complex communication may exist between ovarian cells, because specifically silencing the genes of individual gap junction proteins, such as Gja1, Gja4 or both, has no influence on primordial follicle assembly. On the other hand, non-specific blockers of GJC, such as carbenoxolone (CBX) and 18α-glycyrrhetinic acid (AGA), significantly inhibit mouse primordial follicle assembly. We proved that the temporal window for establishment of GJC in the fetal ovary is from 19.5 dpc to 1 dpp (days postpartum). In addition, the expression of ovarian somatic cell (OSC)-specific genes, such as Notch2, Foxl2 and Irx3, was negatively affected by GJC blockers, whereas oocyte-related genes, such as Ybx2, Nobox and Sohlh1, were hardly affected, implying that the establishment of GJC during this period may be more important to OSCs than to oocytes. In summary, our results indicated that GJC involves in the mouse primordial follicle assembly process at a specific temporal window that needs Notch signaling cross-talking.


Asunto(s)
Comunicación Celular , Uniones Comunicantes/fisiología , Células de la Granulosa/fisiología , Oocitos/fisiología , Folículo Ovárico/fisiología , Parto/fisiología , Animales , Animales Recién Nacidos , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Conexinas/antagonistas & inhibidores , Conexinas/genética , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/genética , Técnicas de Silenciamiento del Gen , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Oocitos/citología , Oocitos/efectos de los fármacos , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Embarazo , ARN Interferente Pequeño/farmacología , Proteína alfa-4 de Unión Comunicante
10.
PLoS One ; 10(6): e0129643, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26076381

RESUMEN

In mammals, the primordial follicle pool represents the entire reproductive potential of a female. The transforming growth factor-ß (TGF-ß) family member activin (ACT) contributes to folliculogenesis, although the exact mechanism is not known. The role of FST288, the strongest ACT-neutralizing isoform of follistatin (FST), during cyst breakdown and primordial follicle formation in the fetal mice ovary was assessed using an in vitro culture system. FST was continuously expressed in the oocytes as well as the cuboidal granulosa cells of growing follicles in perinatal mouse ovaries. Treatment with FST288 delayed germ cell nest breakdown, particularly near the periphery of the ovary, and dramatically decreased the percentage of primordial follicles. In addition, there was a dramatic decrease in proliferation of granulosa cells and somatic cell expression of Notch signaling was impaired. In conclusion, FST288 impacts germ cell nest breakdown and primordial follicle assembly by inhibiting somatic cell proliferation.


Asunto(s)
Quistes/patología , Folistatina/metabolismo , Células Germinativas/citología , Células de la Granulosa/citología , Folículo Ovárico/citología , Ovario/citología , Animales , Apoptosis , Proliferación Celular , Quistes/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Células de la Granulosa/metabolismo , Masculino , Ratones , Folículo Ovárico/metabolismo , Ovario/metabolismo , Transducción de Señal
11.
J Cell Physiol ; 230(12): 2998-3008, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25953201

RESUMEN

In the mammalian ovaries, the primordial follicle pool determines the reproductive capability over the lifetime of a female. The primordial follicle is composed of two cell members, namely the oocyte and the pre-granulosa cells that encircle the oocyte. However, it is unclear what factors are involved in the reorganization of the two distinct cells into one functional unit. This study was performed to address this issue. Firstly, in an in vitro reconstruction system, dispersed ovarian cells from murine fetal ovaries at 19.0 days post coitum (dpc) reassembled into follicle-like structures, independent of the physical distance between the cells, implying that either oocytes or ovarian somatic cells (OSCs) were motile. We then carried out a series of transwell assay experiments, and determined that it was in fact 19.0 dpc OSCs (as opposed to oocytes), which exhibited a significant chemotactic response to both fetal bovine serum and oocytes themselves. We observed that S100A8, a multi-functional chemokine, may participate in the process as it is mainly expressed in oocytes within the cysts/plasmodia. S100A8 significantly promoted the number of migrating OSCs by 2.5 times in vitro, of which 66.9% were FOXL2 protein-positive cells, implying that the majority of motile OSCs were pre-granulosa cells. In addition, an S100A8-specific antibody inhibited the formation of follicle-like reconstruction cell mass in vitro. And, the primordial follicle formation was reduced when S100a8-specific siRNA was applied onto in vitro cultured 17.5 dpc ovary. Therefore, S100A8 could be a chemokine of oocyte origin, which attracts OSCs to form the primordial follicles.


Asunto(s)
Calgranulina A/metabolismo , Quimiotaxis , Células de la Granulosa/metabolismo , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Animales , Calgranulina A/genética , Células Cultivadas , Técnicas de Cocultivo , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Folículo Ovárico/embriología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
12.
Development ; 142(2): 343-51, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25503411

RESUMEN

In mammalian ovaries, a fixed population of primordial follicles forms during the perinatal stage and the oocytes contained within are arrested at the dictyate stage of meiotic prophase I. In the current study, we provide evidence that the level of cyclic AMP (cAMP) in oocytes regulates oocyte meiotic prophase I and primordial folliculogenesis in the perinatal mouse ovary. Our results show that the early meiotic development of oocytes is closely correlated with increased levels of intra-oocyte cAMP. Inhibiting cAMP synthesis in fetal ovaries delayed oocyte meiotic progression and inhibited the disassembly and degradation of synaptonemal complex protein 1. In addition, inhibiting cAMP synthesis in in vitro cultured fetal ovaries prevented primordial follicle formation. Finally, using an in situ oocyte chromosome analysis approach, we found that the dictyate arrest of oocytes is essential for primordial follicle formation under physiological conditions. Taken together, these results suggest a role for cAMP in early meiotic development and primordial follicle formation in the mouse ovary.


Asunto(s)
AMP Cíclico/metabolismo , Profase Meiótica I/fisiología , Oocitos/metabolismo , Organogénesis/fisiología , Folículo Ovárico/embriología , Análisis de Varianza , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Microdisección , Interferencia de ARN , Radioinmunoensayo
13.
J Biol Chem ; 289(12): 8299-311, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24515103

RESUMEN

Physiologically, only a few primordial follicles are activated to enter the growing follicle pool each wave. Recent studies in knock-out mice show that early follicular activation depends on signaling from the tuberous sclerosis complex, the mammalian target of rapamycin complex 1 (mTORC1), phosphatase and tensin homolog deleted on chromosome 10, and phosphatidylinositol 3-kinase (PI3K) pathways. However, the manner in which these pathways are normally regulated, and whether or not TGF-ß acts on them are poorly understood. So, this study aims to identify whether or not TGF-ß acts on the process. Ovary organ culture experiments showed that the culture of 18.5 days post-coitus (dpc) ovaries with TGF-ß1 reduced the total population of oocytes and activated follicles, accelerated oocyte growth was observed in ovaries treated with TGF-ßR1 inhibitor 2-(5-chloro-2-fluorophenyl)pteridin-4-yl]pyridin-4-yl-amine (SD208) compared with control ovaries, the down-regulation of TGF-ßR1 gene expression also activated early primordial follicle oocyte growth. We further showed that there was dramatically more proliferation of granulosa cells in SD208-treated ovaries and less proliferation in TGF-ß1-treated ovaries. Western blot and morphological analyses indicated that TGF-ß signaling manipulated primordial follicle growth through tuberous sclerosis complex/mTORC1 signaling in oocytes, and the mTORC1-specific inhibitor rapamycin could partially reverse the stimulated effect of SD208 on the oocyte growth and decreased the numbers of growing follicles. In conclusion, our results suggest that TGF-ß signaling plays an important physiological role in the maintenance of the dormant pool of primordial follicles, which functions through activation of p70 S6 kinase 1 (S6K1)/ribosomal protein S6 (rpS6) signaling in mouse ovaries.


Asunto(s)
Oocitos/crecimiento & desarrollo , Ovario/citología , Ovario/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Proliferación Celular , Regulación hacia Abajo , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Oocitos/metabolismo , Oocitos/ultraestructura , Técnicas de Cultivo de Órganos , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Folículo Ovárico/ultraestructura , Ovario/metabolismo , Ovario/ultraestructura , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/genética
14.
PLoS One ; 8(4): e61947, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23596531

RESUMEN

The mechanical method to isolate preantral follicle has been reported for many years. However, the culture systems in vitro are still unstable. The aim of this study was to analyze the effect of the culture system of mice preantral follicles on the follicular development in vitro. The results showed that the 96-well plate system was the most effective method for mice follicle development in vitro (volume change: 51.71%; survival rate: 89%, at day 4). Follicle-stimulating hormone (FSH) and Thyroid hormone (TH) are important for normal follicular development and dysregulation of hormones are related with impaired follicular development. To determine the effect of hormone on preantral follicular development, we cultured follicle with hormones in the 96-well plate culture system and found that FSH significantly increased preantral follicular growth on day 4. The FSH-induced growth action was markedly enhanced by T3 although T3 was ineffective alone. We also demonstrated by QRT-PCR that T3 significantly enhanced FSH-induced up-regulation of Xiap mRNA level. Meanwhile, Bad, cell death inducer, was markedly down-regulated by the combination of hormones. Moreover, QRT-PCR results were also consistent with protein regulation which detected by Western Blotting analysis. Taken together, the findings of the present study demonstrate that 96-well plate system is an effective method for preantral follicle development in vitro. Moreover, these results provide insights on the role of thyroid hormone in increasing FSH-induced preantral follicular development, which mediated by up-regulating Xiap and down-regulating Bad.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Triyodotironina/farmacología , Animales , Técnicas de Cultivo de Célula , Femenino , Ratones , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/citología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína Letal Asociada a bcl/metabolismo
15.
PLoS One ; 7(8): e42406, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22870326

RESUMEN

Although the effects of Gonadotropin on ovarian physiology have been known for many decades, its action on glucose uptake in the rat ovary remained poorly understood. Evidence also suggests that glucose uptake is mediated by a number of glucose transporter proteins (Glut). Therefore, we examined the rat ovary for the presence of Glut1-4 and blood glucose level after eCG (equine chorionic gonadotropin) and anti-eCG antiserum treatment. All of the glucose transports were present in the ovarian oocyte, granulosa cells and theca cells in different stage follicles. The expression of Glut in ovary was up-regulated by eCG, however, anti-eCG antiserum reversed eCG action. Western blot analysis also demonstrated the content of Glut1 was higher in eCG treatment group compared with anti-eCG antiserum and control group. The same tendency was shown in other glut isoforms. Moreover, there were no significant difference between the anti-eCG antiserum and control group. In additional, the level of serum glucose in eCG treatment group was significantly higher than others, which is similar with glut expression pattern. High glucose level in blood is correlated with increased expression of glucose transporter proteins in rat ovary. Meanwhile, anti-eCG antiserum increased granulosa cell apoptosis in antral follicle compared with those in eCG group. Our observations provide potential explanation for the effects of Glut on follicular development in rat ovary and a role for eCG in the regulation of ovarian glucose uptake.


Asunto(s)
Apoptosis/efectos de los fármacos , Transportador de Glucosa de Tipo 1/metabolismo , Glucosa/metabolismo , Gonadotropinas/farmacología , Folículo Ovárico/metabolismo , Animales , Apoptosis/fisiología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Femenino , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...