Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Commun Biol ; 4(1): 1133, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34580426

RESUMEN

In early mammalian development, the maturation of follicles containing the immature oocytes is an important biological process as the functional oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. Despite recent work demonstrating the regulatory role of mechanical stress in oocyte growth, quantitative studies of ovarian mechanical properties remain lacking both in vivo and ex vivo. In this work, we quantify the material properties of ooplasm, follicles and connective tissues in intact mouse ovaries at distinct stages of follicle development using Brillouin microscopy, a non-invasive tool to probe mechanics in three-dimensional (3D) tissues. We find that the ovarian cortex and its interior stroma have distinct material properties associated with extracellular matrix deposition, and that intra-follicular mechanical compartments emerge during follicle maturation. Our work provides an alternative approach to study the role of mechanics in follicle morphogenesis and might pave the way for future understanding of mechanotransduction in reproductive biology, with potential implications for infertility diagnosis and treatment.


Asunto(s)
Folículo Ovárico/embriología , Folículo Ovárico/crecimiento & desarrollo , Animales , Fenómenos Biomecánicos , Citoplasma , Femenino , Mecanotransducción Celular , Ratones/embriología , Ratones/crecimiento & desarrollo , Microscopía
2.
Science ; 373(6552)2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34437124

RESUMEN

Oocytes mature in a specialized fluid-filled sac, the ovarian follicle, which provides signals needed for meiosis and germ cell growth. Methods have been developed to generate functional oocytes from pluripotent stem cell-derived primordial germ cell-like cells (PGCLCs) when placed in culture with embryonic ovarian somatic cells. In this study, we developed culture conditions to recreate the stepwise differentiation process from pluripotent cells to fetal ovarian somatic cell-like cells (FOSLCs). When FOSLCs were aggregated with PGCLCs derived from mouse embryonic stem cells, the PGCLCs entered meiosis to generate functional oocytes capable of fertilization and development to live offspring. Generating functional mouse oocytes in a reconstituted ovarian environment provides a method for in vitro oocyte production and follicle generation for a better understanding of mammalian reproduction.


Asunto(s)
Células Madre Embrionarias de Ratones/fisiología , Oocitos/fisiología , Oogénesis , Folículo Ovárico/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Desarrollo Embrionario , Femenino , Fertilización In Vitro , Masculino , Mesodermo/citología , Mesodermo/fisiología , Ratones , Ratones Endogámicos ICR , Células Madre Embrionarias de Ratones/citología , Oocitos/citología , Folículo Ovárico/embriología , Folículo Ovárico/fisiología , RNA-Seq , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Transcriptoma
3.
Theranostics ; 11(10): 4992-5009, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754040

RESUMEN

Rationale: Accumulated evidence indicates that environmental plasticizers are a threat to human and animal fertility. Di (2-ethylhexyl) phthalate (DEHP), a plasticizer to which humans are exposed daily, can trigger reproductive toxicity by acting as an endocrine-disrupting chemical. In mammals, the female primordial follicle pool forms the lifetime available ovarian reserve, which does not undergo regeneration once it is established during the fetal and neonatal period. It is therefore critical to examine the toxicity of DEHP regarding the establishment of the ovarian reserve as it has not been well investigated. Methods: The ovarian cells of postnatal pups, following maternal DEHP exposure, were prepared for single cell-RNA sequencing, and the effects of DEHP on primordial follicle formation were revealed using gene differential expression analysis and single-cell developmental trajectory. In addition, further biochemical experiments, including immunohistochemical staining, apoptosis detection, and Western blotting, were performed to verify the dataset results. Results: Using single-cell RNA sequencing, we revealed the gene expression dynamics of female germ cells and granulosa cells following exposure to DEHP in mice. Regarding germ cells: DEHP impeded the progression of follicle assembly and interfered with their developmental status, while key genes such as Lhx8, Figla, and others, strongly evidenced the reduction. As for granulosa cells: DEHP likely inhibited their proliferative activity, and activated the regulation of cell death. Furthermore, the interaction between ovarian cells mediated by transforming growth factor-beta signaling, was disrupted by DEHP exposure, since the expression of GDF9, BMPR1A, and SMAD3 was affected. In addition, DNA damage and apoptosis were elevated in germ cells and/or somatic cells. Conclusion: These findings offer substantial novel insights into the reproductive toxicity of DEHP exposure during murine germ cell cyst breakdown and primordial follicle formation. These results may enhance the understanding of DEHP exposure on reproductive health.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Óvulo/efectos de los fármacos , Plastificantes/toxicidad , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/efectos de los fármacos , Factor 9 de Diferenciación de Crecimiento/genética , Proteínas con Homeodominio LIM/efectos de los fármacos , Proteínas con Homeodominio LIM/genética , Ratones , Folículo Ovárico/citología , Folículo Ovárico/embriología , Folículo Ovárico/metabolismo , Óvulo/metabolismo , RNA-Seq , Análisis de la Célula Individual , Proteína smad3/efectos de los fármacos , Proteína smad3/genética , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
4.
Development ; 147(22)2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33028611

RESUMEN

The Janus-kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the anterior posterior axis of the Drosophila follicle cells. In the anterior, it activates the bone morphogenetic protein (BMP) signaling pathway through expression of the BMP ligand decapentaplegic (dpp). In the posterior, JAK/STAT works with the epidermal growth factor receptor (EGFR) pathway to express the T-box transcription factor midline (mid). Although MID is necessary for establishing the posterior fate of the egg chamber, we show that it is not sufficient to determine a posterior fate. The ETS-transcription factor pointed (pnt) is expressed in an overlapping domain to mid in the follicle cells. This study shows that pnt is upstream of mid and that it is sufficient to induce a posterior fate in the anterior end, which is characterized by the induction of mid, the prevention of the stretched cells formation and the abrogation of border cell migration. We demonstrate that the anterior BMP signaling is abolished by PNT through dpp repression. However, ectopic DPP cannot rescue the anterior fate formation, suggesting additional targets of PNT participate in the posterior fate determination.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Folículo Ovárico/embriología , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética
5.
Curr Biol ; 30(21): 4213-4226.e4, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32916115

RESUMEN

Encapsulation of germline cells by layers of somatic cells forms the basic unit of female reproduction called primordial follicles in mammals and egg chambers in Drosophila. How germline and somatic tissues are coordinated for the morphogenesis of each separated unit remains poorly understood. Here, using improved live imaging of Drosophila ovaries, we uncovered periodic actomyosin waves at the cortex of germ cells. These contractile waves are associated with pressure release blebs, which project from germ cells into somatic cells. We demonstrate that these cortical activities, together with cadherin-based adhesion, are required to sort each germline cyst as one collective unit. Genetic perturbations of cortical contractility, bleb protrusion, or adhesion between germline and somatic cells induced encapsulation defects resulting from failures to encapsulate any germ cells, or the inclusion of too many germ cells per egg chamber, or even the mechanical split of germline cysts. Live-imaging experiments revealed that reducing contractility or adhesion in the germline reduced the stiffness of germline cysts and their proper anchoring to the somatic cells. Germline cysts can then be squeezed and passively pushed by constricting surrounding somatic cells, resulting in cyst splitting and cyst collisions during encapsulation. Increasing germline cysts activity or blocking somatic cell constriction movements can reveal active forward migration of germline cysts. Our results show that germ cells play an active role in physical coupling with somatic cells to produce the female gamete.


Asunto(s)
Actomiosina/metabolismo , Movimiento Celular/fisiología , Oogénesis/fisiología , Folículo Ovárico/embriología , Animales , Adhesión Celular/fisiología , Drosophila melanogaster , Femenino , Microscopía Intravital , Modelos Animales , Folículo Ovárico/diagnóstico por imagen , Folículo Ovárico/metabolismo
6.
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
7.
Cell Tissue Res ; 381(3): 555-567, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32542407

RESUMEN

In the mouse ovary, interactions between oocytes and somatic cells are essential for folliculogenesis and subsequent follicle development. The polyovular follicle (PF), which contains more than two oocytes in a follicle, can be induced in the neonatal mouse ovary when interactions between oocytes and somatic cells are disrupted by agents such as the potent synthetic estrogen diethylstilbestrol (DES) acting through estrogen receptor (ER) ß. Hedgehog signaling is known to regulate granulosa cell proliferation, thecal cell differentiation, and follicle growth. To investigate the role of hedgehog signaling in the early folliculogenesis and in PF induction by DES, neonatal mouse ovaries were cultured with or without 10 µM cyclopamine (CPA), an inhibitor of hedgehog signaling, and grafted under the kidney capsule of adult ovariectomized host mice. The number and the incidence of PFs were significantly increased in organ-cultured ovaries post-grafting. Expression of procollagen type IV, alpha 1 (Col4a1) in organ-cultured ovaries was significantly reduced by CPA, but not by DES. The expression of two hedgehog ligands, Desert hedgehog (Dhh) and Indian hedgehog (Ihh), and a target gene, Hedgehog interacting protein (Hhip), was significantly increased by DES both in WT and ERß KO mice. Therefore, we infer that DES can affect expression of those genes through ERα but not via suppression of hedgehog signaling. Thus, PFs are induced by DES or CPA, but the induction mechanism is different. Our results revealed an important role of hedgehog signaling in basement membrane remodeling during folliculogenesis even before thecal cell differentiation.


Asunto(s)
Membrana Basal/metabolismo , Proteínas Hedgehog/metabolismo , Folículo Ovárico/embriología , Animales , Animales Recién Nacidos , Proliferación Celular , Femenino , Ratones , Ovario , Transducción de Señal
8.
Cell Death Differ ; 27(8): 2468-2483, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32094512

RESUMEN

KIAA1429 (also known as vir-like m6A methyltransferase-associated protein (VIRMA)), a newly identified component of the RNA m6A methyltransferase complex, plays critical roles in guiding region-selective m6A deposition. However, in mammals, whether KIAA1429 mediates RNA m6A regulatory pathway functions in vivo remains unknown. Here, we show that the Kiaa1429-specific deficiency in oocytes resulted in female infertility with defective follicular development and fully grown germinal vesicle (GV) oocytes failing to undergo germinal vesicle breakdown (GVBD) and consequently losing the ability to resume meiosis. The oocyte growth is accompanied by the accumulation of abundant RNAs and posttranscriptional regulation. We found that the loss of Kiaa1429 could also lead to abnormal RNA metabolism in GV oocytes. RNA-seq profiling revealed that Kiaa1429 deletion altered the expression pattern of the oocyte-derived factors essential for follicular development. In addition, our data show that the conditional depletion of Kiaa1429 decreased the m6A levels in oocytes and mainly affected the alternative splicing of genes associated with oogenesis. In summary, the m6A methyltransferase KIAA1429-mediated RNA metabolism plays critical roles in folliculogenesis and the maintenance of oocyte competence.


Asunto(s)
Metiltransferasas/metabolismo , Oocitos/citología , Oocitos/enzimología , Folículo Ovárico/embriología , Folículo Ovárico/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Empalme Alternativo/genética , Animales , Núcleo Celular/metabolismo , Proliferación Celular , Femenino , Fertilidad , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Metiltransferasas/genética , Ratones Endogámicos C57BL , Modelos Biológicos , Organogénesis/genética , Folículo Ovárico/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina/metabolismo
9.
Development ; 147(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32001440

RESUMEN

Sex determination and differentiation are complex processes controlled by many different factors; however, the relationships among these factors are poorly understood. Zebrafish gonadal differentiation exhibits high plasticity involving multiple factors and pathways, which provides an excellent model for investigating the interactions between them. Ovarian aromatase (cyp19a1a) and dmrt1 are key factors in directing vertebrate ovary and testis differentiation, respectively. Knockout of zebrafish cyp19a1a leads to all-male offspring, whereas the loss of dmrt1 results in a female-biased sex ratio. In the present study, we established dmrt1-/- ;cyp19a1a-/- double mutant zebrafish and discovered that the introduction of the dmrt1 mutation into the cyp19a1a mutant could rescue the all-male phenotype of the latter. Interestingly, despite the lack of aromatase/estrogens, the follicles in the ovary of the rescued cyp19a1a mutant could develop normally up to the previtellogenic stage. Further evidence suggested the ovarian aromatase directed ovarian differentiation by suppressing dmrt1 expression via nuclear estrogen receptors (nERs). Our results provide solid evidence for an interaction between cyp19a1a and dmrt1 in zebrafish gonadal differentiation, and for the dispensability of estrogens in controlling early folliculogenesis.


Asunto(s)
Aromatasa/genética , Aromatasa/fisiología , Folículo Ovárico/embriología , Testículo/embriología , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Alelos , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Estrógenos/fisiología , Femenino , Técnicas de Inactivación de Genes , Genotipo , Heterocigoto , Masculino , Mutación , Fenotipo , Receptores de Estrógenos/fisiología , Procesos de Determinación del Sexo , Diferenciación Sexual , Pez Cebra
10.
Food Chem Toxicol ; 135: 110982, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31747621

RESUMEN

With epidemic of obesity, it affects aspects of female reproduction. Genistein could ameliorate obesity in people and animals, but might exert adverse effects on the female reproductive system. To evaluate the effects of fetal and neonatal genistein exposure on the ovarian health of F1 obese female mice with obesity induced by high-fat diet after weaning, we simulated a diet-induced obesity model to observe and determine biological effects of genistein exposure on the ovarian follicle of overfed female mice. Results showed that F1 female mice with obesity induced by high-fat diet significantly prolonged the estrus cycle, disrupted sex hormonal balance and ovarian follicle development after they were exposed to 25 mg/kg b.w./day of genistein during the fetal and neonatal stages. Genistein significantly up-regulated the ovarian mRNA expression of estrogen receptor beta in F1 obese female mice, and high-fat diet influenced the ovarian mRNA expression of estrogen receptor alpha, luteinizing hormone receptor and follicle-stimulating hormone receptor. Hence, genistein exposure from the fetal stage might increase the risk of reproductive diseases in obese females in later life. Thus, the long-term risks of genistein to obese females should be thoroughly assessed.


Asunto(s)
Dieta Alta en Grasa , Genisteína/efectos adversos , Obesidad/tratamiento farmacológico , Folículo Ovárico/efectos de los fármacos , Animales , Animales Recién Nacidos , Estradiol/metabolismo , Receptor beta de Estrógeno/genética , Ciclo Estral/efectos de los fármacos , Femenino , Feto/efectos de los fármacos , Hormona Folículo Estimulante/metabolismo , Expresión Génica/efectos de los fármacos , Hormona Luteinizante/metabolismo , Ratones Endogámicos ICR , Obesidad/metabolismo , Folículo Ovárico/embriología , Folículo Ovárico/patología , Embarazo , ARN Mensajero/metabolismo
11.
Dev Biol ; 452(2): 127-133, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31071312

RESUMEN

Understanding how cell fate decisions are regulated is a central question in stem cell biology. Recent studies have demonstrated that intracellular pH (pHi) dynamics contribute to this process. Indeed, the pHi of cells within a tissue is not simply a consequence of chemical reactions in the cytoplasm and other cellular activity, but is actively maintained at a specific setpoint in each cell type. We found previously that the pHi of cells in the follicle stem cell (FSC) lineage in the Drosophila ovary increases progressively during differentiation from an average of 6.8 in the FSCs, to 7.0 in newly produced daughter cells, to 7.3 in more differentiated cells. Two major regulators of pHi in this lineage are Drosophila sodium-proton exchanger 2 (dNhe2) and a previously uncharacterized gene, CG8177, that is homologous to mammalian anion exchanger 2 (AE2). Based on this homology, we named the gene anion exchanger 2 (ae2). Here, we generated null alleles of ae2 and found that homozygous mutant flies are viable but have severe defects in ovary development and adult oogenesis. Specifically, we find that ae2 null flies have smaller ovaries, reduced fertility, and impaired follicle formation. In addition, we find that the follicle formation defect can be suppressed by a decrease in dNhe2 copy number and enhanced by the overexpression of dNhe2, suggesting that this phenotype is due to the dysregulation of pHi. These findings support the emerging idea that pHi dynamics regulate cell fate decisions and our studies provide new genetic tools to investigate the mechanisms by which this occurs.


Asunto(s)
Antiportadores/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Oogénesis , Ovario/embriología , Ovario/metabolismo , Animales , Drosophila melanogaster/genética , Epistasis Genética , Femenino , Fertilidad , Mutación/genética , Tamaño de los Órganos , Folículo Ovárico/embriología , Interferencia de ARN , Homología de Secuencia de Aminoácido
12.
J Cell Physiol ; 234(5): 6125-6135, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30246435

RESUMEN

The reserve of primordial follicles, which serves all oocytes for the female reproductive lifespan, is established a few days after birth in mice. During this process, more than half of the oocytes are primarily eliminated by apoptosis. Autophagy, the conserved intracellular process maintaining cellular homeostasis, serves as a protective mechanism for oocyte survival. In the current study, we speculate a new role for autophagy during primordial folliculogenesis. Active autophagy was observed in perinatal ovaries from 16.5 days post coitus to 3 days post parturition. The inhibition of autophagy by 3-methyladenine (3-MA) increased the number of cyst oocytes and delayed follicle formation in vivo and in organ cultures. Furthermore, the reactive oxygen species (ROS) level was elevated in ovaries treated with 3-MA, while N-acetylcysteine, an oxidant, alleviated the inhibitory effect of 3-MA on primordial folliculogenesis. Additionally, the expression of growth differentiation factor 9 and transforming growth factor ß1, which regulates follicle activation, was decreased after 3-MA treatment. These data suggest that the physiological level of autophagy in perinatal ovaries regulates germ cell cyst breakdown and primordial follicle assembly by ROS clearance and exerts extensive effects on further follicular development.


Asunto(s)
Autofagia/fisiología , Folículo Ovárico/embriología , Especies Reactivas de Oxígeno/metabolismo , Animales , Femenino , Ratones , Ovario/embriología
13.
Reprod Sci ; 25(6): 882-892, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28299992

RESUMEN

Prenatal testosterone (T)-treated sheep, similar to women with polycystic ovary syndrome (PCOS), manifests reproductive defects that include multifollicular ovarian phenotype. Women with PCOS manifest increased ovarian matrix metalloproteinases (MMPs) activity. We tested the hypothesis that gestational T excess in sheep would alter ovarian expression of MMPs, tissue inhibitors of MMP (TIMP) and their target proteins laminin B (LAMB), collagen, tumor necrosis factor alpha (TNF), and connexin 43 (GJA1) consistent with increased MMP activity and that these changes are developmentally regulated. The ovarian content of these proteins was quantified by immunohistochemistry in fetal day 90, 140, and adult (21 months of age) ovaries. Prenatal T excess lowered GJA1 protein content in stroma and granulosa cells of primary follicles from fetal day 90 ovaries and decreased stromal MMP9, TIMP1, and LAMB in fetal day 140 ovaries. In the adult, prenatal T-treatment (1) increased MMP9 in theca cells of large preantral follicles and stroma, TNF in granulosa cells of small and large preantral follicles and theca cells of large preantral and antral follicles, and GJA1 in stroma, theca cells of large preantral follicles, and granulosa cells of antral follicles and (2) reduced TIMP1 in stroma, theca cells of large preantral and antral follicles, LAMB in stroma and small prenatral follicles, and collagen content in stroma and around antral follicles. These findings suggest a net increase in MMP activity and its target proteins TNF and GJA1 in prenatal T-treated adult but not in fetal ovaries and their potential involvement in the development of multifollicular morphology.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Ovario/embriología , Ovario/metabolismo , Testosterona/administración & dosificación , Animales , Conexina 43/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Folículo Ovárico/embriología , Folículo Ovárico/metabolismo , Ovario/efectos de los fármacos , Oveja Doméstica , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Sci Rep ; 7(1): 3526, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615648

RESUMEN

The widely-used herbicide atrazine (ATZ) is detected in ground and surface water in many countries. Several studies in animals have demonstrated that ATZ has endocrine-disrupting effects on male and female reproduction in many vertebrate species. In this study, we investigated the effects of ATZ exposure on meiosis, a key step in gametogenesis in mammals. The treatment was initiated before oocyte entry into meiosis, which occurs during the embryonic period in females. We found that embryonic exposure to ATZ increases the level of 8-oxo-guanine in the nucleus of meiotic cells, reflecting oxidative stress and affecting meiotic double-strand break repair, chromosome synapsis and crossover numbers. Finally, embryonic exposure to ATZ reduces the number of primordial follicles and increases the incidence of multi-oocyte follicles in adult mice. Our data demonstrate that embryonic exposure to ATZ disrupts prophase I of meiosis and affects normal follicle formation in female mice.


Asunto(s)
Atrazina/toxicidad , Herbicidas/toxicidad , Meiosis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/embriología , Animales , Femenino , Incidencia , Ratones
15.
Gen Comp Endocrinol ; 249: 82-92, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28495268

RESUMEN

Folliculogenesis is a process that depends on angiogenesis, in which VEGF and Notch signaling pathway members are involved. Although this pathway is present in preantral and antral follicular structures during the second stage of folliculogenesis, this association has not been described. Therefore, this study aimed to identify VEGF and Notch2 in ovary structures of infantile rats after induction of follicular development with a gonadotropin stimulus. In order to explore this possibility we analyzed rat ovary morphology from days 10-25 after birth; subsequently, the transition from preantral follicle to an antral stage was analyzed by the induction of follicular development with equine chorionic gonadotropin (eCG) and VEGF and Notch were identified in the rat ovary by fluorescence. The histological analysis revealed that the ovary of a 10-day-old rat has the highest percentage of preantral follicles and based on this a 10IU eCG dose promoted an increase in the number of antral follicles, as well as a decrease in the number of preantral follicles, related to which there was an increase in ovary weight and size. In addition, a higher concentration of circulating estradiol was observed, proliferation of granulosa cells in both follicle groups was stimulated, and the accumulation of VEGF in granulosa and theca cells and in the antral follicle oocyte was increased (p<0.05), whereas the presence of Notch2 was limited to mural granulosa cells, in granulosa cells that formed the cumulus oophorus and in the oocyte of both groups of follicles. The multiple correspondence analysis allowed us to support an association between VEGF and Notch2 during the transition from preantral to antral follicles in the ovary of an infantile rat.


Asunto(s)
Folículo Ovárico/anatomía & histología , Folículo Ovárico/metabolismo , Receptor Notch2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Recuento de Células , Proliferación Celular/efectos de los fármacos , Gonadotropina Coriónica/farmacología , Estradiol/sangre , Estradiol/metabolismo , Femenino , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Caballos , Folículo Ovárico/embriología , Ratas Wistar
16.
Biomed Res Int ; 2017: 6972030, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28265575

RESUMEN

Primordial follicle formation represents a critical phase of the initiation of embryonic reproductive organ development, while the primordial follicle transition into primary follicle determines whether oestrus or ovulation will occur in female animals. To identify molecular mechanism of new proteins which are involved in ovarian development, we employed 2D-DIGE to compare the protein expression profiles of primordial follicles and primary follicles of fetal ovaries in pigs. Fetal ovaries were collected at distinct time-points of the gestation cycle (g55 and g90). The identified proteins at the g55 time-point are mainly involved in the development of anatomical structures [reticulocalbin-1 (RCN1), reticulocalbin-3 (RCN3)], cell differentiation (actin), and stress response [heterogeneous nuclear ribonucleoprotein K (HNRNPK)]. Meanwhile, at the g90 stage, the isolated proteins with altered expression levels were mainly associated with cell proliferation [major vault protein (MVP)] and stress response [heat shock-related 70 kDa protein 2 (HSPA2)]. In conclusion, our work revealed that primordial follicle formation is regulated by RCN1, RCN3, actin, and HNRNPK, while the primordial follicle transformation to primary follicle is regulated by MVP and HSPA2. Therefore, our results provide further information for the prospective understanding of the molecular mechanism(s) involved in the regulation of the ovarian follicle development.


Asunto(s)
Feto/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Folículo Ovárico/embriología , Proteoma/biosíntesis , Animales , Femenino , Proteómica , Porcinos
17.
Dev Biol ; 422(2): 105-114, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28089848

RESUMEN

The importance of juvenile hormone regulating insect oogenesis suggests looking for genes whose expression is regulated by this hormone. SPARC is a calcium-binding glycoprotein that forms part of the extracellular membranes, which in vertebrates participates in bones mineralization or regulating cell proliferation in some cancer types. This large number of functions described for SPARC in different species might be related to the significant differences in its structure observed when comparing different species-groups. Indeed, these structural differences allow characterizing the different clades. In the cockroach Blattella germanica, a SPARC homolog emerged from ovarian transcriptomes that were constructed to find genes responding to juvenile hormone. In insects, SPARC functions have been studied in oogenesis and in embryo development of Drosophila melanogaster. In the present work, using RNAi approaches, novel functions for SPARC in the B. germanica panoistic ovaries are described. We found that depletion of SPARC does not allow to the follicular cells to complete mitosis, resulting in giant follicular cells nuclei and in a great alteration of the ovarian follicle cytoskeleton. The SPARC contribution to B. germanica oogenesis occurs stabilizing the follicular cell program and helping to maintain the nuclear divisions. Moreover, SPARC is necessary to maintain the cytoskeleton of the follicular cells. Any modification of these key processes disables females for oviposition.


Asunto(s)
Blattellidae/embriología , Citoesqueleto/metabolismo , Epitelio/fisiología , Oogénesis/fisiología , Osteonectina/metabolismo , Folículo Ovárico/embriología , Animales , Proliferación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Hormonas Juveniles/metabolismo , Mitosis/fisiología , Oogénesis/genética , Osteonectina/genética , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Vitelogeninas/biosíntesis
18.
Development ; 143(24): 4631-4642, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27836963

RESUMEN

In the epithelial follicle stem cells (FSCs) of the Drosophila ovary, Epidermal Growth Factor Receptor (EGFR) signaling promotes self-renewal, whereas Notch signaling promotes differentiation of the prefollicle cell (pFC) daughters. We have identified two proteins, Six4 and Groucho (Gro), that link the activity of these two pathways to regulate the earliest cell fate decision in the FSC lineage. Our data indicate that Six4 and Gro promote differentiation towards the polar cell fate by promoting Notch pathway activity. This activity of Gro is antagonized by EGFR signaling, which inhibits Gro-dependent repression via p-ERK mediated phosphorylation. We have found that the phosphorylated form of Gro persists in newly formed pFCs, which may delay differentiation and provide these cells with a temporary memory of the EGFR signal. Collectively, these findings demonstrate that phosphorylated Gro labels a transition state in the FSC lineage and describe the interplay between Notch and EGFR signaling that governs the differentiation processes during this period.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Receptores ErbB/metabolismo , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/genética , Folículo Ovárico/embriología , Receptores de Péptidos de Invertebrados/metabolismo , Receptores Notch/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Células Epiteliales/citología , Femenino , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Folículo Ovárico/citología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Células Madre/citología , Factores de Transcripción/metabolismo
19.
Anat Histol Embryol ; 45(5): 357-66, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27439665

RESUMEN

The aim of this investigation was to study the ultrastructural features during the development of fetal bovine ovaries (crown rump length ranging from 11.4 to 94.0 cm). An interesting observation was the occurrence of big elongated cells containing a variety of electron dense granules and light homogenous vacuoles/bodies. They were located between the stroma cells surrounding the germ cell cord ends, adjacent to the first formed primordial follicles, typically situated near blood vessels. ER alpha and ER beta receptor positive cells could be detected in the same regions by means of immunohistochemistry. Intercellular bridges linked the germ cells nests oogonia. Germ cell cords consisted of centrally located, large, pale oogonia, surrounded by elongated somatic cells with very long cytoplasm extensions. Primordial follicles with flat pale follicular cells could be observed on the inner end of the cords. Extrusions of the outer nuclear membrane could often been recognised in voluminous oocytes.


Asunto(s)
Bovinos/anatomía & histología , Inmunohistoquímica/veterinaria , Oocitos/ultraestructura , Folículo Ovárico/ultraestructura , Células Tecales/citología , Animales , Diferenciación Celular , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Oogénesis , Folículo Ovárico/anatomía & histología , Folículo Ovárico/embriología , Fagocitosis/fisiología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA