Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.906
Filtrar
1.
Sci Data ; 11(1): 383, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38615064

RESUMEN

The rete ovarii (RO) is an epithelial structure that arises during development in close proximity to the ovary and persists throughout adulthood. However, the functional significance of the RO remains elusive, and it is absent from recent discussions of female reproductive anatomy. The RO comprises three regions: the intraovarian rete within the ovary, the extraovarian rete in the periovarian tissue, and the connecting rete linking the two. We hypothesize that the RO plays a pivotal role in ovarian homeostasis and responses to physiological changes. To begin to uncover the nature and function of RO cells, we conducted transcriptomic profiling of the RO. This study presents three datasets, and reports our analysis and quality control approaches for bulk, single-cell, and nucleus-level transcriptomics of the fetal and adult RO tissues using the Pax8-rtTA; Tre-H2B-GFP mouse line, where all RO regions express nuclear GFP. The integration and rigorous validation of these datasets will advance our understanding of the RO's roles in ovarian development, female maturation, and adult female fertility.


Asunto(s)
Ovario , Transcriptoma , Animales , Femenino , Ratones , Feto , Perfilación de la Expresión Génica , Ovario/embriología , Ovario/crecimiento & desarrollo
2.
Nature ; 607(7919): 540-547, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35794482

RESUMEN

Gonadal development is a complex process that involves sex determination followed by divergent maturation into either testes or ovaries1. Historically, limited tissue accessibility, a lack of reliable in vitro models and critical differences between humans and mice have hampered our knowledge of human gonadogenesis, despite its importance in gonadal conditions and infertility. Here, we generated a comprehensive map of first- and second-trimester human gonads using a combination of single-cell and spatial transcriptomics, chromatin accessibility assays and fluorescent microscopy. We extracted human-specific regulatory programmes that control the development of germline and somatic cell lineages by profiling equivalent developmental stages in mice. In both species, we define the somatic cell states present at the time of sex specification, including the bipotent early supporting population that, in males, upregulates the testis-determining factor SRY and sPAX8s, a gonadal lineage located at the gonadal-mesonephric interface. In females, we resolve the cellular and molecular events that give rise to the first and second waves of granulosa cells that compartmentalize the developing ovary to modulate germ cell differentiation. In males, we identify human SIGLEC15+ and TREM2+ fetal testicular macrophages, which signal to somatic cells outside and inside the developing testis cords, respectively. This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis.


Asunto(s)
Linaje de la Célula , Células Germinativas , Ovario , Diferenciación Sexual , Análisis de la Célula Individual , Testículo , Animales , Cromatina/genética , Cromatina/metabolismo , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Inmunoglobulinas , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana , Proteínas de la Membrana , Ratones , Microscopía Fluorescente , Ovario/citología , Ovario/embriología , Factor de Transcripción PAX8 , Embarazo , Primer Trimestre del Embarazo , Segundo Trimestre del Embarazo , Receptores Inmunológicos , Diferenciación Sexual/genética , Testículo/citología , Testículo/embriología , Transcriptoma
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.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34958103

RESUMEN

The specific role of gonadotropin-releasing hormone (GnRH) on brain sexual differentiation remains unclear. To investigate whether gonadotropin and, in turn, testosterone (T) secretion is regulated by GnRH during the critical period for brain differentiation in sheep fetuses, we attempted to selectively suppress pituitary-testicular activation during midgestation with the long-acting GnRH antagonist degarelix. Fetuses received subcutaneous injections of the antagonist or vehicle on day 62 of gestation. After 2 to 3 weeks we examined consequences of the intervention on baseline and GnRH-stimulated plasma luteinizing hormone (LH) and T levels. In addition, we measured the effect of degarelix-treatment on messenger RNA (mRNA) expression for the pituitary gonadotropins and key gonadal steroidogenic enzymes. Baseline and GnRH-stimulated plasma LH levels were significantly suppressed in degarelix-treated male and female fetuses compared to control values. Similarly, T concentrations were suppressed in degarelix-treated males. The percentage of LHß-immunoreactive cells colocalizing c-fos was significantly reduced by degarelix treatment indicating that pituitary sensitivity was inhibited. Degarelix treatment also led to the significant suppression of mRNA expression coding for the pituitary gonadotropin subunits and for the gonadal enzymes involved in androgen synthesis. These findings demonstrate that pharmacologic inhibition of GnRH early in gestation results in suppression of LH secretion and deficits in the plasma T levels of male lamb fetuses. We conclude that GnRH signaling plays a pivotal role for regulating T exposure during the critical period of sheep gestation when the brain is masculinized. Thus, disturbance to gonadotropin secretion during this phase of gestation could have long-term consequence on adult sexual behaviors and fertility.


Asunto(s)
Edad Gestacional , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gonadotropinas Hipofisarias/metabolismo , Oligopéptidos/administración & dosificación , Adenohipófisis/embriología , Ovinos/embriología , Animales , Encéfalo/embriología , Femenino , Sangre Fetal/química , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Liberadora de Gonadotropina/fisiología , Gonadotropinas Hipofisarias/genética , Inyecciones Subcutáneas/veterinaria , Hormona Luteinizante/sangre , Masculino , Ovario/química , Ovario/embriología , Adenohipófisis/química , Adenohipófisis/efectos de los fármacos , Embarazo , ARN Mensajero/análisis , Diferenciación Sexual/fisiología , Testículo/química , Testículo/embriología , Testosterona/sangre
5.
Endocrinology ; 163(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34614143

RESUMEN

AROMATASE is encoded by the CYP19A1 gene and is the cytochrome enzyme responsible for estrogen synthesis in vertebrates. In most mammals, a peak of CYP19A1 gene expression occurs in the fetal XX gonad when sexual differentiation is initiated. To elucidate the role of this peak, we produced 3 lines of TALEN genetically edited CYP19A1 knockout (KO) rabbits that were devoid of any estradiol production. All the KO XX rabbits developed as females with aberrantly small ovaries in adulthood, an almost empty reserve of primordial follicles, and very few large antrum follicles. Ovulation never occurred. Our histological, immunohistological, and transcriptomic analyses showed that the estradiol surge in the XX fetal rabbit gonad is not essential to its determination as an ovary, or for meiosis. However, it is mandatory for the high proliferation and differentiation of both somatic and germ cells, and consequently for establishment of the ovarian reserve.


Asunto(s)
Estrógenos/metabolismo , Ovario/embriología , Ovario/fisiología , Procesos de Determinación del Sexo/fisiología , Animales , Hormona Antimülleriana/metabolismo , Diferenciación Celular , Proliferación Celular , Familia 19 del Citocromo P450/metabolismo , Estradiol/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Gónadas , Mutación INDEL , Folículo Ovárico/fisiología , Ovulación , Fenotipo , Conejos , Diferenciación Sexual/fisiología , Testosterona/metabolismo
6.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34576272

RESUMEN

Follicle stimulating hormone (FSH) is produced by the pituitary gland in a coordinated hypothalamic-pituitary-gonadal (HPG) axis event, plays important roles in reproduction and germ cell development during different phases of reproductive development (fetal, neonatal, puberty, and adult life), and is consequently essential for fertility. FSH is a heterodimeric glycoprotein hormone of two dissociable subunits, α and ß. The FSH ß-subunit (FSHß) function starts upon coupling to its specific receptor: follicle-stimulating hormone receptor (FSHR). FSHRs are localized mainly on the surface of target cells on the testis and ovary (granulosa and Sertoli cells) and have recently been found in testicular stem cells and extra-gonadal tissue. Several reproduction disorders are associated with absent or low FSH secretion, with mutation of the FSH ß-subunit or the FSH receptor, and/or its signaling pathways. However, the influence of FSH on germ cells is still poorly understood; some studies have suggested that this hormone also plays a determinant role in the self-renewal of germinative cells and acts to increase undifferentiated spermatogonia proliferation. In addition, in vitro, together with other factors, it assists the process of differentiation of primordial germ cells (PGCLCs) into gametes (oocyte-like and SSCLCs). In this review, we describe relevant research on the influence of FSH on spermatogenesis and folliculogenesis, mainly in the germ cell of humans and other species. The possible roles of FSH in germ cell generation in vitro are also presented.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Ovario/metabolismo , Células de Sertoli/metabolismo , Animales , Dimerización , Femenino , Fertilidad , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Células Germinativas/metabolismo , Gonadotropinas/metabolismo , Humanos , Masculino , Ratones , Ovario/embriología , Ovario/crecimiento & desarrollo , Hipófisis/embriología , Hipófisis/crecimiento & desarrollo , Hipófisis/metabolismo , Ratas , Receptores de HFE/metabolismo , Reproducción , Maduración Sexual , Espermatogénesis/genética , Espermatogonias/citología
7.
Development ; 148(16)2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34387307

RESUMEN

During early embryogenesis in amniotic vertebrates, the gonads differentiate into either ovaries or testes. The first cell lineage to differentiate gives rise to the supporting cells: Sertoli cells in males and pre-granulosa cells in females. These key cell types direct the differentiation of the other cell types in the gonad, including steroidogenic cells. The gonadal surface epithelium and the interstitial cell populations are less well studied, and little is known about their sexual differentiation programs. Here, we show the requirement of the homeobox transcription factor gene TGIF1 for ovarian development in the chicken embryo. TGIF1 is expressed in the two principal ovarian somatic cell populations: the cortex and the pre-granulosa cells of the medulla. TGIF1 expression is associated with an ovarian phenotype in estrogen-mediated sex reversal experiments. Targeted misexpression and gene knockdown indicate that TGIF1 is required, but not sufficient, for proper ovarian cortex formation. In addition, TGIF1 is identified as the first known regulator of juxtacortical medulla development. These findings provide new insights into chicken ovarian differentiation and development, specifically cortical and juxtacortical medulla formation.


Asunto(s)
Pollos/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Ovario/embriología , Proteínas Represoras/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Embrión de Pollo , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Gónadas/metabolismo , Proteínas de Homeodominio/metabolismo , Masculino , Ovario/citología , Ovario/metabolismo , Proteínas Represoras/metabolismo , Células de Sertoli/metabolismo , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Testículo/metabolismo
8.
Biol Reprod ; 105(2): 491-502, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-33912929

RESUMEN

Sex determination requires the commitment of bipotential gonads to either a testis or an ovarian fate. Gene deletion of the kinase Map3k4 results in gonadal sex reversal in XY mice, and transgenic re-expression of Map3k4 rescues the sex reversal phenotype. Map3k4 encodes a large, multi-functional protein possessing a kinase domain and several, additional protein-protein interaction domains. Although MAP3K4 plays a critical role in male gonadal sex determination, it is unknown if the kinase activity of MAP3K4 is required. Here, we use mice expressing full-length, kinase-inactive MAP3K4 from the endogenous Map3k4 locus to examine the requirement of MAP3K4 kinase activity in sex determination. Although homozygous kinase-inactivation of MAP3K4 (Map3k4KI/KI) is lethal, a small fraction survive to adulthood. We show Map3k4KI/KI adults exhibit a 4:1 female-biased sex ratio. Many adult Map3k4KI/KI phenotypic females have a Y chromosome. XY Map3k4KI/KI adults with sex reversal display female mating behavior, but do not give rise to offspring. Reproductive organs are overtly female, but there is a broad spectrum of ovarian phenotypes, including ovarian absence, primitive ovaries, reduced ovarian size, and ovaries having follicles in all stages of development. Further, XY Map3k4KI/KI adults are smaller than either male or female Map3k4WT/WT mice. Examination of the critical stage of gonadal sex determination at E11.5 shows that loss of MAP3K4 kinase activity results in the loss of Sry expression in XY Map3k4KI/KI embryos, indicating embryonic male gonadal sex reversal. Together, these findings demonstrate the essential role for kinase activity of MAP3K4 in male gonadal sex determination.


Asunto(s)
MAP Quinasa Quinasa Quinasa 4/genética , Ratones/genética , Ovario/embriología , Procesos de Determinación del Sexo/genética , Testículo/embriología , Animales , Femenino , MAP Quinasa Quinasa Quinasa 4/metabolismo , Masculino , Ratones/embriología
9.
PLoS Genet ; 17(3): e1009489, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33780456

RESUMEN

Here we show that multiple modes of Notch signaling activation specify the complexity of spatial cellular interactions necessary for stem cell niche assembly. In particular, we studied the formation of the germline stem cell niche in Drosophila ovaries, which is a two-step process whereby terminal filaments are formed first. Then, terminal filaments signal to the adjacent cap cell precursors, resulting in Notch signaling activation, which is necessary for the lifelong acquisition of stem cell niche cell fate. The genetic data suggest that in order to initiate the process of stem cell niche assembly, Notch signaling is activated among non-equipotent cells via distant induction, where germline Delta is delivered to somatic cells located several diameters away via cellular projections generated by primordial germ cells. At the same time, to ensure the robustness of niche formation, terminal filament cell fate can also be induced by somatic Delta via cis- or trans-inhibition. This exemplifies a double security mechanism that guarantees that the germline stem cell niche is formed, since it is indispensable for the adjacent germline precursor cells to acquire and maintain stemness necessary for successful reproduction. These findings contribute to our understanding of the formation of stem cell niches in their natural environment, which is important for stem cell biology and regenerative medicine.


Asunto(s)
Receptores Notch/metabolismo , Transducción de Señal , Nicho de Células Madre , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Drosophila , Femenino , Células Germinativas/metabolismo , Modelos Biológicos , Mutación , Organogénesis/genética , Ovario/embriología , Ovario/metabolismo , Receptores Notch/genética , Nicho de Células Madre/genética
10.
Sci Rep ; 11(1): 5941, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33723348

RESUMEN

Coccophagus japonicus Compere, an endoparasitoid of Parasaissetia nigra Nietner, has great potential for biological control. To assess the influence of mating on the reproductive performance of this parasitoid, we examined the effects of mating on ovarian development, female longevity and number of eggs laid. The results showed that the egg volume in the ovary of C. japonicus first increased and then decreased with increases in the age of female adults. The peak egg volume in the ovary of mated females occurred 2 days earlier than that of virgin females. Within the female age range of 0-15 days, the numbers of eggs at stages I, II, and III first increased and then decreased with increases in the age of female C. japonicus, whereas the number of eggs at stage IV increased. The duration of the coexistence of females and males significantly influenced the length and width of the female ovaries, and the longest ovary tube and the highest number of eggs were obtained with a coexistence duration of 0 days. C. japonicus female longevity decreased with increases in the number of matings, and the number of eggs laid by females within 15 days decreased with increasing delays in mating. In conclusion, mating can shorten the longevity of C. japonicus females, and selecting newly emerged virgin females for mating can significantly improve the number of eggs laid and the breeding efficiency of the parasitoid.


Asunto(s)
Himenópteros/fisiología , Reproducción , Conducta Sexual Animal , Animales , Femenino , Estadios del Ciclo de Vida , Longevidad , Masculino , Organogénesis , Ovario/embriología , Ovario/crecimiento & desarrollo
11.
Development ; 148(4)2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33526583

RESUMEN

Basement membranes (BM) are extracellular matrices assembled into complex and highly organized networks essential for organ morphogenesis and function. However, little is known about the tissue origin of BM components and their dynamics in vivo Here, we unravel the assembly and role of the BM main component, Collagen type IV (ColIV), in Drosophila ovarian stalk morphogenesis. Stalks are short strings of cells assembled through cell intercalation that link adjacent follicles and maintain ovarian integrity. We show that stalk ColIV has multiple origins and is assembled following a regulated pattern leading to a unique BM organisation. Absence of ColIV leads to follicle fusion, as observed upon ablation of stalk cells. ColIV and integrins are both required to trigger cell intercalation and maintain mechanically strong cell-cell attachment within the stalk. These results show how the dynamic assembly of a mosaic BM controls complex tissue morphogenesis and integrity.


Asunto(s)
Membrana Basal/metabolismo , Comunicación Celular , Drosophila/embriología , Drosophila/metabolismo , Ovario/embriología , Ovario/metabolismo , Animales , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Morfogénesis , Organogénesis , Hipófisis/embriología , Hipófisis/metabolismo
12.
Cell Mol Life Sci ; 78(2): 695-713, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32367190

RESUMEN

Meiosis is one of the most finely orchestrated events during gametogenesis with distinct developmental patterns in males and females. However, the molecular mechanisms involved in this process remain not well known. Here, we report detailed transcriptome analyses of cell populations present in the mouse female gonadal ridges (E11.5) and the embryonic ovaries from E12.5 to E14.5 using single-cell RNA sequencing (scRNA seq). These periods correspond with the initiation and progression of meiosis throughout the first stage of prophase I. We identified 13 transcriptionally distinct cell populations and 7 transcriptionally distinct germ cell subclusters that correspond to mitotic (3 clusters) and meiotic (4 clusters) germ cells. By analysing cluster-specific gene expression profiles, we found four cell clusters correspond to different cell stages en route to meiosis and characterized their detailed transcriptome dynamics. Our scRNA seq analysis here represents a new important resource for deciphering the molecular pathways driving female meiosis initiation.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Meiosis , Ovario/citología , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ovario/embriología
13.
Sci Rep ; 10(1): 20073, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33257723

RESUMEN

Research on avian sex determination has focused on the chicken. In this study, we established the utility of another widely used animal model, the Japanese quail (Coturnix japonica), for clarifying the molecular mechanisms underlying gonadal sex differentiation. In particular, we performed comprehensive gene expression profiling of embryonic gonads at three stages (HH27, HH31 and HH38) by mRNA-seq. We classified the expression patterns of 4,815 genes into nine clusters according to the extent of change between stages. Cluster 2 (characterized by an initial increase and steady levels thereafter), including 495 and 310 genes expressed in males and females, respectively, contained five key genes involved in gonadal sex differentiation. A GO analysis showed that genes in this cluster are related to developmental processes including reproductive structure development and developmental processes involved in reproduction were significant, suggesting that expression profiling is an effective approach to identify novel candidate genes. Based on RNA-seq data and in situ hybridization, the expression patterns and localization of most key genes for gonadal sex differentiation corresponded well to those of the chicken. Our results support the effectiveness of the Japanese quail as a model for studies gonadal sex differentiation in birds.


Asunto(s)
Coturnix/genética , Caracteres Sexuales , Diferenciación Sexual/genética , Animales , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Gónadas/embriología , Masculino , Ovario/embriología , Codorniz/genética , Reproducción/genética , Testículo/embriología , Transcriptoma/genética
14.
Reprod Biol ; 20(4): 555-567, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33191142

RESUMEN

Expression levels of genes involved in the development of germ cells vary throughout the process from bipotential gonadal period to adult gonadal formation. In mice, developments of female and male reproductive system are regulated by germ cell-specific factors and hormones, and determinative days in this regulation are very important. c-Abl is a non-receptor tyrosine kinase with cellular functions including cell proliferation, growth and development. mTERT is involved in maintaining telomerase activity and proliferation of surviving cells. We suggested that c-Abl and mTERT might be important for the healthy development of prenatal and postnatal mouse ovary and testis. We aim to demonstrate localization and expressions of c-Abl and mTERT in crucial days of ovary and testis development in prenatal and postnatal period in mouse by immunofluorescence staining and qRT-PCR, respectively. The importance of c-Abl and mTERT expressions during the healthy gonadal development is indicated in the prenatal and postnatal gonadal development. Also, protein expression levels were detected by Western Blot in only postnatal ovary and testis. Determining the functions of the c-Abl and mTERT throughout the process will be important in terms of understanding the infertility cases in the female and male with future studies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Ovario/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-abl/genética , Telomerasa/genética , Testículo/crecimiento & desarrollo , Animales , Femenino , Células de la Granulosa/enzimología , Masculino , Ratones , Ratones Endogámicos BALB C , Ovario/embriología , Embarazo , Proteínas Proto-Oncogénicas c-abl/análisis , ARN Mensajero/análisis , Telomerasa/análisis , Telomerasa/química , Testículo/embriología
15.
J Assist Reprod Genet ; 37(12): 3077-3087, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33025402

RESUMEN

PURPOSE: The aim of this study was to elucidate whether ovarian tissue is able to withstand a double freezing-thawing procedure. METHODS: Human ovarian cortical biopsies from 4 thawed whole ovaries were divided into 4 experimental subgroups: (a) frozen-thawed non-grafted group, (b) frozen-thawed xenografted group, (c) refrozen-rethawed non-grafted group, and (d) refrozen-rethawed xenografted group. Xenografting was performed using 8 severe combined immunodeficient mice for a total duration of 21 days. The following analyses were conducted: classic hematoxylin and eosin staining, Ki67 immunolabeling, transmission electron microscopy, Masson's green trichrome, and double CD34 immunostaining. RESULTS: Morphologically normal preantral follicles were detected in all groups. We observed a dramatic decline of more than 65% in early preantral follicle survival rates after grafting of both frozen-thawed (p < 0.0001) and refrozen-rethawed (p < 0.0001) ovarian tissue. However, mean follicle densities remained comparable between the frozen-thawed and refrozen-rethawed non-grafted groups, as well as both grafted groups. Equivalent proportions of proliferating early preantral follicles were identified in frozen-thawed and refrozen-rethawed samples, whether the tissue was grafted or not. Furthermore, we did not observe any significant difference in atretic follicle rates between any of the four groups, and the ultrastructural quality of follicles appeared unaffected by the refreezing procedure. Similar proportions of fibrosis were noted in the frozen-thawed and refrozen-rethawed groups, irrespective of grafting. Finally, no significant differences were witnessed in terms of vascularization. CONCLUSION: We were able to demonstrate, for the first time, that refrozen-rethawed ovarian tissue has the same functional characteristics as frozen-thawed ovarian tissue.


Asunto(s)
Criopreservación/métodos , Feto/citología , Oocitos/citología , Folículo Ovárico/citología , Ovario/embriología , Ovario/trasplante , Animales , Células Cultivadas , Femenino , Congelación , Humanos , Ratones , Ratones SCID , Trasplante Heterólogo
16.
EMBO J ; 39(21): e104929, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32954504

RESUMEN

Human germ cells perpetuate human genetic and epigenetic information. However, the underlying mechanism remains elusive, due to a lack of appropriate experimental systems. Here, we show that human primordial germ cell-like cells (hPGCLCs) derived from human-induced pluripotent stem cells (hiPSCs) can be propagated to at least ~106 -fold over a period of 4 months under a defined condition in vitro. During expansion, hPGCLCs maintain an early hPGC-like transcriptome and preserve their genome-wide DNA methylation profiles, most likely due to retention of maintenance DNA methyltransferase activity. These characteristics contrast starkly with those of mouse PGCLCs, which, under an analogous condition, show a limited propagation (up to ~50-fold) and persist only around 1 week, yet undergo cell-autonomous genome-wide DNA demethylation. Importantly, upon aggregation culture with mouse embryonic ovarian somatic cells in xenogeneic-reconstituted ovaries, expanded hPGCLCs initiate genome-wide DNA demethylation and differentiate into oogonia/gonocyte-like cells, demonstrating their germline potential. By creating a paradigm for hPGCLC expansion, our study uncovers critical divergences in expansion potential and the mechanism for epigenetic reprogramming between the human and mouse germ cell lineage.


Asunto(s)
Células Germinativas/metabolismo , Ovario/embriología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Línea Celular , Desmetilación del ADN , Metilación de ADN , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Epigenómica , Femenino , Genoma , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones
17.
FASEB J ; 34(11): 14440-14457, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32892421

RESUMEN

Nerve growth factor (NGF) is critical for the development and maintenance of the peripheral sympathetic neurons. NGF is also involved in the ovarian sympathetic innervation and in the development and maintenance of folliculogenesis. Women with the endocrine disorder, polycystic ovary syndrome (PCOS), have an increased sympathetic nerve activity and increased ovarian NGF levels. The role of ovarian NGF excess in the PCOS pathophysiology and in the PCOS-related features is unclear. Here, using transgenic mice overexpressesing NGF in the ovarian theca cells (17NF mice), we assessed the female embryonic development, and the reproductive and metabolic profile in adult females. Ovarian NGF excess caused growth restriction in the female fetuses, and a delayed gonocyte and primary oocyte maturation. In adulthood, the 17NF mice displayed irregular estrous cycles and altered ovarian expression of steroidogenic and epigenetic markers. They also exhibited an increased sympathetic output with increased circulating dopamine, and metabolic dysfunction reflected by aberrant adipose tissue morphology and function, impaired glucose metabolism, decreased energy expenditure, and hepatic steatosis. These findings indicate that ovarian NGF excess leads to adverse fetal development and to reproductive and metabolic complications in adulthood, mirroring common features of PCOS. This work provides evidence that NGF excess may be implicated in the PCOS pathophysiology.


Asunto(s)
Desarrollo Fetal , Factor de Crecimiento Nervioso/genética , Ovario/metabolismo , Síndrome del Ovario Poliquístico/genética , Animales , Células Cultivadas , Dopamina/metabolismo , Ciclo Estral , Femenino , Ratones , Factor de Crecimiento Nervioso/metabolismo , Oogénesis , Ovario/embriología , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Regulación hacia Arriba
18.
J Morphol ; 281(10): 1160-1172, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32808691

RESUMEN

Pseudoscorpiones (pseudoscorpions, false scorpions) is an order of small terrestrial chelicerates. While most chelicerates are lecithotrophic, that is, embryos develop due to nutrients (mostly yolk) deposited in the oocyte cytoplasm, pseudoscorpions are matrotrophic, that is, embryos are nourished by the female. Pseudoscorpion oocytes contain only a small amount of yolk. The embryos develop within a brood sac carried on the abdominal site of the female and absorb nutrients by a pumping organ. It is believed that in pseudoscorpions nutrients for developing embryos are produced in the ovary during a postovulatory (secretory) phase of the ovarian cycle. The goal of our study was to analyze the structure of the female reproductive system during the secretory phase in the pseudoscorpion Chelifer cancroides, a representative of the family Cheliferidae, considered to be one of the most advanced pseudoscorpion taxa. We use diverse microscopic techniques to document that the nutritive fluid is produced not only in the ovaries but also by the epithelial cells in the oviducts. The secretory active epithelial cells are hypertrophic and polyploid and release their content by fragmentation of apical parts. Our observations also indicate that fertilization occurs in the oviducts. Moreover, in contrast to previous findings, we show that secretion of the nutritive material starts when the fertilized oocytes reach the brood sac and thus precedes formation of the pumping organ. Summing up, we show that C. cancroides exhibits traits of advanced adaptations for matrotrophy due to coordinated secretion of the nutritive fluid by the ovarian and oviductal epithelial cells, which substantially increases the efficiency of nutritive fluid formation. Since the secretion of nutrients starts before formation of the pumping organ, we suggest that the embryos are able to absorb the nutritive fluid also in the early embryonic stages.


Asunto(s)
Adaptación Fisiológica , Arácnidos/anatomía & histología , Genitales Femeninos/anatomía & histología , Animales , Arácnidos/embriología , Arácnidos/ultraestructura , Desarrollo Embrionario , Células Epiteliales/citología , Femenino , Genitales Femeninos/ultraestructura , Lípidos/análisis , Oocitos/citología , Ovario/anatomía & histología , Ovario/embriología , Ovario/ultraestructura , Oviductos/anatomía & histología , Oviductos/ultraestructura , Ovulación , Polisacáridos/análisis , Proteínas/análisis
19.
Gene ; 763: 144956, 2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-32739586

RESUMEN

Sox transcription factors play essential roles in a variety of critical physiological processes. Still, members of the sox gene family have not yet been genome-wide identified in shrimps. In this study, a total of five members of the sox gene family were identified from the genome of Pacific white shrimp Litopenaeus vannamei and classified into three subgroups based on the conserved HMG-box domain. Among them, three belong to the SoxB subgroup (one in B1 and two in B2), one in the SoxC subgroup, and one in the SoxE subgroup. The five sox genes had different sex-biased expression in some tissues. Sox21, soxB1, and sox14 had a higher expression in ovary than in testis. In comparison, sox4 had a male-biased specific expression in the gonad, hepatopancreas, gill, and eyestalk. There was no difference in soxE gene expression between testis and ovary. During embryonic development, the expression level of three sox genes (soxB1, sox21, and soxE) was higher in gastrulation stage compared to previous stages, declined in limb bud stage and then increased in intramembrane nauplius stage; the expression of sox4 was detected in blastula stage and continued to increase in the following two stages and then surged in intramembrane nauplius stage; the highest expression of sox14 was in the fertilized egg stage, and the expression level decreased with the development of the embryo. These results suggest that the shrimp sox gene family may be involved in gametogenesis, tridermogenesis, and neurogenesis.


Asunto(s)
Proteínas de Artrópodos/genética , Penaeidae/genética , Factores de Transcripción SOX/genética , Animales , Proteínas de Artrópodos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Branquias/embriología , Branquias/metabolismo , Hepatopáncreas/embriología , Hepatopáncreas/metabolismo , Masculino , Especificidad de Órganos , Ovario/embriología , Ovario/metabolismo , Penaeidae/embriología , Factores de Transcripción SOX/metabolismo , Testículo/embriología , Testículo/metabolismo
20.
J Assist Reprod Genet ; 37(7): 1613-1622, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32424736

RESUMEN

PURPOSE: The objective of this study was to analyse the expression and cellular localization of FOXO3, pFOXO3 and PTEN throughout human ovary development both before and after birth. METHODS: Foetal, pubertal and adult paraffin-embedded ovarian samples were analysed by immunohistochemistry for cellular localization of FOXO3, pFOXO3 and PTEN proteins. Protein and mRNA expression were analysed by western blot and real time PCR, respectively, from fresh biopsies. RESULTS: PTEN was not detected by immunohistochemistry in germ cells and follicles of foetal, pubertal and adult ovaries. Occasional PTEN immunoreactive granulosa cells were found in atretic antral follicles in the adult ovary. Western blot analysis showed low levels of PTEN protein. Nuclear FOXO3-expressing primordial follicles represented a variable proportion of the ovarian reserve. The presence of FOXO3-expressing primordial follicles was very low in foetal ovary; although always represented in a low proportion, prevalence increased during pubertal and adult life. CONCLUSION: Our results seem to indicate that two subpopulations of primordial follicles, i.e. nuclear FOXO3-expressing and no FOXO3-expressing primordial follicles are found in the postnatal human ovary. This scenario suggests that FOXO3 could be acting as in the mouse model, preventing primordial follicle activation. However, the strategy would not be an "all or nothing" system as in mouse ovary but rather a selected subpopulation of primordial follicles preserved to ensure long-term fertility.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Ovario/embriología , Ovario/fisiología , Fosfohidrolasa PTEN/metabolismo , Adolescente , Adulto , Niño , Femenino , Proteína Forkhead Box O3/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Lactante , Persona de Mediana Edad , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Fosfohidrolasa PTEN/genética , Embarazo , Pubertad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...