Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
FASEB J ; 33(12): 14221-14233, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659914

RESUMEN

Deleted in azoospermia-like (DAZL) is a germ cell RNA-binding protein that is essential for entry and progression through meiosis. The phenotype of the Dazl knockout mouse has extensive germ cell loss because of incomplete meiosis. We have created a Dazl hypomorph model using short interfering RNA knockdown in mouse fetal ovary cultures, allowing investigation of Dazl function in germ cell maturation. Dazl hypomorph ovaries had a phenotype of impaired germ cell nest breakdown with a 66% reduction in total follicle number and an increase in the proportion of primordial follicles (PMFs), with smaller oocytes within these follicles. There was no significant early germ cell loss or meiotic delay. Immunostaining of intercellular bridge component testis-expressed protein (Tex)14 showed ∼59% reduction in foci number and size, without any change in Tex14 mRNA levels. TEX14 expression was also confirmed in the human fetal ovary across gestation. Using 3'UTR-luciferase reporter assays, translational regulation of TEX14 was demonstrated to be DAZL-dependant. Dazl is therefore essential for normal intercellular bridges within germ cell nests and their timely breakdown, with a major impact on subsequent assembly of PMFs.-Rosario, R., Crichton, J. H., Stewart, H. L., Childs, A. J., Adams, I. R., Anderson, R. A. Dazl determines primordial follicle formation through the translational regulation of Tex14.


Asunto(s)
Ovario/crecimiento & desarrollo , Ovario/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Animales , Clonación Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Meiosis/fisiología , Ratones , Interferencia de ARN , ARN Mensajero , Proteínas de Unión al ARN/genética , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética
2.
Cells ; 8(9)2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540096

RESUMEN

C-type natriuretic peptide (CNP) is the most conserved member of the mammalian natriuretic peptide family, and is implicated in the endocrine regulation of growth, metabolism and reproduction. CNP is expressed throughout the body, but is particularly abundant in the central nervous system and anterior pituitary gland. Pituitary gonadotropes are regulated by pulsatile release of gonadotropin releasing hormone (GnRH) from the hypothalamus, to control reproductive function. GnRH and CNP reciprocally regulate their respective signalling pathways in αT3-1 gonadotrope cells, but effects of pulsatile GnRH stimulation on CNP expression has not been explored. Here, we examine the sensitivity of the natriuretic peptide system in LßT2 and αT3-1 gonadotrope cell lines to continuous and pulsatile GnRH stimulation, and investigate putative CNP target genes in gonadotropes. Multiplex RT-qPCR assays confirmed that primary mouse pituitary tissue express Nppc,Npr2 (encoding CNP and guanylyl cyclase B (GC-B), respectively) and Furin (a CNP processing enzyme), but failed to express transcripts for Nppa or Nppb (encoding ANP and BNP, respectively). Pulsatile, but not continuous, GnRH stimulation of LßT2 cells caused significant increases in Nppc and Npr2 expression within 4 h, but failed to alter natriuretic peptide gene expression in αT3-1 cells. CNP enhanced expression of cJun, Egr1, Nr5a1 and Nr0b1, within 8 h in LßT2 cells, but inhibited Nr5a1 expression in αT3-1 cells. Collectively, these data show the gonadotrope natriuretic peptide system is sensitive to pulsatile GnRH signalling, and gonadotrope transcription factors are putative CNP-target genes. Such findings represent additional mechanisms by which CNP may regulate reproductive function.


Asunto(s)
Gonadotrofos/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Células Cultivadas , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Péptido Natriurético Tipo-C/genética
3.
Stem Cell Res ; 21: 193-201, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28434825

RESUMEN

Primordial germ cells undergo three significant processes on their path to becoming primary oocytes: the initiation of meiosis, the formation and breakdown of germ cell nests, and the assembly of single oocytes into primordial follicles. However at the onset of meiosis, the germ cell becomes transcriptionally silenced. Consequently translational control of pre-stored mRNAs plays a central role in coordinating gene expression throughout the remainder of oogenesis; RNA binding proteins are key to this regulation. In this review we examine the role of exemplars of such proteins, namely LIN28, DAZL, BOLL and FMRP, and highlight how their roles during germ cell development are critical to oogenesis and the establishment of the primordial follicle pool.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Células Germinativas/citología , Oogénesis , Proteínas de Unión al ARN/metabolismo , Femenino , Feto/fisiología , Humanos
4.
PLoS One ; 11(10): e0163987, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27695106

RESUMEN

Germ cell development and primordial follicle formation during fetal life is critical in establishing the pool of oocytes that subsequently determines the reproductive lifespan of women. Fragile X-associated primary ovarian insufficiency (FXPOI) is caused by inheritance of the FMR1 premutation allele and approximately 20% of women with the premutation allele develop ovarian dysfunction and premature ovarian insufficiency. However, the underlying disease mechanism remains obscure, and a potential role of FMRP in human ovarian development has not been explored. We have characterised the expression of FMR1 and FMRP in the human fetal ovary at the time of germ cell entry into meiosis through to primordial follicle formation. FMRP expression is exclusively in germ cells in the human fetal ovary. Increased FMRP expression in germ cells coincides with the loss of pluripotency-associated protein expression, and entry into meiosis is associated with FMRP granulation. In addition, we have uncovered FMRP association with components of P-bodies and stress granules, suggesting it may have a role in mRNA metabolism at the time of onset of meiosis. Therefore, this data support the hypothesis that FMRP plays a role regulating mRNAs during pivotal maturational processes in fetal germ cells, and ovarian dysfunction resulting from FMR1 premutation may have its origins during these stages of oocyte development.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Meiosis , Oocitos/metabolismo , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células Germinativas , Humanos , Meiosis/genética , Organogénesis/genética , Ovario/embriología , Ovario/metabolismo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/metabolismo
5.
Mol Hum Reprod ; 22(9): 622-33, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27385727

RESUMEN

STUDY QUESTION: Do changes in the expression of bone morphogenetic proteins (BMPs) 2 and 4, and their antagonists Gremlin1 (GREM1) and Gremlin2 (GREM2) during human fetal ovarian development impact on BMP pathway activity and lead to changes in gene expression that may influence the fate and/or function of ovarian somatic cells? STUDY FINDING: BMPs 2 and 4 differentially regulate gene expression in cultured human fetal ovarian somatic cells. Expression of some, but not all BMP target genes is antagonised by GREM1 and GREM2, indicating the existence of a mechanism to fine-tune BMP signal intensity in the ovary. Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), a marker of immature ovarian somatic cells, is identified as a novel transcriptional target of BMP4. WHAT IS KNOWN ALREADY: Extensive re-organisation of the germ and somatic cell populations in the feto-neonatal ovary culminates in the formation of primordial follicles, which provide the basis for a female's future fertility. BMP growth factors play important roles at many stages of ovarian development and function. GREM1, an extracellular antagonist of BMP signalling, regulates the timing of primordial follicle formation in the mouse ovary, and mRNA levels of BMP4 decrease while those of BMP2 increase prior to follicle formation in the human fetal ovary. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Expression of genes encoding BMP pathway components, BMP antagonists and markers of ovarian somatic cells were determined by quantitative (q)RT-PCR in human fetal ovaries (from 8 to 21 weeks gestation) and fetal ovary-derived somatic cell cultures. Ovarian expression of GREM1 protein was confirmed by immunoblotting. Primary human fetal ovarian somatic cell cultures were derived from disaggregated ovaries by differential adhesion and cultured in the presence of recombinant human BMP2 or BMP4, with or without the addition of GREM1 or GREM2. MAIN RESULTS AND THE ROLE OF CHANCE: We demonstrate that the expression of BMP antagonists GREM1, GREM2 and CHRD  increases in the lead-up to primordial follicle formation in the human fetal ovary, and that the BMP pathway is active in cultured ovarian somatic cells. This leads to differential changes in the expression of a number of genes, some of which are further modulated by GREM1 and/or GREM2. The positive transcriptional regulation of LGR5 (a marker of less differentiated somatic cells) by BMP4 in vitro suggests that increasing levels of GREM1 and reduced levels of BMP4 as the ovary develops in vivo may act to reduce LGR5 levels and allow pre-granulosa cell differentiation. LIMITATIONS, REASONS FOR CAUTION: While we have demonstrated that markers of different somatic cell types are expressed in the cultured ovarian somatic cells, their proportions may not represent the same cells in the intact ovary which also contains germ cells. WIDER IMPLICATIONS OF THE FINDINGS: This study extends previous work identifying germ cells as targets of ovarian BMP signalling, and suggests BMPs may regulate the development of both germ and somatic cells in the developing ovary around the time of follicle formation. LARGE SCALE DATA: Not applicable. STUDY FUNDING/COMPETING INTERESTS: This work was supported by The UK Medical Research Council (Grant No.: G1100357 to RAA), and Medical Research Scotland (Grant No. 345FRG to AJC). The authors have no competing interests to declare.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ovario/citología , Ovario/metabolismo , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Citocinas , Femenino , Regulación de la Expresión Génica/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Humanos , Oocitos/citología , Oocitos/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
8.
J Clin Endocrinol Metab ; 100(9): E1197-205, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26192875

RESUMEN

CONTEXT: Fetal ovarian development and primordial follicle formation underpin future female fertility. Prokineticin (PROK) ligands regulate cell survival, proliferation, and angiogenesis in adult reproductive tissues including the ovary. However, their expression and function during fetal ovarian development remains unclear. OBJECTIVE: This study aimed to investigate expression and localization of the PROK ligands, receptors, and their downstream transcriptional targets in the human fetal ovary. SETTING: This study was conducted at the University of Edinburgh. PARTICIPANTS: Ovaries were collected from 37 morphologically normal human fetuses. DESIGN AND MAIN OUTCOME MEASURES: mRNA and protein expression of PROK ligands and receptors was determined in human fetal ovaries using qRT-PCR, immunoblotting, and immunohistochemistry. Functional studies were performed using a human germ cell tumor line (TCam-2) stably transfected with Prokineticin receptor 1 (PROKR1). RESULTS: Expression of PROK1 and PROKR1 was significantly higher in mid-gestation ovaries (17-20 wk) than at earlier gestations (8-11 and 14-16 wk). PROK2 significantly increased across the gestations examined. PROKR2 expression remained unchanged. PROK ligand and receptor proteins were predominantly localized to germ cells (including oocytes within primordial follicles) and endothelial cells, indicating these cell types to be the targets of PROK signaling in the human fetal ovary. PROK1 treatment of a germ cell line stably expressing PROKR1 resulted in ERK phosphorylation and elevated COX2 expression. CONCLUSIONS: Developmental changes in expression and regulation of COX2 and phosphorylated ERK (pERK) by PROK1 suggest that PROK ligands may be novel regulators of germ cell development in the human fetal ovary, interacting within a network of growth and survival factors prior to primordial follicle formation.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Hormonas Gastrointestinales/metabolismo , Células Germinativas/metabolismo , Neuropéptidos/metabolismo , Ovario/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Ciclooxigenasa 2/genética , Femenino , Desarrollo Fetal , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/farmacología , Células Germinativas/efectos de los fármacos , Humanos , Neuropéptidos/genética , Ovario/embriología , Fosforilación/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/farmacología
9.
PLoS One ; 10(3): e0119819, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25790371

RESUMEN

During human fetal ovary development, the process of primordial follicle formation is immediately preceded by a highly dynamic period of germ cell and somatic cell reorganisation. This is regulated by germ-cell specific transcription regulators, by the conserved RNA binding proteins DAZL and BOLL and by secreted growth factors of the TGFß family, including activin ßA: these all show changing patterns of expression preceding follicle formation. In mice, the transcription factor Nobox is essential for follicle formation and oocyte survival, and NOBOX regulates the expression of GDF9 in humans. We have therefore characterised the expression of GDF9 in relation to these known key factors during follicle formation in the human fetal ovary. mRNA levels of GDF9, BMP15 and NOBOX were quantified by qRT-PCR and showed dramatic increases across gestation. GDF9 protein expression was localised by immunohistochemistry to the same population of germ cells as those expressing activin ßA prior to follicle formation but did not co-localise with either BOLL or DAZL. A novel NOBOX isoform was identified in fetal ovary that was shown to be capable of up-regulating the GDF9 promoter in reporter assays. Thus, during oogenesis in humans, oocytes go through a dynamic and very sharply demarcated sequence of changes in expression of these various proteins, even within individual germ cell nests, likely to be of major functional significance in determining selective germ cell survival at this key stage in ovarian development. Transcriptional variation may contribute to the range of age of onset of POI in women with NOBOX mutations.


Asunto(s)
Factor 9 de Diferenciación de Crecimiento/metabolismo , Proteínas de Homeodominio/metabolismo , Oocitos/metabolismo , Ovario/metabolismo , Factores de Transcripción/metabolismo , Activinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteína Morfogenética Ósea 15/genética , Proteína Morfogenética Ósea 15/metabolismo , Femenino , Feto/metabolismo , Células Germinativas/metabolismo , Factor 9 de Diferenciación de Crecimiento/genética , Células HEK293 , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Ratones , Datos de Secuencia Molecular , Folículo Ovárico/crecimiento & desarrollo , Ovario/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/metabolismo , Alineación de Secuencia , Factores de Transcripción/química , Factores de Transcripción/genética , Regulación hacia Arriba
10.
Hum Reprod ; 29(7): 1471-89, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24847019

RESUMEN

STUDY QUESTION: How does maternal cigarette smoking disturb development of the human fetal ovary? SUMMARY ANSWER: Maternal smoking increases fetal estrogen titres and dysregulates several developmental processes in the fetal ovary. WHAT IS KNOWN ALREADY: Exposure to maternal cigarette smoking during gestation reduces human fetal ovarian cell numbers, germ cell proliferation and subsequent adult fecundity. STUDY DESIGN, SIZE, DURATION: The effects of maternal cigarette smoking on the second trimester human fetal ovary, fetal endocrine signalling and fetal chemical burden were studied. A total of 105 fetuses were studied, 56 from mothers who smoked during pregnancy and 49 from those who did not. PARTICIPANTS/MATERIALS, SETTING METHODS: Ovary, liver and plasma samples were collected from electively terminated, normally progressing, second trimester human fetuses. Circulating fetal hormones, levels of 73 fetal ovarian transcripts, protein localization, density of oocytes/primordial follicles and levels of 16 polycyclic aromatic hydrocarbons (PAHs) in the fetal liver were determined. MAIN RESULTS AND THE ROLE OF CHANCE: Circulating fetal estrogen levels were very high and were increased by maternal smoking (ANOVA, P = 0.055-0.004 versus control). Smoke exposure also dysregulated (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.046-0.023) four fetal ovarian genes (cytochrome P450 scc [CYP11A1], NOBOX oogenesis homeobox [NOBOX], activator of apoptosis harakiri [HRK], nuclear receptor subfamily 2, group E, member 1 [NR2E1]), shifted the ovarian Inhibin ßA/inhibin α ratio (NHBA/INHA) transcript ratio in favour of activin (ANOVA, P = 0.049 versus control) and reduced the proportion of dominant-negative estrogen receptor 2 (ERß: ESR2) isoforms in half the exposed fetuses. PAHs, ligands for the aryl hydrocarbon receptor (AHR), were increased nearly 6-fold by maternal smoking (ANOVA, P = 0.011 versus control). A fifth transcript, COUP transcription factor 1 (nuclear receptor subfamily 2, group F, member 1: NR2F1, which contains multiple AHR-binding sites), was both significantly increased (ANOVA, P = 0.026 versus control) and dysregulated by (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.021) maternal smoking. NR2F1 is associated with repression of FSHR expression and smoke-exposed ovaries failed to show the normal increase in FSHR expression during the second trimester. There was a significantly higher number of DEAD (Asp-Glu-Ala-Asp) box polypeptide 4 (DDX4) VASA-positive (ANOVA, P = 0.016 versus control), but not POU domain, class 1, transcription factor 1 (POU5F1) OCT3/4-positive, oocytes in smoke-exposed fetuses and this matched with a significantly higher number of primordial follicles (ANOVA, P = 0.024 versus control). LIMITATIONS, REASONS FOR CAUTION: The effects of maternal smoking on establishment of the maximum fetal primordial follicle pool cannot be reliably studied in our population since the process is not completed until 28 weeks of gestation and normal fetuses older than 21 weeks of gestation are not available for study. Our data suggest that some fetal ovaries are affected by smoke exposure while others are not, indicating that additional studies, with larger numbers, may show more significant effects. WIDER IMPLICATIONS OF THE FINDINGS: Fetal exposure to chemicals in cigarette smoke is known to lead to reduced fecundity in women. Our study suggests, for the first time, that this occurs via mechanisms involving activation of AHR, disruption of inhibin/activin and estrogen signalling, increased exposure to estrogen and dysregulation of multiple molecular pathways in the exposed human fetal ovary. Our data also suggest that alterations in the ESR2 positive and dominant negative isoforms may be associated with reduced sensitivity of some fetuses to increased estrogens and maternal smoking. STUDY FUNDING/COMPETING INTEREST(S): The study was supported by grants from the Chief Scientist Office (Scottish Executive, CZG/1/109, and CZG/4/742), NHS Grampian Endowments (08/02), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 212885, a Society for Reproduction & Fertility summer studentship, Medical Research Scotland (research grant 354 FRG) and the Medical Research Council (WBS: U.1276.00.002.00001 and G1100357). The authors declare they have no competing interests, be it financial, personal or professional.


Asunto(s)
Exposición Materna/efectos adversos , Ovario/efectos de los fármacos , Fumar/efectos adversos , Adulto , Índice de Masa Corporal , Proliferación Celular , Cotinina/metabolismo , Estrógenos/metabolismo , Femenino , Desarrollo Fetal/efectos de los fármacos , Células Germinativas/citología , Humanos , Inmunohistoquímica , Recién Nacido , Ligandos , Hígado/metabolismo , Oocitos/citología , Folículo Ovárico/embriología , Ovario/embriología , Ovario/patología , Fenotipo , Hidrocarburos Policíclicos Aromáticos , Embarazo , Segundo Trimestre del Embarazo , Transducción de Señal , Productos de Tabaco
11.
Mol Hum Reprod ; 20(1): 42-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23979962

RESUMEN

Fetal life is a critical time for female fertility, when germ cells complete proliferation, initiate meiosis and ultimately form the lifetime stock of primordial follicles. Female fertility may be reduced by in utero exposure to cigarette smoke, which contains ligands for the aryl hydrocarbon receptor (AhR). The AhR is a critical regulator of ovarian germ cell survival in mice; thus activation of this receptor in the ovaries of fetuses exposed to maternal cigarette smoke in utero may provide a mechanism by which female fertility is reduced in later life. We have therefore investigated AhR expression in the human fetal ovary, and examined the effects of an AhR ligand present in cigarette smoke, on germ cells in human fetal ovaries cultured in vitro. The results showed that AHR mRNA expression increased 2-fold between first and late second trimester (P = 0.008). AhR protein was confined to germ cells at all gestations, but varied from expression in most germ cells during the first trimester, to only patchy expression by clusters of germ cells at later gestations. Culture of human fetal ovaries with the AhR ligand 9,10-dimethyl-1,2-benzanthracene-3,4-dihydrodiol (DMBA-DHD; a component of cigarette smoke) did not affect germ cell number in vitro, but significantly reduced the proportion of proliferating germ cells by 29% (as assessed by phospho-histone H3 staining (P = 0.04)). Germ cell apoptosis was not significantly affected. These results reveal that germ cells in the human fetal ovary express AhR from the proliferative stage of development through entry into meiosis and beyond, and demonstrate that AhR ligands found in cigarette smoke have the capacity to impair human fetal ovarian germ cell proliferation.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/análogos & derivados , Células Germinativas/efectos de los fármacos , Ovario/embriología , Receptores de Hidrocarburo de Aril/metabolismo , Humo/efectos adversos , 9,10-Dimetil-1,2-benzantraceno/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Fármacos para la Fertilidad Femenina , Feto/efectos de los fármacos , Células Germinativas/metabolismo , Humanos , Oogénesis/efectos de los fármacos , Ovario/metabolismo , Embarazo , ARN Mensajero/biosíntesis , Receptores de Hidrocarburo de Aril/biosíntesis , Receptores de Hidrocarburo de Aril/genética
12.
PLoS One ; 8(9): e73996, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086306

RESUMEN

The Deleted in Azoospermia gene family encodes three germ cell-specific RNA-binding proteins (DAZ, DAZL and BOLL) that are essential for gametogenesis in diverse species. Targeted disruption of Boll in mice causes male-specific spermiogenic defects, but females are apparently fertile. Overexpression of human BOLL promotes the derivation of germ cell-like cells from genetically female (XX), but not male (XY) human ES cells however, suggesting a functional role for BOLL in regulating female gametogenesis in humans. Whether BOLL is expressed during oogenesis in mammals also remains unclear. We have therefore investigated the expression of BOLL during fetal oogenesis in humans and mice. We demonstrate that BOLL protein is expressed in the germ cells of the human fetal ovary, at a later developmental stage than, and almost mutually-exclusive to, the expression of DAZL. Strikingly, BOLL is downregulated, and DAZL re-expressed, as primordial follicles form, revealing BOLL expression to be restricted to a narrow window during fetal oogenesis. By quantifying the extent of co-expression of DAZL and BOLL with markers of meiosis, we show that this window likely corresponds to the later stages of meiotic prophase I. Finally, we demonstrate that Boll is also transiently expressed during oogenesis in the fetal mouse ovary, but is simultaneously co-expressed within the same germ cells as Dazl. These data reveal significant similarities and differences between the expression of BOLL homologues during oogenesis in humans and mice, and raise questions as to the validity of the Boll(-/-) mouse as a model for understanding BOLL function during human oogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oogénesis/genética , Proteínas de Unión al ARN/genética , Animales , Femenino , Humanos , Masculino , Meiosis , Ratones
13.
Methods Mol Biol ; 957: 59-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23138944

RESUMEN

The development of primordial germ cells into oocytes within primordial follicles involves a complex sequence of proliferation, developmental commitment, entry and arrest in meiosis, and association with surrounding somatic cells. These processes occur over the first few months of development in the human, with multiple stages of development present at any one time point. Immunohistochemistry has been hugely instructive in identifying the various key stages in ovarian development, by allowing simultaneous visualization of different stages of germ cell development, and their spatial arrangement. These studies allow comparison with other species and have identified key differences between human and murine ovarian development as well as giving a basis for functional studies. In this chapter we describe the main methodologies used in immunohistochemistry, using both chromogen and fluorescence approaches, and both single and double antigen detection.


Asunto(s)
Feto , Inmunohistoquímica/métodos , Ovario/citología , Ovario/embriología , Amidinas/metabolismo , Precipitación Química , Compuestos Cromogénicos/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos
14.
PLoS One ; 8(11)2013.
Artículo en Inglés | MEDLINE | ID: mdl-29220838

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0073996.].

16.
Stem Cells Dev ; 21(13): 2343-9, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22296229

RESUMEN

Germ cell development requires timely transition from primordial germ cell (PGC) self-renewal to meiotic differentiation. This is associated with widespread changes in gene expression, including downregulation of stem cell-associated genes, such as OCT4 and KIT, and upregulation of markers of germ cell differentiation and meiosis, such as VASA, STRA8, and SYCP3. The stem cell-expressed RNA-binding protein Lin28 has recently been demonstrated to be essential for PGC specification in mice, and LIN28 is expressed in human germ cell tumors with phenotypic similarities to human fetal germ cells. We have therefore examined the expression of LIN28 during normal germ cell development in the human fetal ovary, from the PGC stage, through meiosis to the initiation of follicle formation. LIN28 transcript levels were highest when the gonad contained only PGCs, and decreased significantly with increasing gestation, coincident with the onset of germ cell differentiation. Immunohistochemistry revealed LIN28 protein expression to be germ cell-specific at all stages examined. All PGCs expressed LIN28, but at later gestations expression was restricted to a subpopulation of germ cells, which we demonstrate to be primordial and premeiotic germ cells based on immunofluorescent colocalization of LIN28 and OCT4, and absence of overlap with the meiosis marker SYCP3. We also demonstrate the expression of the LIN28 target precursor pri-microRNA transcripts pri-LET7a/f/d and pri-LET-7g in the human fetal ovary, and that expression of these is highest at the PGC stage, mirroring that of LIN28. The spatial and temporal restriction of LIN28 expression and coincident peaks of expression of LIN28 and target pri-microRNAs suggest important roles for this protein in the maintenance of the germline stem cell state and the regulation of microRNA activity in the developing human ovary.


Asunto(s)
Feto/citología , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Ovario/embriología , Proteínas de Unión al ARN/metabolismo , Proteínas de Ciclo Celular , Diferenciación Celular , Proteínas de Unión al ADN , Femenino , Feto/embriología , Células Germinativas/metabolismo , Edad Gestacional , Humanos , Inmunohistoquímica , Meiosis , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ovario/citología , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Mol Hum Reprod ; 18(2): 88-95, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21965347

RESUMEN

Fetal ovarian development and primordial follicle formation are imperative for adult fertility in the female. Data suggest the interleukin (IL)6-type cytokines, leukaemia inhibitory factor (LIF), IL6, oncostatin M (OSM) and ciliary neurotrophic factor (CNTF), are able to regulate the survival, proliferation and differentiation of fetal murine germ cells (GCs) in vivo and in vitro. We postulated that these factors may play a similar role during early human GC development and primordial follicle formation. To test this hypothesis, we have investigated the expression and regulation of IL6-type cytokines, using quantitative reverse transcription polymerase chain reaction and immunohistochemistry. Expression of transcripts encoding OSM increased significantly across the gestational range examined (8-20 weeks), while expression of IL6 increased specifically between the first (8-11 weeks) and early second (12-16 weeks) trimesters, co-incident with the initiation of meiosis. LIF and CNTF expression remained unchanged. Expression of the genes encoding the LIF and IL6 receptors, and their common signalling subunit gp130, was also found to be developmentally regulated, with expression increasing significantly with increasing gestation. LIF receptor and gp130 proteins localized exclusively to GCs, including oocytes in primordial follicles, indicating this cell type to be the sole target of IL6-type cytokine signalling in the human fetal ovary. These data establish that IL6-type cytokines and their receptors are expressed in the human fetal ovary and may directly influence GC development at multiple stages of maturation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Folículo Ovárico/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal/genética , Adulto , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Femenino , Feto , Edad Gestacional , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Oncostatina M/genética , Oncostatina M/metabolismo , Oocitos/crecimiento & desarrollo , Folículo Ovárico/crecimiento & desarrollo , Embarazo , Trimestres del Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor Neurotrófico Ciliar/genética , Receptor de Factor Neurotrófico Ciliar/metabolismo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo
18.
Methods Mol Biol ; 825: 199-210, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22144246

RESUMEN

The survival, proliferation, and differentiation of primordial germ cells in the mammalian embryo is regulated by a complex cocktail of growth factors and interactions with surrounding somatic cells, which together form a microenvironment known as the germ cell niche. Extensive insight into the signalling pathways that regulate PGC behaviour has been provided by the study of these cells in rodent models, however little is known about the factors that regulate these processes in human PGCs. In this review, we outline experimental approaches to the culture and manipulation of the first trimester human fetal ovary, and discuss immunohistochemical and stereological approaches to detect changes in human PGC numbers and proliferation in response to treatment with exogenous growth factors.


Asunto(s)
Células Germinativas/citología , Diferenciación Celular , Separación Celular/métodos , Disección/métodos , Embrión de Mamíferos , Femenino , Feto , Humanos , Inmunohistoquímica/métodos , Técnicas de Cultivo de Órganos/métodos , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Embarazo , Cultivo Primario de Células/métodos , Análisis para Determinación del Sexo/métodos
19.
Endocrinology ; 152(11): 4358-67, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21952240

RESUMEN

Proper development of the seminiferous tubules (or testis cords in embryos) is critical for male fertility. Sertoli cells, somatic components of the seminiferous tubules, serve as nurse cells to the male germline, and thus their numbers decide the quantity of sperm output in adulthood. We previously identified activin A, the protein product of the activin ßA (Inhba) gene, as a key regulator of murine Sertoli cell proliferation and testis cord expansion during embryogenesis. Although our genetic studies implicated fetal Leydig cells as the primary producers of testicular activin A, gonocytes are another potential source. To investigate the relative contribution of gonocyte-derived activin A to testis morphogenesis, we compared testis development in the Inhba global knockout mouse, which lacks activin A production in all cells (including the gonocytes), and a steroidogenic factor 1 (Sf1)-specific conditional knockout model in which activin A expression in testicular somatic cells is disrupted but gonocyte expression of activin A remains intact. Surprisingly, testis development was comparable in these two models of activin A insufficiency, with similar reductions in Sertoli cell proliferation and minor differences in testis histology. Thus, our findings suggest activin A from male gonocytes is insufficient to promote Sertoli cell proliferation and testis cord expansion in the absence of somatic cell-derived activin A. Evaluation of adult male mice with fetal disruption of activin A revealed reduced testis size, lowered sperm production, altered testicular histology, and elevated plasma FSH levels, defects reminiscent of human cases of androgen-sufficient idiopathic oligozoospermia.


Asunto(s)
Activinas/metabolismo , Células Intersticiales del Testículo/metabolismo , Morfogénesis/fisiología , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/embriología , Animales , Proliferación Celular , Hormona Folículo Estimulante/sangre , Masculino , Ratones , Ratones Transgénicos , Recuento de Espermatozoides , Testículo/crecimiento & desarrollo , Testículo/metabolismo
20.
PLoS One ; 6(6): e20249, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21674038

RESUMEN

The development of mammalian fetal germ cells along oogenic or spermatogenic fate trajectories is dictated by signals from the surrounding gonadal environment. Germ cells in the fetal testis enter mitotic arrest, whilst those in the fetal ovary undergo sex-specific entry into meiosis, the initiation of which is thought to be mediated by selective exposure of fetal ovarian germ cells to mesonephros-derived retinoic acid (RA). Aspects of this model are hard to reconcile with the spatiotemporal pattern of germ cell differentiation in the human fetal ovary, however. We have therefore examined the expression of components of the RA synthesis, metabolism and signalling pathways, and their downstream effectors and inhibitors in germ cells around the time of the initiation of meiosis in the human fetal gonad. Expression of the three RA-synthesising enzymes, ALDH1A1, 2 and 3 in the fetal ovary and testis was equal to or greater than that in the mesonephros at 8-9 weeks gestation, indicating an intrinsic capacity within the gonad to synthesise RA. Using immunohistochemistry to detect RA receptors RARα, ß and RXRα, we find germ cells to be the predominant target of RA signalling in the fetal human ovary, but also reveal widespread receptor nuclear localization indicative of signalling in the testis, suggesting that human fetal testicular germ cells are not efficiently shielded from RA by the action of the RA-metabolising enzyme CYP26B1. Consistent with this, expression of CYP26B1 was greater in the human fetal ovary than testis, although the sexually-dimorphic expression patterns of the germ cell-intrinsic regulators of meiotic initiation, STRA8 and NANOS2, appear conserved. Finally, we demonstrate that RA induces a two-fold increase in STRA8 expression in cultures of human fetal testis, but is not sufficient to cause widespread meiosis-associated gene expression. Together, these data indicate that while local production of RA within the fetal ovary may be important in regulating the onset of meiosis in the human fetal ovary, mechanisms other than CYP26B1-mediated metabolism of RA may exist to inhibit the entry of germ cells into meiosis in the human fetal testis.


Asunto(s)
Feto/citología , Meiosis , Ovario/citología , Transducción de Señal , Testículo/citología , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Sistema Enzimático del Citocromo P-450/genética , Femenino , Feto/enzimología , Feto/metabolismo , Regulación de la Expresión Génica , Células Germinativas/citología , Células Germinativas/enzimología , Células Germinativas/metabolismo , Humanos , Masculino , Meiosis/genética , Ratones , Ovario/enzimología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Embarazo , Segundo Trimestre del Embarazo/genética , Transporte de Proteínas , Proteínas/genética , Proteínas de Unión al ARN/genética , Receptores de Ácido Retinoico/genética , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Ácido Retinoico 4-Hidroxilasa , Caracteres Sexuales , Transducción de Señal/genética , Testículo/enzimología , Testículo/crecimiento & desarrollo , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA