Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nucleic Acids Res ; 51(12): 5981-5996, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37099375

RESUMEN

Progesterone receptor (PGR) plays diverse roles in reproductive tissues and thus coordinates mammalian fertility. In the ovary, rapid acute induction of PGR is the key determinant of ovulation through transcriptional control of a unique set of genes that culminates in follicle rupture. However, the molecular mechanisms for this specialized PGR function in ovulation is poorly understood. We have assembled a detailed genomic profile of PGR action through combined ATAC-seq, RNA-seq and ChIP-seq analysis in wildtype and isoform-specific PGR null mice. We demonstrate that stimulating ovulation rapidly reprograms chromatin accessibility in two-thirds of sites, correlating with altered gene expression. An ovary-specific PGR action involving interaction with RUNX transcription factors was observed with 70% of PGR-bound regions also bound by RUNX1. These transcriptional complexes direct PGR binding to proximal promoter regions. Additionally, direct PGR binding to the canonical NR3C motif enable chromatin accessibility. Together these PGR actions mediate induction of essential ovulatory genes. Our findings highlight a novel PGR transcriptional mechanism specific to ovulation, providing new targets for infertility treatments or new contraceptives that block ovulation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica , Receptores de Progesterona , Transcripción Genética , Animales , Femenino , Ratones , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Mamíferos/genética , Ratones Noqueados , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo
2.
Development ; 147(6)2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32108023

RESUMEN

Members of the Iroquois B (IrxB) homeodomain cluster genes, specifically Irx3 and Irx5, are crucial for heart, limb and bone development. Recently, we reported their importance for oocyte and follicle survival within the developing ovary. Irx3 and Irx5 expression begins after sex determination in the ovary but remains absent in the fetal testis. Mutually antagonistic molecular signals ensure ovary versus testis differentiation with canonical Wnt/ß-catenin signals paramount for promoting the ovary pathway. Notably, few direct downstream targets have been identified. We report that Wnt/ß-catenin signaling directly stimulates Irx3 and Irx5 transcription in the developing ovary. Using in silico analysis of ATAC- and ChIP-Seq databases in conjunction with mouse gonad explant transfection assays, we identified TCF/LEF-binding sequences within two distal enhancers of the IrxB locus that promote ß-catenin-responsive ovary expression. Meanwhile, Irx3 and Irx5 transcription is suppressed within the developing testis by the presence of H3K27me3 on these same sites. Thus, we resolved sexually dimorphic regulation of Irx3 and Irx5 via epigenetic and ß-catenin transcriptional control where their ovarian presence promotes oocyte and follicle survival vital for future ovarian health.


Asunto(s)
Epigénesis Genética/fisiología , Gónadas/embriología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Ovario/embriología , Ovario/metabolismo , Caracteres Sexuales , Diferenciación Sexual/genética , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/metabolismo
3.
Int J Mol Sci ; 23(22)2022 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-36430923

RESUMEN

Genetic alterations of the RUNX1 gene are associated with a variety of malignancies, including female-related cancers. The role of RUNX1 as either a tumor suppressor gene or an oncogene is tissue-dependent and varies based on the cancer type. Both the amplification and deletion of the RUNX1 gene have been associated with ovarian cancer in humans. In this study, we investigated the effects of Runx1 loss on ovarian pathogenesis in mice. A conditional loss of Runx1 in the somatic cells of the ovary led to an increased prevalence of ovarian tumors in aged mice. By the age of 15 months, 27% of Runx1 knockout (KO) females developed ovarian tumors that presented characteristics of granulosa cell tumors. While ovaries from young adult mice did not display tumors, they all contained abnormal follicle-like lesions. The granulosa cells composing these follicle-like lesions were quiescent, displayed defects in differentiation and were organized in a rosette-like pattern. The RNA-sequencing analysis further revealed differentially expressed genes in Runx1 KO ovaries, including genes involved in metaplasia, ovarian cancer, epithelial cell development, tight junctions, cell-cell adhesion, and the Wnt/beta-catenin pathway. Together, this study showed that Runx1 is required for normal granulosa cell differentiation and prevention of ovarian tumor development in mice.


Asunto(s)
Tumor de Células de la Granulosa , Neoplasias Ováricas , Humanos , Ratones , Femenino , Animales , Lactante , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células de la Granulosa/metabolismo , Neoplasias Ováricas/patología , Tumor de Células de la Granulosa/metabolismo , Carcinoma Epitelial de Ovario/patología
4.
Hum Mol Genet ; 27(24): 4273-4287, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30212841

RESUMEN

The identity of the gonads is determined by which fate, ovarian granulosa cell or testicular Sertoli cell, the bipotential somatic cell precursors choose to follow. In most vertebrates, the conserved transcription factor FOXL2 contributes to the fate of granulosa cells. To understand FOXL2 functions during gonad differentiation, we performed genome-wide analysis of FOXL2 chromatin occupancy in fetal ovaries and established a genetic mouse model that forces Foxl2 expression in the fetal testis. When FOXL2 was ectopically expressed in the somatic cell precursors in the fetal testis, FOXL2 was sufficient to repress Sertoli cell differentiation, ultimately resulting in partial testis-to-ovary sex-reversal. Combining genome-wide analysis of FOXL2 binding in the fetal ovary with transcriptomic analyses of our Foxl2 gain-of-function and previously published Foxl2 loss-of-function models, we identified potential pathways responsible for the feminizing action of FOXL2. Finally, comparison of FOXL2 genome-wide occupancy in the fetal ovary with testis-determining factor SOX9 genome-wide occupancy in the fetal testis revealed extensive overlaps, implying that antagonistic signals between FOXL2 and SOX9 occur at the chromatin level.


Asunto(s)
Proteína Forkhead Box L2/genética , Factor de Transcripción SOX9/genética , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Animales , Cromatina/genética , Femenino , Desarrollo Fetal/genética , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Genoma/genética , Gónadas/crecimiento & desarrollo , Masculino , Ratones , Ovario/crecimiento & desarrollo , Unión Proteica , Testículo/crecimiento & desarrollo , Transcriptoma/genética
5.
Biol Reprod ; 103(5): 966-977, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32945847

RESUMEN

Development and functions of the ovary rely on appropriate signaling and communication between various ovarian cell types. FOXL2, a transcription factor that plays a key role at different stages of ovarian development, is associated with primary ovarian insufficiency and ovarian cancer as a result of its loss-of-function or mutations. In this study, we investigated the impact of aberrant, constitutive expression of FOXL2 in somatic cells of the ovary. Overexpression of FOXL2 that started during fetal life resulted in defects in nest breakdown and consequent formation of polyovular follicles. Granulosa cell differentiation was impaired and recruitment and differentiation of steroidogenic theca cells was compromised. As a consequence, adult ovaries overexpressing FOXL2 exhibited defects in compartmentalization of granulosa and theca cells, significant decreased steroidogenesis and lack of ovulation. These findings demonstrate that fine-tuned expression of FOXL2 is required for proper folliculogenesis and fertility.


Asunto(s)
Proteína Forkhead Box L2/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Proteína Forkhead Box L2/genética , Células de la Granulosa/metabolismo , Ratones , Mutación , Ovario/crecimiento & desarrollo , Células Tecales/metabolismo
6.
Biol Reprod ; 103(5): 951-965, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32948877

RESUMEN

The transcription factor forkhead box L2 (FOXL2) regulates sex differentiation and reproductive function. Elevated levels of this transcription factor have been observed in the diseases of the uterus, such as endometriosis. However, the impact of elevated FOXL2 expression on uterine physiology remains unknown. In order to determine the consequences of altered FOXL2 in the female reproductive axis, we generated mice with over-expression of FOXL2 (FOXL2OE) by crossing Foxl2LsL/+ with the Progesterone receptor Pgrcre model. FOXL2OE uterus showed severe morphological abnormality including abnormal epithelial stratification, blunted adenogenesis, increased endometrial fibrosis, and disrupted myometrial morphology. In contrast, increasing FOXL2 levels specifically in uterine epithelium by crossing the Foxl2LsL/+ with the lactoferrin Ltficre mice resulted in the eFOXL2OE mice with uterine epithelial stratification but without defects in endometrial fibrosis and adenogenesis, demonstrating a role of the endometrial stroma in the uterine abnormalities of the FOXL2OE mice. Transcriptomic analysis of 12 weeks old Pgrcre and FOXL2OE uterus at diestrus stage showed multiple signaling pathways related with cellular matrix, wnt/ß-catenin, and altered cell cycle. Furthermore, we found FOXL2OE mice were sterile. The infertility was caused in part by a disruption of the hypophyseal ovarian axis resulting in an anovulatory phenotype. The FOXL2OE mice failed to show decidual responses during artificial decidualization in ovariectomized mice demonstrating the uterine contribution to the infertility phenotype. These data support that aberrantly increased FOXL2 expressions in the female reproductive tract can disrupt ovarian and uterine functions.


Asunto(s)
Proteína Forkhead Box L2/metabolismo , Anomalías Urogenitales/metabolismo , Útero/anomalías , Útero/metabolismo , Animales , Endometrio/metabolismo , Femenino , Proteína Forkhead Box L2/genética , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología , Transcriptoma , Anomalías Urogenitales/genética
7.
Development ; 143(20): 3700-3710, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27621062

RESUMEN

Testis morphogenesis is a highly orchestrated process involving lineage determination of male germ cells and somatic cell types. Although the origin and differentiation of germ cells are known, the developmental course specific for each somatic cell lineage has not been clearly defined. Here, we construct a comprehensive map of somatic cell lineage progression in the mouse testis. Both supporting and interstitial cell lineages arise from WT1+ somatic progenitor pools in the gonadal primordium. A subpopulation of WT1+ progenitor cells acquire SOX9 expression and become Sertoli cells that form testis cords, whereas the remaining WT1+ cells contribute to progenitor cells in the testis interstitium. Interstitial progenitor cells diversify through the acquisition of HES1, an indication of Notch activation, at the onset of sex determination. HES1+ interstitial progenitors, through the action of Sertoli cell-derived Hedgehog signals, become positive for GLI1. The GLI1+ interstitial cells eventually develop into two cell lineages: steroid-producing fetal Leydig cells and non-steroidogenic cells. The fetal Leydig cell population is restricted by Notch2 signaling from the neighboring somatic cells. The non-steroidogenic progenitor cells retain their undifferentiated state during fetal stage and become adult Leydig cells in post-pubertal testis. These results provide the first lineage progression map that illustrates the sequential establishment of somatic cell populations during testis morphogenesis.


Asunto(s)
Células Madre Adultas/citología , Células Madre/citología , Células Madre/metabolismo , Testículo/embriología , Testículo/metabolismo , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Células de Sertoli/citología , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tamoxifeno/farmacología , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
8.
Genesis ; 55(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28875587

RESUMEN

Recombinase responsive mouse lines expressing diphtheria toxin subunit A (DTA) are well established tools for targeted ablation of genetically defined cell populations. Here we describe a new knock-in allele at the Gt(Rosa)26Sor locus that retains the best features of previously described DTA alleles-including a CAG promoter, attenuated mutant DTA cDNA, and ubiquitous EGFP labeling-with the addition of a Cre-dependent FLEx switch for tight control of expression. The FLEx switch consists of two pairs of antiparallel lox sites requiring Cre-mediated recombination for inversion of the DTA to the proper orientation for transcription. We demonstrate its utility by Cre-dependent ablation of both a broad domain in the embryonic nervous system and a discrete population of cells in the fetal gonads. We conclude that this new DTA line is useful for targeted ablation of genetically-defined cell populations.


Asunto(s)
Toxina Diftérica/genética , Técnicas de Sustitución del Gen/métodos , Animales , Toxina Diftérica/metabolismo , Gónadas/citología , Gónadas/embriología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/genética , Integrasas/metabolismo , Ratones , Sistema Nervioso/citología , Sistema Nervioso/embriología , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Stem Cells ; 34(11): 2772-2783, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27350140

RESUMEN

In this study, we identify a novel and essential role for the Krüppel-like zinc finger transcription factor GLI-similar 3 (GLIS3) in the regulation of postnatal spermatogenesis. We show that GLIS3 is expressed in gonocytes, spermatogonial stem cells (SSCs) and spermatogonial progenitors (SPCs), but not in differentiated spermatogonia and later stages of spermatogenesis or in somatic cells. Spermatogenesis is greatly impaired in GLIS3 knockout mice. Loss of GLIS3 function causes a moderate reduction in the number of gonocytes, but greatly affects the generation of SSCs/SPCs, and as a consequence the development of spermatocytes. Gene expression profiling demonstrated that the expression of genes associated with undifferentiated spermatogonia was dramatically decreased in GLIS3-deficient mice and that the cytoplasmic-to-nuclear translocation of FOXO1, which marks the gonocyte-to-SSC transition and is necessary for SSC self-renewal, is inhibited. These observations suggest that GLIS3 promotes the gonocyte-to-SSC transition and is a critical regulator of the dynamics of early postnatal spermatogenesis. Stem Cells 2016;34:2772-2783.


Asunto(s)
Proteínas Represoras/genética , Espermatocitos/metabolismo , Espermatogénesis/genética , Espermatogonias/metabolismo , Células Madre/metabolismo , Testículo/metabolismo , Transactivadores/genética , Animales , Diferenciación Celular , Proteínas de Unión al ADN , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas , Proteínas Represoras/deficiencia , Espermatocitos/citología , Espermatogonias/citología , Células Madre/citología , Testículo/citología , Transactivadores/deficiencia
10.
Biol Reprod ; 93(2): 35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26108792

RESUMEN

Sex-reversal cases in humans and genetic models in mice have revealed that the fate of the bipotential gonad hinges upon the balance between pro-testis SOX9 and pro-ovary beta-catenin pathways. Our central query was: if SOX9 and beta-catenin define the gonad's identity, then what do the gonads become when both factors are absent? To answer this question, we developed mouse models that lack either Sox9, beta-catenin, or both in the somatic cells of the fetal gonads and examined the morphological outcomes and transcriptome profiles. In the absence of Sox9 and beta-catenin, both XX and XY gonads progressively lean toward the testis fate, indicating that expression of certain pro-testis genes requires the repression of the beta-catenin pathway, rather than a direct activation by SOX9. We also observed that XY double knockout gonads were more masculinized than their XX counterpart. To identify the genes responsible for the initial events of masculinization and to determine how the genetic context (XX vs. XY) affects this process, we compared the transcriptomes of Sox9/beta-catenin mutant gonads and found that early molecular changes underlying the XY-specific masculinization involve the expression of Sry and 21 SRY direct target genes, such as Sox8 and Cyp26b1. These results imply that when both Sox9 and beta-catenin are absent, Sry is capable of activating other pro-testis genes and drive testis differentiation. Our findings not only provide insight into the mechanism of sex determination, but also identify candidate genes that are potentially involved in disorders of sex development.


Asunto(s)
Gónadas/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/fisiología , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología , beta Catenina/genética , beta Catenina/fisiología , Animales , Sistema Enzimático del Citocromo P-450/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Embarazo , Ácido Retinoico 4-Hidroxilasa , Diferenciación Sexual/genética , Proteína de la Región Y Determinante del Sexo/genética , Testículo/embriología , Testículo/crecimiento & desarrollo , Transcriptoma/genética , Cromosoma X/genética , Cromosoma Y/genética
11.
Chromosome Res ; 20(1): 247-58, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22105695

RESUMEN

The chromosome status of the mammalian embryo initiates a multistage process of sexual development in which the bipotential reproductive system establishes itself as either male or female. These events are governed by intricate cell-cell and interorgan communication that is regulated by multiple signaling pathways. The hedgehog signaling pathway was originally identified for its key role in the development of Drosophila, but is now recognized as a critical developmental regulator in many species, including humans. In addition to its developmental roles, the hedgehog signaling pathway also modulates adult organ function, and misregulation of this pathway often leads to diseases, such as cancer. The hedgehog signaling pathway acts through its morphogenetic ligands that signal from ligand-producing cells to target cells over a specified distance. The target cells then respond in a graded manner based on the concentration of the ligands that they are exposed to. Through this unique mechanism of action, the hedgehog signaling pathway elicits cell fate determination, epithelial-mesenchymal interactions, and cellular homeostasis. Here, we review current findings on the roles of hedgehog signaling in the sexually dimorphic development of the reproductive organs with an emphasis on mammals and comparative evidence in other species.


Asunto(s)
Proteínas Hedgehog/genética , Diferenciación Sexual , Sexo , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Desarrollo Fetal , Regulación del Desarrollo de la Expresión Génica , Genes , Células Germinativas/citología , Células Germinativas/crecimiento & desarrollo , Células Germinativas/metabolismo , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Mamíferos , Reproducción , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo
12.
Front Endocrinol (Lausanne) ; 13: 910964, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846302

RESUMEN

In the 1940s, Alfred Jost demonstrated the necessity of testicular secretions, particularly androgens, for male internal and external genitalia differentiation. Since then, our knowledge of androgen impacts on differentiation of the male internal (Wolffian duct) and external genitalia (penis) has been drastically expanded upon. Between these two morphologically and functionally distinct organs, divergent signals facilitate the establishment of tissue-specific identities. Conversely, conserved actions of androgen signaling are present in both tissues and are largely responsible for the growth and expansion of the organs. In this review we synthesize the existing knowledge of the cell type-specific, organ specific, and conserved signaling mechanisms of androgens. Mechanistic studies on androgen signaling in the Wolffian duct and male external genitalia have largely been conducted in mouse model organisms. Therefore, the majority of the review is focused on mouse model studies.


Asunto(s)
Andrógenos , Receptores Androgénicos , Animales , Genitales Masculinos , Masculino , Ratones , Transducción de Señal , Sistema Urogenital
13.
Front Cell Dev Biol ; 10: 944776, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36158204

RESUMEN

Differentiation of the bipotential gonadal primordium into ovaries and testes is a common process among vertebrate species. While vertebrate ovaries eventually share the same functions of producing oocytes and estrogens, ovarian differentiation relies on different morphogenetic, cellular, and molecular cues depending on species. The aim of this review is to highlight the conserved and divergent features of ovarian differentiation through an evolutionary perspective. From teleosts to mammals, each clade or species has a different story to tell. For this purpose, this review focuses on three specific aspects of ovarian differentiation: ovarian morphogenesis, the evolution of the role of estrogens on ovarian differentiation and the molecular pathways involved in granulosa cell determination and maintenance.

14.
Hum Mol Genet ; 18(3): 405-17, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18981061

RESUMEN

Sexually dimorphic development of the gonads is controlled by positive and negative regulators produced by somatic cells. Many Wnt ligands, including ones that signal via the canonical beta-catenin pathway, are expressed in fetal gonads. beta-catenin, a key transcriptional regulator of the canonical Wnt pathway and an element of the cell adhesion complex, is essential for various aspects of embryogenesis. To study the involvement of beta-catenin in sex determination, we ablated beta-catenin specifically in the SF1-positive population of somatic cells. Although beta-catenin was present in gonads of both sexes, it was necessary only for ovarian differentiation but dispensable for testis development. Loss of beta-catenin in fetal testes did not affect Sertoli cell differentiation, testis morphogenesis or masculinization of the embryos. However, we observed molecular and morphological defects in ovaries lacking beta-catenin, including formation of testis-specific coelomic vessel, appearance of androgen-producing adrenal-like cells and loss of female germ cells. These phenotypes were strikingly similar to those found in the R-spondin1 (Rspo1) and Wnt4 knockout ovaries. In the absence of beta-catenin, expression of Wnt4 was down-regulated while that of Rspo1 was not affected, placing beta-catenin as a component in between Rspo1 and Wnt4. Our results demonstrate that beta-catenin is responsible for transducing sex-specific signals in the SF1-positive somatic cell population during mouse gonadal development.


Asunto(s)
Gónadas/crecimiento & desarrollo , Diferenciación Sexual , beta Catenina/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/embriología , Gónadas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factores de Empalme de ARN , Transducción de Señal , Especificidad de la Especie , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/genética
15.
Biol Reprod ; 84(5): 894-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21209421

RESUMEN

Appearance of mouse fetal Leydig cells requires activation of the Hedgehog pathway. Upon binding to the membrane-bound receptor patched, Hedgehog ligands induce intracellular responses via a combined effect of Gli transcription factors. Szczepny et al. (Biol Reprod 2009; 80:258-263) found that Gli1, one of the three Gli transcription factors, is present in the fetal testis and that its expression is suppressed by the Hedgehog inhibitor cyclopamine. In this study, we investigated the involvement of the Gli1 and Gli2 factors in mouse fetal Leydig cell differentiation. The Gli1 and Gli2 transcription factors showed an overlapping expression pattern in the testis interstitium at the time when fetal Leydig cells appear. Despite their similar expression, Gli1 and Gli2 patterns were differentially regulated. Initial Gli1 and Gli2 expression depends upon an active Hedgehog pathway; however, maintenance of only Gli1, but not Gli2, expression requires activation of the pathway. Inactivation of either the Gli1 or Gli2 gene did not affect fetal Leydig cell development and testis morphology, suggesting a functional redundancy. When the transcriptional activity of both GLI1 and GLI2 was suppressed by a selective inhibitor, GANT61, in cultured fetal testes before the appearance of fetal Leydig cells, Gli1 and Gli2 expression and steroidogenic marker activity were completely abolished. However at later stages when Leydig cells were already present, GANT61 treatment inhibited Gli1 expression but had no effects on Gli2 expression and fetal Leydig cell appearance. Our results reveal overlapping and redundant Gli1 and Gli2 roles in fetal Leydig cell differentiation and a novel regulation of Gli2 expression in the fetal testis.


Asunto(s)
Diferenciación Celular , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Genes Reporteros , Proteínas Hedgehog/antagonistas & inhibidores , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel/genética , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Piridinas/farmacología , Pirimidinas/farmacología , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptor Smoothened , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
16.
Dev Biol ; 329(1): 96-103, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19268447

RESUMEN

Proper cell fate determination in mammalian gonads is critical for the establishment of sexual identity. The Hedgehog (Hh) pathway has been implicated in cell fate decision for various organs, including gonads. Desert Hedgehog (Dhh), one of the three mammalian Hh genes, has been implicated with other genes in the establishment of mouse fetal Leydig cells. To investigate whether Hh alone is sufficient to induce fetal Leydig cell differentiation, we ectopically activated the Hh pathway in Steroidogenic factor 1 (SF1)-positive somatic cell precursors of fetal ovaries. Hh activation transformed SF1-positive somatic ovarian cells into functional fetal Leydig cells. These ectopic fetal Leydig cells produced androgens and insulin-like growth factor 3 (INLS3) that cause virilization of female embryos and ovarian descent. However, the female reproductive system remained intact, indicating a typical example of female pseudohermaphroditism. The appearance of fetal Leydig cells was a direct consequence of Hh activation as evident by the absence of other testicular components in the affected ovary. This study provides not only insights into mechanisms of cell lineage specification in gonads, but also a model to understand defects in sexual differentiation.


Asunto(s)
Trastornos del Desarrollo Sexual/metabolismo , Feto , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/metabolismo , Ovario/metabolismo , Animales , Diferenciación Celular/genética , Trastornos del Desarrollo Sexual/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/fisiología , Masculino , Ratones , Ratones Transgénicos , Diferenciación Sexual , Transducción de Señal/genética
17.
BMC Dev Biol ; 10: 66, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20540774

RESUMEN

BACKGROUND: The synthesis of microRNA (miRNA) is a multi-step process that requires the action of the ribonuclease Dicer1. Dicer1 is responsible for the final processing of miRNA and has been implicated in cellular processes such as proliferation, apoptosis, and differentiation. Mouse embryos lacking Dicer1 die in early embryogenesis. In this study, we investigated whether Dicer1 is required for development of adrenal, testis, and ovary in mouse embryos. RESULTS: To target Dicer1 deletion specifically in developing adrenals and gonads, we used Steroidogenic factor 1-cre (Sf1/Cre) line in which Cre recombinase is active in the progenitor cells of adrenals and gonads. Lack of Dicer1 in the SF1-positive cells did not affect formation and early differentiation of the adrenals and gonads. However, increasing numbers of apoptotic cells were first detected in the Dicer1 knockout adrenal cortex at 18.5 days post coitum (dpc), followed by apoptosis of somatic cells and germ cells in the testis at postnatal day 0. Affected adrenal and testes underwent complete degeneration 48 hrs after the onset of apoptosis. However, ovaries were not affected at least until postnatal day 5, when the animals died due to adrenal insufficiency. CONCLUSIONS: Dicer1 is dispensable for formation and differentiation of fetal tissues derived from the SF1-positive adrenogonadal primordium. Dicer1 is essential for maintaining cell survival in adrenal and testis; however, development of the ovary from fetal stages to postnatal day 5 does not require the presence of Dicer1. Our results reveal a tissue-specific requirement of Dicer1 and microRNAs. Future research is needed to understand how the tissue-specific role of Dicer1 is established.


Asunto(s)
Glándulas Suprarrenales/embriología , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/metabolismo , Ovario/embriología , Factor Esteroidogénico 1/metabolismo , Testículo/embriología , Glándulas Suprarrenales/citología , Glándulas Suprarrenales/metabolismo , Animales , Supervivencia Celular , Femenino , Masculino , Ratones , Ratones Noqueados , Ovario/citología , Ovario/metabolismo , Ribonucleasa III , Testículo/citología , Testículo/metabolismo
18.
Toxicol Appl Pharmacol ; 242(2): 224-30, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19874833

RESUMEN

Any insult that affects survival of ovarian antral follicles can cause abnormal estradiol production and fertility problems. Phthalate esters (PEs) are plasticizers used in a wide range of consumer and industrial products. Exposure to these chemicals has been linked to reduced fertility in humans and animal models. Di-(2-ethylhexyl) phthalate (DEHP) and mono-(2-ethylhexyl) phthalate (MEHP) decrease serum estradiol levels and aromatase (Arom) expression, prolong estrous cycles, and cause anovulation in animal and culture models. These observations suggest PEs directly target antral follicles. We therefore tested the hypothesis that DEHP (1-100 microg/ml) and MEHP (0.1-10 microg/ml) directly inhibit antral follicular growth and estradiol production. Antral follicles from adult mice were cultured with DEHP or MEHP, and/or estradiol for 96 h. During culture, follicle size was measured every 24 h as a measurement of follicle growth. After culture, media were collected for measurement of estradiol levels and follicles were subjected to measurement of cylin-D-2 (Ccnd2), cyclin-dependent-kinase-4 (Cdk4), and Arom. We found that DEHP and MEHP inhibited growth of follicles and decreased estradiol production compared to controls at the highest doses. DEHP and MEHP also decreased mRNA expression of Ccnd2, Cdk4, and Arom at the highest dose. Addition of estradiol to the culture medium prevented the follicles from DEHP- and MEHP-induced inhibition of growth, reduction in estradiol levels, and decreased Ccnd2 and Cdk4 expression. Collectively, our results indicate that DEHP and MEHP may directly inhibit antral follicle growth via a mechanism that partially includes reduction in levels of estradiol production and decreased expression of cell cycle regulators.


Asunto(s)
División Celular/efectos de los fármacos , Dietilhexil Ftalato/análogos & derivados , Dietilhexil Ftalato/farmacología , Estradiol/metabolismo , Folículo Ovárico/efectos de los fármacos , Animales , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Folículo Ovárico/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
19.
Sex Dev ; 14(1-6): 51-59, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33684916

RESUMEN

Sexually dimorphic establishment of the reproductive tract system requires sex-specific regression of the Wolffian duct and Müllerian duct in the mesonephros. In an XX embryo, the Wolffian duct regresses under the control of the mesenchymal transcription factor COUP-TFII. To understand cellular and molecular actions underlying Wolffian duct regression, we performed transcriptomic analyses of XX mesonephroi with or without Coup-tfII and genome-wide analysis of COUP-TFII chromatin occupancy in XX mesonephroi. The integrative analysis of COUP-TFII genome-wide binding and transcriptomic analysis revealed the suppression of muscle differentiation and extracellular matrix genes by COUP-TFII and identified a group of potential transcriptional partners of COUP-TFII in the mesenchyme that potentially facilitate Wolffian duct regression. These findings provide insights into the molecular action of COUP-TFII in the Wolffian duct mesenchyme and identify a list of biologically relevant candidate genes and pathways for future functional analyses in sexual differentiation of reproductive tracts.

20.
Front Genet ; 11: 511286, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193599

RESUMEN

Single-cell RNA sequencing (scRNA-seq) technologies have precipitated the development of bioinformatic tools to reconstruct cell lineage specification and differentiation processes with single-cell precision. However, current start-up costs and recommended data volumes for statistical analysis remain prohibitively expensive, preventing scRNA-seq technologies from becoming mainstream. Here, we introduce single-cell amalgamation by latent semantic analysis (SALSA), a versatile workflow that combines measurement reliability metrics with latent variable extraction to infer robust expression profiles from ultra-sparse sc-RNAseq data. SALSA uses a matrix focusing approach that starts by identifying facultative genes with expression levels greater than experimental measurement precision and ends with cell clustering based on a minimal set of Profiler genes, each one a putative biomarker of cluster-specific expression profiles. To benchmark how SALSA performs in experimental settings, we used the publicly available 10X Genomics PBMC 3K dataset, a pre-curated silver standard from human frozen peripheral blood comprising 2,700 single-cell barcodes, and identified 7 major cell groups matching transcriptional profiles of peripheral blood cell types and driven agnostically by < 500 Profiler genes. Finally, we demonstrate successful implementation of SALSA in a replicative scRNA-seq scenario by using previously published DropSeq data from a multi-batch mouse retina experimental design, thereby identifying 10 transcriptionally distinct cell types from > 64,000 single cells across 7 independent biological replicates based on < 630 Profiler genes. With these results, SALSA demonstrates that robust pattern detection from scRNA-seq expression matrices only requires a fraction of the accrued data, suggesting that single-cell sequencing technologies can become affordable and widespread if meant as hypothesis-generation tools to extract large-scale differential expression effects.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA