Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36912416

RESUMEN

During mammalian development, gonadal sex determination results from the commitment of bipotential supporting cells to Sertoli or granulosa cell fates. Typically, this decision is coordinated across the gonad to ensure commitment to a single organ fate. When unified commitment fails in an XY mouse, an ovotestis forms in which supporting cells in the center of the gonad typically develop as Sertoli cells, while supporting cells in the poles develop as granulosa cells. This central bias for Sertoli cell fate was thought to result from the initial expression of the drivers of Sertoli cell fate, SRY and/or SOX9, in the central domain, followed by paracrine expansion to the poles. However, we show here that the earliest cells expressing SRY and SOX9 are widely distributed across the gonad. In addition, Sertoli cell fate does not spread among supporting cells through paracrine relay. Instead, we uncover a center-biased pattern of supporting cell precursor ingression that occurs in both sexes and results in increased supporting cell density in the central domain. Our findings prompt a new model of gonad patterning in which a density-dependent organizing principle dominates Sertoli cell fate stabilization.


Asunto(s)
Gónadas , Procesos de Determinación del Sexo , Femenino , Ratones , Masculino , Animales , Gónadas/metabolismo , Células de Sertoli/metabolismo , Diferenciación Celular , Desarrollo Embrionario , Factor de Transcripción SOX9/metabolismo , Testículo/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Mamíferos/metabolismo
2.
PLoS Genet ; 19(3): e1010656, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36857387

RESUMEN

DND1 is essential to maintain germ cell identity. Loss of Dnd1 function results in germ cell differentiation to teratomas in some inbred strains of mice or to somatic fates in zebrafish. Using our knock-in mouse line in which a functional fusion protein between DND1 and GFP is expressed from the endogenous locus (Dnd1GFP), we distinguished two male germ cell (MGC) populations during late gestation cell cycle arrest (G0), consistent with recent reports of heterogeneity among MGCs. Most MGCs express lower levels of DND1-GFP (DND1-GFP-lo), but some MGCs express elevated levels of DND1-GFP (DND1-GFP-hi). A RNA-seq time course confirmed high Dnd1 transcript levels in DND1-GFP-hi cells along with 5-10-fold higher levels for multiple epigenetic regulators. Using antibodies against DND1-GFP for RNA immunoprecipitation (RIP)-sequencing, we identified multiple epigenetic and translational regulators that are binding targets of DND1 during G0 including DNA methyltransferases (Dnmts), histone deacetylases (Hdacs), Tudor domain proteins (Tdrds), actin dependent regulators (Smarcs), and a group of ribosomal and Golgi proteins. These data suggest that in DND1-GFP-hi cells, DND1 hosts coordinating mRNA regulons that consist of functionally related and localized groups of epigenetic enzymes and translational components.


Asunto(s)
Espermatogonias , Pez Cebra , Animales , Femenino , Masculino , Ratones , Embarazo , Cromatina/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Unión al ARN/genética , Espermatogonias/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
3.
Development ; 148(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-33795229

RESUMEN

During development of the mouse urogenital complex, the gonads undergo changes in three-dimensional structure, body position and spatial relationship with the mesonephric ducts, kidneys and adrenals. The complexity of genital ridge development obscures potential connections between morphogenesis and gonadal sex determination. To characterize the morphogenic processes implicated in regulating gonad shape and fate, we used whole-embryo tissue clearing and light sheet microscopy to assemble a time course of gonad development in native form and context. Analysis revealed that gonad morphology is determined through anterior-to-posterior patterns as well as increased rates of growth, rotation and separation in the central domain that may contribute to regionalization of the gonad. We report a close alignment of gonad and mesonephric duct movements as well as delayed duct development in a gonad dysgenesis mutant, which together support a mechanical dependency linking gonad and mesonephric duct morphogenesis.


Asunto(s)
Gónadas/fisiología , Morfogénesis/fisiología , Conductos Mesonéfricos/fisiología , Animales , Embrión de Mamíferos/fisiología , Femenino , Edad Gestacional , Riñón/fisiología , Masculino , Mesonefro/fisiología , Ratones , Ratones Endogámicos C57BL , Diferenciación Sexual/fisiología
4.
Nat Rev Genet ; 18(11): 675-689, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28804140

RESUMEN

The discovery of the Sry gene in 1990 triggered a revolution in our understanding of sex determination. More recently, advances in non-model organisms have been fuelled by the rapid evolution of affordable genome and transcriptome technologies. This Review considers the unusual plasticity in the bipotential system of sex determination and some of the diverse mechanisms that have evolved to control this critical developmental decision, including strong genetic pathways, environmental influences and epigenetic regulation. Ideas emerging from model and non-model organisms that suggest that sex determination operates as an antagonistic network with the emergent property of bistability are discussed.


Asunto(s)
Epigénesis Genética/fisiología , Modelos Genéticos , Procesos de Determinación del Sexo/fisiología , Vertebrados/fisiología , Animales , Perfilación de la Expresión Génica
5.
Annu Rev Cell Dev Biol ; 25: 457-82, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19807280

RESUMEN

A critical element of successful sexual reproduction is the generation of sexually dimorphic adult reproductive organs, the testis and ovary, which produce functional gametes. Examination of different vertebrate species shows that the adult gonad is remarkably similar in its morphology across different phylogenetic classes. Surprisingly, however, the cellular and molecular programs employed to create similar organs are not evolutionarily conserved. We highlight the mechanisms used by different vertebrate model systems to generate the somatic architecture necessary to support gametogenesis. In addition, we examine the different vertebrate patterns of germ cell migration from their site of origin to colonize the gonad and highlight their roles in sex-specific morphogenesis. We also discuss the plasticity of the adult gonad and consider how different genetic and environmental conditions can induce transitions between testis and ovary morphology.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Ovario/embriología , Testículo/embriología , Vertebrados/embriología , Animales , Movimiento Celular , Femenino , Humanos , Masculino , Morfogénesis , Ovario/metabolismo , Diferenciación Sexual , Testículo/metabolismo
6.
Development ; 146(19)2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31253634

RESUMEN

The adult spermatogonial stem cell population arises from pluripotent primordial germ cells (PGCs) that enter the fetal testis around embryonic day (E)10.5. PGCs undergo rapid mitotic proliferation, then enter prolonged cell cycle arrest (G1/G0), during which they transition to pro-spermatogonia. In mice homozygous for the Ter mutation in the RNA-binding protein Dnd1 (Dnd1Ter/Ter ), many male germ cells (MGCs) fail to enter G1/G0 and instead form teratomas: tumors containing many embryonic cell types. To investigate the origin of these tumors, we sequenced the MGC transcriptome in Dnd1Ter/Ter mutants at E12.5, E13.5 and E14.5, immediately prior to teratoma formation, and correlated this information with DO-RIP-Seq-identified DND1 direct targets. Consistent with previous results, we found DND1 controls downregulation of many genes associated with pluripotency and active cell cycle, including mTor, Hippo and Bmp/Nodal signaling pathway elements. However, DND1 targets also include genes associated with male differentiation, including a large group of chromatin regulators activated in wild-type but not mutant MGCs during the E13.5 and E14.5 transition. Results suggest multiple DND1 functions and link DND1 to initiation of epigenetic modifications in MGCs.


Asunto(s)
Reprogramación Celular/genética , Epigénesis Genética , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/genética , Secuencia de Bases , Ciclo Celular/genética , Cromatina/metabolismo , Elementos Transponibles de ADN/genética , Regulación hacia Abajo/genética , Embrión de Mamíferos/citología , Femenino , Homocigoto , Masculino , Ratones , Mutación/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Transcripción Genética , Regulación hacia Arriba/genética
7.
Proc Natl Acad Sci U S A ; 116(12): 5570-5575, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30819894

RESUMEN

Testes and ovaries undergo sex-specific morphogenetic changes and adopt strikingly different morphologies, despite the fact that both arise from a common precursor, the bipotential gonad. Previous studies showed that recruitment of vasculature is critical for testis patterning. However, vasculature is not recruited into the early ovary. Peripheral innervation is involved in patterning development of many organs but has been given little attention in gonad development. In this study, we show that while innervation in the male reproductive complex is restricted to the epididymis and vas deferens and never invades the interior of the testis, neural crest-derived innervation invades the interior of the ovary around E16.5. Individual neural crest cells colonize the ovary, differentiate into neurons and glia, and form a dense neural network within the ovarian medulla. Using a sex-reversing mutant mouse line, we show that innervation is specific to ovary development, is not dependent on the genetic sex of gonadal or neural crest cells, and may be blocked by repressive guidance signals elevated in the male pathway. This study reveals another aspect of sexually dimorphic gonad development, establishes a precise timeline and structure of ovarian innervation, and raises many questions for future research.


Asunto(s)
Gónadas/inervación , Ovario/inervación , Testículo/inervación , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Gónadas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Morfogénesis , Cresta Neural/inervación , Neuronas/fisiología , Ovario/citología , Caracteres Sexuales , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología , Diferenciación Sexual/genética , Testículo/citología
8.
PLoS Genet ; 15(5): e1007895, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31116734

RESUMEN

XX and XY fetal gonads are initially bipotential, poised between the ovary and testis fate. Multiple lines of evidence suggest that commitment to testis fate requires the repression of genes associated with ovary fate. It was previously shown that loss of CBX2, the subunit of the Polycomb Repressive Complex 1 (PRC1) that binds H3K27me3 and mediates silencing, leads to ovary development in XY mice and humans. While it had been proposed that CBX2 is an activator of the testis-determining gene Sry, we investigated the alternative possibility that CBX2 has a direct role as a repressor of the antagonistic ovary-promoting pathway. To investigate this possibility, we developed a quantitative genome-wide profile of the repressive histone mark H3K27me3 and its active counterpart H3K4me3 in isolated XY and XX gonadal supporting cells before and after sex determination. We show that testis and ovary sex-determining (SD) genes are bivalent before sex determination, providing insight into how the bipotential state of the gonad is established at the epigenetic level. After sex determination, many SD genes of the alternate pathway remain bivalent, possibly contributing to the ability of these cells to transdifferentiate even in adults. The finding that many genes in the Wnt signaling pathway were targeted for H3K27me3-mediated repression in Sertoli cells led us to test whether deletion of Wnt4 could rescue testis development in Cbx2 mutants. We show that Sry expression and testis development were rescued in XY Cbx2-/-;Wnt4-/- mice. Furthermore, we show that CBX2 directly binds the downstream Wnt signaler Lef1, an ovary-promoting gene that remains bivalent in Sertoli cells. Our results suggest that stabilization of the testis fate requires CBX2-mediated repression of bivalent ovary-determining genes, which would otherwise block testis development.


Asunto(s)
Epigénesis Genética , Ovario/metabolismo , Complejo Represivo Polycomb 1/genética , Procesos de Determinación del Sexo , Testículo/metabolismo , Vía de Señalización Wnt/genética , Animales , Embrión de Mamíferos , Femenino , Factor 9 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Ovario/citología , Ovario/crecimiento & desarrollo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Complejo Represivo Polycomb 1/deficiencia , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Diferenciación Sexual , Testículo/citología , Testículo/crecimiento & desarrollo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
9.
Biol Reprod ; 105(4): 958-975, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34007995

RESUMEN

Testis differentiation is initiated when Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. Sertoli cells are essential for testis development, but cell types within the interstitial compartment, such as immune and endothelial cells, are also critical for organ formation. Our previous work implicated macrophages in fetal testis morphogenesis, but little is known about genes underlying immune cell development during organogenesis. Here, we examine the role of the immune-associated genes Mafb and Maf in mouse fetal gonad development, and we demonstrate that deletion of these genes leads to aberrant hematopoiesis manifested by supernumerary gonadal monocytes. Mafb; Maf double knockout embryos underwent initial gonadal sex determination normally, but exhibited testicular hypervascularization, testis cord formation defects, Leydig cell deficit, and a reduced number of germ cells. In general, Mafb and Maf alone were dispensable for gonad development; however, when both genes were deleted, we observed significant defects in testicular morphogenesis, indicating that Mafb and Maf work redundantly during testis differentiation. These results demonstrate previously unappreciated roles for Mafb and Maf in immune and vascular development and highlight the importance of interstitial cells in gonadal differentiation.


Asunto(s)
Factor de Transcripción MafB/genética , Células Mieloides/metabolismo , Organogénesis/genética , Proteínas Proto-Oncogénicas c-maf/genética , Testículo/embriología , Animales , Embrión de Mamíferos/embriología , Factor de Transcripción MafB/metabolismo , Masculino , Ratones , Proteínas Proto-Oncogénicas c-maf/metabolismo
10.
Biol Reprod ; 104(4): 861-874, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33394034

RESUMEN

In vertebrates, the RNA-binding protein (RBP) dead end 1 (DND1) is essential for primordial germ cell (PGC) survival and maintenance of cell identity. In multiple species, Dnd1 loss or mutation leads to severe PGC loss soon after specification or, in some species, germ cell transformation to somatic lineages. Our investigations into the role of DND1 in PGC specification and differentiation have been limited by the absence of an available antibody. To address this problem, we used CRISPR/Cas9 gene editing to establish a transgenic mouse line carrying a DND1GFP fusion allele. We present imaging analysis of DND1GFP expression showing that DND1GFP expression is heterogeneous among male germ cells (MGCs) and female germ cells (FGCs). DND1GFP was detected in MGCs throughout fetal life but lost from FGCs at meiotic entry. In postnatal and adult testes, DND1GFP expression correlated with classic markers for the premeiotic spermatogonial population. Utilizing the GFP tag for RNA immunoprecipitation (RIP) analysis in MGCs validated this transgenic as a tool for identifying in vivo transcript targets of DND1. The DND1GFP mouse line is a novel tool for isolation and analysis of embryonic and fetal germ cells, and the spermatogonial population of the postnatal and adult testis.


Asunto(s)
Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas de Neoplasias/genética , Alelos , Animales , Diferenciación Celular/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Células Germinativas/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión/genética
11.
Reproduction ; 162(2): 141-147, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34085952

RESUMEN

Sertoli cells proliferate and construct seminiferous tubules during fetal life, then undergo differentiation and maturation in the prepubertal testes. In the adult testes, mature Sertoli cells maintain spermatogonia and support spermatogenesis during the entire lifetime. Although Sertoli-like cells have been derived from iPS cells, they tend to remain immature. To investigate whether Sertoli cells can spontaneously acquire the ability to support spermatogenesis when transferred into the adult testis, we transplanted mouse fetal testicular cells into a Sertoli-depleted adult testis. We found that donor E12.5, E14.5 and E16.5 Sertoli cells colonized adult seminiferous tubules and supported host spermatogenesis 2 months after transplantation, demonstrating that immature fetal Sertoli cells can undergo sufficient maturation in the adult testis to become functional. This technique will be useful to analyze the developmental process of Sertoli cell maturation and to investigate the potential of iPS-derived Sertoli cells to colonize, undergo maturation, and support spermatogenesis within the testis environment.


Asunto(s)
Diferenciación Celular , Feto/citología , Células de Sertoli/citología , Células de Sertoli/trasplante , Espermatogénesis , Testículo/citología , Animales , Femenino , Masculino , Ratones , Embarazo , Túbulos Seminíferos/citología , Túbulos Seminíferos/fisiología , Maduración Sexual , Testículo/fisiología
12.
Development ; 144(9): 1607-1618, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28360133

RESUMEN

During early gonadogenesis, proliferating cells in the coelomic epithelium (CE) give rise to most of the somatic cells in both XX and XY gonads. Previous dye-labeling experiments showed that a single CE cell could give rise to additional CE cells and to both supporting and interstitial cell lineages, implying that cells in the CE domain are multipotent progenitors, and suggesting that an asymmetric division is involved in the acquisition of gonadal cell fates. We found that NUMB is asymmetrically localized in CE cells, suggesting that it might be involved. To test this hypothesis, we conditionally deleted Numb on a Numbl mutant background just prior to gonadogenesis. Mutant gonads showed a loss of cell polarity in the surface epithelial layers, large interior cell patches expressing the undifferentiated cell marker LHX9, and a loss of differentiated cells in somatic cell lineages. These results indicate that NUMB is necessary for establishing polarity in CE cells, and that asymmetric divisions resulting from CE polarity are required for commitment to differentiated somatic cell fates. Surprisingly, germ cells, which do not arise from the CE, were also affected in mutants, which may be a direct or indirect effect of loss of Numb.


Asunto(s)
Linaje de la Célula , Gónadas/embriología , Gónadas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Organogénesis , Animales , División Celular Asimétrica/efectos de los fármacos , Recuento de Células , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Linaje de la Célula/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Polaridad Celular/genética , Células Cultivadas , Dipéptidos/farmacología , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gónadas/efectos de los fármacos , Gónadas/patología , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Homeodominio LIM/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Modelos Biológicos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Organogénesis/efectos de los fármacos , Organogénesis/genética , Fenotipo , Receptores Notch/genética , Receptores Notch/metabolismo , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
13.
Development ; 144(12): 2222-2233, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506988

RESUMEN

The molecular mechanism underlying temperature-dependent sex determination (TSD) has been a long-standing mystery; in particular, the thermosensitive genetic triggers for gonadal sex differentiation are largely unknown. Here, we have characterized a conserved DM domain gene, Dmrt1, in the red-eared slider turtle Trachemys scripta (T. scripta), which exhibits TSD. We found that Dmrt1 has a temperature-dependent, sexually dimorphic expression pattern, preceding gonadal sex differentiation, and is capable of responding rapidly to temperature shifts and aromatase inhibitor treatment. Most importantly, loss- and gain-of-function analyses provide solid evidence that Dmrt1 is both necessary and sufficient to initiate male development in T. scripta Furthermore, the DNA methylation dynamics of the Dmrt1 promoter are tightly correlated with temperature and could mediate the impact of temperature on sex determination. Collectively, our findings demonstrate that Dmrt1 is a candidate master male sex-determining gene in this TSD species, consistent with the idea that DM domain genes are conserved during the evolution of sex determination mechanisms.


Asunto(s)
Procesos de Determinación del Sexo/genética , Factores de Transcripción/genética , Tortugas/embriología , Tortugas/genética , Animales , Animales Modificados Genéticamente , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Lentivirus/genética , Masculino , Filogenia , Regiones Promotoras Genéticas , Caracteres Sexuales , Procesos de Determinación del Sexo/fisiología , Temperatura , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Tortugas/metabolismo
14.
Development ; 144(4): 720-730, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087634

RESUMEN

A current goal of molecular biology is to identify transcriptional networks that regulate cell differentiation. However, identifying functional gene regulatory elements has been challenging in the context of developing tissues where material is limited and cell types are mixed. To identify regulatory sites during sex determination, we subjected Sertoli cells from mouse fetal testes to DNaseI-seq and ChIP-seq for H3K27ac. DNaseI-seq identified putative regulatory sites around genes enriched in Sertoli and pregranulosa cells; however, active enhancers marked by H3K27ac were enriched proximal to only Sertoli-enriched genes. Sequence analysis identified putative binding sites of known and novel transcription factors likely controlling Sertoli cell differentiation. As a validation of this approach, we identified a novel Sertoli cell enhancer upstream of Wt1, and used it to drive expression of a transgenic reporter in Sertoli cells. This work furthers our understanding of the complex genetic network that underlies sex determination and identifies regions that potentially harbor non-coding mutations underlying disorders of sexual development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Elementos Reguladores de la Transcripción , Células de Sertoli/metabolismo , Animales , Sitios de Unión , Diferenciación Celular , Desoxirribonucleasa I/metabolismo , Elementos de Facilitación Genéticos , Genes Reporteros , Genoma , Histonas/metabolismo , Homocigoto , Masculino , Ratones , Mutación , Regiones Promotoras Genéticas , Procesos de Determinación del Sexo , Testículo/embriología , Transgenes
15.
Biol Reprod ; 102(5): 1080-1089, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31965156

RESUMEN

At any given time, the ovary contains a number of follicles in distinct growth stages, each with a set of identifying characteristics. Although follicle counting and staging using histological stains on paraffin-embedded ovary sections has been the gold standard in assessing ovarian health in fertility studies, the final counts rely on extrapolation factors that diverge greatly among studies. These methods also limit our ability to investigate spatial aspects of ovary organization. Recent advances in optical tissue clearing and lightsheet microscopy have permitted comprehensive analysis of intact tissues. In this study, we set out to determine the best clearing and imaging methods to generate 3D images of the complete adult mouse ovary that could be used for accurate assessments of ovarian follicles. We found that a combination of iDISCO and CUBIC was the best method to clear the immunostained ovary. Using lightsheet microscopy, we generated 3D images of the intact ovary and performed qualitative assessments of follicles at all stages of development. This study is an important step toward developing quantitative computational models that allow rapid and accurate assessments of growing and quiescent primordial follicles, and to investigate the integrity of extrinsic ovarian components including vascular and neuronal networks.


Asunto(s)
Microscopía Fluorescente/métodos , Ovario/anatomía & histología , Manejo de Especímenes/métodos , Animales , Femenino , Colorantes Fluorescentes/metabolismo , Genes Transgénicos Suicidas , Ratones , Ratones Endogámicos C57BL
16.
Reproduction ; 158(6): F101-F111, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31265995

RESUMEN

Work that established the testis as the driver of male development, and the Y chromosome as the bearer of the male-determining gene, established a working model, and set the stage for the molecular age of mammalian sex determination. The discovery and characterization of Sry/SRY at the top of the hierarchy in mammals launched the field in two major directions. The first was to identify the downstream transcription factors and other molecular players that drive the bifurcation of Sertoli and granulosa cell differentiation. The second major direction was to understand organogenesis of the early bipotential gonad, and how divergence of its two distinct morphogenetic pathways (testis and ovary) is regulated at the cellular level. This review will summarize the early discoveries soon after Sry was identified and focus on my study of the gonad as a model of organogenesis.


Asunto(s)
Investigación Biomédica/historia , Diferenciación Celular , Organogénesis , Ovario/citología , Reproducción , Testículo/citología , Cromosoma Y/genética , Animales , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Masculino , Ratones , Ovario/metabolismo , Testículo/metabolismo , Estados Unidos
17.
Dev Biol ; 420(1): 166-177, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27671871

RESUMEN

Temperature dependent sex determination (TSD) is the process by which the environmental temperature experienced during embryogenesis influences the sex of an organism, as in the red-eared slider turtle Trachemys scripta elegans. In accord with current paradigms of vertebrate sex determination, temperature is believed to exert its effects on sexual development in T. scripta entirely within the middle third of development, when the gonad is forming. However, whether temperature regulates the transcriptome in T. scripta early embryos in a manner that could influence secondary sex characteristics or establish a pro-male or pro-female environment has not been investigated. In addition, apart from a handful of candidate genes, very little is known about potential similarities between the expression cascade during TSD and the genetic cascade that drives mammalian sex determination. Here, we conducted an unbiased transcriptome-wide analysis of the effects of male- and female-promoting temperatures on the turtle embryo prior to gonad formation, and on the gonad during the temperature sensitive period. We found sexually dimorphic expression reflecting differences in steroidogenic enzymes and brain development prior to gonad formation. Within the gonad, we mapped a cascade of differential expression similar to the genetic cascade established in mammals. Using a Hidden Markov Model based clustering approach, we identified groups of genes that show heterochronic shifts between M. musculus and T. scripta. We propose a model in which multiple factors influenced by temperature accumulate during early gonadogenesis, and converge on the antagonistic regulation of aromatase to canalize sex determination near the end of the temperature sensitive window of development.


Asunto(s)
Gónadas/crecimiento & desarrollo , Desarrollo Sexual , Temperatura , Tortugas/crecimiento & desarrollo , Animales , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Hormonas/biosíntesis , Masculino , Mamíferos/genética , Cadenas de Markov , Ratones , Especificidad de Órganos , Organogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Desarrollo Sexual/genética , Especificidad de la Especie , Esteroides/biosíntesis , Factores de Tiempo , Transcriptoma/genética , Tortugas/genética
18.
Proc Natl Acad Sci U S A ; 111(23): E2384-93, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912173

RESUMEN

Organogenesis of the testis is initiated when expression of Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. The cells in the early bipotential gonad undergo de novo organization to form testis cords that enclose germ cells inside tubules lined by epithelial Sertoli cells. Although Sertoli cells are a driving force in the de novo formation of testis cords, recent studies in mouse showed that reorganization of the vasculature and of interstitial cells also play critical roles in testis cord morphogenesis. However, the mechanism driving reorganization of the vasculature during fetal organogenesis remained unclear. Here we demonstrate that fetal macrophages are associated with nascent gonadal and mesonephric vasculature during the initial phases of testis morphogenesis. Macrophages mediate vascular reorganization and prune errant germ cells and somatic cells after testis architecture is established. We show that gonadal macrophages are derived from primitive yolk-sac hematopoietic progenitors and exhibit hallmarks of M2 activation status, suggestive of angiogenic and tissue remodeling functions. Depletion of macrophages resulted in impaired vascular reorganization and abnormal cord formation. These findings reveal a previously unappreciated role for macrophages in testis morphogenesis and suggest that macrophages are an intermediary between neovascularization and organ architecture during fetal organogenesis.


Asunto(s)
Macrófagos/metabolismo , Morfogénesis , Testículo/irrigación sanguínea , Testículo/embriología , Animales , Receptor 1 de Quimiocinas CX3C , Linaje de la Célula , Feto/irrigación sanguínea , Feto/citología , Feto/embriología , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Células Mieloides/metabolismo , Técnicas de Cultivo de Órganos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Testículo/citología , Factores de Tiempo , Saco Vitelino/metabolismo
19.
Genes Dev ; 23(21): 2521-36, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19884258

RESUMEN

Despite the identification of some key genes that regulate sex determination, most cases of disorders of sexual development remain unexplained. Evidence suggests that the sexual fate decision in the developing gonad depends on a complex network of interacting factors that converge on a critical threshold. To elucidate the transcriptional network underlying sex determination, we took the first expression quantitative trait loci (eQTL) approach in a developing organ. We identified reproducible differences in the transcriptome of the embryonic day 11.5 (E11.5) XY gonad between C57BL/6J (B6) and 129S1/SvImJ (129S1), indicating that the reported sensitivity of B6 to sex reversal is consistent with a higher expression of a female-like transcriptome in B6. Gene expression is highly variable in F2 XY gonads from B6 and 129S1 intercrosses, yet strong correlations emerged. We estimated the F2 coexpression network and predicted roles for genes of unknown function based on their connectivity and position within the network. A genetic analysis of the F2 population detected autosomal regions that control the expression of many sex-related genes, including Sry (sex-determining region of the Y chromosome) and Sox9 (Sry-box containing gene 9), the key regulators of male sex determination. Our results reveal the complex transcription architecture underlying sex determination, and provide a mechanism by which individuals may be sensitized for sex reversal.


Asunto(s)
Trastornos del Desarrollo Sexual , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Procesos de Determinación del Sexo , Animales , Cruzamiento , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Variación Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Sitios de Carácter Cuantitativo/genética , Factor de Transcripción SOX9/metabolismo , Cromosoma X/genética , Cromosoma Y/genética
20.
Mol Hum Reprod ; 22(12): 842-851, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27698028

RESUMEN

STUDY QUESTION: Can host fertility be rescued by grafting of a fragment of a healthy ovary soon after chemotherapy? SUMMARY ANSWER: We found that grafting a green fluorescent protein (GFP)-positive fragment from a healthy isogenic ovary to the left ovary of a chemo-treated host rescued function and fertility of the grafted host ovary, and resulted in the production of host-derived offspring as late as the sixth litter after chemotherapy (CTx) treatment, whereas none of the ungrafted controls produced a second litter. WHAT IS KNOWN ALREADY: In women and girls undergoing chemotherapy, infertility and premature ovarian failure are frequent outcomes. There are accumulating reports of improved endocrine function after autotransplantation of an ovarian fragment, raising the possibility that the transplant is beneficial to the endogenous ovary. STUDY DESIGN, SIZE, DURATION: We first established a CTx treatment regimen that resulted in the permanent loss of fertility in 100% of female mice of the FVB inbred strain. We grafted an isogenic ovary fragment from a healthy female homozygous for a GFP transgene to the left ovary of 100 CTx-treated hosts, and compared fertility to 39 ungrafted controls in 6 months of continuous matings, using GFP to distinguish offspring derived from the graft, and those derived from the host. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immunofluoresece and western blot analysis of 39 treated ovaries during and 15 days after CTx treatment revealed elevated apoptosis, rapid loss of granulosa cells and an increased recruitment of growing follicles. Using immunofluorescence and confocal imaging, we tracked the outcome of the grafted tissue over 4 months and its effect on the adjacent and contralateral ovary of the host. MAIN RESULTS AND THE ROLE OF CHANCE: Fifty-three percent of grafted females produced a second litter whereas none of the ungrafted females produced a second litter. The likelihood that this could occur by chance is very low (P < 0.0001). LIMITATIONS, REASONS FOR CAUTION: These results are shown only in mice, and whether or how they might apply to chemotherapy patients subjected to different CTx regimens is not yet clear. WIDER IMPLICATIONS OF THE FINDINGS: Our experiments prove that rescue of a chemo-treated ovary is possible, and establish a system to investigate the mechanism of rescue and to identify the factors responsible with the long-term goal of developing therapies for preservation of ovarian endocrine function and fertility in women undergoing chemotherapy. LARGE SCALE DATA: No large datasets were produced. STUDY FUNDING/COMPETING INTERESTS: Duke University Medical Center Chancellor's Discovery Grant to BC; ESJ was supported by an NRSA 5F31CA165545; SK was supported by NIH RO1 GM08033; RWT was supported by the Duke University School of Medicine Ovarian Cancer Research Fellowship; XBM was supported by CONICYT. The authors have no conflicts of interest to declare.


Asunto(s)
Ovario/trasplante , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Busulfano/efectos adversos , Ciclofosfamida/efectos adversos , Femenino , Preservación de la Fertilidad/métodos , Células de la Granulosa/efectos de los fármacos , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Masculino , Ratones , Oocitos/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/fisiología , Ovario/cirugía , Insuficiencia Ovárica Primaria/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA