Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 18(2): e1010088, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35192609

RESUMEN

The mammalian nuclear hormone receptors LRH1 (NR5A2) and SF1 (NR5A1) are close paralogs that can bind the same DNA motif and play crucial roles in gonadal development and function. Lrh1 is essential for follicle development in the ovary and has been proposed to regulate steroidogenesis in the testis. Lrh1 expression in the testis is highly elevated by loss of the sex regulator Dmrt1, which triggers male-to-female transdifferentiation of Sertoli cells. While Sf1 has a well-defined and crucial role in testis development, no function for Lrh1 in the male gonad has been reported. Here we use conditional genetics to examine Lrh1 requirements both in gonadal cell fate reprogramming and in normal development of the three major cell lineages of the mouse testis. We find that loss of Lrh1 suppresses sexual transdifferentiation, confirming that Lrh1 can act as a key driver in reprogramming sexual cell fate. In otherwise wild-type testes, we find that Lrh1 is dispensable in Leydig cells but is required in Sertoli cells for their proliferation, for seminiferous tubule morphogenesis, for maintenance of the blood-testis barrier, for feedback regulation of androgen production, and for support of spermatogenesis. Expression profiling identified misexpressed genes likely underlying most aspects of the Sertoli cell phenotype. In the germ line we found that Lrh1 is required for maintenance of functional spermatogonia, and hence mutants progressively lose spermatogenesis. Reduced expression of the RNA binding factor Nxf2 likely contributes to the SSC defect. Unexpectedly, however, over time the Lrh1 mutant germ line recovered abundant spermatogenesis and fertility. This finding indicates that severe germ line depletion triggers a response allowing mutant spermatogonia to recover the ability to undergo complete spermatogenesis. Our results demonstrate that Lrh1, like Sf1, is an essential regulator of testis development and function but has a very distinct repertoire of functions.


Asunto(s)
Células de Sertoli , Testículo , Animales , Femenino , Masculino , Mamíferos , Ratones , Diferenciación Sexual , Espermatogénesis/genética , Espermatogonias , Testículo/metabolismo
2.
Nucleic Acids Res ; 49(11): 6144-6164, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34096593

RESUMEN

Mammalian sexual development commences when fetal bipotential progenitor cells adopt male Sertoli (in XY) or female granulosa (in XX) gonadal cell fates. Differentiation of these cells involves extensive divergence in chromatin state and gene expression, reflecting distinct roles in sexual differentiation and gametogenesis. Surprisingly, differentiated gonadal cell fates require active maintenance through postnatal life to prevent sexual transdifferentiation and female cell fate can be reprogrammed by ectopic expression of the sex regulator DMRT1. Here we examine how DMRT1 reprograms granulosa cells to Sertoli-like cells in vivo and in culture. We define postnatal sex-biased gene expression programs and identify three-dimensional chromatin contacts and differentially accessible chromatin regions (DARs) associated with differentially expressed genes. Using a conditional transgene we find DMRT1 only partially reprograms the ovarian transcriptome in the absence of SOX9 and its paralog SOX8, indicating that these factors functionally cooperate with DMRT1. ATAC-seq and ChIP-seq show that DMRT1 induces formation of many DARs that it binds with SOX9, and DMRT1 is required for binding of SOX9 at most of these. We suggest that DMRT1 can act as a pioneer factor to open chromatin and allow binding of SOX9, which then cooperates with DMRT1 to reprogram sexual cell fate.


Asunto(s)
Reprogramación Celular/genética , Células de la Granulosa/metabolismo , Factor de Transcripción SOX9/metabolismo , Células de Sertoli/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , ADN/metabolismo , Femenino , Masculino , Ratones , Elementos Reguladores de la Transcripción , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Transcriptoma
3.
EMBO Rep ; 20(8): e48577, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31267656

RESUMEN

In most animals, sexual reproduction results in a 1:1 ratio of females to males. For several sectors of agriculture, for example, milk or egg production, only a single sex is needed. Biasing the sex ratio so that only offspring of the desired sex are produced has the potential to increase breeding efficiency. In this issue of EMBO Reports, Yosef et al [1] demonstrate a genetic approach to bias the sex ratio in mice by specifically disrupting essential genes in male embryos. Their approach is an important first step toward generating sex-ratio biasing applications for agriculture.


Asunto(s)
Mamíferos , Razón de Masculinidad , Animales , Cruzamiento , Femenino , Técnicas Genéticas , Masculino , Ratones , Reproducción
4.
Development ; 144(22): 4137-4147, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28982686

RESUMEN

The mitosis-meiosis transition is essential for spermatogenesis. Specific and timely downregulation of the transcription factor DMRT1, and consequent induction of Stra8 expression, is required for this process in mammals, but the molecular mechanism has remained unclear. Here, we show that ß-TrCP, the substrate recognition component of an E3 ubiquitin ligase complex, targets DMRT1 for degradation and thereby controls the mitosis-meiosis transition in mouse male germ cells. Conditional inactivation of ß-TrCP2 in male germ cells of ß-TrCP1 knockout mice resulted in sterility due to a lack of mature sperm. The ß-TrCP-deficient male germ cells did not enter meiosis, but instead underwent apoptosis. The induction of Stra8 expression was also attenuated in association with the accumulation of DMRT1 at the Stra8 promoter in ß-TrCP-deficient testes. DMRT1 contains a consensus ß-TrCP degron sequence that was found to bind ß-TrCP. Overexpression of ß-TrCP induced the ubiquitylation and degradation of DMRT1. Heterozygous deletion of Dmrt1 in ß-TrCP-deficient spermatogonia increased meiotic cells with a concomitant reduction of apoptosis. Collectively, our data indicate that ß-TrCP regulates the transition from mitosis to meiosis in male germ cells by targeting DMRT1 for degradation.


Asunto(s)
Meiosis , Mitosis , Espermatozoides/citología , Espermatozoides/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Fertilidad , Eliminación de Gen , Marcación de Gen , Heterocigoto , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteolisis , Túbulos Seminíferos/patología , Espermatogénesis , Especificidad por Sustrato , Testículo/patología , Factores de Transcripción/metabolismo , Ubiquitinación , Proteínas con Repetición de beta-Transducina/química , Proteínas con Repetición de beta-Transducina/metabolismo
5.
J Neurosci ; 38(42): 9105-9121, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30143575

RESUMEN

Specification of dorsoventral regional identity in progenitors of the developing telencephalon is a first pivotal step in the development of the cerebral cortex and basal ganglia. Previously, we demonstrated that the two zinc finger doublesex and mab-3 related (Dmrt) genes, Dmrt5 (Dmrta2) and Dmrt3, which are coexpressed in high caudomedial to low rostrolateral gradients in the cerebral cortical primordium, are separately needed for normal formation of the cortical hem, hippocampus, and caudomedial neocortex. We have now addressed the role of Dmrt3 and Dmrt5 in controlling dorsoventral division of the telencephalon in mice of either sex by comparing the phenotypes of single knock-out (KO) with double KO embryos and by misexpressing Dmrt5 in the ventral telencephalon. We find that DMRT3 and DMRT5 act as critical regulators of progenitor cell dorsoventral identity by repressing ventralizing regulators. Early ventral fate transcriptional regulators expressed in the dorsal lateral ganglionic eminence, such as Gsx2, are upregulated in the dorsal telencephalon of Dmrt3;Dmrt5 double KO embryos and downregulated when ventral telencephalic progenitors express ectopic Dmrt5 Conditional overexpression of Dmrt5 throughout the telencephalon produces gene expression and structural defects that are highly consistent with reduced GSX2 activity. Further, Emx2;Dmrt5 double KO embryos show a phenotype similar to Dmrt3;Dmrt5 double KO embryos, and both DMRT3, DMRT5 and the homeobox transcription factor EMX2 bind to a ventral telencephalon-specific enhancer in the Gsx2 locus. Together, our findings uncover cooperative functions of DMRT3, DMRT5, and EMX2 in dividing dorsal from ventral in the telencephalon.SIGNIFICANCE STATEMENT We identified the DMRT3 and DMRT5 zinc finger transcription factors as novel regulators of dorsoventral patterning in the telencephalon. Our data indicate that they have overlapping functions and compensate for one another. The double, but not the single, knock-out produces a dorsal telencephalon that is ventralized, and olfactory bulb tissue takes over most remaining cortex. Conversely, overexpressing Dmrt5 throughout the telencephalon causes expanded expression of dorsal gene determinants and smaller olfactory bulbs. Furthermore, we show that the homeobox transcription factor EMX2 that is coexpressed with DMRT3 and DMRT5 in cortical progenitors cooperates with them to maintain dorsoventral patterning in the telencephalon. Our study suggests that DMRT3/5 function with EMX2 in positioning the pallial-subpallial boundary by antagonizing the ventral homeobox transcription factor GSX2.


Asunto(s)
Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Telencéfalo/embriología , Factores de Transcripción/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Telencéfalo/metabolismo , Factores de Transcripción/genética
6.
Cereb Cortex ; 28(2): 493-509, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031177

RESUMEN

Mice that are constitutively null for the zinc finger doublesex and mab-3 related (Dmrt) gene, Dmrt5/Dmrta2, show a variety of patterning abnormalities in the cerebral cortex, including the loss of the cortical hem, a powerful cortical signaling center. In conditional Dmrt5 gain of function and loss of function mouse models, we generated bidirectional changes in the neocortical area map without affecting the hem. Analysis indicated that DMRT5, independent of the hem, directs the rostral-to-caudal pattern of the neocortical area map. Thus, DMRT5 joins a small number of transcription factors shown to control directly area size and position in the neocortex. Dmrt5 deletion after hem formation also reduced hippocampal size and shifted the position of the neocortical/paleocortical boundary. Dmrt3, like Dmrt5, is expressed in a gradient across the cortical primordium. Mice lacking Dmrt3 show cortical patterning defects akin to but milder than those in Dmrt5 mutants, perhaps in part because Dmrt5 expression increases in the absence of Dmrt3. DMRT5 upregulates Dmrt3 expression and negatively regulates its own expression, which may stabilize the level of DMRT5. Together, our findings indicate that finely tuned levels of DMRT5, together with DMRT3, regulate patterning of the cerebral cortex.


Asunto(s)
Desarrollo Embrionario/fisiología , Hipocampo/metabolismo , Neocórtex/metabolismo , Factores de Transcripción/biosíntesis , Animales , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Neurogénesis/fisiología
7.
Nucleic Acids Res ; 45(12): 7191-7211, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28472341

RESUMEN

In mammalian embryonic gonads, SOX9 is required for the determination of Sertoli cells that orchestrate testis morphogenesis. To identify genetic networks directly regulated by SOX9, we combined analysis of SOX9-bound chromatin regions from murine and bovine foetal testes with sequencing of RNA samples from mouse testes lacking Sox9. We found that SOX9 controls a conserved genetic programme that involves most of the sex-determining genes. In foetal testes, SOX9 modulates both transcription and directly or indirectly sex-specific differential splicing of its target genes through binding to genomic regions with sequence motifs that are conserved among mammals and that we called 'Sertoli Cell Signature' (SCS). The SCS is characterized by a precise organization of binding motifs for the Sertoli cell reprogramming factors SOX9, GATA4 and DMRT1. As SOX9 biological role in mammalian gonads is to determine Sertoli cells, we correlated this genomic signature with the presence of SOX9 on chromatin in foetal testes, therefore equating this signature to a genomic bar code of the fate of foetal Sertoli cells. Starting from the hypothesis that nuclear factors that bind to genomic regions with SCS could functionally interact with SOX9, we identified TRIM28 as a new SOX9 partner in foetal testes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Factor de Transcripción SOX9/genética , Células de Sertoli/metabolismo , Transcriptoma , Animales , Bovinos , Cromatina/química , Cromatina/metabolismo , Embrión de Mamíferos , Femenino , Feto , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Redes Reguladoras de Genes , Masculino , Ratones , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Represoras/metabolismo , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ARN , Células de Sertoli/citología , Procesos de Determinación del Sexo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito
8.
PLoS Genet ; 12(9): e1006293, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27583450

RESUMEN

Male mammals produce sperm for most of postnatal life and therefore require a robust germ line stem cell system, with precise balance between self-renewal and differentiation. Prior work established doublesex- and mab-3-related transcription factor 1 (Dmrt1) as a conserved transcriptional regulator of male sexual differentiation. Here we investigate the role of Dmrt1 in mouse spermatogonial stem cell (SSC) homeostasis. We find that Dmrt1 maintains SSCs during steady state spermatogenesis, where it regulates expression of Plzf, another transcription factor required for SSC maintenance. We also find that Dmrt1 is required for recovery of spermatogenesis after germ cell depletion. Committed progenitor cells expressing Ngn3 normally do not contribute to SSCs marked by the Id4-Gfp transgene, but do so when spermatogonia are chemically depleted using busulfan. Removal of Dmrt1 from Ngn3-positive germ cells blocks the replenishment of Id4-GFP-positive SSCs and recovery of spermatogenesis after busulfan treatment. Our data therefore reveal that Dmrt1 supports SSC maintenance in two ways: allowing SSCs to remain in the stem cell pool under normal conditions; and enabling progenitor cells to help restore the stem cell pool after germ cell depletion.


Asunto(s)
Espermatogénesis/genética , Espermatogonias/metabolismo , Factores de Transcripción/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Espermatogonias/citología , Factores de Transcripción/metabolismo
9.
Dev Biol ; 424(2): 208-220, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28274610

RESUMEN

Retinoic acid (RA) is a potent inducer of cell differentiation and plays an essential role in sex-specific germ cell development in the mammalian gonad. RA is essential for male gametogenesis and hence fertility. However, RA can also disrupt sexual cell fate in somatic cells of the testis, promoting transdifferentiation of male Sertoli cells to female granulosa-like cells when the male sexual regulator Dmrt1 is absent. The feminizing ability of RA in the Dmrt1 mutant somatic testis suggests that RA might normally play a role in somatic cell differentiation or cell fate maintenance in the ovary. To test for this possibility we disrupted RA signaling in somatic cells of the early fetal ovary using three genetic strategies and one pharmaceutical approach. We found that deleting all three RA receptors (RARs) in the XX somatic gonad at the time of sex determination did not significantly affect ovarian differentiation, follicle development, or female fertility. Transcriptome analysis of adult triple mutant ovaries revealed remarkably little effect on gene expression in the absence of somatic RAR function. Likewise, deletion of three RA synthesis enzymes (Aldh1a1-3) at the time of sex determination did not masculinize the ovary. A dominant-negative RAR transgene altered granulosa cell proliferation, likely due to interference with a non-RA signaling pathway, but did not prevent granulosa cell specification and oogenesis or abolish fertility. Finally, culture of fetal XX gonads with an RAR antagonist blocked germ cell meiotic initiation but did not disrupt sex-biased gene expression. We conclude that RA signaling, although crucial in the ovary for meiotic initiation, is not required for granulosa cell specification, differentiation, or reproductive function.


Asunto(s)
Ovario/embriología , Ovario/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Familia de Aldehído Deshidrogenasa 1 , Animales , Linaje de la Célula/efectos de los fármacos , Femenino , Feto/embriología , Feto/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Genes Dominantes , Isoenzimas/metabolismo , Masculino , Mamíferos , Meiosis/efectos de los fármacos , Mesonefro/efectos de los fármacos , Mesonefro/embriología , Mesonefro/metabolismo , Ratones , Ovario/efectos de los fármacos , Receptores de Ácido Retinoico/metabolismo , Retinal-Deshidrogenasa/metabolismo , Retinoides/farmacología , Procesos de Determinación del Sexo/efectos de los fármacos , Técnicas de Cultivo de Tejidos
10.
Nat Rev Genet ; 13(3): 163-74, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22310892

RESUMEN

Most animals reproduce sexually, but the genetic and molecular mechanisms that determine the eventual sex of each embryo vary remarkably. DM domain genes, which are related to the insect gene doublesex, are integral to sexual development and its evolution in many metazoans. Recent studies of DM domain genes reveal mechanisms by which new sexual dimorphisms have evolved in invertebrates and show that one gene, Dmrt1, was central to multiple evolutionary transitions between sex-determining mechanisms in vertebrates. In addition, Dmrt1 coordinates a surprising array of distinct cell fate decisions in the mammalian gonad and even guards against transdifferentiation of male cells into female cells in the adult testis.


Asunto(s)
Evolución Biológica , Genitales/embriología , Células Germinativas/fisiología , Procesos de Determinación del Sexo , Factores de Transcripción/fisiología , Adulto , Animales , Femenino , Humanos , Masculino
11.
Development ; 141(19): 3662-71, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25249458

RESUMEN

In mammals, a key transition in spermatogenesis is the exit from spermatogonial differentiation and mitotic proliferation and the entry into spermatocyte differentiation and meiosis. Although several genes that regulate this transition have been identified, how it is controlled and coordinated remains poorly understood. Here, we examine the role in male gametogenesis of the Doublesex-related gene Dmrt6 (Dmrtb1) in mice and find that Dmrt6 plays a crucial role in directing germ cells through the mitotic-to-meiotic germ cell transition. DMRT6 protein is expressed in late mitotic spermatogonia. In mice of the C57BL/6J strain, a null mutation in Dmrt6 disrupts spermatogonial differentiation, causing inappropriate expression of spermatogonial differentiation factors, including SOHLH1, SOHLH2 and DMRT1 as well as the meiotic initiation factor STRA8, and causing most late spermatogonia to undergo apoptosis. In mice of the 129Sv background, most Dmrt6 mutant germ cells can complete spermatogonial differentiation and enter meiosis, but they show defects in meiotic chromosome pairing, establishment of the XY body and processing of recombination foci, and they mainly arrest in mid-pachynema. mRNA profiling of Dmrt6 mutant testes together with DMRT6 chromatin immunoprecipitation sequencing suggest that DMRT6 represses genes involved in spermatogonial differentiation and activates genes required for meiotic prophase. Our results indicate that Dmrt6 plays a key role in coordinating the transition in gametogenic programs from spermatogonial differentiation and mitosis to spermatocyte development and meiosis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Meiosis/fisiología , Mitosis/fisiología , Espermatogénesis/fisiología , Espermatogonias/metabolismo , Factores de Transcripción/metabolismo , Animales , Bromodesoxiuridina , Inmunoprecipitación de Cromatina , Biología Computacional , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica/genética , Vectores Genéticos/genética , Genotipo , Factores de Diferenciación de Crecimiento/metabolismo , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia de ARN , Espermatogonias/citología , Factores de Transcripción/genética
12.
Nature ; 476(7358): 101-4, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21775990

RESUMEN

Sex in mammals is determined in the fetal gonad by the presence or absence of the Y chromosome gene Sry, which controls whether bipotential precursor cells differentiate into testicular Sertoli cells or ovarian granulosa cells. This pivotal decision in a single gonadal cell type ultimately controls sexual differentiation throughout the body. Sex determination can be viewed as a battle for primacy in the fetal gonad between a male regulatory gene network in which Sry activates Sox9 and a female network involving WNT/ß-catenin signalling. In females the primary sex-determining decision is not final: loss of the FOXL2 transcription factor in adult granulosa cells can reprogram granulosa cells into Sertoli cells. Here we show that sexual fate is also surprisingly labile in the testis: loss of the DMRT1 transcription factor in mouse Sertoli cells, even in adults, activates Foxl2 and reprograms Sertoli cells into granulosa cells. In this environment, theca cells form, oestrogen is produced and germ cells appear feminized. Thus Dmrt1 is essential to maintain mammalian testis determination, and competing regulatory networks maintain gonadal sex long after the fetal choice between male and female. Dmrt1 and Foxl2 are conserved throughout vertebrates and Dmrt1-related sexual regulators are conserved throughout metazoans. Antagonism between Dmrt1 and Foxl2 for control of gonadal sex may therefore extend beyond mammals. Reprogramming due to loss of Dmrt1 also may help explain the aetiology of human syndromes linked to DMRT1, including disorders of sexual differentiation and testicular cancer.


Asunto(s)
Caracteres Sexuales , Procesos de Determinación del Sexo/fisiología , Diferenciación Sexual/fisiología , Testículo/metabolismo , Factores de Transcripción/metabolismo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Transdiferenciación Celular , Femenino , Feminización/genética , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Masculino , Ratones , Modelos Biológicos , Ovario/citología , Ovario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/metabolismo , Células de Sertoli/citología , Células de Sertoli/metabolismo , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Testículo/citología , Células Tecales/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
13.
Mol Biol Evol ; 32(5): 1296-309, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25657328

RESUMEN

Sex chromosomes have evolved many times in animals and studying these replicate evolutionary "experiments" can help broaden our understanding of the general forces driving the origin and evolution of sex chromosomes. However this plan of study has been hindered by the inability to identify the sex chromosome systems in the large number of species with cryptic, homomorphic sex chromosomes. Restriction site-associated DNA sequencing (RAD-seq) is a critical enabling technology that can identify the sex chromosome systems in many species where traditional cytogenetic methods have failed. Using newly generated RAD-seq data from 12 gecko species, along with data from the literature, we reinterpret the evolution of sex-determining systems in lizards and snakes and test the hypothesis that sex chromosomes can routinely act as evolutionary traps. We uncovered between 17 and 25 transitions among gecko sex-determining systems. This is approximately one-half to two-thirds of the total number of transitions observed among all lizards and snakes. We find support for the hypothesis that sex chromosome systems can readily become trap-like and show that adding even a small number of species from understudied clades can greatly enhance hypothesis testing in a model-based phylogenetic framework. RAD-seq will undoubtedly prove useful in evaluating other species for male or female heterogamety, particularly the majority of fish, amphibian, and reptile species that lack visibly heteromorphic sex chromosomes, and will significantly accelerate the pace of biological discovery.


Asunto(s)
Evolución Molecular , Lagartos/genética , Procesos de Determinación del Sexo , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Lagartos/fisiología , Masculino , Filogenia , Mapeo Restrictivo , Cromosomas Sexuales/genética , Serpientes/genética , Serpientes/fisiología
14.
Proc Natl Acad Sci U S A ; 110(40): 16033-8, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24046365

RESUMEN

How sexual regulators translate global sexual fate into appropriate local sexual differentiation events is perhaps the least understood aspect of sexual development. Here we have used ChIP followed by deep sequencing (ChIP-seq) to identify direct targets of the nematode global sexual regulator Transformer 1 (TRA-1), a transcription factor acting at the interface between organism-wide and cell-specific sexual regulation to control all sex-specific somatic differentiation events. We identified 184 TRA-1-binding sites in Caenorhabditis elegans, many with temporal- and/or tissue-specific TRA-1 association. We also identified 78 TRA-1-binding sites in the related nematode Caenorhabditis briggsae, 19 of which are conserved between the two species. Some DNA segments containing TRA-1-binding sites drive male-specific expression patterns, and RNAi depletion of some genes adjacent to TRA-1-binding sites results in defects in male sexual development. TRA-1 binds to sites adjacent to a number of heterochronic regulatory genes, some of which drive male-specific expression, suggesting that TRA-1 imposes sex specificity on developmental timing. We also found evidence for TRA-1 feedback regulation of the global sex-determination pathway: TRA-1 binds its own locus and those of multiple upstream masculinizing genes, and most of these associations are conserved in C. briggsae. Thus, TRA-1 coordinates sexual development by reinforcing the sex-determination decision and directing downstream sexual differentiation events.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Unión al ADN/fisiología , Retroalimentación Fisiológica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Procesos de Determinación del Sexo/fisiología , Diferenciación Sexual/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Caenorhabditis elegans/genética , Inmunoprecipitación de Cromatina , Biología Computacional , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Interferencia de ARN , Análisis de Secuencia de ADN , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Factores de Transcripción/genética
15.
Dev Biol ; 389(2): 160-72, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24576538

RESUMEN

DMRT1 encodes a conserved transcription factor with an essential role in gonadal function. In the chicken, DMRT1 in located on the Z sex chromosome and is currently the best candidate master regulator of avian gonadal sex differentiation. We previously showed that knockdown of DMRT1 expression during the period of sexual differentiation induces feminisation of male embryonic chicken gonads. This gene is therefore necessary for proper testis development in the chicken. However, whether it is sufficient to induce testicular differentiation has remained unresolved. We show here that over-expression of DMRT1 induces male pathway genes and antagonises the female pathway in embryonic chicken gonads. Ectopic DMRT1 expression in female gonads induces localised SOX9 and AMH expression. It also induces expression of the recently identified Z-linked male factor, Hemogen (HEMGN). Masculinised gonads show evidence of cord-like structures and retarded female-type cortical development. Furthermore, expression of the critical feminising enzyme, aromatase, is reduced in the presence of over-expressed DMRT1. These data indicate that DMRT1 is an essential sex-linked regulator of gonadal differentiation in avians, and that it likely acts via a dosage mechanism established through the lack of global Z dosage compensation in birds.


Asunto(s)
Gónadas/embriología , Gónadas/metabolismo , Procesos de Determinación del Sexo , Factores de Transcripción/metabolismo , Animales , Hormona Antimülleriana/metabolismo , Aromatasa/genética , Aromatasa/metabolismo , Embrión de Pollo , Electroporación , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Factor de Transcripción SOX9/metabolismo , Testículo/embriología , Testículo/metabolismo , Factores de Tiempo
16.
Dev Biol ; 377(1): 67-78, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23473982

RESUMEN

Dmrt1 (doublesex and mab-3 related transcription factor (1) is a regulator of testis development in vertebrates that has been implicated in testicular germ cell tumors of mouse and human. In the fetal mouse testis Dmrt1 regulates germ cell pluripotency in a strain-dependent manner. Loss of Dmrt1 in 129Sv strain mice results in a >90% incidence of testicular teratomas, tumors consisting cells of multiple germ layers; by contrast, these tumors have never been observed in Dmrt1 mutants of C57BL/6J (B6) or mixed genetic backgrounds. To further investigate the interaction between Dmrt1 and genetic background we compared mRNA expression in wild type and Dmrt1 mutant fetal testes of 129Sv and B6 mice at embryonic day 15.5 (E15.5), prior to overt tumorigenesis. Loss of Dmrt1 caused misexpression of overlapping but distinct sets of mRNAs in the two strains. The mRNAs that were selectively affected included some that changed expression only in one strain or the other and some that changed in both strains but to a greater degree in one versus the other. In particular, loss of Dmrt1 in 129Sv testes caused a more severe failure to silence regulators of pluripotency than in B6 testes. A number of genes misregulated in 129Sv mutant testes also are misregulated in human testicular germ cell tumors (TGCTs), suggesting similar etiology between germ cell tumors in mouse and man. Expression profiling showed that DMRT1 also regulates pluripotency genes in the fetal ovary, although Dmrt1 mutant females do not develop teratomas. Pathway analysis indicated disruption of several signaling pathways in Dmrt1 mutant fetal testes, including Nodal, Notch, and GDNF. We used a Nanos3-cre knock-in allele to perform conditional gene targeting, testing the GDNF coreceptors Gfra1 and Ret for effects on teratoma susceptibility. Conditional deletion of Gfra1 but not Ret in fetal germ cells of animals outcrossed to 129Sv caused a modest but significant elevation in tumor incidence. Despite some variability in genetic background in these crosses, this result is consistent with previous genetic mapping of teratoma susceptibility loci to the region containing Gfra1. Using Nanos3-cre we also uncovered a strong genetic interaction between Dmrt1 and Nanos3, suggesting parallel functions for these two genes in fetal germ cells. Finally, we used chromatin immunoprecipitation (ChIP-seq) analysis to identify a number of potentially direct DMRT1 targets. This analysis suggested that DMRT1 controls pluripotency via transcriptional repression of Esrrb, Nr5a2/Lrh1, and Sox2. Given the strong evidence for involvement of DMRT1 in human TGCT, the downstream genes and pathways identified in this study provide potentially useful candidates for roles in the human disease.


Asunto(s)
Feto/patología , Células Germinativas/patología , Neoplasias/embriología , Neoplasias/patología , Células Madre Pluripotentes/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Proliferación Celular , ADN/metabolismo , ADN-Citosina Metilasas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neoplasias/metabolismo , Proteína Nodal/genética , Proteína Nodal/metabolismo , Ovario/embriología , Ovario/enzimología , Ovario/patología , Células Madre Pluripotentes/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Especificidad de la Especie , Testículo/embriología , Testículo/metabolismo , Testículo/patología
17.
Cytogenet Genome Res ; 143(4): 251-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25227445

RESUMEN

Evaluating homology between the sex chromosomes of different species is an important first step in deducing the origins and evolution of sex-determining mechanisms in a clade. Here, we describe the preparation of Z and W chromosome paints via chromosome microdissection from the Australian marbled gecko (Christinus marmoratus) and their subsequent use in evaluating sex chromosome homology with the ZW chromosomes of the Kwangsi gecko (Gekko hokouensis) from eastern Asia. We show that the ZW sex chromosomes of C. marmoratus and G. hokouensis are not homologous and represent independent origins of female heterogamety within the Gekkonidae. We also show that the C. marmoratus Z and W chromosomes are genetically similar to each other as revealed by C-banding, comparative genomic hybridization, and the reciprocal painting of Z and W chromosome probes. This implies that sex chromosomes in C. marmoratus are at an early stage of differentiation, suggesting a recent origin.


Asunto(s)
Lagartos/genética , Cromosomas Sexuales/genética , Animales , Células Cultivadas , Bandeo Cromosómico , Hibridación Genómica Comparativa , Evolución Molecular , Femenino , Hibridación Fluorescente in Situ , Masculino , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie
18.
Cereb Cortex ; 23(11): 2552-67, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22923088

RESUMEN

Regional patterning of the cerebral cortex is initiated by morphogens secreted by patterning centers that establish graded expression of transcription factors within cortical progenitors. Here, we show that Dmrt5 is expressed in cortical progenitors in a high-caudomedial to low-rostrolateral gradient. In its absence, the cortex is strongly reduced and exhibits severe abnormalities, including agenesis of the hippocampus and choroid plexus and defects in commissural and thalamocortical tracts. Loss of Dmrt5 results in decreased Wnt and Bmp in one of the major telencephalic patterning centers, the dorsomedial telencephalon, and in a reduction of Cajal-Retzius cells. Expression of the dorsal midline signaling center-dependent transcription factors is downregulated, including Emx2, which promotes caudomedial fates, while the rostral determinant Pax6, which is inhibited by midline signals, is upregulated. Consistently, Dmrt5(-/-) brains exhibit patterning defects with a dramatic reduction of the caudomedial cortex. Dmrt5 is increased upon the activation of Wnt signaling and downregulated in Gli3(xt/xt) mutants. We conclude that Dmrt5 is a novel Wnt-dependent transcription factor required for early cortical development and that it may regulate initial cortical patterning by promoting dorsal midline signaling center formation and thereby helping to establish the graded expression of the other transcription regulators of cortical identity.


Asunto(s)
Corteza Cerebral/embriología , Factores de Transcripción/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Corteza Cerebral/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética , Proteínas Wnt/metabolismo
19.
Proc Natl Acad Sci U S A ; 107(30): 13360-5, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20616082

RESUMEN

The DM domain proteins Doublesex- and MAB-3-related transcription factors (DMRTs) are widely conserved in metazoan sex determination and sexual differentiation. One of these proteins, DMRT1, plays diverse and essential roles in development of the vertebrate testis. In mammals DMRT1 is expressed and required in both germ cells and their supporting Sertoli cells. Despite its critical role in testicular development, little is known about how DMRT1 functions as a transcription factor or what genes it binds and regulates. We combined ChIP methods with conditional gene targeting and mRNA expression analysis and identified almost 1,400 promoter-proximal regions bound by DMRT1 in the juvenile mouse testis and determined how expression of the associated mRNAs is affected when Dmrt1 is selectively mutated in germ cells or Sertoli cells. These analyses revealed that DMRT1 is a bifunctional transcriptional regulator, activating some genes and repressing others. ChIP analysis using conditional mutant testes showed that DNA binding and transcriptional regulation of individual target genes can differ between germ cells and Sertoli cells. Genes bound by DMRT1 in vivo were enriched for a motif closely resembling the sequence DMRT1 prefers in vitro. Differential response of genes to loss of DMRT1 corresponded to differences in the enriched motif, suggesting that other transacting factors may modulate DMRT1 activity. DMRT1 bound its own promoter and those of six other Dmrt genes, indicating auto- and cross-regulation of these genes. Many of the DMRT1 target genes identified here are known to be important for a variety of functions in testicular development; the others are candidates for further investigation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genoma , Testículo/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Unión Proteica , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética , Activación Transcripcional , Transfección
20.
Dev Biol ; 356(1): 63-70, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21621532

RESUMEN

Dmrt1 belongs to the DM domain gene family of conserved sexual regulators. In the mouse Dmrt1 is expressed in the genital ridge (the gonadal primordium) in both sexes and then becomes testis-specific shortly after sex determination. The essential role of DMRT1 in testicular differentiation is well established, and includes transcriptional repression of the meiotic inducer Stra8. However Dmrt1 mutant females are fertile and the role of Dmrt1 in the ovary has not been studied. Here we show in the mouse that most Dmrt1 mutant germ cells in the fetal ovary have greatly reduced expression of STRA8, and fail to properly localize SYCP3 and γH2AX during meiotic prophase. Lack of DMRT1 in the fetal ovary results in the formation of many fewer primordial follicles in the juvenile ovary, although these are sufficient for fertility. Genome-wide chromatin immunoprecipitiation (ChIP-chip) and quantitative ChIP (qChIP) combined with mRNA expression profiling suggests that transcriptional activation of Stra8 in fetal germ cells is the main function of DMRT1 in females, and that this regulation likely is direct. Thus DMRT1 controls Stra8 sex-specifically, activating it in the fetal ovary and repressing it in the adult testis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oogénesis , Ovario/crecimiento & desarrollo , Óvulo/metabolismo , Proteínas/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Femenino , Histonas/metabolismo , Profase Meiótica I , Ratones , Mutación , Proteínas Nucleares/metabolismo , Ovario/citología , Ovario/metabolismo , Óvulo/citología , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA