Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
PLoS Genet ; 17(3): e1009468, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33788836

RESUMEN

Doublesex (Dsx) and Fruitless (Fru) are the two downstream transcription factors that actuate Drosophila sex determination. While Dsx assists Fru to regulate sex-specific behavior, whether Fru collaborates with Dsx in regulating other aspects of sexual dimorphism remains unknown. One important aspect of sexual dimorphism is found in the gonad stem cell (GSC) niches, where male and female GSCs are regulated to create large numbers of sperm and eggs. Here we report that Fru is expressed male-specifically in the GSC niche and plays important roles in the development and maintenance of these cells. Unlike previously-studied aspects of sex-specific Fru expression, which are regulated by Transformer (Tra)-mediated alternative splicing, we show that male-specific expression of fru in the gonad is regulated downstream of dsx, and is independent of tra. fru genetically interacts with dsx to support maintenance of the niche throughout development. Ectopic expression of fru inhibited female niche formation and partially masculinized the ovary. fru is also required autonomously for cyst stem cell maintenance and cyst cell survival. Finally, we identified a conserved Dsx binding site upstream of fru promoter P4 that regulates fru expression in the niche, indicating that fru is likely a direct target for transcriptional regulation by Dsx. These findings demonstrate that fru acts outside the nervous system to influence sexual dimorphism and reveal a new mechanism for regulating sex-specific expression of fru that is regulated at the transcriptional level by Dsx, rather than by alternative splicing by Tra.


Asunto(s)
Proteínas de Drosophila/genética , Regulación de la Expresión Génica , Gónadas/citología , Gónadas/metabolismo , Proteínas del Tejido Nervioso/genética , Caracteres Sexuales , Procesos de Determinación del Sexo/genética , Nicho de Células Madre/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Proteínas de Drosophila/metabolismo , Evolución Molecular , Femenino , Técnica del Anticuerpo Fluorescente , Orden Génico , Sitios Genéticos , Masculino , Proteínas del Tejido Nervioso/metabolismo , Testículo , Factores de Transcripción/metabolismo
2.
Development ; 146(11)2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31043421

RESUMEN

Sex-specific development of the gonads is a key aspect of sexual dimorphism that is regulated by Doublesex/Mab3-related transcription factors (DMRTs) in diverse animal species. We find that in mutants for Drosophila dsx, important components of the male and female gonad stem cell niches (hubs and terminal filaments/cap cells, respectively) still form. Initially, gonads in all dsx mutants (both XX and XY) initiate the male program of development, but later half of these gonads switch to form female stem cell niche structures. One individual can have both male-type and female-type gonad niches; however, male and female niches are usually not observed in the same gonad, indicating that cells make a 'group decision' about which program to follow. We conclude that dsx does not act in an instructive manner to regulate male versus female niche formation, as these structures form in the absence of dsx function. Instead, dsx acts to 'tip the balance' between the male or female programs, which are then executed independently of dsx We show that bric a brac acts downstream of dsx to control the male versus female niche decision. These results indicate that, in both flies and mammals, the sexual fate of the somatic gonad is remarkably plastic and is controlled by a combination of autonomous and non-autonomous cues.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster , Gónadas/citología , Gónadas/metabolismo , Procesos de Determinación del Sexo/genética , Nicho de Células Madre/genética , Animales , Animales Modificados Genéticamente , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Femenino , Gónadas/embriología , Masculino , Especificidad de Órganos/genética , Factores de Transcripción/fisiología
3.
PLoS Genet ; 15(7): e1007617, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31329582

RESUMEN

For sexually reproducing organisms, production of male or female gametes depends on specifying the correct sexual identity in the germline. In D. melanogaster, Sex lethal (Sxl) is the key gene that controls sex determination in both the soma and the germline, but how it does so in the germline is unknown, other than that it is different than in the soma. We conducted an RNA expression profiling experiment to identify direct and indirect germline targets of Sxl specifically in the undifferentiated germline. We find that, in these cells, Sxl loss does not lead to a global masculinization observed at the whole-genome level. In contrast, Sxl appears to affect a discrete set of genes required in the male germline, such as Phf7. We also identify Tudor domain containing protein 5-like (Tdrd5l) as a target for Sxl regulation that is important for male germline identity. Tdrd5l is repressed by Sxl in female germ cells, but is highly expressed in male germ cells where it promotes proper male fertility and germline differentiation. Additionally, Tdrd5l localizes to cytoplasmic granules with some characteristics of RNA Processing (P-) Bodies, suggesting that it promotes male identity in the germline by regulating post-transcriptional gene expression.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ARN/métodos , Procesos de Determinación del Sexo , Animales , Diferenciación Celular , Gránulos Citoplasmáticos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de Unión al ARN/genética , Dominio Tudor
4.
Development ; 138(16): 3357-66, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21752937

RESUMEN

Male and female germ cells follow distinct developmental paths with respect to germline stem cell (GSC) production and the types of differentiated progeny they produce (sperm versus egg). An essential aspect of germline development is how sexual identity is used to differentially regulate the male and female germ cell genomes to allow for these distinct outcomes. Here, we identify a gene, no child left behind (nclb), that plays very different roles in the male versus female germline in Drosophila. In particular, nclb is required for GSC maintenance in males, but not in females. Male GSCs mutant for nclb are rapidly lost from the niche, and begin to differentiate but cannot complete spermatogenesis. We further find that nclb encodes a member of a new family of conserved chromatin-associated proteins. NCLB interacts with chromatin in a specific manner and is associated with sites of active transcription. Thus, NCLB appears to be a novel chromatin regulator that exhibits very different effects on the male and female germ cell genomes.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Espermatozoides/metabolismo , Células Madre/metabolismo , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Mutación , Oogénesis , Óvulo/citología , Óvulo/metabolismo , Espermatogénesis , Espermatozoides/citología , Células Madre/citología , Transcripción Genética
5.
Nat Cell Biol ; 9(12): 1344-5, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18059357

RESUMEN

Stem cells, and the microenvironment or 'niche' that influences them, must often reside in a particular location within a tissue to perform their function. Integrin-mediated adhesion is now shown to regulate the location of the stem-cell niche in the Drosophila testis.


Asunto(s)
Drosophila/citología , Células Madre/citología , Animales , Adhesión Celular , Diferenciación Celular , Linaje de la Célula , Drosophila/embriología , Drosophila/crecimiento & desarrollo , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Masculino , Células Madre/fisiología , Testículo/citología , Testículo/embriología , Testículo/crecimiento & desarrollo
6.
bioRxiv ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38187640

RESUMEN

In vertebrate sexual development, two important steroid hormones, testosterone and estrogen, regu-late the sex-specific development of many tissues. In contrast, invertebrates utilize a single steroid hormone, ecdysone, to regulate developmental timing in both sexes. However, here we show that in Drosophila melanogaster, sex-specific ecdysone (E) signaling controls important aspects of gonad sexual dimorphism. Rather than being regulated at the level of hormone production, hor-mone activity is regulated cell-autonomously through sex-specific hormone reception. Ecdysone receptor (EcR) expression is restricted to the developing ovary and is repressed in the testis at a time when ecdysone initiates ovary morphogenesis. Interestingly, EcR expression is regulated downstream of the sex determination factor Doublesex (Dsx), the founding member of the Dsx/Mab3 Related Transcription Factor (DMRT) family that regulates gonad development in all animals. E signaling is required for normal ovary development1,2, and ectopic activation of E sig-naling in the testis antagonized stem cell niche identity and feminized somatic support cells, which were transformed into follicle-like cells. This work demonstrates that invertebrates can also use steroid hormone signaling to control sex-specific development. Further, it may help explain recent work showing that vertebrate sexual development is surprisingly cell-autonomous. For example, chickens utilize testosterone and estrogen to control sex-specific development, but when they have a mixture of cells with male and female genotypes, the male cells develop as male and the female cells develop as female despite exposure to the same circulating hormones3. Sex-specific regulation of steroid hormone signaling may well underly such cell-autonomous sexual fate choices in verte-brates as it does in Drosophila.

7.
Dev Biol ; 367(2): 114-25, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22575490

RESUMEN

To form a gonad, germ cells (GCs) and somatic gonadal precursor cells (SGPs) must migrate to the correct location in the developing embryo and establish the cell-cell interactions necessary to create proper gonad architecture. During gonad morphogenesis, SGPs send out cellular extensions to ensheath the individual GCs and promote their development. We have identified mutations in the raw gene that result in a failure of the SGPs to ensheath the GCs, leading to defects in GC development. Using genetic analysis and gene expression studies, we find that Raw negatively regulates JNK signaling during gonad morphogenesis, and increased JNK signaling is sufficient to cause ensheathment defects. In particular, Raw functions upstream of the Drosophila Jun-related transcription factor to regulate its subcellular localization. Since JNK signaling regulates cell adhesion during the morphogenesis of many tissues, we examined the relationship between raw and the genes encoding Drosophila E-cadherin and ß-catenin, which function together in cell adhesion. We find that loss of DE-cadherin strongly enhances the raw mutant gonad phenotype, while increasing DE-cadherin function rescues this phenotype. Further, loss of raw results in mislocalization of ß-catenin away from the cell surface. Therefore, cadherin-based cell adhesion, likely at the level of ß-catenin, is a primary mechanism by which Raw regulates germline-soma interaction.


Asunto(s)
Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriología , Drosophila/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Gónadas/embriología , Gónadas/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Adhesión Celular , Proteínas del Citoesqueleto/genética , Cartilla de ADN/genética , Drosophila/genética , Proteínas de Drosophila/genética , Genes de Insecto , Gónadas/citología , Mutación
8.
Dev Cell ; 14(2): 275-86, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18267095

RESUMEN

Sex determination in Drosophila is commonly thought to be a cell-autonomous process, where each cell decides its own sexual fate based on its sex chromosome constitution (XX versus XY). This is in contrast to sex determination in mammals, which largely acts nonautonomously through cell-cell signaling. Here we examine how sexual dimorphism is created in the Drosophila gonad by investigating the formation of the pigment cell precursors, a male-specific cell type in the embryonic gonad. Surprisingly, we find that sex determination in the pigment cell precursors, as well as the male-specific somatic gonadal precursors, is non-cell autonomous. Male-specific expression of Wnt2 within the somatic gonad triggers pigment cell precursor formation from surrounding cells. Our results indicate that nonautonomous sex determination is important for creating sexual dimorphism in the Drosophila gonad, similar to the manner in which sex-specific gonad formation is controlled in mammals.


Asunto(s)
Drosophila melanogaster/embriología , Gónadas/embriología , Caracteres Sexuales , Procesos de Determinación del Sexo , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Cuerpo Adiposo/citología , Cuerpo Adiposo/metabolismo , Gónadas/citología , Masculino , Modelos Biológicos , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Factor de Transcripción SOX9 , Células Madre/citología , Células Madre/metabolismo , Testículo/citología , Testículo/embriología , Testículo/metabolismo , Proteínas Wnt/metabolismo
9.
Curr Biol ; 33(11): R442-R444, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37279665

RESUMEN

The piRNA pathway represses transposon activity to protect the germline genome for future generations. A new study shows how germline sex determination influences the production of different piRNAs in male and female germ cells.


Asunto(s)
Proteínas de Drosophila , Femenino , Masculino , Animales , Proteínas de Drosophila/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN de Interacción con Piwi , Células Germinativas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Elementos Transponibles de ADN/genética , Drosophila melanogaster/genética , Proteínas Argonautas/genética
10.
Dev Biol ; 353(2): 217-28, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21377458

RESUMEN

Organogenesis is a complex process requiring multiple cell types to associate with one another through correct cell contacts and in the correct location to achieve proper organ morphology and function. To better understand the mechanisms underlying gonad formation, we performed a mutagenesis screen in Drosophila and identified twenty-four genes required for gonadogenesis. These genes affect all different aspects of gonad formation and provide a framework for understanding the molecular mechanisms that control these processes. We find that gonad formation is regulated by multiple, independent pathways; some of these regulate the key cell adhesion molecule DE-cadherin, while others act through distinct mechanisms. In addition, we discover that the Slit/Roundabout pathway, best known for its role in regulating axonal guidance, is essential for proper gonad formation. Our findings shed light on the complexities of gonadogenesis and the genetic regulation required for proper organ formation.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/embriología , Drosophila/genética , Genes de Insecto , Gónadas/embriología , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Animales , Animales Modificados Genéticamente , Cadherinas/genética , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Gónadas/citología , Mutagénesis , Mutación , Fenotipo , Transducción de Señal , Proteínas Roundabout
11.
Development ; 136(22): 3821-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19855024

RESUMEN

The establishment of sexual identity is a crucial step of germ cell development in sexually reproducing organisms. Sex determination in the germline is controlled differently than in the soma, and often depends on communication from the soma. To investigate how sexual identity is established in the Drosophila germline, we first conducted a molecular screen for genes expressed in a sex-specific manner in embryonic germ cells. Sex-specific expression of these genes is initiated at the time of gonad formation (stage 15), indicating that sexual identity in the germline is established by this time. Experiments where the sex of the soma was altered relative to that of the germline (by manipulating transformer) reveal a dominant role for the soma in regulating initial germline sexual identity. Germ cells largely take on the sex of the surrounding soma, although the sex chromosome constitution of the germ cells still plays some role at this time. The male soma signals to the germline through the JAK/STAT pathway, while the nature of the signal from the female soma remains unknown. We also find that the genes ovo and ovarian tumor (otu) are expressed in a female-specific manner in embryonic germ cells, consistent with their role in promoting female germline identity. However, removing the function of ovo and otu, or reducing germline function of Sex lethal, had little effect on establishment of germline sexual identity. This is consistent with our findings that signals from the soma are dominant over germline autonomous cues at the initial stage of germline sex determination.


Asunto(s)
Drosophila melanogaster/embriología , Drosophila melanogaster/fisiología , Células Germinativas/fisiología , Procesos de Determinación del Sexo , Animales , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/citología , Gónadas/fisiología , Masculino , Factores de Transcripción/fisiología
12.
Sex Dev ; 16(5-6): 323-328, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35259743

RESUMEN

BACKGROUND: The establishment of male or female identity (sex determination) is essential for creating the anatomical, physiological, and behavioral differences between 2 sexes of the same species (sexual dimorphism). In many organisms, including mammals and Drosophila, sex is determined by inheritance of sex chromosomes, while in other animals, sex is determined by environmental factors. Arguably the most important consequence of sex determination is the production of healthy gametes necessary for reproduction: female oocytes and male spermatids. SUMMARY: The generation of sperm and oocytes requires cooperation between 2 different cell types within the gonad: germ cells and somatic cells. Defects in sex determination in either the somatic gonad or germline lead to disorders of sexual development and infertility. In Drosophila, the gene Sex lethal (Sxl) is the key determinant of sex in both the soma and the germline. However, how Sxl controls sex determination is much more well understood in the soma than the germline. Key Mesage: This review will focus on Sxl in the germline, how it is activated specifically in female germ cells, and how it regulates germline sex determination and sexual development.

13.
Cell Rep ; 39(1): 110620, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385723

RESUMEN

Establishing germ cell sexual identity is critical for development of male and female germline stem cells (GSCs) and production of sperm or eggs. Germ cells depend on signals from the somatic gonad to determine sex, but in organisms such as flies, mice, and humans, the sex chromosome genotype of the germ cells is also important for germline sexual development. How somatic signals and germ-cell-intrinsic cues combine to regulate germline sex determination is thus a key question. We find that JAK/STAT signaling in the GSC niche promotes male identity in germ cells, in part by activating the chromatin reader Phf7. Further, we find that JAK/STAT signaling is blocked in XX (female) germ cells through the action of the sex determination gene Sex lethal to preserve female identity. Thus, an important function of germline sexual identity is to control how GSCs respond to signals in their niche environment.


Asunto(s)
Proteínas de Drosophila , Células Germinativas , Procesos de Determinación del Sexo , Células Madre , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Células Germinativas/metabolismo , Proteínas de Homeodominio/metabolismo , Quinasas Janus/metabolismo , Masculino , Factores de Transcripción STAT/metabolismo , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología , Transducción de Señal/fisiología , Nicho de Células Madre , Células Madre/metabolismo
14.
Nature ; 436(7050): 563-7, 2005 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16049490

RESUMEN

Germ cells must develop along distinct male or female paths to produce the sperm or eggs required for sexual reproduction. In both mouse and Drosophila, the sexual identity of germ cells is influenced by the sex of the surrounding somatic tissue (for example, refs 1, 2, reviewed in refs 3, 4); however, little is known about how the soma controls germline sex determination. Here we show that the janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway provides a sex-specific signal from the soma to the germ line in Drosophila embryonic gonads. The somatic gonad expresses a JAK/STAT ligand, unpaired (upd), in a male-specific manner, and activates the JAK/STAT pathway in male germ cells at the time of gonad formation. Furthermore, the JAK/STAT pathway is necessary for male-specific germ cell behaviour during early gonad development, and is sufficient to activate aspects of male germ cell behaviour in female germ cells. Our findings provide direct evidence that the JAK/STAT pathway mediates a key signal from the somatic gonad that regulates male germline sexual development.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Diferenciación Sexual , Transducción de Señal , Transactivadores/metabolismo , Animales , División Celular , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Activación Enzimática , Femenino , Hibridación in Situ , Janus Quinasa 1 , Larva/citología , Larva/metabolismo , Masculino , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción STAT , Caracteres Sexuales
15.
Dev Biol ; 334(2): 335-44, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19643104

RESUMEN

Germline stem cells (GSCs) in Drosophila are descendants of primordial germ cells (PGCs) specified during embryogenesis. The precise timing of GSC establishment in the testis has not been determined, nor is it known whether mechanisms that control GSC maintenance in the adult are involved in GSC establishment. Here, we determine that PGCs in the developing male gonad first become GSCs at the embryo to larval transition. This coincides with formation of the embryonic hub; the critical signaling center that regulates adult GSC behavior within the stem cell microenvironment (niche). We find that the Jak-STAT signaling pathway is activated in a subset of PGCs that associate with the newly-formed embryonic hub. These PGCs express GSC markers and function like GSCs, while PGCs that do not associate with the hub begin to differentiate. In the absence of Jak-STAT activation, PGCs adjacent to the hub fail to exhibit the characteristics of GSCs, while ectopic activation of the Jak-STAT pathway prevents differentiation. These findings show that stem cell formation is closely linked to development of the stem cell niche, and suggest that Jak-STAT signaling is required for initial establishment of the GSC population in developing testes.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Células Madre Embrionarias/citología , Quinasas Janus/fisiología , Factores de Transcripción STAT/fisiología , Espermatozoides/citología , Testículo/embriología , Factores de Transcripción/fisiología , Animales , Adhesión Celular , Polaridad Celular , Proteínas de Drosophila/análisis , Drosophila melanogaster/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Células Madre Embrionarias/metabolismo , Activación Enzimática , Larva , Masculino , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal/fisiología , Espermatogonias/citología , Espermatozoides/metabolismo , Testículo/citología , Proteínas Supresoras de Tumor/fisiología
16.
Dev Cell ; 5(2): 205-16, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12919673

RESUMEN

Sexually dimorphic development of the gonad is essential for germ cell development and sexual reproduction. We have found that the Drosophila embryonic gonad is already sexually dimorphic at the time of initial gonad formation. Male-specific somatic gonadal precursors (msSGPs) contribute only to the testis and express a Drosophila homolog of Sox9 (Sox100B), a gene essential for testis formation in humans. The msSGPs are specified in both males and females, but are only recruited into the developing testis. In females, these cells are eliminated via programmed cell death dependent on the sex determination regulatory gene doublesex. Our work furthers the hypotheses that a conserved pathway controls gonad sexual dimorphism in diverse species and that sex-specific cell recruitment and programmed cell death are common mechanisms for creating sexual dimorphism.


Asunto(s)
Apoptosis/fisiología , Drosophila melanogaster/embriología , Caracteres Sexuales , Diferenciación Sexual/fisiología , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Femenino , Genes Homeobox , Gónadas/citología , Gónadas/embriología , Gónadas/fisiología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Proteínas Nucleares/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción SOX9 , Cromosomas Sexuales/metabolismo , Factores de Transcripción/metabolismo
17.
Genetics ; 206(4): 1939-1949, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28588035

RESUMEN

A key aspect of germ cell development is to establish germline sexual identity and initiate a sex-specific developmental program to promote spermatogenesis or oogenesis. Previously, we have identified the histone reader Plant Homeodomain Finger 7 (PHF7) as an important regulator of male germline identity. To understand how PHF7 directs sexual differentiation of the male germline, we investigated the downstream targets of PHF7 by combining transcriptome analyses, which reveal genes regulated by Phf7, with genomic profiling of histone H3K4me2, the chromatin mark that is bound by PHF7. Through these genomic experiments, we identify a novel spermatocyte factor Receptor Accessory Protein Like 1 (REEPL1) that can promote spermatogenesis and whose expression is kept off by PHF7 in the spermatogonial stage. Loss of Reepl1 significantly rescues the spermatogenesis defects in Phf7 mutants, indicating that regulation of Reepl1 is an essential aspect of PHF7 function. Further, increasing REEPL1 expression facilitates spermatogenic differentiation. These results indicate that PHF7 controls spermatogenesis by regulating the expression patterns of important male germline genes.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Homeodominio/genética , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Masculino , Espermatocitos/citología
18.
Mech Dev ; 121(11): 1323-33, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15454263

RESUMEN

Sexual dimorphism requires the integration of positional information in the embryo with the sex determination pathway. Homeotic genes are a major source of positional information responsible for patterning along the anterior-posterior axis in embryonic development, and are likely to play a critical role in sexual dimorphism. Here, we investigate the role of homeotic genes in the sexually dimorphic development of the gonad in Drosophila. We have found that Abdominal-B (ABD-B) is expressed in a sexually dimorphic manner in the embryonic gonad. Furthermore, Abd-B is necessary and sufficient for specification of a sexually dimorphic cell type, the male-specific somatic gonadal precursors (msSGPs). In Abd-B mutants, the msSGPs are not specified and male gonads now resemble female gonads with respect to these cells. Ectopic expression of Abd-B is sufficient to induce formation of extra msSGPs in additional segments of the embryo. Abd-B works together with abdominal-A to pattern the non-sexually dimorphic somatic gonad in both sexes, while Abd-B alone specifies the msSGPs. Our results indicate that Abd-B acts at multiple levels to regulate gonad development and that Abd-B class homeotic genes are conserved factors in establishing gonad sexual dimorphism in diverse species.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/embriología , Gónadas/embriología , Proteínas de Homeodominio/fisiología , Procesos de Determinación del Sexo , Animales , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/análisis , Proteínas de Drosophila/genética , Embrión no Mamífero/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/genética , Gónadas/química , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/genética , Masculino , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Caracteres Sexuales , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/fisiología
19.
G3 (Bethesda) ; 4(6): 979-81, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24939183

RESUMEN

In this commentary, Michelle Arbeitman et al., examine the topic of the Genetics of Sex as explored in this month's issues of GENETICS and G3: Genes|Genomes|Genetics. These inaugural articles are part of a joint Genetics of Sex collection (ongoing) in the GSA journals.


Asunto(s)
Procesos de Determinación del Sexo/genética , Animales , Femenino , Células Germinativas , Humanos , Masculino , Selección Genética , Cromosomas Sexuales
20.
Dev Cell ; 31(6): 761-73, 2014 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-25535918

RESUMEN

Primary sex-determination "switches" evolve rapidly, but Doublesex (DSX)-related transcription factors (DMRTs) act downstream of these switches to control sexual development in most animal species. Drosophila dsx encodes female- and male-specific isoforms (DSX(F) and DSX(M)), but little is known about how dsx controls sexual development, whether DSX(F) and DSX(M) bind different targets, or how DSX proteins direct different outcomes in diverse tissues. We undertook genome-wide analyses to identify DSX targets using in vivo occupancy, binding site prediction, and evolutionary conservation. We find that DSX(F) and DSX(M) bind thousands of the same targets in multiple tissues in both sexes, yet these targets have sex- and tissue-specific functions. Interestingly, DSX targets show considerable overlap with targets identified for mouse DMRT1. DSX targets include transcription factors and signaling pathway components providing for direct and indirect regulation of sex-biased expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Animales , Animales Modificados Genéticamente , Sitios de Unión , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma , Estudio de Asociación del Genoma Completo , Masculino , Ratones , Fenotipo , Interferencia de ARN , Análisis de Secuencia de ADN , Factores Sexuales , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA