Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 33(11): R442-R444, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37279665

RESUMO

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.


Assuntos
Proteínas de Drosophila , Feminino , Masculino , Animais , Proteínas de Drosophila/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA de Interação com Piwi , Células Germinativas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Elementos de DNA Transponíveis/genética , Drosophila melanogaster/genética , Proteínas Argonautas/genética
2.
Cell Rep ; 39(1): 110620, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35385723

RESUMO

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.


Assuntos
Proteínas de Drosophila , Células Germinativas , Processos de Determinação Sexual , Células-Tronco , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Células Germinativas/metabolismo , Proteínas de Homeodomínio/metabolismo , Janus Quinases/metabolismo , Masculino , Fatores de Transcrição STAT/metabolismo , Processos de Determinação Sexual/genética , Processos de Determinação Sexual/fisiologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco , Células-Tronco/metabolismo
3.
Sex Dev ; 16(5-6): 323-328, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35259743

RESUMO

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.

4.
PLoS Genet ; 17(3): e1009468, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788836

RESUMO

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.


Assuntos
Proteínas de Drosophila/genética , Regulação da Expressão Gênica , Gônadas/citologia , Gônadas/metabolismo , Proteínas do Tecido Nervoso/genética , Caracteres Sexuais , Processos de Determinação Sexual/genética , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Drosophila/metabolismo , Evolução Molecular , Feminino , Imunofluorescência , Ordem dos Genes , Loci Gênicos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Testículo , Fatores de Transcrição/metabolismo
5.
PLoS Genet ; 15(7): e1007617, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31329582

RESUMO

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.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Ligação a RNA/metabolismo , Análise de Sequência de RNA/métodos , Processos de Determinação Sexual , Animais , Diferenciação Celular , Grânulos Citoplasmáticos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Ligação a RNA/genética , Domínio Tudor
6.
Development ; 146(11)2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31043421

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Gônadas/citologia , Gônadas/metabolismo , Processos de Determinação Sexual/genética , Nicho de Células-Tronco/genética , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Feminino , Gônadas/embriologia , Masculino , Especificidade de Órgãos/genética , Fatores de Transcrição/fisiologia
7.
Genetics ; 206(4): 1939-1949, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28588035

RESUMO

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.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Homeodomínio/genética , Espermatócitos/metabolismo , Espermatogênese/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Masculino , Espermatócitos/citologia
8.
Dev Cell ; 31(6): 761-73, 2014 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-25535918

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Estudo de Associação Genômica Ampla , Masculino , Camundongos , Fenótipo , Interferência de RNA , Análise de Sequência de DNA , Fatores Sexuais , Fatores de Transcrição/metabolismo
10.
G3 (Bethesda) ; 4(6): 979-81, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24939183

RESUMO

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.


Assuntos
Processos de Determinação Sexual/genética , Animais , Feminino , Células Germinativas , Humanos , Masculino , Seleção Genética , Cromossomos Sexuais
11.
Spermatogenesis ; 2(3): 129-136, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23087832

RESUMO

The creation of sexual dimorphism in the gonads is essential for producing the male and female gametes required for sexual reproduction. Sexual development of the gonads involves both somatic cells and germ cells, which often undergo sex determination by different mechanisms. While many sex-specific characteristics evolve rapidly and are very different between animal species, gonad function and the formation of sperm and eggs appear more similar and may be more conserved. Consistent with this, the doublesex/mab3 Related Transcription factors (DMRTs) are important for gonad sexual dimorphism in a wide range of animals, including flies, worms and mammals. Here we explore how sexual dimorphism is regulated in the Drosophila gonad, focusing on recent discoveries relating to testis development. We will discuss how sex determination in both the germline and the soma are utilized to create a testis, including the role of the key somatic sex determination factor doublesex.

12.
Dev Cell ; 22(5): 1041-51, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595675

RESUMO

Establishment of germline sexual identity is critical for production of male and female germline stem cells, as well as sperm versus eggs. Here we identify PHD Finger Protein 7 (PHF7) as an important factor for male germline sexual identity in Drosophila. PHF7 exhibits male-specific expression in early germ cells, germline stem cells, and spermatogonia. It is important for germline stem cell maintenance and gametogenesis in males, whereas ectopic expression in female germ cells ablates the germline. Strikingly, expression of PHF7 promotes spermatogenesis in XX germ cells when they are present in a male soma. PHF7 homologs are also specifically expressed in the mammalian testis, and human PHF7 rescues Drosophila Phf7 mutants. PHF7 associates with chromatin, and both the human and fly proteins bind histone H3 N-terminal tails with a preference for dimethyl lysine 4 (H3K4me2). We propose that PHF7 acts as a conserved epigenetic "reader" that activates the male germline sexual program.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Células Germinativas/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Processos de Determinação Sexual , Animais , Cromatina/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Mutação , Especificidade de Órgãos , Ovário/citologia , Ovário/metabolismo , Alinhamento de Sequência , Espermatogênese/genética , Células-Tronco/metabolismo , Testículo/citologia , Testículo/metabolismo
13.
Dev Biol ; 367(2): 114-25, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22575490

RESUMO

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.


Assuntos
Caderinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Drosophila/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Gônadas/embriologia , Gônadas/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Animais Geneticamente Modificados , Sequência de Bases , Adesão Celular , Proteínas do Citoesqueleto/genética , Primers do DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Genes de Insetos , Gônadas/citologia , Mutação
15.
Development ; 138(16): 3357-66, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21752937

RESUMO

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.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Espermatozoides/metabolismo , Células-Tronco/metabolismo , Animais , Proteínas Cromossômicas não Histona/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Epigênese Genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mutação , Oogênese , Óvulo/citologia , Óvulo/metabolismo , Espermatogênese , Espermatozoides/citologia , Células-Tronco/citologia , Transcrição Gênica
16.
Dev Biol ; 353(2): 217-28, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21377458

RESUMO

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.


Assuntos
Proteínas de Drosophila/genética , Drosophila/embriologia , Drosophila/genética , Genes de Insetos , Gônadas/embriologia , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Animais , Animais Geneticamente Modificados , Caderinas/genética , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Gônadas/citologia , Mutagênese , Mutação , Fenótipo , Transdução de Sinais , Proteínas Roundabout
18.
Curr Opin Cell Biol ; 22(6): 722-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21030233

RESUMO

Sex determination is regulated very differently in the soma vs. the germline, yet both processes are critical for the creation of the male and female gametes. In general, the soma plays an essential role in regulating sexual identity of the germline. However, in some species, such as Drosophila and mouse, the sex chromosome constitution of the germ cells makes an autonomous contribution to germline sexual development. Here we review how the soma and germline cooperate to determine germline sexual identity for some important model systems, the fly, the worm and the mouse, and discuss some of the implications of 'dual control' (soma plus germline) as compared to species where germline sex is dictated only by the surrounding soma.


Assuntos
Células Germinativas/fisiologia , Processos de Determinação Sexual/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Masculino , Oócitos/fisiologia , Espermatozoides/fisiologia
19.
Development ; 136(22): 3821-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19855024

RESUMO

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.


Assuntos
Drosophila melanogaster/embriologia , Drosophila melanogaster/fisiologia , Células Germinativas/fisiologia , Processos de Determinação Sexual , Animais , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/citologia , Gônadas/fisiologia , Masculino , Fatores de Transcrição/fisiologia
20.
Dev Biol ; 334(2): 335-44, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19643104

RESUMO

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.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Células-Tronco Embrionárias/citologia , Janus Quinases/fisiologia , Fatores de Transcrição STAT/fisiologia , Espermatozoides/citologia , Testículo/embriologia , Fatores de Transcrição/fisiologia , Animais , Adesão Celular , Polaridade Celular , Proteínas de Drosophila/análise , Drosophila melanogaster/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células-Tronco Embrionárias/metabolismo , Ativação Enzimática , Larva , Masculino , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais/fisiologia , Espermatogônias/citologia , Espermatozoides/metabolismo , Testículo/citologia , Proteínas Supressoras de Tumor/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...