Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
PLoS Genet ; 17(3): e1009468, 2021 03.
Article in English | MEDLINE | ID: mdl-33788836

ABSTRACT

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.


Subject(s)
Drosophila Proteins/genetics , Gene Expression Regulation , Gonads/cytology , Gonads/metabolism , Nerve Tissue Proteins/genetics , Sex Characteristics , Sex Determination Processes/genetics , Stem Cell Niche/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Animals , Drosophila Proteins/metabolism , Evolution, Molecular , Female , Fluorescent Antibody Technique , Gene Order , Genetic Loci , Male , Nerve Tissue Proteins/metabolism , Testis , Transcription Factors/metabolism
2.
Development ; 146(11)2019 06 10.
Article in English | MEDLINE | ID: mdl-31043421

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , Gonads/cytology , Gonads/metabolism , Sex Determination Processes/genetics , Stem Cell Niche/genetics , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Female , Gonads/embryology , Male , Organ Specificity/genetics , Transcription Factors/physiology
3.
PLoS Genet ; 12(9): e1006295, 2016 09.
Article in English | MEDLINE | ID: mdl-27599372

ABSTRACT

Deletions, commonly referred to as deficiencies by Drosophila geneticists, are valuable tools for mapping genes and for genetic pathway discovery via dose-dependent suppressor and enhancer screens. More recently, it has become clear that deviations from normal gene dosage are associated with multiple disorders in a range of species including humans. While we are beginning to understand some of the transcriptional effects brought about by gene dosage changes and the chromosome rearrangement breakpoints associated with them, much of this work relies on isolated examples. We have systematically examined deficiencies of the left arm of chromosome 2 and characterize gene-by-gene dosage responses that vary from collapsed expression through modest partial dosage compensation to full or even over compensation. We found negligible long-range effects of creating novel chromosome domains at deletion breakpoints, suggesting that cases of gene regulation due to altered nuclear architecture are rare. These rare cases include trans de-repression when deficiencies delete chromatin characterized as repressive in other studies. Generally, effects of breakpoints on expression are promoter proximal (~100bp) or in the gene body. Effects of deficiencies genome-wide are in genes with regulatory relationships to genes within the deleted segments, highlighting the subtle expression network defects in these sensitized genetic backgrounds.


Subject(s)
Chromatin/genetics , Drosophila melanogaster/genetics , Gene Dosage , Gene Regulatory Networks , Animals , Chromosome Breakpoints , Chromosomes, Insect/genetics , Gene Deletion
4.
Genetics ; 213(3): 877-895, 2019 11.
Article in English | MEDLINE | ID: mdl-31558581

ABSTRACT

Heterochromatin-mediated repression is essential for controlling the expression of transposons and for coordinated cell type-specific gene regulation. The small ovary (sov) locus was identified in a screen for female-sterile mutations in Drosophila melanogaster, and mutants show dramatic ovarian morphogenesis defects. We show that the null sov phenotype is lethal and map the locus to the uncharacterized gene CG14438, which encodes a nuclear zinc-finger protein that colocalizes with the essential Heterochromatin Protein 1 (HP1a). We demonstrate Sov functions to repress inappropriate gene expression in the ovary, silence transposons, and suppress position-effect variegation in the eye, suggesting a central role in heterochromatin stabilization.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Heterochromatin/metabolism , Animals , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/metabolism , DNA Transposable Elements , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Heterochromatin/genetics , Loss of Function Mutation , Ovary/growth & development , Ovary/metabolism , Zinc Fingers
5.
Genetics ; 206(4): 1939-1949, 2017 08.
Article in English | MEDLINE | ID: mdl-28588035

ABSTRACT

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.


Subject(s)
Drosophila Proteins/genetics , Homeodomain Proteins/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Histones/metabolism , Homeodomain Proteins/metabolism , Male , Spermatocytes/cytology
6.
mBio ; 8(4)2017 08 15.
Article in English | MEDLINE | ID: mdl-28811341

ABSTRACT

The U.S. Culture Collection Network held a meeting to share information about how culture collections are responding to the requirements of the recently enacted Nagoya Protocol on Access to Genetic Resources and the Fair and Equitable Sharing of Benefits Arising from their Utilization to the Convention on Biological Diversity (CBD). The meeting included representatives of many culture collections and other biological collections, the U.S. Department of State, U.S. Department of Agriculture, Secretariat of the CBD, interested scientific societies, and collection groups, including Scientific Collections International and the Global Genome Biodiversity Network. The participants learned about the policies of the United States and other countries regarding access to genetic resources, the definition of genetic resources, and the status of historical materials and genetic sequence information. Key topics included what constitutes access and how the CBD Access and Benefit-Sharing Clearing-House can help guide researchers through the process of obtaining Prior Informed Consent on Mutually Agreed Terms. U.S. scientists and their international collaborators are required to follow the regulations of other countries when working with microbes originally isolated outside the United States, and the local regulations required by the Nagoya Protocol vary by the country of origin of the genetic resource. Managers of diverse living collections in the United States described their holdings and their efforts to provide access to genetic resources. This meeting laid the foundation for cooperation in establishing a set of standard operating procedures for U.S. and international culture collections in response to the Nagoya Protocol.


Subject(s)
Biodiversity , Biological Specimen Banks , Biotechnology/legislation & jurisprudence , Environmental Microbiology , Agriculture/legislation & jurisprudence , Agriculture/organization & administration , Biological Specimen Banks/legislation & jurisprudence , Biological Specimen Banks/organization & administration , Biotechnology/organization & administration , Databases, Genetic/legislation & jurisprudence , Models, Genetic , United States , United States Department of Agriculture
7.
Genome Biol ; 15(6): 118, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-25002081

ABSTRACT

Recent work in the silkworm Bombyx mori has uncovered a novel Piwi-interacting RNA regulator of the sex determination switch doublesex.


Subject(s)
Bombyx/genetics , RNA, Small Interfering/genetics , Sex Characteristics , Sex Determination Processes/genetics , Animals , Female , Male
8.
Dev Cell ; 31(6): 761-73, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25535918

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , Animals , Animals, Genetically Modified , Binding Sites , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome , Genome-Wide Association Study , Male , Mice , Phenotype , RNA Interference , Sequence Analysis, DNA , Sex Factors , Transcription Factors/metabolism
9.
Spermatogenesis ; 2(3): 129-136, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-23087832

ABSTRACT

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.

10.
Curr Top Dev Biol ; 83: 65-107, 2008.
Article in English | MEDLINE | ID: mdl-19118664

ABSTRACT

Animals have evolved a fascinating array of mechanisms for conducting sexual reproduction. These include producing the sex-specific gametes, as well as mechanisms for attracting a mate, courting a mate, and getting the gametes together. These processes require that males and females take on dramatically different forms (sexual dimorphism). Here, we will explore the problem of how sex is determined in Drosophila, and pay particular attention to how information about sexual identity is used to instruct males and females to develop differently. Along the way, we will highlight new work that challenges some of the traditional views about sex determination. In Drosophila, it is commonly thought that every cell decides its own sex based on its sex chromosome constitution (XX vs. XY). However, we now know that many cell types undergo nonautonomous sex determination, where they are told what sex to be through signals from surrounding cells, independent of their own chromosomal content. Further, it now appears that not all cells even "know" their sex, since key members of the sex determination pathway are not expressed in all cells. Thus, our understanding of how sex is determined, and how sexual identity is used to create sexual dimorphism, has changed considerably.


Subject(s)
Drosophila/anatomy & histology , Drosophila/embryology , Sex Characteristics , Sex Determination Processes , Sex Differentiation , Animals , Drosophila/cytology , Drosophila/genetics , Female , Genes, Lethal , Gonads/embryology , Gonads/metabolism , Humans , Male , Sex Differentiation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL