Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37314324

RESUMEN

Coordinated regulation of gene activity by transcriptional and translational mechanisms poise stem cells for a timely cell-state transition during differentiation. Although important for all stemness-to-differentiation transitions, mechanistic understanding of the fine-tuning of gene transcription is lacking due to the compensatory effect of translational control. We used intermediate neural progenitor (INP) identity commitment to define the mechanisms that fine-tune stemness gene transcription in fly neural stem cells (neuroblasts). We demonstrate that the transcription factor FruitlessC (FruC) binds cis-regulatory elements of most genes uniquely transcribed in neuroblasts. Loss of fruC function alone has no effect on INP commitment but drives INP dedifferentiation when translational control is reduced. FruC negatively regulates gene expression by promoting low-level enrichment of the repressive histone mark H3K27me3 in gene cis-regulatory regions. Identical to fruC loss-of-function, reducing Polycomb Repressive Complex 2 activity increases stemness gene activity. We propose low-level H3K27me3 enrichment fine-tunes gene transcription in stem cells, a mechanism likely conserved from flies to humans.


From neurons to sperm, our bodies are formed of a range of cells tailored to perform a unique role. However, organisms also host small reservoirs of unspecialized 'stem cells' that retain the ability to become different kinds of cells. When these stem cells divide, one daughter cell remains a stem cell while the other undergoes a series of changes that allows it to mature into a specific cell type. This 'differentiation' process involves quickly switching off the stem cell programme, the set of genes that give a cell the ability to keep dividing while maintaining an unspecialized state. Failure to do so can result in the differentiating cell reverting towards its initial state and multiplying uncontrollably, which can lead to tumours and other health problems. While scientists have a good understanding of how the stem cell programme is turned off during differentiation, controlling these genes is a balancing act that starts even before division: if the program is over-active in the 'mother' stem cell, for instance, the systems that switch it off in its daughter can become overwhelmed. The mechanisms presiding over these steps are less well-understood. To address this knowledge gap, Rajan, Anhezini et al. set out to determine how stem cells present in the brains of fruit flies could control the level of activity of their own stem cell programme. RNA sequencing and other genetic analyses revealed that a protein unique to these cells, called Fruitless, was responsible for decreasing the activity of the programme. Biochemical experiments then showed that Fruitless performed this role by attaching a small amount of chemical modifications (called methyl groups) to the proteins that 'package' the DNA near genes involved in the stem cell programme. High levels of methyl groups present near a gene will switch off this sequence completely; however, the amount of methyl groups that Fruitless helped to deposit is multiple folds lower. Consequently, Fruitless 'fine-tunes' the activity of the stem cell programme instead, dampening it just enough to stop it from overpowering the 'off' mechanism that would take place later in the daughter cell. These results shed new light on how stem cells behave ­ and how our bodies stop them from proliferating uncontrollably. In the future, Rajan, Anhezini et al. hope that this work will help to understand and treat diseases caused by defective stem cell differentiation.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Humanos , Histonas/metabolismo , Drosophila melanogaster/genética , Proteínas de Drosophila/metabolismo , Código de Histonas , Células-Madre Neurales/metabolismo , Transcripción Genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/metabolismo
2.
J Neurogenet ; 35(3): 285-294, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34338589

RESUMEN

The identification of mutations in the gene fruitless (fru) paved the way for understanding the genetic basis of male sexual behavior in the vinegar fly Drosophila melanogaster. D. melanogaster males perform an elaborate courtship display to the female, ultimately leading to copulation. Mutations in fru have been shown to disrupt most aspects of the male's behavioral display, rendering males behaviorally sterile. The fru genomic locus encodes for multiple transcription factor isoforms from several promoters; only those under the regulation of the most distal P1 promoter are under the control of the sex determination hierarchy and play a role in male-specific behaviors. In this study, we used CRISPR/Cas9-based targeted genome editing of the fru gene, to remove the P1 promoter region. We have shown that removal of the P1 promoter leads to a dramatic decrease in male courtship displays towards females and male-specific sterility. We have expanded the analysis of fru P1-dependent behaviors, examining male's response to courtship song and general activity levels during12-hour light: dark cycles. Our novel allele expands the mutant repertoire available for future studies of fru P1-derived function in D. melanogaster. Our fruΔP1 mutant will be useful for future studies of fru P1-derived function, as it can be homozygosed without disrupting additional downstream promoter function and can be utilized in heterozygous combinations with other extant fru alleles.


Asunto(s)
Cortejo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas del Tejido Nervioso/genética , Conducta Sexual Animal/fisiología , Factores de Transcripción/genética , Animales , Masculino , Mutación , Regiones Promotoras Genéticas
3.
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
4.
Curr Biol ; 31(6): 1175-1191.e6, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33508219

RESUMEN

Although males and females largely share the same genome and nervous system, they differ profoundly in reproductive investments and require distinct behavioral, morphological, and physiological adaptations. How can the nervous system, while bound by both developmental and biophysical constraints, produce these sex differences in behavior? Here, we uncover a novel dimorphism in Drosophila melanogaster that allows deployment of completely different behavioral repertoires in males and females with minimum changes to circuit architecture. Sexual differentiation of only a small number of higher order neurons in the brain leads to a change in connectivity related to the primary reproductive needs of both sexes-courtship pursuit in males and communal oviposition in females. This study explains how an apparently similar brain generates distinct behavioral repertoires in the two sexes and presents a fundamental principle of neural circuit organization that may be extended to other species.


Asunto(s)
Drosophila melanogaster , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Olfato/fisiología , Visión Ocular/fisiología , Animales , Encéfalo/citología , Encéfalo/fisiología , Cortejo , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Femenino , Masculino , Neuronas/fisiología , Oviposición , Estimulación Luminosa
5.
Cell Rep ; 33(11): 108516, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326795

RESUMEN

Sexual dimorphism in Drosophila courtship circuits requires the male-specific transcription factor fruM, which is alternatively spliced to encode the FruMA, FruMB, and FruMC isoforms. Most fruM-positive neurons express multiple variants; however, the functional significance of their co-expression remains undetermined. Do co-expressed isoforms each play unique roles to jointly regulate dimorphism? By focusing on fruM-positive olfactory receptor neurons (ORNs), here, we show that FruMB and FruMC are both required for males' age-dependent sensitization to aphrodisiac olfactory cues in a cell-autonomous manner. Interestingly, FruMB expression is upregulated with age in Or47b and Ir84a ORNs, and its overexpression mimics the effect of age in elevating olfactory responses. Mechanistically, FruMB and FruMC synergistically mediate response sensitization through cooperation of their respective downstream effectors, namely, PPK25 and PPK23, which are both required for forming a functional amplification channel in ORNs. Together, these results provide critical mechanistic insight into how co-expressed FruM isoforms jointly coordinate dimorphic neurophysiology.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Factores de Transcripción/metabolismo , Animales , Drosophila , Isoformas de Proteínas/metabolismo
6.
Elife ; 92020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32314735

RESUMEN

The Drosophila ventral nerve cord (VNC) receives and processes descending signals from the brain to produce a variety of coordinated locomotor outputs. It also integrates sensory information from the periphery and sends ascending signals to the brain. We used single-cell transcriptomics to generate an unbiased classification of cellular diversity in the VNC of five-day old adult flies. We produced an atlas of 26,000 high-quality cells, representing more than 100 transcriptionally distinct cell types. The predominant gene signatures defining neuronal cell types reflect shared developmental histories based on the neuroblast from which cells were derived, as well as their birth order. The relative position of cells along the anterior-posterior axis could also be assigned using adult Hox gene expression. This single-cell transcriptional atlas of the adult fly VNC will be a valuable resource for future studies of neurodevelopment and behavior.


Asunto(s)
Sistema Nervioso Central/citología , Drosophila melanogaster/citología , Neuronas/citología , Transcriptoma , Animales
7.
Genetics ; 205(2): 761-773, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28007892

RESUMEN

Nutrient acquisition and energy storage are critical parts of achieving metabolic homeostasis. The foraging gene in Drosophila melanogaster has previously been implicated in multiple feeding-related and metabolic traits. Before foraging's functions can be further dissected, we need a precise genetic null mutant to definitively map its amorphic phenotypes. We used homologous recombination to precisely delete foraging, generating the for0 null allele, and used recombineering to reintegrate a full copy of the gene, generating the {forBAC} rescue allele. We show that a total loss of foraging expression in larvae results in reduced larval path length and food intake behavior, while conversely showing an increase in triglyceride levels. Furthermore, varying foraging gene dosage demonstrates a linear dose-response on these phenotypes in relation to foraging gene expression levels. These experiments have unequivocally proven a causal, dose-dependent relationship between the foraging gene and its pleiotropic influence on these feeding-related traits. Our analysis of foraging's transcription start sites, termination sites, and splicing patterns using rapid amplification of cDNA ends (RACE) and full-length cDNA sequencing, revealed four independent promoters, pr1-4, that produce 21 transcripts with nine distinct open reading frames (ORFs). The use of alternative promoters and alternative splicing at the foraging locus creates diversity and flexibility in the regulation of gene expression, and ultimately function. Future studies will exploit these genetic tools to precisely dissect the isoform- and tissue-specific requirements of foraging's functions and shed light on the genetic control of feeding-related traits involved in energy homeostasis.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Conducta Alimentaria , Dosificación de Gen , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Pleiotropía Genética , Fenotipo , Regiones Promotoras Genéticas , Empalme del ARN , Triglicéridos/metabolismo
8.
Elife ; 52016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27855059

RESUMEN

Copulation is the goal of the courtship process, crucial to reproductive success and evolutionary fitness. Identifying the circuitry underlying copulation is a necessary step towards understanding universal principles of circuit operation, and how circuit elements are recruited into the production of ordered action sequences. Here, we identify key sex-specific neurons that mediate copulation in Drosophila, and define a sexually dimorphic motor circuit in the male abdominal ganglion that mediates the action sequence of initiating and terminating copulation. This sexually dimorphic circuit composed of three neuronal classes - motor neurons, interneurons and mechanosensory neurons - controls the mechanics of copulation. By correlating the connectivity, function and activity of these neurons we have determined the logic for how this circuitry is coordinated to generate this male-specific behavior, and sets the stage for a circuit-level dissection of active sensing and modulation of copulatory behavior.


Asunto(s)
Copulación , Drosophila/fisiología , Interneuronas/fisiología , Neuronas Motoras/fisiología , Vías Nerviosas , Células Receptoras Sensoriales/fisiología , Animales , Masculino , Redes Neurales de la Computación
9.
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
10.
Fly (Austin) ; 8(2): 95-100, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483248

RESUMEN

Courtship is pivotal to successful reproduction throughout the animal kingdom. Sexual differences in the nervous system are thought to underlie courtship behavior. Male courtship behavior in Drosophila is in large part regulated by the gene fruitless (fru). fru has been reported to encode at least three putative BTB-zinc-finger transcription factors predicted to have different DNA-binding specificities. Although a large number of previous studies have demonstrated that fru plays essential roles in male courtship behavior, we know little about the function of Fru isoforms at the molecular level. Our recent study revealed that male-specific Fru isoforms are expressed in highly overlapping subsets of neurons in the male brain and ventral nerve cord. Fru isoforms play both distinct and redundant roles in male courtship behavior. Importantly, we have identified for the first time, by means of the DamID technique, direct Fru transcriptional target genes. Fru target genes overwhelmingly represent genes previously reported to be involved in the nervous system development, such as CadN, lola and pdm2. Our study provides important insight into how the sexually dimorphic neural circuits underlying reproductive behavior are established.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/crecimiento & desarrollo , Conducta Sexual Animal/fisiología , Factores de Transcripción/genética , Animales , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Proteínas del Tejido Nervioso/metabolismo , Isoformas de Proteínas/metabolismo , Caracteres Sexuales , Desarrollo Sexual , Factores de Transcripción/metabolismo
11.
Curr Biol ; 24(6): R243-4, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24650914

RESUMEN

Animals are constantly receiving information about their environment that must be filtered to ensure that they respond in the appropriate manner. New data have revealed how neurons in male Drosophila promote a heightened state of aggression in response to a rival male.


Asunto(s)
Drosophila melanogaster/fisiología , Neuronas/metabolismo , Taquicininas/metabolismo , Animales , Femenino , Masculino
12.
Curr Biol ; 24(7): 725-30, 2014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24631243

RESUMEN

Mating elicits profound behavioral and physiological changes in many species that are crucial for reproductive success. After copulation, Drosophila melanogaster females reduce their sexual receptivity and increase egg laying [1, 2]. Transfer of male sex peptide (SP) during copulation mediates these postmating responses [1, 3-6] via SP sensory neurons in the uterus defined by coexpression of the proprioceptive neuronal marker pickpocket (ppk) and the sex-determination genes doublesex (dsx) and fruitless (fru) [7-9]. Although neurons expressing dsx downstream of SP signaling have been shown to regulate postmating behaviors [9], how the female nervous system coordinates the change from pre- to postcopulatory states is unknown. Here, we show a role of the neuromodulator octopamine (OA) in the female postmating response. Lack of OA disrupts postmating responses in mated females, while increase of OA induces postmating responses in virgin females. Using a novel dsx(FLP) allele, we uncovered dsx neuronal elements associated with OA signaling involved in modulation of postmating responses. We identified a small subset of sexually dimorphic OA/dsx(+) neurons (approximately nine cells in females) in the abdominal ganglion. Our results are consistent with a model whereby OA neuronal signaling increases after copulation, which in turn modulates changes in female behavior and physiology in response to reproductive state.


Asunto(s)
Drosophila/fisiología , Octopamina/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Femenino , Datos de Secuencia Molecular , Octopamina/farmacología , Octopamina/fisiología , Caracteres Sexuales
13.
Curr Biol ; 24(3): 229-41, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24440396

RESUMEN

BACKGROUND: In Drosophila, male courtship behavior is regulated in large part by the gene fruitless (fru). fru encodes a set of putative transcription factors that promote male sexual behavior by controlling the development of sexually dimorphic neuronal circuitry. Little is known about how Fru proteins function at the level of transcriptional regulation or the role that isoform diversity plays in the formation of a male-specific nervous system. RESULTS: To characterize the roles of sex-specific Fru isoforms in specifying male behavior, we generated novel isoform-specific mutants and used a genomic approach to identify direct Fru isoform targets during development. We demonstrate that all Fru isoforms directly target genes involved in the development of the nervous system, with individual isoforms exhibiting unique binding specificities. We observe that fru behavioral phenotypes are specified by either a single isoform or a combination of isoforms. Finally, we illustrate the utility of these data for the identification of novel sexually dimorphic genomic enhancers and novel downstream regulators of male sexual behavior. CONCLUSIONS: These findings suggest that Fru isoform diversity facilitates both redundancy and specificity in gene expression, and that the regulation of neuronal developmental genes may be the most ancient and conserved role of fru in the specification of a male-specific nervous system.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Factores de Transcripción/genética , Animales , Secuencia de Bases , Sistema Nervioso Central/metabolismo , Drosophila melanogaster/metabolismo , Técnicas de Inactivación de Genes , Masculino , Datos de Secuencia Molecular , Isoformas de Proteínas/genética
14.
Nat Neurosci ; 13(4): 458-66, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20305646

RESUMEN

Doublesex proteins, which are part of the structurally and functionally conserved Dmrt gene family, are important for sex determination throughout the animal kingdom. We inserted Gal4 into the doublesex (dsx) locus of Drosophila melanogaster, allowing us to visualize and manipulate cells expressing dsx in various tissues. In the nervous system, we detected differences between the sexes in dsx-positive neuronal numbers, axonal projections and synaptic density. We found that dsx was required for the development of male-specific neurons that coexpressed fruitless (fru), a regulator of male sexual behavior. We propose that dsx and fru act together to form the neuronal framework necessary for male sexual behavior. We found that disrupting dsx neuronal function had profound effects on male sexual behavior. Furthermore, our results suggest that dsx-positive neurons are involved in pre- to post-copulatory female reproductive behaviors.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Diferenciación Sexual/genética , Conducta Sexual Animal/fisiología , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Femenino , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...