Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters











Publication year range
2.
Elife ; 122023 Jun 14.
Article in English | MEDLINE | ID: mdl-37314324

ABSTRACT

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.


Subject(s)
Drosophila Proteins , Neural Stem Cells , Animals , Humans , Histones/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Histone Code , Neural Stem Cells/metabolism , Transcription, Genetic , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism
3.
Science ; 375(6584): eabk2432, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35239393

ABSTRACT

For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Transcriptome , Animals , Cell Nucleus/metabolism , Databases, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Gene Expression Regulation , Gene Regulatory Networks , Genes, Insect , Male , RNA-Seq , Sex Characteristics , Single-Cell Analysis , Transcription Factors/genetics
4.
Annu Rev Cell Dev Biol ; 37: 519-547, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34613817

ABSTRACT

Male and female brains display anatomical and functional differences. Such differences are observed in species across the animal kingdom, including humans, but have been particularly well-studied in two classic animal model systems, the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans. Here we summarize recent advances in understanding how the worm and fly brain acquire sexually dimorphic features during development. We highlight the advantages of each system, illustrating how the precise anatomical delineation of sexual dimorphisms in worms has enabled recent analysis into how these dimorphisms become specified during development, and how focusing on sexually dimorphic neurons in the fly has enabled an increasingly detailed understanding of sex-specific behaviors.


Subject(s)
Drosophila melanogaster , Nervous System , Animals , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Female , Male , Neurons/physiology , Sex Characteristics
5.
J Neurogenet ; 35(3): 285-294, 2021 09.
Article in English | MEDLINE | ID: mdl-34338589

ABSTRACT

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.


Subject(s)
Courtship , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Nerve Tissue Proteins/genetics , Sexual Behavior, Animal/physiology , Transcription Factors/genetics , Animals , Male , Mutation , Promoter Regions, Genetic
7.
Curr Biol ; 31(6): 1175-1191.e6, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33508219

ABSTRACT

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.


Subject(s)
Drosophila melanogaster , Sex Characteristics , Sexual Behavior, Animal/physiology , Smell/physiology , Vision, Ocular/physiology , Animals , Brain/cytology , Brain/physiology , Courtship , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Female , Male , Neurons/physiology , Oviposition , Photic Stimulation
8.
Cell ; 184(2): 507-520.e16, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33382967

ABSTRACT

Aggression involves both sexually monomorphic and dimorphic actions. How the brain implements these two types of actions is poorly understood. We have identified three cell types that regulate aggression in Drosophila: one type is sexually shared, and the other two are sex specific. Shared common aggression-promoting (CAP) neurons mediate aggressive approach in both sexes, whereas functionally downstream dimorphic but homologous cell types, called male-specific aggression-promoting (MAP) neurons in males and fpC1 in females, control dimorphic attack. These symmetric circuits underlie the divergence of male and female aggressive behaviors, from their monomorphic appetitive/motivational to their dimorphic consummatory phases. The strength of the monomorphic → dimorphic functional connection is increased by social isolation in both sexes, suggesting that it may be a locus for isolation-dependent enhancement of aggression. Together, these findings reveal a circuit logic for the neural control of behaviors that include both sexually monomorphic and dimorphic actions, which may generalize to other organisms.


Subject(s)
Aggression/physiology , Drosophila melanogaster/physiology , Logic , Sex Characteristics , Sexual Behavior, Animal/physiology , Animals , Female , Male , Nerve Net/physiology , Neurons/physiology , Social Isolation , Tachykinins/metabolism
9.
Cell Rep ; 33(11): 108516, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326795

ABSTRACT

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.


Subject(s)
Drosophila Proteins/metabolism , Nerve Tissue Proteins/metabolism , Olfactory Receptor Neurons/metabolism , Transcription Factors/metabolism , Animals , Drosophila , Protein Isoforms/metabolism
10.
Curr Biol ; 30(19): 3736-3748.e5, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32795437

ABSTRACT

Communication between male and female fruit flies during courtship is essential for successful mating, but, as with many other species, it is the female who decides whether to mate. Here, we show a novel role for ovipositor extrusion in promoting male copulation attempts in virgin and mated females and signaling acceptance in virgins. We first show that ovipositor extrusion is only displayed by sexually mature females, exclusively during courtship and in response to the male song. We identified a pair of descending neurons that controls ovipositor extrusion in mated females. Genetic silencing of the descending neurons shows that ovipositor extrusion stimulates the male to attempt copulation. A detailed behavioral analysis revealed that during courtship, the male repeatedly licks the female genitalia, independently of ovipositor extrusion, and that licking an extruded ovipositor prompts a copulation attempt. However, if the ovipositor is not subsequently retracted, copulation is prevented, as it happens with mated females. In this study, we reveal a dual function of the ovipositor: while its extrusion is necessary for initiating copulation by the male, its retraction signals female acceptance. We thus uncover the significance of the communication between male and female that initiates the transition from courtship to copulation.


Subject(s)
Drosophila melanogaster/anatomy & histology , Oviposition/physiology , Sexual Behavior, Animal/physiology , Animals , Copulation/physiology , Courtship , Female , Genitalia, Female/physiology , Male
11.
Elife ; 92020 04 21.
Article in English | MEDLINE | ID: mdl-32314735

ABSTRACT

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.


Subject(s)
Central Nervous System/cytology , Drosophila melanogaster/cytology , Neurons/cytology , Transcriptome , Animals
12.
Curr Opin Insect Sci ; 35: 48-53, 2019 10.
Article in English | MEDLINE | ID: mdl-31336357

ABSTRACT

When Drosophila males encounter another fly, they have to make a rapid assessment to ensure the appropriate response: should they court, fight or pursue a different action entirely? Previous work has focused on the significance of sensory cues detected by the male during these encounters; however, recent evidence highlights the importance of the male's own internal state in shaping his responses. Additionally, once triggered, courtship is not a rigid sequence of motor actions, but rather a finely tuned behavioural display that must continually update in response to sensory feedback. Here, we review recent findings highlighting how sensory information and internal states are integrated ensuring appropriate action selection, and how they sustain and fine-tune motor output. We further discuss recent advances in our understanding of species differences in sensory processing that may contribute to reproductive isolation.


Subject(s)
Drosophila/physiology , Perception , Sexual Behavior, Animal , Aggression , Animals , Courtship , Decision Making/physiology , Drosophila/drug effects , Female , Male , Pheromones , Species Specificity
13.
Proc Biol Sci ; 285(1892)2018 11 28.
Article in English | MEDLINE | ID: mdl-30487307

ABSTRACT

Sex differences in lifespan are ubiquitous, but the underlying causal factors remain poorly understood. Inter- and intrasexual social interactions are well known to influence lifespan in many taxa, but it has proved challenging to separate the role of sex-specific behaviours from wider physiological differences between the sexes. To address this problem, we genetically manipulated the sexual identity of the nervous system-and hence sexual behaviour-in Drosophila melanogaster, and measured lifespan under varying social conditions. Consistent with previous studies, masculinization of the nervous system in females induced male-specific courtship behaviour and aggression, while nervous system feminization in males induced male-male courtship and reduced aggression. Control females outlived males, but masculinized female groups displayed male-like lifespans and male-like costs of group living. By varying the mixture of control and masculinized females within social groups, we show that male-specific behaviours are costly to recipients, even when received from females. However, consistent with recent findings, our data suggest courtship expression to be surprisingly low cost. Overall, our study indicates that nervous system-mediated expression of sex-specific behaviour per se-independent of wider physiological differences between the sexes, or the receipt of aggression or courtship-plays a limited role in mediating sex differences in lifespan.


Subject(s)
Drosophila melanogaster/physiology , Sexual Behavior, Animal/physiology , Animals , Longevity/physiology , Nervous System Physiological Phenomena , Sex Characteristics , Sex Factors , Social Behavior
14.
Curr Biol ; 28(15): R840-R842, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30086319

ABSTRACT

Animals rely on sensory cues to help them find suitable mates. Visual cues are particularly useful for locating mates during the day. A new study has revealed key visual neurons in male Drosophila used to identify and pursue potential mates.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Courtship , Love , Male , Neurons , Sexual Behavior, Animal
15.
Biol Open ; 7(6)2018 Jun 11.
Article in English | MEDLINE | ID: mdl-29666051

ABSTRACT

Males in numerous animal species use mating songs to attract females and intimidate competitors. We demonstrate that modulations in song amplitude are behaviourally relevant in the fruit fly Drosophila We show that Drosophilamelanogaster females prefer amplitude modulations that are typical of melanogaster song over other modulations, which suggests that amplitude modulations are processed auditorily by D. melanogaster Our work demonstrates that receivers can decode messages in amplitude modulations, complementing the recent finding that male flies actively control song amplitude. To describe amplitude modulations, we propose the concept of song amplitude structure (SAS) and discuss similarities and differences to amplitude modulation with distance (AMD).This article has an associated First Person interview with the first author of the paper.

16.
Adv Genet ; 99: ix, 2017.
Article in English | MEDLINE | ID: mdl-29050556
17.
Nat Ecol Evol ; 1(6): 0154, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28580431

ABSTRACT

Female aggression towards other females is associated with reproduction in many taxa, and traditionally thought to be related to the protection or provisioning of offspring, such as through increased resource acquisition. However, the underlying reproductive factors causing aggressive behaviour in females remain unknown. Here we show that female aggression in the fruit fly Drosophila melanogaster is strongly stimulated by the receipt of sperm at mating, and in part by an associated seminal fluid protein, the sex peptide. We further show that the post-mating increase in female aggression is decoupled from the costs of egg production and from post-mating decreases in sexual receptivity. Our results suggest that male ejaculates can have a surprisingly direct influence on aggression in recipient females. Male ejaculate traits thus influence the female social competitive environment with potentially far-reaching ecological and evolutionary consequences.

18.
Elife ; 52016 11 15.
Article in English | MEDLINE | ID: mdl-27855059

ABSTRACT

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.


Subject(s)
Copulation , Drosophila/physiology , Interneurons/physiology , Motor Neurons/physiology , Neural Pathways , Sensory Receptor Cells/physiology , Animals , Male , Neural Networks, Computer
19.
Curr Biol ; 26(18): 2508-2515, 2016 09 26.
Article in English | MEDLINE | ID: mdl-27568592

ABSTRACT

Courtship in Drosophila melanogaster offers a powerful experimental paradigm for the study of innate sexually dimorphic behaviors [1, 2]. Fruit fly males exhibit an elaborate courtship display toward a potential mate [1, 2]. Females never actively court males, but their response to the male's display determines whether mating will actually occur. Sex-specific behaviors are hardwired into the nervous system via the actions of the sex determination genes doublesex (dsx) and fruitless (fru) [1]. Activation of male-specific dsx/fru(+) P1 neurons in the brain initiates the male's courtship display [3, 4], suggesting that neurons unique to males trigger this sex-specific behavior. In females, dsx(+) neurons play a pivotal role in sexual receptivity and post-mating behaviors [1, 2, 5-9]. Yet it is still unclear how dsx(+) neurons and dimorphisms in these circuits give rise to the different behaviors displayed by males and females. Here, we manipulated the function of dsx(+) neurons in the female brain to investigate higher-order neurons that drive female behaviors. Surprisingly, we found that activation of female dsx(+) neurons in the brain induces females to behave like males by promoting male-typical courtship behaviors. Activated females display courtship toward conspecific males or females, as well other Drosophila species. We uncovered specific dsx(+) neurons critical for driving male courtship and identified pheromones that trigger such behaviors in activated females. While male courtship behavior was thought to arise from male-specific central neurons, our study shows that the female brain is equipped with latent courtship circuitry capable of inducing this male-specific behavioral program.


Subject(s)
Courtship , Drosophila melanogaster/physiology , Neurons/physiology , Animals , Brain/physiology , Female
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL