Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.026
Filtrar
1.
J Cell Biol ; 223(2)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38261271

RESUMEN

The nuclear lamina (NL) plays various roles and participates in nuclear integrity, chromatin organization, and transcriptional regulation. Lamin proteins, the main components of the NL, form a homogeneous meshwork structure under the nuclear envelope. Lamins are essential, but it is unknown whether their homogeneous distribution is important for nuclear function. Here, we found that PIGB, an enzyme involved in glycosylphosphatidylinositol (GPI) synthesis, is responsible for the homogeneous lamin meshwork in Drosophila. Loss of PIGB resulted in heterogeneous distributions of B-type lamin and lamin-binding proteins in larval muscles. These phenotypes were rescued by expression of PIGB lacking GPI synthesis activity. The PIGB mutant exhibited changes in lamina-associated domains that are large heterochromatic genomic regions in the NL, reduction of nuclear stiffness, and deformation of muscle fibers. These results suggest that PIGB maintains the homogeneous meshwork of the NL, which may be essential for chromatin distribution and nuclear mechanical properties.


Asunto(s)
Proteínas de Drosophila , Drosophila , Músculo Esquelético , Lámina Nuclear , Animales , Lamina Tipo B/genética , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Lámina Nuclear/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Glicosilfosfatidilinositoles/metabolismo
2.
Curr Biol ; 34(3): 473-488.e6, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38181792

RESUMEN

Complex behaviors depend on the precise developmental specification of neuronal circuits, but the relationship between genetic programs for neural development, circuit structure, and behavioral output is often unclear. The central complex (CX) is a conserved sensory-motor integration center in insects, which governs many higher-order behaviors and largely derives from a small number of type II neural stem cells (NSCs). Here, we show that Imp, a conserved IGF-II mRNA-binding protein expressed in type II NSCs, plays a role in specifying essential components of CX olfactory navigation circuitry. We show the following: (1) that multiple components of olfactory navigation circuitry arise from type II NSCs. (2) Manipulating Imp expression in type II NSCs alters the number and morphology of many of these circuit elements, with the most potent effects on neurons targeting the ventral layers of the fan-shaped body (FB). (3) Imp regulates the specification of Tachykinin-expressing ventral FB input neurons. (4) Imp is required in type II NSCs for establishing proper morphology of the CX neuropil structures. (5) Loss of Imp in type II NSCs abolishes upwind orientation to attractive odor while leaving locomotion and odor-evoked regulation of movement intact. Taken together, our findings establish that a temporally expressed gene can regulate the expression of a complex behavior by developmentally regulating the specification of multiple circuit components and provides a first step toward a developmental dissection of the CX and its roles in behavior.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Células-Madre Neurales , Proteínas de Unión al ARN , Olfato , Navegación Espacial , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Células-Madre Neurales/metabolismo , Neuronas/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología
3.
J Cell Biol ; 223(2)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38126997

RESUMEN

Lattice cells (LCs) in the developing Drosophila retina change shape before attaining final form. Previously, we showed that repeated contraction and expansion of apical cell contacts affect these dynamics. Here, we describe another factor, the assembly of a Rho1-dependent medioapical actomyosin ring formed by nodes linked by filaments that contract the apical cell area. Cell area contraction alternates with relaxation, generating pulsatile changes in cell area that exert force on neighboring LCs. Moreover, Rho1 signaling is sensitive to mechanical changes, becoming active when tension decreases and cells expand, while the negative regulator RhoGAP71E accumulates when tension increases and cells contract. This results in cycles of cell area contraction and relaxation that are reciprocally synchronized between adjacent LCs. Thus, mechanically sensitive Rho1 signaling controls pulsatile medioapical actomyosin contraction and coordinates cell behavior across the epithelium. Disrupting the kinetics of pulsing can lead to developmental errors, suggesting this process controls cell shape and tissue integrity during epithelial morphogenesis of the retina.


Asunto(s)
Actomiosina , Drosophila , Ojo , Animales , Citoesqueleto de Actina/fisiología , Actomiosina/fisiología , Citocinesis , Drosophila/embriología , Morfogénesis , Ojo/embriología , Proteínas de Unión al GTP rho/fisiología , Proteínas de Drosophila/fisiología , Retina/citología
4.
Neural Dev ; 18(1): 9, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38031099

RESUMEN

The generation of neuronal diversity remains incompletely understood. In Drosophila, the central brain is populated by neural stem cells derived from progenitors called neuroblasts (NBs). There are two types of NBs, type 1 and 2. T1NBs have a relatively simple lineage, whereas T2NBs expand and diversify the neural population with the generation of intermediate neural progenitors (INPs), contributing many neurons to the adult central complex, a brain region essential for navigation. However, it is not fully understood how neural diversity is created in T2NB and INP lineages. Imp, an RNA-binding protein, is expressed in T2NBs in a high-to-low temporal gradient, while the RNA-binding protein Syncrip forms an opposing gradient. It remains unknown if Imp expression is carried into INPs; whether it forms a gradient similar to NBs; and whether INP expression of Imp is required for generating neuronal identity or morphology. Here, we show that Imp/Syp are both present in INPs, but not always in opposing gradients. We find that newborn INPs adopt their Imp/Syp levels from their parental T2NBs; that Imp and Syp are expressed in stage-specific high-to-low gradients in INPs. In addition, there is a late INP pulse of Imp. We find that neurons born from old INPs (E-PG and PF-R neurons) have altered morphology following both Imp knock-down and Imp overexpression. We conclude that Imp functions in INPs and newborn neurons to determine proper neuronal morphology and central complex neuropil organization.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Proteínas de Drosophila/fisiología , Linaje de la Célula/fisiología , Neuronas , Drosophila , Neurópilo/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
5.
Sci Rep ; 13(1): 12502, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37532734

RESUMEN

To investigate the effect of the antioxidant N-acetylcysteine (NAC) on the proliferation and apoptosis in CG8005 gene-interfering Drosophila S2 embryonic cells by scavenging intracellular reactive oxygen species (ROS). The interfering efficiency of CG8005 gene in Drosophila S2 embryonic cells was verified by real-time quantitative PCR (qRT-PCR). Different concentrations of NAC and phosphate buffered saline (PBS) were used to affect the Drosophila S2 embryonic cells. The growth state of Drosophila S2 embryonic cells was observed by light microscope. Two probes dihydroethidium (DHE) and 2,7-dichlorodihydrofluorescein-acetoacetate (DCFH-DA) were used to observe the ROS production in each group after immunofluorescence staining. TUNEL staining and flow cytometry were used to investigate the apoptosis level of Drosophila S2 embryos, and CCK-8 (Cell Counting Kit-8) was used to detect the cell viability of Drosophila S2 embryos. The knockdown efficiency of siCG8005-2 fragment was high and stable, which was verified by interference efficiency (P < 0.05). There was no significant change in the growth of Drosophila S2 embryonic cells after the treatment of NAC as compared to PBS group. Moreover, knockdowning CG8005 gene resulted in an increase in ROS and apoptosis in Drosophila S2 embryonic cells (P < 0.05) and a decrease in proliferation activity (P < 0.05). In addition, the pretreatment of antioxidant NAC could inhibit ROS production in Drosophila S2 embryonic cells (P < 0.05), reduce cell apoptosis (P < 0.05), and improve cell survival (P < 0.05). The CG8005 gene in Drosophila S2 embryonic cells could regulate the proliferation and apoptosis of S2 embryonic cells by disrupting the redox homeostasis, and antioxidant NAC could inhibit cell apoptosis and promotes cell proliferation by scavenging ROS in Drosophila S2 embryonic cells, which is expected to provide novel insights for the pathogenesis of male infertility and spermatogenesis.


Asunto(s)
Acetilcisteína , Antioxidantes , Proteínas de Drosophila , Drosophila , Animales , Masculino , Acetilcisteína/farmacología , Antioxidantes/farmacología , Apoptosis , Proliferación Celular , Drosophila/embriología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología
6.
PLoS One ; 18(8): e0290048, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37561803

RESUMEN

Drosophila melanogaster is a popular model organism in the study of memory due to a wide arsenal of methods used to analyze neuronal activity. The most commonly used tests in research of behavioral plasticity are shock avoidance associated with chemosensory cues and courtship suppression after mating failure. Many authors emphasize the value of courtship suppression as a model of behavior most appropriate to natural conditions. However, researchers often investigate courtship suppression using immobilized and decapitated females as targets of courtship by males, which makes the data obtained from such flies less valuable. In our study, we evaluate courtship suppression towards immature mobile non-receptive females after training with mated or immature females combined with an aversive stimulus (quinine). We have shown that the previously described mechanisms of courtship suppression, as a result of the association of the courtship object with the repellent, as well as due to increased sensitivity to the anti-aphrodisiac cVA after mating failure, are not confirmed when immature mobile females are used. We discuss the reasons for the discrepancies between our results and literature data, define the conditions to be met in the courtship suppression test if the aim is to analyze the natural forms of behavioral plasticity, and present data on the test modifications to approximate conditions to natural ones.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Masculino , Femenino , Drosophila melanogaster/fisiología , Proteínas de Drosophila/fisiología , Cortejo , Conducta Sexual Animal/fisiología
7.
Dev Cell ; 58(14): 1299-1313.e5, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37295436

RESUMEN

Heart development begins with the formation of a tube as cardiac progenitors migrate from opposite sides of the embryo. Abnormal cardiac progenitor movements cause congenital heart defects. However, the mechanisms of cell migration during early heart development remain poorly understood. Using quantitative microscopy, we found that in Drosophila embryos, cardiac progenitors (cardioblasts) migrated through a sequence of forward and backward steps. Cardioblast steps were associated with oscillatory non-muscle myosin II waves that induced periodic shape changes and were necessary for timely heart tube formation. Mathematical modeling predicted that forward cardioblast migration required a stiff boundary at the trailing edge. Consistent with this, we found a supracellular actin cable at the trailing edge of the cardioblasts that limited the amplitude of the backward steps, thus biasing the direction of cell movement. Our results indicate that periodic shape changes coupled with a polarized actin cable produce asymmetrical forces that promote cardioblast migration.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Proteínas de Drosophila/fisiología , Actinas , Corazón , Miosinas , Morfogénesis , Drosophila melanogaster
8.
Curr Biol ; 33(13): 2702-2716.e3, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37352854

RESUMEN

Sleep is essential, but animals may forgo sleep to engage in other critical behaviors, such as feeding and reproduction. Previous studies have shown that female flies exhibit decreased sleep after mating, but our understanding of the process is limited. Here, we report that postmating nighttime sleep loss is modulated by diet and sleep deprivation, demonstrating a complex interaction among sleep, reproduction, and diet. We also find that female-specific pC1 neurons and sleep-promoting dorsal fan-shaped body (dFB) neurons are required for postmating sleep plasticity. Activating pC1 neurons leads to sleep suppression on standard fly culture media but has little sleep effect on sucrose-only food. Published connectome data suggest indirect, inhibitory connections among pC1 subtypes. Using calcium imaging, we show that activating the pC1e subtype inhibits dFB neurons. We propose that pC1 and dFB neurons integrate the mating status, food context, and sleep drive to modulate postmating sleep plasticity.


Asunto(s)
Proteínas de Drosophila , Trastornos del Inicio y del Mantenimiento del Sueño , Animales , Femenino , Drosophila/fisiología , Proteínas de Drosophila/fisiología , Sueño/fisiología , Privación de Sueño , Drosophila melanogaster/fisiología
9.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37218457

RESUMEN

Female insects can enter reproductive diapause, a state of suspended egg development, to conserve energy under adverse environments. In many insects, including the fruit fly, Drosophila melanogaster, reproductive diapause, also frequently called reproductive dormancy, is induced under low-temperature and short-day conditions by the downregulation of juvenile hormone (JH) biosynthesis in the corpus allatum (CA). In this study, we demonstrate that neuropeptide Diuretic hormone 31 (DH31) produced by brain neurons that project into the CA plays an essential role in regulating reproductive dormancy by suppressing JH biosynthesis in adult D. melanogaster. The CA expresses the gene encoding the DH31 receptor, which is required for DH31-triggered elevation of intracellular cAMP in the CA. Knocking down Dh31 in these CA-projecting neurons or DH31 receptor in the CA suppresses the decrease of JH titer, normally observed under dormancy-inducing conditions, leading to abnormal yolk accumulation in the ovaries. Our findings provide the first molecular genetic evidence demonstrating that CA-projecting peptidergic neurons play an essential role in regulating reproductive dormancy by suppressing JH biosynthesis.


Asunto(s)
Drosophila melanogaster , Hormonas de Insectos , Animales , Femenino , Corpora Allata , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Hormonas Juveniles , Neuronas , Hormonas de Insectos/genética , Hormonas de Insectos/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Reproducción
10.
Curr Biol ; 33(10): 2095-2103.e3, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37098339

RESUMEN

In a heterogeneous and changing environment, oviposition site selection strongly affects the survival and fitness of the offspring.1,2 Similarly, competition between larvae affects their prospects.3 However, little is known about the involvement of pheromones in regulating these processes.4,5,6,7,8 Here, we show that mated females of Drosophila melanogaster prefer to lay eggs on substrates containing extracts of conspecific larvae. After analyzing these extracts chemically, we test each compound in an oviposition assay and find that mated females display a dose-dependent preference to lay eggs on substrates spiked with (Z)-9-octadecenoic acid ethyl ester (OE). This egg-laying preference relies on gustatory receptor Gr32a and tarsal sensory neurons expressing this receptor. The concentration of OE also regulates larval place choice in a dose-dependent manner. Physiologically, OE activates female tarsal Gr32a+ neurons. In conclusion, our results reveal a cross-generation communication strategy essential for oviposition site selection and regulation of larval density.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Femenino , Drosophila melanogaster/fisiología , Oviposición/fisiología , Feromonas , Proteínas de Drosophila/fisiología , Larva/fisiología
11.
Neurosci Bull ; 39(9): 1396-1410, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36941515

RESUMEN

Females increase aggression for mating opportunities and for acquiring reproductive resources. Although the close relationship between female aggression and mating status is widely appreciated, whether and how female aggression is regulated by mating-related cues remains poorly understood. Here we report an interesting observation that Drosophila virgin females initiate high-frequency attacks toward mated females. We identify 11-cis-vaccenyl acetate (cVA), a male-derived pheromone transferred to females during mating, which promotes virgin female aggression. We subsequently reveal a cVA-responsive neural circuit consisting of four orders of neurons, including Or67d, DA1, aSP-g, and pC1 neurons, that mediate cVA-induced virgin female aggression. We also determine that aSP-g neurons release acetylcholine (ACh) to excite pC1 neurons via the nicotinic ACh receptor nAChRα7. Together, beyond revealing cVA as a mating-related inducer of virgin female aggression, our results identify a neural circuit linking the chemosensory perception of mating-related cues to aggressive behavior in Drosophila females.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Masculino , Femenino , Drosophila/fisiología , Proteínas de Drosophila/fisiología , Señales (Psicología) , Conducta Sexual Animal/fisiología , Agresión/fisiología , Drosophila melanogaster/fisiología
12.
J Cell Biol ; 222(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36409222

RESUMEN

In Drosophila melanogaster, the anterior-posterior body axis is maternally established and governed by differential localization of partitioning defective (Par) proteins within the oocyte. At mid-oogenesis, Par-1 accumulates at the oocyte posterior end, while Par-3/Bazooka is excluded there but maintains its localization along the remaining oocyte cortex. Past studies have proposed the need for somatic cells at the posterior end to initiate oocyte polarization by providing a trigger signal. To date, neither the molecular identity nor the nature of the signal is known. Here, we provide evidence that mechanical contact of posterior follicle cells (PFCs) with the oocyte cortex causes the posterior exclusion of Bazooka and maintains oocyte polarity. We show that Bazooka prematurely accumulates exclusively where posterior follicle cells have been mechanically detached or ablated. Furthermore, we provide evidence that PFC contact maintains Par-1 and oskar mRNA localization and microtubule cytoskeleton polarity in the oocyte. Our observations suggest that cell-cell contact mechanics modulates Par protein binding sites at the oocyte cortex.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Folículo Ovárico , Animales , Femenino , Tipificación del Cuerpo , Polaridad Celular , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/fisiología , Oocitos/fisiología , Folículo Ovárico/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología
13.
Proc Natl Acad Sci U S A ; 119(37): e2201513119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067320

RESUMEN

Most animal species display dimorphic sexual behaviors and male-biased aggressiveness. Current models have focused on the male-specific product from the fruitless (fruM) gene, which controls male courtship and male-specific aggression patterns in fruit flies, and describe a male-specific mechanism underlying sexually dimorphic behaviors. Here we show that the doublesex (dsx) gene, which expresses male-specific DsxM and female-specific DsxF transcription factors, functions in the nervous system to control both male and female sexual and aggressive behaviors. We find that Dsx is not only required in central brain neurons for male and female sexual behaviors, but also functions in approximately eight pairs of male-specific neurons to promote male aggressiveness and approximately two pairs of female-specific neurons to inhibit female aggressiveness. DsxF knockdown females fight more frequently, even with males. Our findings reveal crucial roles of dsx, which is broadly conserved from worms to humans, in a small number of neurons in both sexes to establish dimorphic sexual and aggressive behaviors.


Asunto(s)
Agresión , Cortejo , Proteínas de Unión al ADN , Proteínas de Drosophila , Drosophila melanogaster , Conducta Sexual Animal , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Femenino , Masculino
14.
Proc Natl Acad Sci U S A ; 119(38): e2204229119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095217

RESUMEN

Forgetting is an essential component of the brain's memory management system, providing a balance to memory formation processes by removing unused or unwanted memories, or by suppressing their expression. However, the molecular, cellular, and circuit mechanisms underlying forgetting are poorly understood. Here we show that the memory suppressor gene, sickie, functions in a single dopamine neuron (DAn) by supporting the process of active forgetting in Drosophila. RNAi knockdown (KD) of sickie impairs forgetting by reducing the Ca2+ influx and DA release from the DAn that promotes forgetting. Coimmunoprecipitation/mass spectrometry analyses identified cytoskeletal and presynaptic active zone (AZ) proteins as candidates that physically interact with Sickie, and a focused RNAi screen of the candidates showed that Bruchpilot (Brp)-a presynaptic AZ protein that regulates calcium channel clustering and neurotransmitter release-impairs active forgetting like sickie KD. In addition, overexpression of brp rescued the impaired forgetting of sickie KD, providing evidence that they function in the same process. Moreover, we show that sickie KD in the DAn reduces the abundance and size of AZ markers but increases their number, suggesting that Sickie controls DAn activity for forgetting by modulating the presynaptic AZ structure. Our results identify a molecular and circuit mechanism for normal levels of active forgetting and reveal a surprising role of Sickie in maintaining presynaptic AZ structure for neurotransmitter release.


Asunto(s)
Dopamina , Proteínas de Drosophila , Drosophila melanogaster , Memoria , Proteínas del Tejido Nervioso , Animales , Dopamina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Terminales Presinápticos/fisiología , Transmisión Sináptica
15.
PLoS One ; 17(8): e0269208, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35969522

RESUMEN

The Ajuba LIM protein Jub mediates regulation of Hippo signaling by cytoskeletal tension through interaction with the kinase Warts and participates in feedback regulation of junctional tension through regulation of the cytohesin Steppke. To investigate how Jub interacts with and regulates its distinct partners, we investigated the ability of Jub proteins missing different combinations of its three LIM domains to rescue jub phenotypes and to interact with α-catenin, Warts and Steppke. Multiple regions of Jub contribute to its ability to bind α-catenin and to localize to adherens junctions in Drosophila wing imaginal discs. Co-immunoprecipitation experiments in cultured cells identified a specific requirement for LIM2 for binding to Warts. However, in vivo, both LIM1 and LIM2, but not LIM3, were required for regulation of wing growth, Yorkie activity, and Warts localization. Conversely, LIM2 and LIM3, but not LIM1, were required for regulation of cell shape and Steppke localization in vivo, and for maximal Steppke binding in co-immunoprecipitation experiments. These observations identify distinct functions for the different LIM domains of Jub.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/metabolismo , Proteínas con Dominio LIM/fisiología , Animales , Citoesqueleto/química , Citoesqueleto/fisiología , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/análisis , Proteínas de Drosophila/genética , Proteínas con Dominio LIM/análisis , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM/análisis , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/fisiología , Transducción de Señal , Factores de Transcripción/metabolismo , Alas de Animales/crecimiento & desarrollo , alfa Catenina/metabolismo
16.
Proc Biol Sci ; 289(1981): 20220837, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35975433

RESUMEN

The insect gut is frequently exposed to pathogenic threats and must not only clear these potential infections, but also tolerate relatively high microbe loads. In contrast to the mechanisms that eliminate pathogens, we currently know less about the mechanisms of disease tolerance. We investigated how well-described mechanisms that prevent, signal, control or repair damage during infection contribute to the phenotype of disease tolerance. We established enteric infections with the bacterial pathogen Pseudomonas entomophila in transgenic lines of Drosophila melanogaster fruit flies affecting dcy (a major component of the peritrophic matrix), upd3 (a cytokine-like molecule), irc (a negative regulator of reactive oxygen species) and egfr1 (epithelial growth factor receptor). Flies lacking dcy experienced the highest mortality, while loss of function of either irc or upd3 reduced tolerance in both sexes. The disruption of egfr1 resulted in a severe loss in tolerance in male flies but had no substantial effect on the ability of female flies to tolerate P. entomophila infection, despite carrying greater microbe loads than males. Together, our findings provide evidence for the role of damage limitation mechanisms in disease tolerance and highlight how sexual dimorphism in these mechanisms could generate sex differences in infection outcomes.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Bacterias/metabolismo , Drosophila , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Receptores ErbB , Femenino , Masculino
17.
J Cell Biol ; 221(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-35976098

RESUMEN

Modulation of presynaptic actin dynamics is fundamental to synaptic growth and functional plasticity; yet the underlying molecular and cellular mechanisms remain largely unknown. At Drosophila NMJs, the presynaptic Rac1-SCAR pathway mediates BMP-induced receptor macropinocytosis to inhibit BMP growth signaling. Here, we show that the Rho-type GEF Vav acts upstream of Rac1 to inhibit synaptic growth through macropinocytosis. We also present evidence that Vav-Rac1-SCAR signaling has additional roles in tetanus-induced synaptic plasticity. Presynaptic inactivation of Vav signaling pathway components, but not regulators of macropinocytosis, impairs post-tetanic potentiation (PTP) and enhances synaptic depression depending on external Ca2+ concentration. Interfering with the Vav-Rac1-SCAR pathway also impairs mobilization of reserve pool (RP) vesicles required for tetanus-induced synaptic plasticity. Finally, treatment with an F-actin-stabilizing drug completely restores RP mobilization and plasticity defects in Vav mutants. We propose that actin-regulatory Vav-Rac1-SCAR signaling independently regulates structural and functional presynaptic plasticity by driving macropinocytosis and RP mobilization, respectively.


Asunto(s)
Actinas , Proteínas de Drosophila , Factores de Intercambio de Guanina Nucleótido , Plasticidad Neuronal , Sinapsis , Actinas/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas/fisiología , Calcio , Drosophila/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Microfilamentos/fisiología , Unión Neuromuscular/fisiología , Transducción de Señal , Sinapsis/fisiología , Tétanos/metabolismo , Proteínas de Unión al GTP rac/fisiología
18.
Neurosci Bull ; 38(11): 1277-1291, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35788510

RESUMEN

The choice of females to accept or reject male courtship is a critical decision for animal reproduction. Serotonin (5-hydroxytryptamine; 5-HT) has been found to regulate sexual behavior in many species, but it is unclear how 5-HT and its receptors function to regulate different aspects of sexual behavior. Here we used Drosophila melanogaster as the model animal to investigate how 5-HT and its receptors modulate female sexual receptivity. We found that knockout of tryptophan hydroxylase (Trh), which is involved in the biosynthesis of 5-HT, severely reduced virgin female receptivity without affecting post-mating behaviors. We identified a subset of sexually dimorphic Trh neurons that co-expressed fruitless (fru), in which the activity was correlated with sexual receptivity in females. We also found that 5-HT1A and 5-HT7 receptors regulate virgin female receptivity. Our findings demonstrate how 5-HT functions in sexually dimorphic neurons to promote virgin female receptivity through two of its receptors.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Masculino , Femenino , Drosophila/fisiología , Drosophila melanogaster/fisiología , Serotonina , Proteínas de Drosophila/fisiología , Conducta Sexual Animal/fisiología , Factores de Transcripción , Proteínas del Tejido Nervioso
19.
Proc Natl Acad Sci U S A ; 119(31): e2123467119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35881788

RESUMEN

Capicua (Cic) proteins are conserved HMG-box transcriptional repressors that control receptor tyrosine kinase (RTK) signaling responses and are implicated in human neurological syndromes and cancer. While Cic is known to exist as short (Cic-S) and long (Cic-L) isoforms with identical HMG-box and associated core regions but distinct N termini, most previous studies have focused on Cic-S, leaving the function of Cic-L unexplored. Here we show that Cic-L acts in two capacities during Drosophila oogenesis: 1) as a canonical sensor of RTK signaling in somatic follicle cells, and 2) as a regulator of postmitotic growth in germline nurse cells. In these latter cells, Cic-L behaves as a temporal signal that terminates endoreplicative growth before they dump their contents into the oocyte. We show that Cic-L is necessary and sufficient for nurse cell endoreplication arrest and induces both stabilization of CycE and down-regulation of Myc. Surprisingly, this function depends mainly on the Cic-L-specific N-terminal module, which is capable of acting independently of the Cic HMG-box-containing core. Mirroring these observations, basal metazoans possess truncated Cic-like proteins composed only of Cic-L N-terminal sequences, suggesting that this module plays unique, ancient roles unrelated to the canonical function of Cic.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Proteínas HMGB , Oogénesis , Proteínas Represoras , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Proteínas HMGB/genética , Proteínas HMGB/fisiología , Oogénesis/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/fisiología
20.
Dev Dyn ; 251(11): 1780-1797, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35656583

RESUMEN

BACKGROUND: POLR1D is a subunit of RNA Polymerases I and III, which synthesize ribosomal RNAs. Dysregulation of these polymerases cause several types of diseases, including ribosomopathies. The craniofacial disorder Treacher Collins Syndrome (TCS) is a ribosomopathy caused by mutations in several subunits of RNA Polymerase I, including POLR1D. Here, we characterized the effect of a missense mutation in POLR1D and RNAi knockdown of POLR1D on Drosophila development. RESULTS: We found that a missense mutation in Drosophila POLR1D (G30R) reduced larval rRNA levels, slowed larval growth, and arrested larval development. Remarkably, the G30R substitution is at an orthologous glycine in POLR1D that is mutated in a TCS patient (G52E). We showed that the G52E mutation in human POLR1D, and the comparable substitution (G30E) in Drosophila POLR1D, reduced their ability to heterodimerize with POLR1C in vitro. We also found that POLR1D is required early in the development of Drosophila neural cells. Furthermore, an RNAi screen revealed that POLR1D is also required for development of non-neural Drosophila cells, suggesting the possibility of defects in other cell types. CONCLUSIONS: These results establish a role for POLR1D in Drosophila development, and present Drosophila as an attractive model to evaluate the molecular defects of TCS mutations in POLR1D.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , Proteínas de Drosophila , Drosophila , Disostosis Mandibulofacial , Animales , Humanos , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/fisiología , Drosophila/embriología , Drosophila/genética , Disostosis Mandibulofacial/genética , Mutación , Fosfoproteínas/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA