Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(1): 18-20, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29958107

RESUMEN

A new study combines detailed biochemical characterization with whole-animal genetics and computational transcriptome data mining to reveal how the LRRC33 milieu molecule imposes an exquisite level of spatial control on TGF-ß signaling in the CNS.


Asunto(s)
Factor de Crecimiento Transformador beta/genética , Animales , Comunicación Celular , Microglía , Transducción de Señal , Transcriptoma
2.
Cell ; 163(4): 907-19, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26544939

RESUMEN

Steroid hormones are a large family of cholesterol derivatives regulating development and physiology in both the animal and plant kingdoms, but little is known concerning mechanisms of their secretion from steroidogenic tissues. Here, we present evidence that in Drosophila, endocrine release of the steroid hormone ecdysone is mediated through a regulated vesicular trafficking mechanism. Inhibition of calcium signaling in the steroidogenic prothoracic gland results in the accumulation of unreleased ecdysone, and the knockdown of calcium-mediated vesicle exocytosis components in the gland caused developmental defects due to deficiency of ecdysone. Accumulation of synaptotagmin-labeled vesicles in the gland is observed when calcium signaling is disrupted, and these vesicles contain an ABC transporter that functions as an ecdysone pump to fill vesicles. We propose that trafficking of steroid hormones out of endocrine cells is not always through a simple diffusion mechanism as presently thought, but instead can involve a regulated vesicle-mediated release process.


Asunto(s)
Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Vesículas Secretoras/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Difusión , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Glándulas Endocrinas/metabolismo , Exocitosis , Técnicas de Silenciamiento del Gen , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Larva/citología , Larva/metabolismo , Sinaptotagminas/metabolismo
3.
PLoS Biol ; 20(5): e3001660, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35594316

RESUMEN

In polarized epithelial cells, receptor-ligand interactions can be restricted by different spatial distributions of the 2 interacting components, giving rise to an underappreciated layer of regulatory complexity. We explored whether such regulation occurs in the Drosophila wing disc, an epithelial tissue featuring the TGF-ß family member Decapentaplegic (Dpp) as a morphogen controlling growth and patterning. Dpp protein has been observed in an extracellular gradient within the columnar cell layer of the disc, but also uniformly in the disc lumen, leading to the question of how graded signaling is achieved in the face of 2 distinctly localized ligand pools. We find the Dpp Type II receptor Punt, but not the Type I receptor Tkv, is enriched at the basolateral membrane and depleted at the junctions and apical surface. Wit, a second Type II receptor, shows a markedly different behavior, with the protein detected on all membrane regions but enriched at the apical side. Mutational studies identified a short juxtamembrane sequence required for basolateral restriction of Punt in both wing discs and mammalian Madin-Darby canine kidney (MDCK) cells. This basolateral targeting (BLT) determinant can dominantly confer basolateral localization on an otherwise apical receptor. Rescue of punt mutants with transgenes altered in the targeting motif showed that flies expressing apicalized Punt due to the lack of a functional BLT displayed developmental defects, female sterility, and significant lethality. We also show that apicalized Punt does not produce an ectopic signal, indicating that the apical pool of Dpp is not a significant signaling source even when presented with Punt. Instead, we find that basolateral presentation of Punt is required for optimal signaling. Finally, we present evidence that the BLT acts through polarized sorting machinery that differs between types of epithelia. This suggests a code whereby each epithelial cell type may differentially traffic common receptors to enable distinctive responses to spatially localized pools of extracellular ligands.


Asunto(s)
Proteínas de Drosophila , Drosophila , Factor de Crecimiento Transformador beta , Animales , Membrana Celular/metabolismo , Perros , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Ligandos , Células de Riñón Canino Madin Darby , Proteínas Serina-Treonina Quinasas , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
4.
Development ; 148(1)2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33234715

RESUMEN

The Myostatin/Activin branch of the TGF-ß superfamily acts as a negative regulator of vertebrate skeletal muscle size, in part, through downregulation of insulin/insulin-like growth factor 1 (IGF-1) signaling. Surprisingly, recent studies in Drosophila indicate that motoneuron-derived Activin signaling acts as a positive regulator of muscle size. Here we demonstrate that Drosophila Activin signaling promotes the growth of muscle cells along all three axes: width, thickness and length. Activin signaling positively regulates the insulin receptor (InR)/TORC1 pathway and the level of Myosin heavy chain (Mhc), an essential sarcomeric protein, via increased Pdk1 and Akt1 expression. Enhancing InR/TORC1 signaling in the muscle of Activin pathway mutants restores Mhc levels close to those of the wild type, but only increases muscle width. In contrast, hyperactivation of the Activin pathway in muscles increases overall larval body and muscle fiber length, even when Mhc levels are lowered by suppression of TORC1. Together, these results indicate that the Drosophila Activin pathway regulates larval muscle geometry and body size via promoting InR/TORC1-dependent Mhc production and the differential assembly of sarcomeric components into either pre-existing or new sarcomeric units depending on the balance of InR/TORC1 and Activin signals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Desarrollo de Músculos , Receptor de Insulina/metabolismo , Transducción de Señal , Animales , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Larva/metabolismo , Modelos Biológicos , Músculos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas/metabolismo , Sarcómeros/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
5.
Development ; 145(6)2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29467242

RESUMEN

Adult size and fitness are controlled by a combination of genetics and environmental cues. In Drosophila, growth is confined to the larval phase and final body size is impacted by the duration of this phase, which is under neuroendocrine control. The neuropeptide prothoracicotropic hormone (PTTH) has been proposed to play a central role in controlling the length of the larval phase through regulation of ecdysone production, a steroid hormone that initiates larval molting and metamorphosis. Here, we test this by examining the consequences of null mutations in the Ptth gene for Drosophila development. Loss of Ptth causes several developmental defects, including a delay in developmental timing, increase in critical weight, loss of coordination between body and imaginal disc growth, and reduced adult survival in suboptimal environmental conditions such as nutritional deprivation or high population density. These defects are caused by a decrease in ecdysone production associated with altered transcription of ecdysone biosynthetic genes. Therefore, the PTTH signal contributes to coordination between environmental cues and the developmental program to ensure individual fitness and survival.


Asunto(s)
Adaptación Fisiológica/genética , Plasticidad de la Célula/fisiología , Drosophila/crecimiento & desarrollo , Hormonas de Insectos/fisiología , Adaptación Fisiológica/fisiología , Animales , Plasticidad de la Célula/genética , Señales (Psicología) , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/metabolismo , Ecdisona/biosíntesis , Ambiente , Inmunohistoquímica , Hormonas de Insectos/genética , Larva/metabolismo , Larva/fisiología , Metamorfosis Biológica/fisiología , Mutagénesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
6.
J Chem Phys ; 155(20): 204103, 2021 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-34852495

RESUMEN

We present OrbNet Denali, a machine learning model for an electronic structure that is designed as a drop-in replacement for ground-state density functional theory (DFT) energy calculations. The model is a message-passing graph neural network that uses symmetry-adapted atomic orbital features from a low-cost quantum calculation to predict the energy of a molecule. OrbNet Denali is trained on a vast dataset of 2.3 × 106 DFT calculations on molecules and geometries. This dataset covers the most common elements in biochemistry and organic chemistry (H, Li, B, C, N, O, F, Na, Mg, Si, P, S, Cl, K, Ca, Br, and I) and charged molecules. OrbNet Denali is demonstrated on several well-established benchmark datasets, and we find that it provides accuracy that is on par with modern DFT methods while offering a speedup of up to three orders of magnitude. For the GMTKN55 benchmark set, OrbNet Denali achieves WTMAD-1 and WTMAD-2 scores of 7.19 and 9.84, on par with modern DFT functionals. For several GMTKN55 subsets, which contain chemical problems that are not present in the training set, OrbNet Denali produces a mean absolute error comparable to those of DFT methods. For the Hutchison conformer benchmark set, OrbNet Denali has a median correlation coefficient of R2 = 0.90 compared to the reference DLPNO-CCSD(T) calculation and R2 = 0.97 compared to the method used to generate the training data (ωB97X-D3/def2-TZVP), exceeding the performance of any other method with a similar cost. Similarly, the model reaches chemical accuracy for non-covalent interactions in the S66x10 dataset. For torsional profiles, OrbNet Denali reproduces the torsion profiles of ωB97X-D3/def2-TZVP with an average mean absolute error of 0.12 kcal/mol for the potential energy surfaces of the diverse fragments in the TorsionNet500 dataset.

7.
J Chem Phys ; 153(15): 154105, 2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33092381

RESUMEN

The ability to understand and engineer molecular structures relies on having accurate descriptions of the energy as a function of atomic coordinates. Here, we outline a new paradigm for deriving energy functions of hyperdimensional molecular systems, which involves generating data for low-dimensional systems in virtual reality (VR) to then efficiently train atomic neural networks (ANNs). This generates high-quality data for specific areas of interest within the hyperdimensional space that characterizes a molecule's potential energy surface (PES). We demonstrate the utility of this approach by gathering data within VR to train ANNs on chemical reactions involving fewer than eight heavy atoms. This strategy enables us to predict the energies of much higher-dimensional systems, e.g., containing nearly 100 atoms. Training on datasets containing only 15k geometries, this approach generates mean absolute errors around 2 kcal mol-1. This represents one of the first times that an ANN-PES for a large reactive radical has been generated using such a small dataset. Our results suggest that VR enables the intelligent curation of high-quality data, which accelerates the learning process.

8.
J Chem Phys ; 150(22): 220901, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31202243

RESUMEN

As molecular scientists have made progress in their ability to engineer nanoscale molecular structure, we face new challenges in our ability to engineer molecular dynamics (MD) and flexibility. Dynamics at the molecular scale differs from the familiar mechanics of everyday objects because it involves a complicated, highly correlated, and three-dimensional many-body dynamical choreography which is often nonintuitive even for highly trained researchers. We recently described how interactive molecular dynamics in virtual reality (iMD-VR) can help to meet this challenge, enabling researchers to manipulate real-time MD simulations of flexible structures in 3D. In this article, we outline various efforts to extend immersive technologies to the molecular sciences, and we introduce "Narupa," a flexible, open-source, multiperson iMD-VR software framework which enables groups of researchers to simultaneously cohabit real-time simulation environments to interactively visualize and manipulate the dynamics of molecular structures with atomic-level precision. We outline several application domains where iMD-VR is facilitating research, communication, and creative approaches within the molecular sciences, including training machines to learn potential energy functions, biomolecular conformational sampling, protein-ligand binding, reaction discovery using "on-the-fly" quantum chemistry, and transport dynamics in materials. We touch on iMD-VR's various cognitive and perceptual affordances and outline how these provide research insight for molecular systems. By synergistically combining human spatial reasoning and design insight with computational automation, technologies such as iMD-VR have the potential to improve our ability to understand, engineer, and communicate microscopic dynamical behavior, offering the potential to usher in a new paradigm for engineering molecules and nano-architectures.


Asunto(s)
Simulación de Dinámica Molecular , Programas Informáticos , Realidad Virtual , Benzamidinas/metabolismo , Ciclofilina A/química , Humanos , Subtipo H7N9 del Virus de la Influenza A/enzimología , Relaciones Interpersonales , Ligandos , Redes Neurales de la Computación , Neuraminidasa/metabolismo , Compuestos Orgánicos/química , Oseltamivir/metabolismo , Unión Proteica , Conformación Proteica , Teoría Cuántica , Tripsina/metabolismo
9.
Genes Dev ; 25(14): 1459-63, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21764850

RESUMEN

Nitric oxide (NO) is an important second messenger involved in numerous biological processes, but how it regulates gene expression is not well understood. In this issue of Genes & Development, Cáceres and colleagues (pp. 1476-1485) report a critical requirement of NO as a direct regulator of gene expression through its binding to a heme-containing nuclear receptor in Drosophila. This may be an anciently evolved mechanism to coordinate behavior and metabolism during animal development.


Asunto(s)
Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Metamorfosis Biológica/fisiología , Óxido Nítrico/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster/genética , Ecdisteroides/biosíntesis , Proteínas de Insectos/metabolismo , Metamorfosis Biológica/genética , Unión Proteica , Receptores de Esteroides/metabolismo
10.
Dev Biol ; 430(1): 166-176, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782527

RESUMEN

Steroid hormones regulate life stage transitions, allowing animals to appropriately follow a developmental timeline. During insect development, the steroid hormone ecdysone is synthesized and released in a regulated manner by the prothoracic gland (PG) and then hydroxylated to the active molting hormone, 20-hydroxyecdysone (20E), in peripheral tissues. We manipulated ecdysteroid titers, through temporally controlled over-expression of the ecdysteroid-inactivating enzyme, CYP18A1, in the PG using the GeneSwitch-GAL4 system in the fruit fly Drosophila melanogaster. We monitored expression of a 20E-inducible glue protein gene, Salivary gland secretion 3 (Sgs3), using a Sgs3:GFP fusion transgene. In wild type larvae, Sgs3-GFP expression is activated at the midpoint of the third larval instar stage in response to the rising endogenous level of 20E. By first knocking down endogenous 20E levels during larval development and then feeding 20E to these larvae at various stages, we found that Sgs3-GFP expression could be triggered at an inappropriate developmental stage after a certain time lag. This stage-precocious activation of Sgs3 required expression of the Broad-complex, similar to normal Sgs3 developmental regulation, and a small level of nutritional input. We suggest that these studies provide evidence for a tissue-autonomic regulatory system for a metamorphic event independent from the primary 20E driven developmental progression.


Asunto(s)
Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Ecdisterona/metabolismo , Proteínas del Pegamento Salivar de Drosophila/metabolismo , Estructuras Animales/efectos de los fármacos , Estructuras Animales/metabolismo , Animales , Proteínas Fluorescentes Verdes/metabolismo , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Mifepristona/farmacología , Modelos Biológicos , Progesterona/análogos & derivados , Transducción de Señal , Factores de Tiempo , Transgenes
11.
PLoS Genet ; 11(12): e1005712, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26658797

RESUMEN

Steroid hormones are crucial for many biological events in multicellular organisms. In insects, the principal steroid hormones are ecdysteroids, which play essential roles in regulating molting and metamorphosis. During larval and pupal development, ecdysteroids are synthesized in the prothoracic gland (PG) from dietary cholesterol via a series of hydroxylation and oxidation steps. The expression of all but one of the known ecdysteroid biosynthetic enzymes is restricted to the PG, but the transcriptional regulatory networks responsible for generating such exquisite tissue-specific regulation is only beginning to be elucidated. Here, we report identification and characterization of the C2H2-type zinc finger transcription factor Ouija board (Ouib) necessary for ecdysteroid production in the PG in the fruit fly Drosophila melanogaster. Expression of ouib is predominantly limited to the PG, and genetic null mutants of ouib result in larval developmental arrest that can be rescued by administrating an active ecdysteroid. Interestingly, ouib mutant animals exhibit a strong reduction in the expression of one ecdysteroid biosynthetic enzyme, spookier. Using a cell culture-based luciferase reporter assay, Ouib protein stimulates transcription of spok by binding to a specific ~15 bp response element in the spok PG enhancer element. Most remarkable, the developmental arrest phenotype of ouib mutants is rescued by over-expression of a functionally-equivalent paralog of spookier. These observations imply that the main biological function of Ouib is to specifically regulate spookier transcription during Drosophila development.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ecdisteroides/genética , Factores de Transcripción/genética , Dedos de Zinc/genética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Ecdisteroides/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Larva/genética , Larva/crecimiento & desarrollo , Metamorfosis Biológica , Fenotipo , Unión Proteica
12.
Proc Natl Acad Sci U S A ; 111(15): 5729-34, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706779

RESUMEN

The ability to maintain cellular and physiological metabolic homeostasis is key for the survival of multicellular organisms in changing environmental conditions. However, our understanding of extracellular signaling pathways that modulate metabolic processes remains limited. In this study we show that the Activin-like ligand Dawdle (Daw) is a major regulator of systemic metabolic homeostasis and cellular metabolism in Drosophila. We find that loss of canonical Smad signaling downstream of Daw leads to defects in sugar and systemic pH homeostasis. Although Daw regulates sugar homeostasis by positively influencing insulin release, we find that the effect of Daw on pH balance is independent of its role in insulin signaling and is caused by accumulation of organic acids that are primarily tricarboxylic acid (TCA) cycle intermediates. RNA sequencing reveals that a number of TCA cycle enzymes and nuclear-encoded mitochondrial genes including genes involved in oxidative phosphorylation and ß-oxidation are up-regulated in the daw mutants, indicating either a direct or indirect role of Daw in regulating these genes. These findings establish Activin signaling as a major metabolic regulator and uncover a functional link between TGF-ß signaling, insulin signaling, and metabolism in Drosophila.


Asunto(s)
Activinas/metabolismo , Metabolismo de los Hidratos de Carbono/fisiología , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/fisiología , Regulación de la Expresión Génica/fisiología , Homeostasis/fisiología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Western Blotting , Proteínas Portadoras , Drosophila melanogaster/metabolismo , Fluorescencia , Regulación de la Expresión Génica/genética , Concentración de Iones de Hidrógeno , Larva/crecimiento & desarrollo , Larva/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética
13.
PLoS Genet ; 10(6): e1004343, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945799

RESUMEN

Specialized endocrine cells produce and release steroid hormones that govern development, metabolism and reproduction. In order to synthesize steroids, all the genes in the biosynthetic pathway must be coordinately turned on in steroidogenic cells. In Drosophila, the steroid producing endocrine cells are located in the prothoracic gland (PG) that releases the steroid hormone ecdysone. The transcriptional regulatory network that specifies the unique PG specific expression pattern of the ecdysone biosynthetic genes remains unknown. Here, we show that two transcription factors, the POU-domain Ventral veins lacking (Vvl) and the nuclear receptor Knirps (Kni), have essential roles in the PG during larval development. Vvl is highly expressed in the PG during embryogenesis and is enriched in the gland during larval development, suggesting that Vvl might function as a master transcriptional regulator in this tissue. Vvl and Kni bind to PG specific cis-regulatory elements that are required for expression of the ecdysone biosynthetic genes. Knock down of either vvl or kni in the PG results in a larval developmental arrest due to failure in ecdysone production. Furthermore, Vvl and Kni are also required for maintenance of TOR/S6K and prothoracicotropic hormone (PTTH) signaling in the PG, two major pathways that control ecdysone biosynthesis and PG cell growth. We also show that the transcriptional regulator, Molting defective (Mld), controls early biosynthetic pathway steps. Our data show that Vvl and Kni directly regulate ecdysone biosynthesis by transcriptional control of biosynthetic gene expression and indirectly by affecting PTTH and TOR/S6K signaling. This provides new insight into the regulatory network of transcription factors involved in the coordinated regulation of steroidogenic cell specific transcription, and identifies a new function of Vvl and Knirps in endocrine cells during post-embryonic development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Ecdisona/biosíntesis , Hormonas de Insectos/biosíntesis , Proteínas Nucleares/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Represoras/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/biosíntesis , Animales , Sitios de Unión , Transporte Biológico/genética , Colesterol/metabolismo , Proteínas de Unión al ADN , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Ecdisona/genética , Ecdisona/metabolismo , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hormonas de Insectos/metabolismo , Proteínas de la Membrana/biosíntesis , Factores del Dominio POU/biosíntesis , Factores del Dominio POU/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Serina-Treonina Quinasas TOR/biosíntesis , Transcripción Genética
14.
Development ; 140(3): 649-59, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293296

RESUMEN

Imaginal disc development in Drosophila requires coordinated cellular proliferation and tissue patterning. In our studies of TGFß superfamily signaling components, we found that a protein null mutation of Smad2, the only Activin subfamily R-Smad in the fruit fly, produces overgrown wing discs that resemble gain of function for BMP subfamily signaling. The wing discs are expanded specifically along the anterior-posterior axis, with increased proliferation in lateral regions. The morphological defect is not observed in mutants for the TGFß receptor baboon, and epistasis tests showed that baboon is epistatic to Smad2 for disc overgrowth. Rescue experiments indicate that Baboon binding, but not canonical transcription factor activity, of Smad2 is required for normal disc growth. Smad2 mutant discs generate a P-Mad stripe that is narrower and sharper than the normal gradient, and activation targets are correspondingly expressed in narrowed domains. Repression targets of P-Mad are profoundly mis-regulated, with brinker and pentagone reporter expression eliminated in Smad2 mutants. Loss of expression requires a silencer element previously shown to be controlled by BMP signaling. Epistasis experiments show that Baboon, Mad and Schnurri are required to mediate the ectopic silencer output in the absence of Smad2. Taken together, our results show that loss of Smad2 permits promiscuous Baboon activity, which represses genes subject to control by Mad-dependent silencer elements. The absence of Brinker and Pentagone in Smad2 mutants explains the compound wing disc phenotype. Our results highlight the physiological relevance of substrate inhibition of a kinase, and reveal a novel interplay between the Activin and BMP pathways.


Asunto(s)
Receptores de Activinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Discos Imaginales/fisiología , Proteína Smad2/metabolismo , Alas de Animales/fisiología , Receptores de Activinas/genética , Alelos , Animales , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Tamaño de la Célula , Cruzamientos Genéticos , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Epistasis Genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Discos Imaginales/metabolismo , Inmunohistoquímica , Masculino , Mutagénesis Sitio-Dirigida , Mutación , Tamaño de los Órganos , Fenotipo , Interferencia de ARN , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Smad Reguladas por Receptores , Proteína Smad2/genética , Factores de Tiempo , Alas de Animales/metabolismo
15.
Development ; 140(23): 4730-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24173800

RESUMEN

Steroid hormones trigger the onset of sexual maturation in animals by initiating genetic response programs that are determined by steroid pulse frequency, amplitude and duration. Although steroid pulses coordinate growth and timing of maturation during development, the mechanisms generating these pulses are not known. Here we show that the ecdysone steroid pulse that drives the juvenile-adult transition in Drosophila is determined by feedback circuits in the prothoracic gland (PG), the major steroid-producing tissue of insect larvae. These circuits coordinate the activation and repression of hormone synthesis, the two key parameters determining pulse shape (amplitude and duration). We show that ecdysone has a positive-feedback effect on the PG, rapidly amplifying its own synthesis to trigger pupariation as the onset of maturation. During the prepupal stage, a negative-feedback signal ensures the decline in ecdysone levels required to produce a temporal steroid pulse that drives developmental progression to adulthood. The feedback circuits rely on a developmental switch in the expression of Broad isoforms that transcriptionally activate or silence components in the ecdysone biosynthetic pathway. Remarkably, our study shows that the same well-defined genetic program that stimulates a systemic downstream response to ecdysone is also utilized upstream to set the duration and amplitude of the ecdysone pulse. Activation of this switch-like mechanism ensures a rapid, self-limiting PG response that functions in producing steroid oscillations that can guide the decision to terminate growth and promote maturation.


Asunto(s)
Drosophila melanogaster/embriología , Ecdisona/metabolismo , Hormonas de Insectos/metabolismo , Maduración Sexual/genética , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Ecdisona/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Larva/crecimiento & desarrollo , Larva/metabolismo , Metamorfosis Biológica/genética , Regiones Promotoras Genéticas , Transducción de Señal
16.
Methods ; 68(1): 183-93, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24680699

RESUMEN

The TGF-ß pathway is an evolutionarily conserved signal transduction module that mediates diverse biological processes in animals. In Drosophila, both the BMP and Activin branches are required for viability. Studies rooted in classical and molecular genetic approaches continue to uncover new developmental roles for TGF-ß signaling. We present an overview of the secreted ligands, transmembrane receptors and cellular Smad transducer proteins that compose the core pathway in Drosophila. An assortment of tools have been developed to conduct tissue-specific loss- and gain-of-function experiments for these pathway components. We discuss the deployment of these reagents, with an emphasis on appropriate usage and limitations of the available tools. Throughout, we note reagents that are in need of further improvement or development, and signaling features requiring further study. A general theme is that comparison of phenotypes for ligands, receptors, and Smads can be used to map tissue interactions, and to separate canonical and non-canonical signaling activities. Core TGF-ß signaling components are subject to multiple layers of regulation, and are coupled to context-specific inputs and outputs. In addition to fleshing out how TGF-ß signaling serves the fruit fly, we anticipate that future studies will uncover new regulatory nodes and modes and will continue to advance paradigms for how TGF-ß signaling regulates general developmental processes.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Activinas/genética , Activinas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Biología Evolutiva/métodos , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Fosforilación , Factor de Crecimiento Transformador beta/metabolismo
17.
Development ; 138(13): 2693-703, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21613324

RESUMEN

In insects, initiation of metamorphosis requires a surge in the production of the steroid hormone 20-hydroxyecdysone from the prothoracic gland, the primary endocrine organ of juvenile larvae. Here, we show that blocking TGFß/Activin signaling, specifically in the Drosophila prothoracic gland, results in developmental arrest prior to metamorphosis. The terminal, giant third instar larval phenotype results from a failure to induce the large rise in ecdysteroid titer that triggers metamorphosis. We further demonstrate that activin signaling regulates competence of the prothoracic gland to receive PTTH and insulin signals, and that these two pathways act at the mRNA and post-transcriptional levels, respectively, to control ecdysone biosynthetic enzyme expression. This dual regulatory circuitry may provide a cross-check mechanism to ensure that both developmental and nutritional inputs are synchronized before initiating the final genetic program leading to reproductive adult development. As steroid hormone production in C. elegans and mammals is also influenced by TGFß/Activin signaling, this family of secreted factors may play a general role in regulating developmental transitions across phyla.


Asunto(s)
Activinas/metabolismo , Sistemas Neurosecretores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Drosophila , Ecdisteroides/metabolismo , Glándulas Endocrinas/metabolismo , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Hormonas de Insectos/metabolismo , Metamorfosis Biológica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
Annu Rev Entomol ; 58: 497-516, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23072462

RESUMEN

The steroid hormone ecdysone is the central regulator of insect developmental transitions. Recent new advances in our understanding of ecdysone action have relied heavily on the application of Drosophila melanogaster molecular genetic tools to study insect metamorphosis. In this review, we focus on three major aspects of Drosophila ecdysone biology: (a) factors that regulate the timing of ecdysone release, (b) molecular basis of stage- and tissue-specific responses to ecdysone, and (c) feedback regulation and coordination of ecdysone signaling.


Asunto(s)
Drosophila melanogaster/fisiología , Ecdisona/metabolismo , Transducción de Señal , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Metamorfosis Biológica
19.
PLoS One ; 19(6): e0305696, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38913612

RESUMEN

In Drosophila coordinated proliferation of two neural stem cells, neuroblasts (NB) and neuroepithelial (NE) cells, is pivotal for proper larval brain growth that ultimately determines the final size and performance of an adult brain. The larval brain growth displays two phases based on behaviors of NB and NEs: the first one in early larval stages, influenced by nutritional status and the second one in the last larval stage, promoted by ecdysone signaling after critical weight checkpoint. Mutations of the baboon (babo) gene that produces three isoforms (BaboA-C), all acting as type-I receptors of Activin-type transforming growth factor ß (TGF-ß) signaling, cause a small brain phenotype due to severely reduced proliferation of the neural stem cells. In this study we show that loss of babo function severely affects proliferation of NBs and NEs as well as conversion of NEs from both phases. By analyzing babo-null and newly generated isoform-specific mutants by CRISPR mutagenesis as well as isoform-specific RNAi knockdowns in a cell- and stage-specific manner, our data support differential contributions of the isoforms for these cellular events with BaboA playing the major role. Stage-specific expression of EcR-B1 in the brain is also regulated primarily by BaboA along with function of the other isoforms. Blocking EcR function in both neural stem cells results in a small brain phenotype that is more severe than baboA-knockdown alone. In summary, our study proposes that the Babo-mediated signaling promotes proper behaviors of the neural stem cells in both phases and achieves this by acting upstream of EcR-B1 expression in the second phase.


Asunto(s)
Encéfalo , Proliferación Celular , Proteínas de Drosophila , Larva , Células-Madre Neurales , Células Neuroepiteliales , Isoformas de Proteínas , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Larva/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Encéfalo/metabolismo , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Transducción de Señal , Receptores de Activinas/metabolismo , Receptores de Activinas/genética
20.
Genetics ; 226(2)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-37981656

RESUMEN

The fourth chromosome is the final frontier for genetic analysis in Drosophila. Small, heterochromatic, and devoid of recombination the fourth has long been ignored. Nevertheless, its long arm contains 79 protein-coding genes. The Fourth Chromosome Resource Project (FCRP) has a goal of facilitating the investigation of genes on this neglected chromosome. The project has 446 stocks publicly available at the Bloomington and Kyoto stock centers with phenotypic data curated by the FlyBase and FlyPush resources. Four of the five stock sets are nearly complete: (1) UAS.fly cDNAs, (2) UAS.human homolog cDNAs, (3) gene trap mutants and protein traps, and (4) stocks promoting meiotic and mitotic recombination on the fourth. Ongoing is mutagenesis of each fourth gene on a new FRT-bearing chromosome for marked single-cell clones. Beyond flies, FCRP facilitates the creation and analysis of humanized fly stocks. These provide opportunities to apply Drosophila genetics to the analysis of human gene interaction and function. In addition, the FCRP provides investigators with confidence through stock validation and an incentive via phenotyping to tackle genes on the fourth that have never been studied. Taken together, FCRP stocks will facilitate all manner of genetic and molecular studies. The resource is readily available to researchers to enhance our understanding of metazoan biology, including conserved molecular mechanisms underlying health and disease.


Asunto(s)
Cromosomas , Drosophila , Animales , Humanos , Drosophila/genética , Mutagénesis , Drosophila melanogaster/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA