Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 137(7): 1331-42, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19563762

RESUMEN

Dorsal closure is a tissue-modeling process in the developing Drosophila embryo during which an epidermal opening is closed. It begins with the appearance of a supracellular actin cable that surrounds the opening and provides a contractile force. Amnioserosa cells that fill the opening produce an additional critical force pulling on the surrounding epidermal tissue. We show that this force is not gradual but pulsed and occurs long before dorsal closure starts. Quantitative analysis, combined with laser cutting experiments and simulations, reveals that tension-based dynamics and cell coupling control the force pulses. These constitutively pull the surrounding epidermal tissue dorsally, but the displacement is initially transient. It is translated into dorsal-ward movement only with the help of the actin cable, which acts like a ratchet, counteracting ventral-ward epidermis relaxation after force pulses. Our work uncovers a sophisticated mechanism of cooperative force generation between two major forces driving morphogenesis.


Asunto(s)
Drosophila melanogaster/embriología , Actinas/metabolismo , Animales , Fenómenos Biomecánicos , Drosophila melanogaster/citología , Ectodermo/citología , Ectodermo/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Morfogénesis
2.
PLoS Biol ; 16(4): e2004718, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29702642

RESUMEN

Sarcomeres are stereotyped force-producing mini-machines of striated muscles. Each sarcomere contains a pseudocrystalline order of bipolar actin and myosin filaments, which are linked by titin filaments. During muscle development, these three filament types need to assemble into long periodic chains of sarcomeres called myofibrils. Initially, myofibrils contain immature sarcomeres, which gradually mature into their pseudocrystalline order. Despite the general importance, our understanding of myofibril assembly and sarcomere maturation in vivo is limited, in large part because determining the molecular order of protein components during muscle development remains challenging. Here, we applied polarization-resolved microscopy to determine the molecular order of actin during myofibrillogenesis in vivo. This method revealed that, concomitantly with mechanical tension buildup in the myotube, molecular actin order increases, preceding the formation of immature sarcomeres. Mechanistically, both muscle and nonmuscle myosin contribute to this actin order gain during early stages of myofibril assembly. Actin order continues to increase while myofibrils and sarcomeres mature. Muscle myosin motor activity is required for the regular and coordinated assembly of long myofibrils but not for the high actin order buildup during sarcomere maturation. This suggests that, in muscle, other actin-binding proteins are sufficient to locally bundle or cross-link actin into highly regular arrays.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/metabolismo , Drosophila melanogaster/ultraestructura , Miofibrillas/ultraestructura , Pupa/ultraestructura , Sarcómeros/ultraestructura , Citoesqueleto de Actina/metabolismo , Actinas/ultraestructura , Animales , Fenómenos Biomecánicos , Conectina/metabolismo , Conectina/ultraestructura , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Vuelo Animal/fisiología , Microscopía de Polarización/métodos , Miofibrillas/metabolismo , Miosinas/metabolismo , Miosinas/ultraestructura , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Sarcómeros/metabolismo
3.
Elife ; 102021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33404503

RESUMEN

Skeletal muscles are composed of gigantic cells called muscle fibers, packed with force-producing myofibrils. During development, the size of individual muscle fibers must dramatically enlarge to match with skeletal growth. How muscle growth is coordinated with growth of the contractile apparatus is not understood. Here, we use the large Drosophila flight muscles to mechanistically decipher how muscle fiber growth is controlled. We find that regulated activity of core members of the Hippo pathway is required to support flight muscle growth. Interestingly, we identify Dlg5 and Slmap as regulators of the STRIPAK phosphatase, which negatively regulates Hippo to enable post-mitotic muscle growth. Mechanistically, we show that the Hippo pathway controls timing and levels of sarcomeric gene expression during development and thus regulates the key components that physically mediate muscle growth. Since Dlg5, STRIPAK and the Hippo pathway are conserved a similar mechanism may contribute to muscle or cardiomyocyte growth in humans.


Asunto(s)
Drosophila melanogaster/fisiología , Regulación de la Expresión Génica , Vía de Señalización Hippo/fisiología , Fibras Musculares Esqueléticas/fisiología , Miofibrillas/metabolismo , Sarcómeros/genética , Animales , Drosophila melanogaster/genética
4.
Methods Mol Biol ; 1889: 331-348, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30367424

RESUMEN

RNA interference (RNAi) is the method of choice to systematically test for gene function in an intact organism. The model organism Drosophila has the advantage that RNAi is cell autonomous, meaning it does not spread from one cell to the next. Hence, RNAi can be performed in a tissue-specific manner by expressing short or long inverted repeat constructs (hairpins) designed to target mRNAs from one specific target gene. This achieves tissue-specific knock-down of a target gene of choice. Here, we detail the methodology to test gene function in Drosophila muscle tissue by expressing hairpins in a muscle-specific manner using the GAL4-UAS system. We further discuss the systematic RNAi resource collections available which also permit large scale screens in a muscle-specific manner. The full power of such screens is revealed by combination of high-throughput assays followed by detailed morphological assays. Together, this chapter should be a practical guide to enable the reader to either test a few candidate genes, or large gene sets for particular functions in Drosophila muscle tissue and provide first insights into the biological process the gene might be important for in muscle.


Asunto(s)
Drosophila/embriología , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Desarrollo de Músculos/genética , Interferencia de ARN , Animales , Técnicas de Inactivación de Genes , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento , ARN Mensajero , ARN Interferente Pequeño
5.
Methods Mol Biol ; 1478: 117-143, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27730578

RESUMEN

RNAi technologies enable the testing of gene function in a cell-type- and stage-specific manner in Drosophila. The development of genome-wide RNAi libraries has allowed expansion of this approach to the genome scale and supports identification of most genes required for a given process in a cell type of choice. However, a large-scale RNAi approach also harbors many potential pitfalls that can complicate interpretation of the results. Here, we summarize published screens and provide a guide on how to optimally plan and perform a large-scale, in vivo RNAi screen. We highlight the importance of assay design and give suggestions on how to optimize the assay conditions by testing positive and negative control genes. These genes are used to estimate false-negative and false-positive rates of the screen data. We discuss the planning and logistics of a large-scale screen in detail and suggest bioinformatics platforms to identify and select gene groups of interest for secondary assays. Finally, we review various options to confirm RNAi knock-down specificity and thus identify high confidence genes for more detailed case-by-case studies in the future.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Genoma , Ensayos Analíticos de Alto Rendimiento , ARN Helicasas/genética , Interferencia de ARN , Ribonucleasa III/genética , Factores de Transcripción/genética , Animales , Biología Computacional/métodos , Bases de Datos Genéticas , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica , Técnicas Genéticas , Biblioteca Genómica , ARN Helicasas/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Curr Biol ; 24(7): 705-16, 2014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24631244

RESUMEN

BACKGROUND: Higher animals generate an elaborate muscle-tendon network to perform their movements. To build a functional network, developing muscles must establish stable connections with tendons and assemble their contractile apparatuses. Current myofibril assembly models do not consider the impact of muscle-tendon attachment on myofibrillogenesis. However, if attachment and myofibrillogenesis are not properly coordinated, premature muscle contractions can destroy an unstable myotendinous system, leading to severe myopathies. RESULTS: Here, we use Drosophila indirect flight muscles to investigate how muscle-tendon attachment and myofibrillogenesis are coordinated. We find that flight muscles first stably attach to tendons and then assemble their myofibrils. Interestingly, this myofibril assembly is triggered simultaneously throughout the entire muscle, suggesting a self-assembly mechanism. By applying laser-cutting experiments, we show that muscle attachment coincides with an increase in mechanical tension before periodic myofibrils can be detected. We manipulated tension buildup within the myotendinous system either by genetically compromising attachment initiation and integrin recruitment to the myotendinous junction or by optically severing tendons from muscle. Both treatments cause strong myofibrillogenesis defects. We find that myosin motor activity is required for both tension formation and myofibril assembly, suggesting that myofibril assembly itself contributes to tension buildup. CONCLUSIONS: Our results demonstrate that force-resistant attachment enables a stark tension increase in the myotendinous system. Subsequently, this tension increase triggers simultaneous myofibril self-assembly throughout the entire muscle fiber. As myofibril and sarcomeric architecture as well as their molecular components are evolutionarily conserved, we propose a similar tension-based mechanism to regulate myofibrillogenesis in vertebrates.


Asunto(s)
Drosophila/anatomía & histología , Vuelo Animal/fisiología , Desarrollo de Músculos , Animales , Fenómenos Biomecánicos , Drosophila/fisiología , Músculos/anatomía & histología , Músculos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA