Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(3)2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35159130

RESUMO

Muscle fibers are multinucleated cells that arise during embryogenesis through the fusion of mononucleated myoblasts. Myoblast fusion is a lifelong process that is crucial for the growth and regeneration of muscles. Understanding the molecular mechanism of myoblast fusion may open the way for novel therapies in muscle wasting and weakness. Recent reports in Drosophila and mammals have provided new mechanistic insights into myoblast fusion. In Drosophila, muscle formation occurs twice: during embryogenesis and metamorphosis. A fundamental feature is the formation of a cell-cell communication structure that brings the apposing membranes into close proximity and recruits possible fusogenic proteins. However, genetic studies suggest that myoblast fusion in Drosophila is not a uniform process. The complexity of the players involved in myoblast fusion can be modulated depending on the type of muscle that is formed. In this review, we introduce the different types of multinucleated muscles that form during Drosophila development and provide an overview in advances that have been made to understand the mechanism of myoblast fusion. Finally, we will discuss conceptual frameworks in cell-cell fusion in Drosophila and mammals.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Animais , Comunicação Celular , Fusão Celular , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mamíferos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo
2.
J Cell Sci ; 129(18): 3426-36, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27521427

RESUMO

The fusion of founder cells and fusion-competent myoblasts (FCMs) is crucial for muscle formation in Drosophila Characteristic events of myoblast fusion include the recognition and adhesion of myoblasts, and the formation of branched F-actin by the Arp2/3 complex at the site of cell-cell contact. At the ultrastructural level, these events are reflected by the appearance of finger-like protrusions and electron-dense plaques that appear prior to fusion. Severe defects in myoblast fusion are caused by the loss of Kette (a homolog of Nap1 and Hem-2, also known as NCKAP1 and NCKAP1L, respectively), a member of the regulatory complex formed by Scar or WAVE proteins (represented by the single protein, Scar, in flies). kette mutants form finger-like protrusions, but the electron-dense plaques are extended. Here, we show that the electron-dense plaques in wild-type and kette mutant myoblasts resemble other electron-dense structures that are known to function as cellular junctions. Furthermore, analysis of double mutants and attempts to rescue the kette mutant phenotype with N-cadherin, wasp and genes of members of the regulatory Scar complex revealed that Kette has two functions during myoblast fusion. First, Kette controls the dissolution of electron-dense plaques. Second, Kette controls the ratio of the Arp2/3 activators Scar and WASp in FCMs.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Junções Intercelulares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Animais , Caderinas/metabolismo , Fusão Celular , Modelos Biológicos , Mutação/genética , Mioblastos/ultraestrutura , Fenótipo , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
J Cell Sci ; 129(3): 604-20, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26675239

RESUMO

Wiskott-Aldrich syndrome proteins (WASPs) are nucleation-promoting factors (NPF) that differentially control the Arp2/3 complex. In Drosophila, three different family members, SCAR (also known as WAVE), WASP and WASH (also known as CG13176), have been analyzed so far. Here, we characterized WHAMY, the fourth Drosophila WASP family member. whamy originated from a wasp gene duplication and underwent a sub-neofunctionalization. Unlike WASP, we found that WHAMY specifically interacted with activated Rac1 through its two CRIB domains, which were sufficient for targeting WHAMY to lamellipodial and filopodial tips. Biochemical analyses showed that WHAMY promoted exceptionally fast actin filament elongation, although it did not activate the Arp2/3 complex. Loss- and gain-of-function studies revealed an important function of WHAMY in membrane protrusions and cell migration in macrophages. Genetic data further implied synergistic functions between WHAMY and WASP during morphogenesis. Double mutants were late-embryonic lethal and showed severe defects in myoblast fusion. Trans-heterozygous mutant animals showed strongly increased defects in sensory cell fate specification. Thus, WHAMY is a novel actin polymerase with an initial partitioning of ancestral WASP functions in development and subsequent acquisition of a new function in cell motility during evolution.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Macrófagos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/metabolismo , Organogênese/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Drosophila/fisiologia , Morfogênese/fisiologia , Desenvolvimento Muscular/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
4.
J Neurogenet ; 28(3-4): 302-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24957080

RESUMO

Membrane fusion is essential for the communication of membrane-defined compartments, development of multicellular organisms and tissue homeostasis. Although membrane fusion has been studied extensively, still little is known about the molecular mechanisms. Especially the intercellular fusion of cells during development and tissue homeostasis is poorly understood. Somatic muscle formation in Drosophila depends on the intercellular fusion of myoblasts. In this process, myoblasts recognize each other and adhere, thereby triggering a protein machinery that leads to electron-dense plaques, vesicles and F-actin formation at apposing membranes. Two models of how local membrane stress is achieved to induce the merging of the myoblast membranes have been proposed: the electron-dense vesicles transport and release a fusogen and F-actin bends the plasma membrane. In this review, we highlight cell-adhesion molecules and intracellular proteins known to be involved in myoblast fusion. The cell-adhesion proteins also mediate the recognition and adhesion of other cell types, such as neurons that communicate with each other via special intercellular junctions, termed chemical synapses. At these synapses, neurotransmitters are released through the intracellular fusion of synaptic vesicles with the plasma membrane. As the targeting of electron-dense vesicles in myoblasts shares some similarities with the targeting of synaptic vesicle fusion, we compare molecules required for synaptic vesicle fusion to recently identified molecules involved in myoblast fusion.


Assuntos
Membrana Celular/metabolismo , Mioblastos/metabolismo , Sinapses/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Drosophila/metabolismo , Exocitose/fisiologia , Vesículas Sinápticas/metabolismo
5.
Dev Genes Evol ; 223(3): 159-69, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23111653

RESUMO

Fibroblast growth factor receptors (FGFR) are highly conserved receptor tyrosine kinases, and evolved early in metazoan evolution. In order to investigate their functional conservation, we asked whether the Kringelchen FGFR in the freshwater polyp Hydra vulgaris, is able to functionally replace FGFR in fly embryos. In Drosophila, two endogenous FGFR, Breathless (Btl) and Heartless (Htl), ensure formation of the tracheal system and mesodermal cell migration as well as formation of the heart. Using UAS-kringelchen-5xmyc transgenic flies and targeted expression, we show that Kringelchen is integrated correctly into the cell membrane of mesodermal and tracheal cells in Drosophila. Nevertheless, Kringelchen expression driven in tracheal cells failed to rescue the btl (LG19) mutant. The Hydra FGFR was able to substitute for Heartless in the htl (AB42) null mutant; however, this occurred only during early mesodermal cell migration. Our data provide evidence for functional conservation of this early-diverged FGFR across these distantly related phyla, but also selectivity for the Htl FGFR in the Drosophila system.


Assuntos
Drosophila/genética , Hydra/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Evolução Molecular , Dados de Sequência Molecular , Mutação , Filogenia , Receptores de Fatores de Crescimento de Fibroblastos/química , Homologia de Sequência de Aminoácidos
6.
J Cell Sci ; 126(Pt 1): 360-72, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22992459

RESUMO

The formation of the larval body wall musculature of Drosophila depends on the asymmetric fusion of two myoblast types, founder cells (FCs) and fusion-competent myoblasts (FCMs). Recent studies have established an essential function of Arp2/3-based actin polymerization during myoblast fusion, formation of a dense actin focus at the site of fusion in FCMs, and a thin sheath of actin in FCs and/or growing muscles. The formation of these actin structures depends on recognition and adhesion of myoblasts that is mediated by cell surface receptors of the immunoglobulin superfamily. However, the connection of the cell surface receptors with Arp2/3-based actin polymerization is poorly understood. To date only the SH2-SH3 adaptor protein Crk has been suggested to link cell adhesion with Arp2/3-based actin polymerization in FCMs. Here, we propose that the SH2-SH3 adaptor protein Dock, like Crk, links cell adhesion with actin polymerization. We show that Dock is expressed in FCs and FCMs and colocalizes with the cell adhesion proteins Sns and Duf at cell-cell contact points. Biochemical data in this study indicate that different domains of Dock are involved in binding the cell adhesion molecules Duf, Rst, Sns and Hbs. We emphasize the importance of these interactions by quantifying the enhanced myoblast fusion defects in duf dock, sns dock and hbs dock double mutants. Additionally, we show that Dock interacts biochemically and genetically with Drosophila Scar, Vrp1 and WASp. Based on these data, we propose that Dock links cell adhesion in FCs and FCMs with either Scar- or Vrp1-WASp-dependent Arp2/3 activation.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Drosophila , Proteínas de Drosophila/genética , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/genética , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas do Tecido Nervoso/genética , Proteína da Síndrome de Wiskott-Aldrich/genética
7.
Dev Biol ; 368(1): 18-27, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22595515

RESUMO

Myoblast fusion is a key process in multinucleated muscle formation. Prior to fusion, myoblasts recognize and adhere to each other with the aid of cell-adhesion proteins integrated into the membrane. Their intracellular domains participate in signal transduction by binding to cytoplasmic proteins. Here we identified the calcium-dependent cell-adhesion protein N-cadherin as the binding partner of the guanine-nucleotide exchange factor Schizo/Loner in Drosophila melanogaster. N-cadherin was expressed in founder cells and fusion-competent myoblasts of Drosophila during the first fusion phase. Our genetic analyses demonstrated that the myoblast fusion defect of schizo/loner mutants is rescued in part by the loss-of-function mutation of N-cadherin, which suggests that Schizo/Loner is a negative regulator of N-cadherin. Based on our findings, we propose a model where N-cadherin must be removed from the myoblast membrane to induce a protein-free zone at the cell-cell contact point to permit fusion.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Caderinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mioblastos/metabolismo , Fator 1 de Ribosilação do ADP/genética , Animais , Animais Geneticamente Modificados , Sítios de Ligação/genética , Caderinas/genética , Fusão Celular , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Imuno-Histoquímica , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Mutação , Mioblastos/citologia , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
8.
Dev Dyn ; 238(6): 1513-25, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19418445

RESUMO

In Drosophila, as in mammals, myoblast fusion is fundamental for development. This fusion process has two distinct phases that share common ultrastructural features and at least some molecular players between Drosophila and vertebrates. Here, we integrate the latest data on the key molecular players and ultrastructural features found during myoblast fusion into a new working model to explain this fundamental cellular process. At cell-cell contact sites, a protein complex (FuRMAS) serves as a signalling centre and might restrict the area of membrane fusion. The FuRMAS consists of a ring of cell adhesion molecules, signalling proteins, and F-actin. Regulated F-actin branching plays a pivotal role in myoblast fusion with regard to vesicle transport, fusion pore formation, and expansion as well as the integration of the fusion-competent myoblast into the growing myotube. Interestingly, local F-actin accumulation is a typical feature of other transient adhesive structures such as the immunological synapse, podosomes, and invadopodia. Developmental Dynamics 238:1513-1525, 2009. (c) 2009 Wiley-Liss, Inc.


Assuntos
Fusão Celular , Drosophila melanogaster/fisiologia , Complexos Multiproteicos/metabolismo , Mioblastos/fisiologia , Actinas/metabolismo , Animais , Padronização Corporal , Membrana Celular/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Mioblastos/ultraestrutura , Transdução de Sinais/fisiologia
9.
J Cell Sci ; 121(Pt 8): 1303-13, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18388318

RESUMO

Myoblast fusion takes place in two steps in mammals and in Drosophila. First, founder cells (FCs) and fusion-competent myoblasts (FCMs) fuse to form a trinucleated precursor, which then recruits further FCMs. This process depends on the formation of the fusion-restricted myogenic-adhesive structure (FuRMAS), which contains filamentous actin (F-actin) plugs at the sites of cell contact. Fusion relies on the HEM2 (NAP1) homolog Kette, as well as Blow and WASP, a member of the Wiskott-Aldrich-syndrome protein family. Here, we show the identification and characterization of schwächling--a new Arp3-null allele. Ultrastructural analyses demonstrate that Arp3 schwächling mutants can form a fusion pore, but fail to integrate the fusing FCM. Double-mutant experiments revealed that fusion is blocked completely in Arp3 and wasp double mutants, suggesting the involvement of a further F-actin regulator. Indeed, double-mutant analyses with scar/WAVE and with the WASP-interacting partner vrp1 (sltr, wip)/WIP show that the F-actin regulator scar also controls F-actin formation during myoblast fusion. Furthermore, the synergistic phenotype observed in Arp3 wasp and in scar vrp1 double mutants suggests that WASP and SCAR have distinct roles in controlling F-actin formation. From these findings we derived a new model for actin regulation during myoblast fusion.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Animais , Sequência de Bases , Primers do DNA , Drosophila , Proteínas de Drosophila/genética , Proteínas dos Microfilamentos/genética , Microscopia Eletrônica , Reação em Cadeia da Polimerase , Proteína da Síndrome de Wiskott-Aldrich/genética
10.
Dev Biol ; 304(2): 664-74, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17306790

RESUMO

In higher organisms, mononucleated myoblasts fuse to form multinucleated myotubes. During this process, myoblasts undergo specific changes in cell morphology and cytoarchitecture. Previously, we have shown that the actin regulator Kette (Hem-2/Nap-1) is essential for myoblast fusion. In this study, we describe the role of the evolutionary conserved Wiskott-Aldrich syndrome protein that serves as a regulator for the Arp2/3 complex for myoblast fusion. By screening an EMS mutagenesis collection, we discovered a new wasp allele that does not complete fusion during myogenesis. Interestingly, this new wasp3D3-035 allele is characterized by a disruption of fusion after precursor formation. The molecular lesion in this wasp allele leads to a stop codon preventing translation of the CA domain. Usually, the WASP protein exerts its function through the Arp2/3-interacting CA domain. Accordingly, a waspDeltaCA that is expressed in a wild-type background acts as dominant-negative during the fusion process. Furthermore, we show that the myoblast fusion phenotype of kette mutant embryos can be suppressed by reducing the gene dose of wasp3D3-035. Thus, Kette antagonizes WASP function during myoblast fusion.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Alelos , Animais , Fusão Celular , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas dos Microfilamentos/genética , Mutação , Proteína da Síndrome de Wiskott-Aldrich/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...