Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 11(3)2022 01 19.
Article in English | MEDLINE | ID: mdl-35159130

ABSTRACT

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.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Cell Communication , Cell Fusion , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Mammals/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism
2.
Biol Open ; 2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34897417

ABSTRACT

Guanine nucleotide exchange factors (GEF) of the BRAG subfamily activate small Arf GTPases, which are pivotal regulators of intracellular membrane traffic and actin dynamics. Consequently, BRAG proteins have been implicated to regulate the surface levels of adhesive and signaling receptors. However, not much is known about the mechanism leading to the regulation of these surface proteins. In this study we found that the Drosophila BRAG GEF Schizo interacts physically with the Abl-interactor (Abi). schizo mutants display severe defects in myoblast fusion during syncytial muscle formation and show increased amounts of the cell adhesion protein N-cadherin. We demonstrate that the schizo myoblast fusion phenotype can be rescued by the expression of the Schizo GEF (Sec7) and membrane-binding (pleckstrin homology) domain. Furthermore, the expression of the Sec7-PH domain in a wild-type background decreases the amounts of N-cadherin and impairs myoblast fusion. These findings support the notion that the Sec7-PH domain serves as a constitutive-active form of Schizo. Using a yeast-two hybrid assay, we show that the SH3 domain of Abi interacts with the N-terminal region of Schizo. This region is also able to bind to the cytodomain of the cell adhesion molecule N-cadherin. To shed light on the function of Schizo and Abi in N-cadherin removal, we employed epistasis experiments in different developmental contexts of Drosophila. These studies point towards a new model for the regulation of Schizo. We propose that the binding of Abi to the N-terminal part of Schizo antagonizes Schizo function to inhibit N-cadherin removal.

3.
Pharmacol Res Perspect ; 4(5): e00248, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27713820

ABSTRACT

Fibromyalgia is characterized by widespread musculoskeletal pain, fatigue, and depression. The aim was to analyze potential mitochondrial dysfunction or autophagy in mice after exposure to intermittent cold stress (ICS). Muscle and liver specimens were obtained from 36 mice. Lactate dehydrogenase (LDH) activity was measured. Microtubule-associated protein light chain 3 (MAP1LC3B) and glycogen content were determined histologically; muscle ultrastructure by electron microscopy. Mitochondrial- and autophagy-related markers were analyzed by RT-qPCR and Western blotting. ATP level, cytotoxicity, and caspase 3 activity were measured in murine C2C12 myoblasts after ICS exposure. Coenzyme Q10B (COQ10B) transcript was up-regulated in limb muscle of ICS mice, whereas its protein content was stable. Cytochrome C oxidase 4 (COX4I1) and LDH activity increased in limb muscle of male ICS mice. Glycogen content was lower in muscle and liver tissue of male ICS mice. Electron micrographs of ICS mice specimens showed mitochondrial damage and autophagic vesicles. A significant up-regulation of autophagic transcripts of MAP1LC3B and BECLIN 1 (BECN1) was observed. Map1lc3b protein showed an aggregated distribution in ICS mice and SqSTM1/p62 (p62) protein level was stable. Furthermore, ATP level and caspase activity, detected as apoptotic marker, were significantly lowered after ICS exposure in differentiated C2C12 myoblasts. The present study shows that ICS mice are characterized by mitochondrial dysfunction, autophagic processes, and metabolic alterations. Further investigations could dissect autophagy process in the proposed model and link these mechanisms to potential therapeutic options for fibromyalgia.

4.
J Cell Sci ; 129(18): 3426-36, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27521427

ABSTRACT

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.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Intercellular Junctions/metabolism , Microfilament Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Cadherins/metabolism , Cell Fusion , Models, Biological , Mutation/genetics , Myoblasts/ultrastructure , Phenotype , rac1 GTP-Binding Protein/metabolism
5.
J Cell Sci ; 129(3): 604-20, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26675239

ABSTRACT

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.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Drosophila Proteins/metabolism , Drosophila/metabolism , Macrophages/metabolism , Microfilament Proteins/metabolism , Myoblasts/metabolism , Organogenesis/physiology , Actin Cytoskeleton/metabolism , Animals , Drosophila/physiology , Morphogenesis/physiology , Muscle Development/physiology , Wiskott-Aldrich Syndrome Protein/metabolism
6.
BMC Cell Biol ; 15: 27, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25000973

ABSTRACT

BACKGROUND: The visceral musculature of Drosophila larvae comprises circular visceral muscles tightly interwoven with longitudinal visceral muscles. During myogenesis, the circular muscles arise by one-to-one fusion of a circular visceral founder cell (FC) with a visceral fusion-competent myoblast (FCM) from the trunk visceral mesoderm, and longitudinal muscles arise from FCs of the caudal visceral mesoderm. Longitudinal FCs migrate anteriorly under guidance of fibroblast growth factors during embryogenesis; it is proposed that they fuse with FCMs from the trunk visceral mesoderm to give rise to syncytia containing up to six nuclei. RESULTS: Using fluorescence in situ hybridization and immunochemical analyses, we investigated whether these fusion events during migration use the same molecular repertoire and cellular components as fusion-restricted myogenic adhesive structure (FuRMAS), the adhesive signaling center that mediates myoblast fusion in the somatic mesoderm. Longitudinal muscles were formed by the fusion of one FC with Sns-positive FCMs, and defects in FCM specification led to defects in longitudinal muscle formation. At the fusion sites, Duf/Kirre and the adaptor protein Rols7 accumulated in longitudinal FCs, and Blow and F-actin accumulated in FCMs. The accumulation of these four proteins at the fusion sites argues for FuRMAS-like adhesion and signaling centers. Longitudinal fusion was disturbed in rols and blow single, and scar wip double mutants. Mutants of wasp or its interaction partner wip had no defects in longitudinal fusion. CONCLUSIONS: Our results indicated that all embryonic fusion events depend on the same cell-adhesion molecules, but that the need for Rols7 and regulators of F-actin distinctly differs. Rols7 was required for longitudinal visceral and somatic myoblast fusion but not for circular visceral fusion. Importantly, longitudinal fusion depended on Kette and SCAR/Wave but was independent of WASp-dependent Arp2/3 activation. Thus, the complexity of the players involved in muscle formation increases from binucleated circular muscles to longitudinal visceral muscles to somatic muscles.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Myoblasts/cytology , Animals , Animals, Genetically Modified , Cell Movement , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , Muscle Development , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/embryology , Muscles/metabolism , Myoblasts/metabolism
7.
J Neurogenet ; 28(3-4): 302-15, 2014.
Article in English | MEDLINE | ID: mdl-24957080

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Myoblasts/metabolism , Synapses/metabolism , Animals , Cell Adhesion Molecules/metabolism , Drosophila/metabolism , Exocytosis/physiology , Synaptic Vesicles/metabolism
8.
Dev Genes Evol ; 223(3): 159-69, 2013 May.
Article in English | MEDLINE | ID: mdl-23111653

ABSTRACT

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.


Subject(s)
Drosophila/genetics , Hydra/genetics , Receptors, Fibroblast Growth Factor/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Evolution, Molecular , Molecular Sequence Data , Mutation , Phylogeny , Receptors, Fibroblast Growth Factor/chemistry , Sequence Homology, Amino Acid
9.
J Cell Sci ; 126(Pt 1): 360-72, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22992459

ABSTRACT

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.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Nerve Tissue Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Drosophila , Drosophila Proteins/genetics , Immunoglobulins/genetics , Immunoglobulins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins/genetics , Muscle Development/genetics , Muscle Development/physiology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/genetics , Wiskott-Aldrich Syndrome Protein/genetics
10.
Dev Biol ; 368(1): 18-27, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22595515

ABSTRACT

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.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Cadherins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Myoblasts/metabolism , ADP-Ribosylation Factor 1/genetics , Animals , Animals, Genetically Modified , Binding Sites/genetics , Cadherins/genetics , Cell Fusion , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mutation , Myoblasts/cytology , Protein Binding , Two-Hybrid System Techniques
11.
J Cell Sci ; 125(Pt 2): 461-72, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22302994

ABSTRACT

Here we report on the generation and in vivo analysis of a series of loss-of-function mutants for the Drosophila ArfGEF, Gartenzwerg. The Drosophila gene gartenzwerg (garz) encodes the orthologue of mammalian GBF1. garz is expressed ubiquitously in embryos with substantially higher abundance in cells forming diverse tubular structures such as salivary glands, trachea, proventriculus or hindgut. In the absence of functional Garz protein, the integrity of the Golgi complex is impaired. As a result, both vesicle transport of cargo proteins and directed apical membrane delivery are severely disrupted. Dysfunction of the Arf1-COPI machinery caused by a loss of Garz leads to perturbations in establishing a polarized epithelial architecture of tubular organs. Furthermore, insufficient apical transport of proteins and other membrane components causes incomplete luminal diameter expansion and deficiencies in extracellular matrix assembly. The fact that homologues of Garz are present in every annotated metazoan genome indicates that secretion processes mediated by the GBF-type ArfGEFs play a universal role in animal development.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Guanine Nucleotide Exchange Factors/physiology , Secretory Pathway , Animals , Cell Line , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Mutation , Salivary Glands/embryology , Salivary Glands/ultrastructure , Trachea/embryology , Trachea/metabolism , Trachea/ultrastructure
12.
Dev Dyn ; 238(6): 1513-25, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19418445

ABSTRACT

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.


Subject(s)
Cell Fusion , Drosophila melanogaster/physiology , Multiprotein Complexes/metabolism , Myoblasts/physiology , Actins/metabolism , Animals , Body Patterning , Cell Membrane/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Myoblasts/ultrastructure , Signal Transduction/physiology
13.
J Cell Sci ; 121(Pt 8): 1303-13, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18388318

ABSTRACT

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.


Subject(s)
Actin-Related Protein 2-3 Complex/physiology , Drosophila Proteins/physiology , Microfilament Proteins/physiology , Wiskott-Aldrich Syndrome Protein/physiology , Actin-Related Protein 2-3 Complex/genetics , Animals , Base Sequence , DNA Primers , Drosophila , Drosophila Proteins/genetics , Microfilament Proteins/genetics , Microscopy, Electron , Polymerase Chain Reaction , Wiskott-Aldrich Syndrome Protein/genetics
14.
Dev Biol ; 304(2): 664-74, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17306790

ABSTRACT

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.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Microfilament Proteins/metabolism , Myoblasts/physiology , Wiskott-Aldrich Syndrome Protein/metabolism , Alleles , Animals , Cell Fusion , Drosophila/embryology , Drosophila Proteins/genetics , Microfilament Proteins/genetics , Mutation , Wiskott-Aldrich Syndrome Protein/genetics
15.
Development ; 131(11): 2587-94, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15148300

ABSTRACT

The CNS of bilateral symmetric organisms is characterized by intensive contralateral axonal connections. Genetic screens in Drosophila have identified only a few genes required for guiding commissural growth cones toward and across the midline. Two evolutionarily conserved signaling molecules, Netrin and Slit, are expressed in the CNS midline cells. Netrin acts primarily as an attractive signaling cue, whereas Slit mediates repulsive functions. Here, we describe a detailed analysis of the Drosophila gene schizo, which is required for commissure formation. schizo leads to a commissural phenotype reminiscent of netrin mutant embryos. Double-mutant analyses indicate that Netrin and Schizo act independently. The schizo mutant phenotype can be suppressed by either expressing netrin in the CNS midline cells or by a reduction of the slit gene dose, indicating that the balance of attractive and repulsive signaling is impaired in schizo mutants. Overexpression of the schizo RNA in the CNS midline using the GAL4/UAS system leads to a slit phenocopy, suggesting that schizo primarily antagonizes Slit signaling. This is further supported by cell type-specific rescue experiments. The schizo gene generates at least two proteins containing a conserved Sec7 and a pleckstrin homology domain (PH) characteristic for guanine nucleotide exchange factors (GEF) acting on ARF GTPases, which are known to regulate endocytosis. In support of the notion that schizo regulates Slit expression via endocytosis, we found that block of endocytosis leads to a schizo-like phenotype. We thus propose that the balance of the two signaling cues Netrin and Slit can be regulated, controlling membrane dynamics.


Subject(s)
ADP-Ribosylation Factors/genetics , Central Nervous System/embryology , Drosophila Proteins/genetics , Drosophila/embryology , Guanine Nucleotide Exchange Factors/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Axons , Blood Proteins/metabolism , Body Patterning/genetics , Central Nervous System/physiology , Drosophila/genetics , Drosophila Proteins/metabolism , Dynamins/genetics , Dynamins/metabolism , Embryo, Nonmammalian , Endocytosis/physiology , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/metabolism , Mutation , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin-1 , Phosphoproteins/metabolism , Protein Structure, Tertiary , Signal Transduction , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...