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










Publication year range
1.
J Insect Physiol ; 117: 103900, 2019.
Article in English | MEDLINE | ID: mdl-31202852

ABSTRACT

Imaging of living, intact Drosophila larvae is challenged if normal bodily function must be observed or when healthy larvae must be recovered for subsequent studies. Here, we describe a simple and short protocol that employs transient airborne chloroform or desflurane (1,2,2,2-tetrafluoroethyl difluoromethyl ether) to efficiently immobilize larvae without the use of manipulation devices, vaporizers or imaging chambers. This non-lethal method allows the use of anesthetics while allowing tracking of individual Drosophila into adulthood for follow-up experiments. At dosages sufficient to immobilize larvae, Desflurane, but not chloroform reduced the central nervous system response to auditory stimulus. Desflurane doses were sufficient to arrest the heart, however significant rapid recovery was observed. With our method, chloroform provided more rapid anesthesia but slower recovery than Desflurane. Without specialized hardware, this technique allows for repeated imaging of living Drosophila larvae.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Chloroform/administration & dosage , Desflurane/administration & dosage , Drosophila/drug effects , Immobilization/methods , Animals , Central Nervous System/drug effects , Larva/drug effects , Microscopy, Confocal
2.
Dev Biol ; 454(1): 52-65, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31228417

ABSTRACT

Hematopoietic cell lineages support organismal needs by responding to positional and systemic signals that balance proliferative and differentiation events. Drosophila provides an excellent genetic model to dissect these signals, where the activity of cues in the hemolymph or substrate can be traced to determination and differentiation events of well characterized hemocyte types. Plasmatocytes in third instar larvae increase in number in response to infection and in anticipation of metamorphosis. Here we characterize hemocyte clustering, proliferation and transdifferentiation on the heart or dorsal vessel. Hemocytes accumulate on the inner foldings of the heart basement membrane, where they move with heart contraction, and are in proximity to the heart ostia and pericardial nephrocytes. The numbers of hemocytes vary, but increase transiently before pupariation, and decrease by 4 h before pupa formation. During their accumulation at the heart, plasmatocytes can proliferate and can transdifferentiate into crystal cells. Serrate expressing cells as well as lamellocyte-like, Atilla expressing ensheathing cells are associated with some, but not all hemocyte clusters. Hemocyte aggregation is enhanced by the presence of a heart specific Collagen, Pericardin, but not the associated pericardial cells. The varied and transient number of hemocytes in the pericardial compartment suggests that this is not a hematopoietic hub, but a niche supporting differentiation and rapid dispersal in response to systemic signals.


Subject(s)
Collagen Type IV/metabolism , Drosophila Proteins/metabolism , Hematopoiesis/physiology , Hemocytes/physiology , Animals , Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Collagen/metabolism , Collagen/physiology , Collagen Type IV/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Heart/physiology , Hemolymph/metabolism , Larva/metabolism , Metamorphosis, Biological/physiology , Pupa/metabolism
4.
Vet Sci ; 4(2)2017 Apr 24.
Article in English | MEDLINE | ID: mdl-29056682

ABSTRACT

Congenital heart defects, clinically identified in both small and large animals, are multifactorial and complex. Although heritable factors are known to have a role in cardiovascular disease, the full genetic aetiology remains unclear. Model organism research has proven valuable in providing a deeper understanding of the essential factors in heart development. For example, mouse knock-out studies reveal a role for the Integrin adhesion receptor in cardiac tissue. Recent research in Drosophila melanogaster (the fruit fly), a powerful experimental model, has demonstrated that the link between the extracellular matrix and the cell, mediated by Integrins, is required for multiple aspects of cardiogenesis. Here we test the hypothesis that Integrins signal to the heart cells through Src42A kinase. Using the powerful genetics and cell biology analysis possible in Drosophila, we demonstrate that Src42A acts in early events of heart tube development. Careful examination of mutant heart tissue and genetic interaction data suggests that Src42A's role is independent of Integrin and the Integrin-related Focal Adhesion Kinase. Rather, Src42A acts non-autonomously by promoting programmed cell death of the amnioserosa, a transient tissue that neighbors the developing heart.

5.
Vet Sci ; 4(2)2017 Apr 24.
Article in English | MEDLINE | ID: mdl-29056683

ABSTRACT

The extracellular matrix (ECM) is a dynamic scaffold within organs and tissues that enables cell morphogenesis and provides structural support. Changes in the composition and organisation of the cardiac ECM are required for normal development. Congenital and age-related cardiac diseases can arise from mis-regulation of structural ECM proteins (Collagen, Laminin) or their receptors (Integrin). Key regulators of ECM turnover include matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitors of matrix metalloproteinases (TIMPs). MMP expression is increased in mice, pigs, and dogs with cardiomyopathy. The complexity and longevity of vertebrate animals makes a short-lived, genetically tractable model organism, such as Drosophila melanogaster, an attractive candidate for study. We survey ECM macromolecules and their role in heart development and growth, which are conserved between Drosophila and vertebrates, with focus upon the consequences of altered expression or distribution. The Drosophila heart resembles that of vertebrates during early development, and is amenable to in vivo analysis. Experimental manipulation of gene function in a tissue- or temporally-regulated manner can reveal the function of adhesion or ECM genes in the heart. Perturbation of the function of ECM proteins, or of the MMPs that facilitate ECM remodelling, induces cardiomyopathies in Drosophila, including cardiodilation, arrhythmia, and cardia bifida, that provide mechanistic insight into cardiac disease in mammals.

6.
PLoS One ; 12(2): e0171905, 2017.
Article in English | MEDLINE | ID: mdl-28192468

ABSTRACT

Matrix Metalloproteinases (Mmps) degrade glycoproteins and proteoglycans of the extracellular matrix (ECM) or cell surface and are crucial for morphogenesis. Mmps and their inhibitors are expressed during early stages of cardiac development in vertebrates and expression is altered in multiple congenital cardiomyopathies such as cardia bifida. Drosophila genome encodes two copies of Mmps, Mmp1 and Mmp2 whereas in humans up to 25 Mmps have been identified with overlapping functions. We investigated the role of Mmps during embryonic heart development in Drosophila, a process which is morphogenetically similar to early heart tube formation in vertebrates. We demonstrate that the two Mmps in Drosophila have distinct and overlapping roles in cell motility, cell adhesion and cardiac lumenogenesis. We determined that Mmp1 and Mmp2 promote Leading Edge membrane dynamics of cardioblasts during collective migration. Mmp2 is essential for cardiac lumen formation, and mutants generate a cardia bifida phenotype. Mmp1 is required for luminal expansion. Mmp1 and Mmp2 both localise to the basal domains of cardiac cells, however, occupy non-overlapping domains apically. Mmp1 and Mmp2 regulate the proteoglycan composition and size of the apical and basal ECM, yet only Mmp2 is required to restrict ECM assembly to the lumen. Mmp1 negatively regulates the size of the adhesive Cadherin cell surface domain, whereas in a complementary fashion, Mmp2 negatively regulates the size of the Integrin-ECM domain and thereby prescribes the domain to establish and restrict Slit morphogen signalling. Inhibition of Mmp activity through ectopic expression of Tissue Inhibitor of Metalloproteinase in the ectoderm blocks lumen formation. Therefore, Mmp expression and function identifies ECM differentiation and remodelling as a key element for cell polarisation and organogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion/genetics , Cell Movement/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Extracellular Matrix/metabolism , Heart/embryology , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 2/genetics , Microscopy, Confocal , Morphogenesis/genetics , Mutation , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Myocardium/cytology , Myocardium/enzymology , Myocardium/metabolism , Phenotype , RNA Interference
7.
Dev Biol ; 419(2): 285-297, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27618756

ABSTRACT

Collective cell migration is the coordinated movement of cells, which organize tissues during morphogenesis, repair and some cancers. The motile cell membrane of the advancing front in collective cell migration is termed the Leading Edge. The embryonic development of the vertebrate and Drosophila hearts are both characterized by the coordinated medial migration of a bilateral cluster of mesodermal cells. In Drosophila, the cardioblasts form cohesive bilateral rows that migrate collectively as a unit towards the dorsal midline to form the dorsal vessel. We have characterized the collective cell migration of cardioblasts as an in vivo quantitative model to study the behaviour of the Leading Edge. We investigated whether guidance signalling through Slit and Netrin pathways plays a role in cell migration during heart development. Through time-lapse imaging and quantitative assessment of migratory behaviour of the cardioblasts in loss-of-function mutants, we demonstrate that both Slit and Netrin mediated signals are autonomously and concomitantly required to maximize migration velocity, filopodial and lamellipodial activities. Additionally, we show that another Slit and Netrin receptor, Dscam1, the role of which during heart development was previously unknown, is required for both normal migration of cardioblasts and luminal expansion. Leading edge behaviour analysis revealed a dosage dependent genetic interaction between Slit and Netrin receptors suggesting that downstream signalling through these receptors converge on a common output that increases leading edge activity of the cardioblasts. Finally, we found that guidance signalling maintains the balance between epithelial and mesenchymal characteristics of the migrating cardioblasts.


Subject(s)
Cell Movement/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Heart/embryology , Myocytes, Cardiac/cytology , Pseudopodia/physiology , Stem Cells/cytology , Animals , Axon Guidance , Cell Adhesion Molecules , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Epithelial-Mesenchymal Transition , Luminescent Proteins/analysis , Morphogenesis , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Netrin Receptors , Netrin-1 , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/physiology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Signal Transduction , Time-Lapse Imaging , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Roundabout Proteins
8.
PLoS One ; 10(6): e0131238, 2015.
Article in English | MEDLINE | ID: mdl-26110760

ABSTRACT

Mechanotransduction of tension can govern the remodeling of cardiomyocytes during growth or cardiomyopathy. Tension is signaled through the integrin adhesion complexes found at muscle insertions and costameres but the relative importance of signalling during cardiomyocyte growth versus remodelling has not been assessed. Employing the Drosophila cardiomyocyte as a genetically amenable model, we depleted the levels of Talin, a central component of the integrin adhesion complex, at different stages of heart growth and remodeling. We demonstrate a continuous requirement for Talin during heart growth to maintain the one-to-one apposition of myofibril ends between cardiomyocytes. Retracted myofibrils cannot regenerate appositions to adjacent cells after restoration of normal Talin expression, and the resulting deficit reduces heart contraction and lifespan. Reduction of Talin during heart remodeling after hatching or during metamorphosis results in pervasive degeneration of cell contacts, myofibril length and number, for which restored Talin expression is insufficient for regeneration. Resultant dilated cardiomyopathy results in a fibrillating heart with poor rhythmicity. Cardiomyocytes have poor capacity to regenerate deficits in myofibril orientation and insertion, despite an ongoing capacity to remodel integrin based adhesions.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Heart/growth & development , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Talin/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Larva/cytology , Larva/growth & development , Larva/physiology , Male , Organogenesis , Talin/deficiency , Talin/genetics
9.
Dev Biol ; 405(2): 189-201, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25958089

ABSTRACT

Fluid- and gas-transporting tubular organs are critical to metazoan development and homeostasis. Tubulogenesis involves cell polarization and morphogenesis to specify the luminal, adhesive, and basal cell domains and to establish an open lumen. We explore a requirement for Talin, a cytoplasmic integrin adapter, during Drosophila melanogaster embryonic heart tube development. Talin marks the presumptive luminal domain and is required to orient and develop an open luminal space within the heart. Genetic analysis demonstrates that loss of zygotic or maternal-and-zygotic Talin disrupts heart cell migratory dynamics, morphogenesis, and polarity. Talin is essential for subsequent polarization of luminal determinants Slit, Robo, and Dystroglycan as well as stabilization of extracellular and intracellular integrin adhesion factors. In the absence of Talin function, mini-lumens enriched in luminal factors form in ectopic locations. Rescue experiments performed with mutant Talin transgenes suggest that actin-binding is required for normal lumen formation, but not for initial heart cell polarization. We propose that Talin provides instructive cues to position the luminal domain and coordinate the actin cytoskeleton during Drosophila heart lumen development.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Heart/embryology , Talin/physiology , Actins/metabolism , Animals , Body Patterning , Cell Movement , Crosses, Genetic , Cytoskeleton/metabolism , Green Fluorescent Proteins/metabolism , Heart/physiology , Immunohistochemistry , Morphogenesis/genetics , Mutation , Protein Binding , Protein Structure, Tertiary , Pseudopodia/metabolism , Transgenes
10.
BMC Dev Biol ; 12: 8, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22353787

ABSTRACT

BACKGROUND: The formation of a tubular organ, such as the heart, requires the communication of positional and polarity signals between migratory cells. Key to this process is the establishment of a new luminal domain on the cell surface, generally from the apical domain of a migratory cell. This domain will also acquire basal properties, as it will produce a luminal extracellular matrix. Integrin receptors are the primary means of cell adhesion and adhesion signaling with the extracellular matrix. Here we characterise the requirement of Integrins in a genetic model of vasculogenesis, the formation of the heart in Drosophila. RESULTS: As with vertebrates, the Drosophila heart arises from lateral mesoderm that migrates medially to meet their contralateral partners, to then assemble a midline vessel. During migration, Integrins are among the first proteins restricted to the presumptive luminal domain of cardioblasts. Integrins are required for normal levels of leading edge membrane motility. Apical accumulation of Integrins is enhanced by Robo, and reciprocally, apicalisation of luminal factors like Slit and Robo requires Integrin function. Integrins may provide a template for the formation of a lumen by stabilising lumen factors like Robo. Subsequent to migration, Integrin is required for normal cardioblast alignment and lumen formation. This phenotype is most readily modified by other mutations that affect adhesion, such as Talin and extracellular matrix ligands. CONCLUSION: Our findings reveal an instructive role for Integrins in communicating polarising information to cells during migration, and during transition to an epithelial tube structure.


Subject(s)
Cell Polarity , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Heart/embryology , Integrin alpha Chains/physiology , Myoblasts, Cardiac/physiology , Myocardium/cytology , Animals , Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Regulatory Networks , Integrin alpha Chains/metabolism , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/ultrastructure , Myocardium/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Roundabout Proteins
11.
Dev Biol ; 356(2): 279-90, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21565181

ABSTRACT

The transmembrane proteoglycan Syndecan contributes to cell surface signaling of diverse ligands in mammals, yet in Drosophila, genetic evidence links Syndecan only to the Slit receptor Roundabout and to the receptor tyrosine phosphatase LAR. Here we characterize the requirement for syndecan in the determination and morphogenesis of the Drosophila heart, and reveal two phases of activity, indicating that Syndecan is a co-factor in at least two signaling events in this tissue. There is a stochastic failure to determine heart cell progenitors in a subset of abdominal hemisegments in embryos mutant for syndecan, and subsequent to Syndecan depletion by RNA interference. This phenotype is sensitive to gene dosage in the FGF receptor (Heartless), its ligand, Pyramus, as well as BMP-ligand Decapentaplegic (Dpp) and co-factor Sara. Syndecan is also required for lumen formation during assembly of the heart vessel, a phenotype shared with mutations in the Slit and Integrin signaling pathways. Phenotypic interactions of syndecan with slit and Integrin mutants suggest intersecting function, consistent with Syndecan acting as a co-receptor for Slit in the Drosophila heart.


Subject(s)
Drosophila melanogaster/embryology , Heart/embryology , Syndecans/physiology , Animals , Cell Polarity , Drosophila Proteins/physiology , Integrins/physiology , Morphogenesis , Nerve Tissue Proteins/physiology
12.
BMC Dev Biol ; 8: 99, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18847477

ABSTRACT

BACKGROUND: Scaffolding proteins belonging to the membrane associated guanylate kinase (MAGUK) superfamily function as adapters linking cytoplasmic and cell surface proteins to the cytoskeleton to regulate cell-cell adhesion, cell-cell communication and signal transduction. We characterize here a Drosophila MAGUK member, Varicose (Vari), the homologue of vertebrate scaffolding protein PALS2. RESULTS: Varicose localizes to pleated septate junctions (pSJs) of all embryonic, ectodermally-derived epithelia and peripheral glia. In vari mutants, essential SJ proteins NeurexinIV and FasciclinIII are mislocalized basally and epithelia develop a leaky paracellular seal. In addition, vari mutants display irregular tracheal tube diameters and have reduced lumenal protein accumulation, suggesting involvement in tracheal morphogenesis. We found that Vari is distributed in the cytoplasm of the optic lobe neuroepithelium, as well as in a subset of neuroblasts and differentiated neurons of the nervous system. We reduced vari function during the development of adult epithelia with a partial rescue, RNA interference and generation of genetically mosaic tissue. All three approaches demonstrate that vari is required for the patterning and morphogenesis of adult epithelial hairs and bristles. CONCLUSION: Varicose is involved in scaffold assembly at the SJ and has a role in patterning and morphogenesis of adult epithelia.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Epithelium/embryology , Epithelium/physiology , Guanylate Cyclase/physiology , Membrane Proteins/physiology , Nervous System/embryology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Epithelium/metabolism , Eye/embryology , Eye/growth & development , Eye/metabolism , Gene Expression Regulation, Developmental , Genes, Lethal , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nervous System/metabolism , Nervous System Physiological Phenomena/genetics , Wings, Animal/embryology , Wings, Animal/growth & development , Wings, Animal/metabolism
13.
Dev Biol ; 293(1): 154-64, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16516189

ABSTRACT

Slit is a secreted guidance cue that conveys repellent or attractive signals from target and guidepost cells. In Drosophila, responsive cells express one or more of three Robo receptors. The cardial cells of the developing heart express both Slit and Robo2. This is the first report of coincident expression of a Robo and its ligand. In slit mutants, cardial cell alignment, polarization and uniform migration are disrupted. The heart phenotype of robo2 mutants is similar, with fewer migration defects. In the guidance of neuronal growth cones in Drosophila, there is a phenotypic interaction between slit and robo heterozygotes, and also with genes required for Robo signaling. In contrast, in the heart, slit has little or no phenotypic interaction with Robo-related genes, including Robo2, Nck2, and Disabled. However, there is a strong phenotypic interaction with Integrin genes and their ligands, including Laminin and Collagen, and intracellular messengers, including Talin and ILK. This indicates that Slit participates in adhesion or adhesion signaling during heart development.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Heart/embryology , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Animals , Animals, Genetically Modified , Drosophila/ultrastructure , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila Proteins/ultrastructure , Integrins/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/ultrastructure , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Immunologic/ultrastructure
14.
Oncogene ; 22(13): 1916-26, 2003 Apr 03.
Article in English | MEDLINE | ID: mdl-12673197

ABSTRACT

The ErbB-2/Neu receptor tyrosine kinase plays a causal role in tumorigenesis in mammals. Neu's carboxyl terminus contains five phosphorylated tyrosines that mediate transformation through interaction with cytoplasmic SH2 or PTB containing adaptor proteins. We show that Drosophila adaptors signal from individual phosphotyrosine sites of rat Neu. Activated Neu expression in the midline glia suppressed apoptosis, similar to that seen with activated Drosophila EGF-R expression. Expression in eye and wing tissues generated graded phenotypes suitable for dosage-sensitive modifier genetics. Suppression of ErbB-2/Neu-induced phenotypes in tissues haplosufficient for genes encoding adaptor protein or second messengers suggests that pTyr 1227(YD) signals require Shc, and that pTyr 1253 (YE) signalling does not employ Ras, but does require Raf function. Signalling from pTyr (YB) was affected by a haplosufficiency in drk (Grb-2), and in genes thought to function downstream of Grb-2: dab, sos, csw (Shp-2), and dos (Gab-1). These data demonstrate the power of Drosophila genetics to unmask the molecules that signal from oncogenic ErbB-2/Neu.


Subject(s)
Adaptor Proteins, Vesicular Transport , Drosophila Proteins/physiology , Phosphotyrosine/chemistry , Protein Kinases , Protein Processing, Post-Translational , Receptor, ErbB-2/chemistry , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Apoptosis/genetics , Apoptosis/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , ErbB Receptors/physiology , Eye/growth & development , Eye Proteins/genetics , Eye Proteins/physiology , Gene Dosage , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Morphogenesis/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Phenotype , Phosphorylation , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Protein Tyrosine Phosphatases, Non-Receptor , Proteins/genetics , Proteins/physiology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/physiology , Rats , Receptor, ErbB-2/physiology , Receptors, Invertebrate Peptide/physiology , Recombinant Fusion Proteins/physiology , Shc Signaling Adaptor Proteins , Signal Transduction/genetics , Son of Sevenless Protein, Drosophila/genetics , Son of Sevenless Protein, Drosophila/physiology , Structure-Activity Relationship , Wings, Animal/growth & development
15.
J Neurosci ; 22(11): 4448-55, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12040052

ABSTRACT

Integrins are concentrated within growth cones, but their contribution to axon extension and pathfinding is unclear. Genetic lesion of individual integrins does not stop growth cone extension or motility, but does increase axon defasciculation and axon tract displacement. In this study, we document a dosage-dependent phenotypic interaction between genes for the integrins, their ligands, and the midline growth cone repellent, Slit, but not for the midline attractant, Netrin. Longitudinal tract axons in Drosophila embryos doubly heterozygous for slit and an integrin gene, encoding alphaPS1, alphaPS2, alphaPS3, or betaPS1, take ectopic trajectories across the midline of the CNS. Drosophila doubly heterozygous for slit and the genes encoding the integrin ligands Laminin A and Tiggrin reveal similar errors in midline axon guidance. We propose that the strength of adhesive signaling from integrins influences the threshold of response by growth cones to repellent axon guidance cues.


Subject(s)
Axons/metabolism , Drosophila Proteins , Integrins/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Animals , Drosophila , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/innervation , Embryo, Nonmammalian/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Dosage , Gene Expression , Growth Cones/metabolism , Heterozygote , Immunohistochemistry , Integrins/genetics , Laminin/genetics , Laminin/metabolism , Ligands , Mutation , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Netrin-1 , Netrins , Penetrance , Phenotype , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...