Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Curr Protoc Neurosci ; 91(1): e90, 2020 03.
Article in English | MEDLINE | ID: mdl-31971665

ABSTRACT

Mosaic analysis with a repressible cell marker (MARCM)-related technologies are positive genetic mosaic labeling systems that have been widely applied in studies of Drosophila brain development and neural circuit formation to identify diverse neuronal types, reconstruct neural lineages, and investigate the function of genes and molecules. Two types of MARCM-related technologies have been developed: single-colored and twin-colored. Single-colored MARCM technologies label one of two twin daughter cells in otherwise unmarked background tissues through site-specific recombination of homologous chromosomes during mitosis of progenitors. On the other hand, twin-colored genetic mosaic technologies label both twin daughter cells with two distinct colors, enabling the retrieval of useful information from both progenitor-derived cells and their subsequent clones. In this overview, we describe the principles and usage guidelines for MARCM-related technologies in order to help researchers employ these powerful genetic mosaic systems in their investigations of intricate neurobiological topics. © 2020 by John Wiley & Sons, Inc.


Subject(s)
Drosophila melanogaster/genetics , Neurons/ultrastructure , Animals , Cell Division , Cell Lineage , Clone Cells/ultrastructure , Color , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Gene Expression , Genes, Insect , Genes, Reporter , Genes, Suppressor , Imaginal Discs/ultrastructure , Mosaicism , Neural Stem Cells/cytology , RNA Interference , Recombinases , Recombination, Genetic
2.
Dev Cell ; 51(3): 299-312.e4, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31607650

ABSTRACT

Tissue folding is a fundamental process that shapes epithelia into complex 3D organs. The initial positioning of folds is the foundation for the emergence of correct tissue morphology. Mechanisms forming individual folds have been studied, but the precise positioning of folds in complex, multi-folded epithelia is less well-understood. We present a computational model of morphogenesis, encompassing local differential growth and tissue mechanics, to investigate tissue fold positioning. We use the Drosophila wing disc as our model system and show that there is spatial-temporal heterogeneity in its planar growth rates. This differential growth, especially at the early stages of development, is the main driver for fold positioning. Increased apical layer stiffness and confinement by the basement membrane drive fold formation but influence positioning to a lesser degree. The model successfully predicts the in vivo morphology of overgrowth clones and wingless mutants via perturbations solely on planar differential growth in silico.


Subject(s)
Drosophila melanogaster/growth & development , Epithelium/growth & development , Morphogenesis , Animals , Basement Membrane/ultrastructure , Clone Cells , Computer Simulation , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epithelium/anatomy & histology , Epithelium/ultrastructure , Imaginal Discs/anatomy & histology , Imaginal Discs/ultrastructure , Models, Biological , Mutation/genetics , Time Factors , Wings, Animal/anatomy & histology , Wings, Animal/ultrastructure , Wnt1 Protein/genetics
3.
Bull Exp Biol Med ; 166(2): 233-236, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30488212

ABSTRACT

The genotoxic effects of heterocyclic compounds were evaluated on the basis of genetic and toxicological characteristics of a biological model of Drosophila melanogaster. Analysis of the viability parameters (fertility, progeny mortality) showed that of 6 tested substance, substance No. 3 exhibited minimum toxicity. After application of substances No. 1 and No. 5 in the studied concentrations, the number of survived flies was insufficient for further analysis, which attested to high toxicity of these substances. The intensity of apoptosis was studied in response to substances Nos. 2, 4, and 6. Substance No. 4 proved to be optimal by the parameter toxicity/apoptosis (low toxicity/high apoptosis), while substance No. 3 exhibited low toxicity, which manifested in low apoptosis intensity.


Subject(s)
Benzocaine/toxicity , Drosophila melanogaster/drug effects , Fertility/drug effects , Longevity/drug effects , Quinoxalines/toxicity , Toxicity Tests , Animals , Apoptosis/drug effects , Clutch Size/drug effects , DNA Damage , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Imaginal Discs/anatomy & histology , Imaginal Discs/drug effects , Imaginal Discs/ultrastructure , Larva/drug effects , Larva/genetics , Larva/growth & development , Longevity/genetics , Mutagenicity Tests , Pefloxacin , Predictive Value of Tests , Pupa/drug effects , Pupa/genetics , Pupa/growth & development
4.
Brain ; 141(6): 1622-1636, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29718187

ABSTRACT

Several genes related to mitochondrial functions have been identified as causative genes of neuropathy or ataxia. Cytochrome c oxidase assembly factor 7 (COA7) may have a role in assembling mitochondrial respiratory chain complexes that function in oxidative phosphorylation. Here we identified four unrelated patients with recessive mutations in COA7 among a Japanese case series of 1396 patients with Charcot-Marie-Tooth disease (CMT) or other inherited peripheral neuropathies, including complex forms of CMT. We also found that all four patients had characteristic neurological features of peripheral neuropathy and ataxia with cerebellar atrophy, and some patients showed leukoencephalopathy or spinal cord atrophy on MRI scans. Validated mutations were located at highly conserved residues among different species and segregated with the disease in each family. Nerve conduction studies showed axonal sensorimotor neuropathy. Sural nerve biopsies showed chronic axonal degeneration with a marked loss of large and medium myelinated fibres. An immunohistochemical assay with an anti-COA7 antibody in the sural nerve from the control patient showed the positive expression of COA7 in the cytoplasm of Schwann cells. We also observed mildly elevated serum creatine kinase levels in all patients and the presence of a few ragged-red fibres and some cytochrome c oxidase-negative fibres in a muscle biopsy obtained from one patient, which was suggestive of subclinical mitochondrial myopathy. Mitochondrial respiratory chain enzyme assay in skin fibroblasts from the three patients showed a definitive decrease in complex I or complex IV. Immunocytochemical analysis of subcellular localization in HeLa cells indicated that mutant COA7 proteins as well as wild-type COA7 were localized in mitochondria, which suggests that mutant COA7 does not affect the mitochondrial recruitment and may affect the stability or localization of COA7 interaction partners in the mitochondria. In addition, Drosophila COA7 (dCOA7) knockdown models showed rough eye phenotype, reduced lifespan, impaired locomotive ability and shortened synaptic branches of motor neurons. Our results suggest that loss-of-function COA7 mutation is responsible for the phenotype of the presented patients, and this new entity of disease would be referred to as spinocerebellar ataxia with axonal neuropathy type 3.


Subject(s)
Electron Transport Complex IV/genetics , Hereditary Sensory and Motor Neuropathy/complications , Hereditary Sensory and Motor Neuropathy/genetics , Mutation/genetics , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/genetics , Adolescent , Animals , Animals, Genetically Modified , Brain/diagnostic imaging , Cells, Cultured , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Family Health , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Predisposition to Disease/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Humans , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Locomotion/drug effects , Locomotion/genetics , Male , Middle Aged , Motor Neurons/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Psychomotor Performance/physiology , RNA Interference/physiology , Spinal Cord/diagnostic imaging , Spinocerebellar Ataxias/diagnostic imaging , Young Adult
5.
Methods Mol Biol ; 1642: 195-209, 2017.
Article in English | MEDLINE | ID: mdl-28815502

ABSTRACT

The fruit fly, Drosophila melanogaster, has been a favorite experimental system of developmental biologists for more than a century. One of the most attractive features of this model system is the clarity by which one can analyze mutant phenotypes. Most genes are found in single copies, and loss-of-function mutants often have obvious phenotypes that can be analyzed during development and in adulthood. As with all metazoans, a significant fraction of Drosophila genes are used during both embryonic and postembryonic development, and null mutants often die during embryogenesis thereby precluding the analysis of postembryonic tissues. For several decades researchers worked around this problem by either studying gynandromorphs or irradiating chromosomes carrying mutations in the hope of inducing mitotic recombination which would then allow for the analysis of mutant phenotypes in smaller populations of cells. The former method suffers from the fact that mutations in the gene of interest are often lethal when generated in large sectors, which is a hallmark of gynandromorphs. Clonal induction with the latter method occurs at relatively low frequencies making this method laborious. The introduction of the yeast FRT System/FRT site-directed recombination system to Drosophila has made generating loss-of-function mosaic clones simple and easy. Over the years several variants of this method have allowed developmental biologists to remove genes, overexpress genes, and even express one gene in patches of cells that are mutant for a second gene. In this review we will briefly discuss some of various FRT System/FRT-based approaches that are being used to manipulate gene expression in Drosophila. The individual FRT System/FRT-based methods are described in the papers that are cited herein. We will outline the procedure that our lab uses to prepare and analyze mosaic clones in Drosophila eye-antennal imaginal discs.


Subject(s)
DNA Nucleotidyltransferases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Recombination, Genetic , Animals , Animals, Genetically Modified , Chromosomes, Insect/chemistry , Chromosomes, Insect/metabolism , Clone Cells , DNA Nucleotidyltransferases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaginal Discs/growth & development , Imaginal Discs/ultrastructure , Larva/genetics , Larva/growth & development , Larva/metabolism , Mosaicism , Mutation , Phenotype , Tissue Fixation/methods
6.
Methods Mol Biol ; 1478: 203-213, 2016.
Article in English | MEDLINE | ID: mdl-27730583

ABSTRACT

The ex vivo cultivation and live imaging of wing discs open exciting new research avenues by overcoming the limitations of end-point analysis of fixed tissues. Here we describe how to prepare an optimized wing disc culture medium (WM1) and how to dissect and arrange wing discs for cultivation and live imaging. This protocol enables the study of dynamic phenomena such as cell division and delamination as well as the use of pharmacological compounds and biosensors. Wing discs cultured and imaged as described here, maintain constant levels of proliferation during the first ten hours of culture.


Subject(s)
Imaginal Discs/ultrastructure , Optical Imaging/methods , Tissue Culture Techniques , Wings, Animal/ultrastructure , Animals , Complex Mixtures/chemistry , Culture Media/pharmacology , Drosophila melanogaster/drug effects , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drosophila melanogaster/ultrastructure , Imaginal Discs/drug effects , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Insulin/pharmacology , Larva/drug effects , Larva/growth & development , Larva/metabolism , Larva/ultrastructure , Wings, Animal/drug effects , Wings, Animal/growth & development , Wings, Animal/metabolism
7.
J Cell Sci ; 129(14): 2726-31, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27235419

ABSTRACT

The uncoordinated (unc) gene product, a potential ortholog of mammalian orofaciodigital syndrome 1 (Ofd1), is involved in the assembly of the ciliary axoneme in Drosophila and it is, therefore, constrained to cell types that have ciliary structures, namely type 1 sensory neurons and male germ cells. Here, we show that evenly spaced Unc-GFP spots are present in the eye imaginal discs of third-instar larvae. These spots are restricted to the R8 photoreceptor cell of each ommatidium in association with mother centrioles. This finding is unexpected because the Drosophila eye is of the rhabdomeric type and would be expected to lack ciliary structures.


Subject(s)
Cilia/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Green Fluorescent Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Animals , Centrioles/metabolism , Cilia/ultrastructure , Drosophila melanogaster/ultrastructure , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Mutation/genetics , Photoreceptor Cells, Invertebrate/ultrastructure
8.
FEBS J ; 282(24): 4727-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26411401

ABSTRACT

OSCP1/NOR1 (organic solute carrier partner 1/oxidored nitrodomain-containing protein 1) is a known tumor suppressor protein. OSCP1 has been reported to mediate transport of various organic solutes into cells; however, its role during development has not yet been addressed. Here we report the results of studies on dOSCP1 (the Drosophila ortholog of hOSCP1) to elucidate the role of OSCP1/NOR1 during development. Knockdown of dOSCP1 in the eye imaginal discs induced a rough-eye phenotype in adult flies. This phenotype resulted from induction of caspase-dependent apoptosis followed by a compensatory cell proliferation and generation of reactive oxygen species in eye imaginal discs. The induction of apoptosis appears to be associated with down-regulation of the anti-apoptotic Buffy gene and up-regulation of the pro-apoptotic Debcl gene. These effects of knockdown of dOSCP1 lead to mitochondrial fragmentation, degradation, and a shortfall in ATP production. We also found that knockdown of dOSCP1 causes a defect in cone cell and pigment cell differentiation in pupal retinae. Moreover, mutations in epidermal growth factor receptor pathway-related genes, such as Spitz and Drk, enhanced the rough-eye phenotype induced by dOSCP1 knockdown. These results suggest that dOSCP1 positively regulates the epidermal growth factor receptor signaling pathway. Overall, our findings indicate that dOSCP1 plays multiple roles during eye development in Drosophila.


Subject(s)
Apoptosis , Drosophila Proteins/agonists , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , ErbB Receptors/agonists , Eye/metabolism , Membrane Transport Proteins/metabolism , Reactive Oxygen Species/metabolism , Receptors, Invertebrate Peptide/agonists , Signal Transduction , Animals , Animals, Genetically Modified , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , ErbB Receptors/genetics , ErbB Receptors/metabolism , Eye/cytology , Eye/growth & development , Eye/ultrastructure , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Imaginal Discs/cytology , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Larva/genetics , Larva/physiology , Membrane Transport Proteins/genetics , Microscopy, Electron, Scanning , Mutation , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/ultrastructure , RNA Interference , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism
9.
Elife ; 4: e06567, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25826608

ABSTRACT

The Hippo pathway controls tissue growth through a core kinase cascade that impinges on the transcription of growth-regulatory genes. Understanding how this pathway is regulated in development remains a major challenge. Recent studies suggested that Hippo signaling can be modulated by cytoskeletal tension through a Rok-myosin II pathway. How cytoskeletal tension is regulated or its relationship to the other known upstream regulators of the Hippo pathway remains poorly defined. In this study, we identify spectrin, a contractile protein at the cytoskeleton-membrane interface, as an upstream regulator of the Hippo signaling pathway. We show that, in contrast to canonical upstream regulators such as Crumbs, Kibra, Expanded, and Merlin, spectrin regulates Hippo signaling in a distinct way by modulating cortical actomyosin activity through non-muscle myosin II. These results uncover an essential mediator of Hippo signaling by cytoskeleton tension, providing a new entry point to dissecting how mechanical signals regulate Hippo signaling in living tissues.


Subject(s)
Actomyosin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ovarian Follicle/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spectrin/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actomyosin/genetics , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eye/cytology , Eye/growth & development , Eye/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaginal Discs/growth & development , Imaginal Discs/ultrastructure , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mitosis , Ovarian Follicle/growth & development , Ovarian Follicle/ultrastructure , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Pupa/cytology , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , Spectrin/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
PLoS One ; 8(10): e76775, 2013.
Article in English | MEDLINE | ID: mdl-24146926

ABSTRACT

The DYRKs (dual-specificity tyrosine phosphorylation-regulated kinases) are a conserved family of protein kinases that are associated with a number of neurological disorders, but whose biological targets are poorly understood. Drosophila encodes three Dyrks: minibrain/Dyrk1A, DmDyrk2, and DmDyrk3. Here we describe the creation and characterization of a DmDyrk2 null allele, DmDyrk2(1w17) . We provide evidence that the smell impaired allele smi35A(1) , is likely to encode DmDyrk2. We also demonstrate that DmDyrk2 is expressed late in the developing third antennal segment, an anatomical structure associated with smell. In addition, we find that DmDyrk2 is expressed in the morphogenetic furrow of the developing eye, that loss of DmDyrk2 in the eye produced a subtle but measurable defect, and that ectopic DmDyrk2 expression in the eye produced a strong rough eye phenotype characterized by increased secondary, tertiary and bristle interommatidial cells. This phenotype was dependent on DmDyrk2 kinase activity and was only manifest when expressed in post-mitotic non-neuronal progenitors. Together, these data indicate that DmDyrk2 is expressed in developing sensory systems, that it is required for the development of the visual system, and that the eye is a good model to identify DmDyrk2 targets.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Protein Serine-Threonine Kinases/metabolism , Visual Pathways/growth & development , Visual Pathways/metabolism , Alleles , Animals , Arthropod Antennae/metabolism , Arthropod Antennae/ultrastructure , Avoidance Learning , Body Patterning , DNA Transposable Elements/genetics , Drosophila Proteins/deficiency , Electroretinography , Fluorescent Antibody Technique , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Mitosis , Morphogenesis , Odorants , Phenotype , Protein Serine-Threonine Kinases/deficiency , Pupa/cytology , Pupa/metabolism , Retina/cytology , Retina/ultrastructure , Stem Cells/cytology
11.
Tohoku J Exp Med ; 231(2): 117-25, 2013 10.
Article in English | MEDLINE | ID: mdl-24126234

ABSTRACT

Heme oxygenase (HO) is a rate-limiting step of heme degradation, which catalyzes the conversion of heme into biliverdin, iron, and CO. HO has been characterized in microorganisms, insects, plants, and mammals. The mammalian enzyme participates in adaptive and protective responses to oxidative stress and various inflammatory stimuli. The present study reports that eye imaginal disc-specific knockdown of the Drosophila HO homologue (dHO) conferred serious abnormal eye morphology in adults, resulting in the generation of reactive oxygen species and apoptosis in third-instar larvae. Oxidative stress frequently induces DNA lesions that are recognized by damage sensors, including ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and rad3-related (ATR) proteins. The knockdown of dHO took place in G0/G1-arrested cells posterior to the morphogenetic furrow and thus prevented these cells from entering S-phase, with an increase in the level of histone H2A.V, a DNA damage marker. Moreover, the knockdown of dHO resulted in the enhancement of the rough eye phenotype in ATM-deficient flies or was lethal in ATR-deficient flies. These results indicate that dHO functions in control of the signal pathway of DNA damage. On the other hand, genetic crosses with a collection of Drosophila deficiency stocks allowed us to identify eight genomic regions, each deletion of which caused suppression of the rough eye phenotype induced by dHO knockdown. This information should facilitate the identification of HO regulators in Drosophila and clarification of the roles of HO in eye development.


Subject(s)
Compound Eye, Arthropod/growth & development , DNA Damage/genetics , Drosophila melanogaster/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Signal Transduction/genetics , Animals , Animals, Genetically Modified , Bromodeoxyuridine , Cell Cycle/genetics , Cell Cycle/physiology , Compound Eye, Arthropod/abnormalities , Crosses, Genetic , DNA Damage/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Knockdown Techniques , HEK293 Cells , Heme Oxygenase (Decyclizing)/genetics , Humans , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Larva/enzymology , Larva/growth & development , Microscopy, Electron, Scanning , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species , Signal Transduction/physiology
12.
Science ; 340(6139): 1445-7, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23704373

ABSTRACT

Although intercellular bridges resulting from incomplete cytokinesis were discovered in somatic Drosophila tissues decades ago, the impact of these structures on intercellular communication and tissue biology is largely unknown. In this work, we demonstrate that the ~250-nanometer-diameter somatic ring canals permit diffusion of cytoplasmic contents between connected cells and across mitotic clone boundaries and enable the equilibration of protein between transcriptionally mosaic follicle cells in the Drosophila ovary. We obtained similar, although more restricted, results in the larval imaginal discs. Our work illustrates the lack of cytoplasmic autonomy in these tissues and suggests a role for somatic ring canals in promoting homogeneous protein expression within the tissue.


Subject(s)
Cytoplasm/metabolism , Cytoplasmic Structures/metabolism , Cytoplasmic Structures/ultrastructure , Drosophila Proteins/metabolism , Green Fluorescent Proteins/metabolism , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Protein Transport , Animals , Cell Cycle , Diffusion , Drosophila , Female , Giant Cells/ultrastructure , Microscopy, Electron , Mitosis , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovarian Follicle/ultrastructure , Recombination, Genetic , Transcription, Genetic , Transgenes
13.
Dev Biol ; 377(1): 113-25, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23428616

ABSTRACT

Ommatidial rotation is one of the most important events for correct patterning of the Drosophila eye. Although several signaling pathways are involved in this process, few genes have been shown to specifically affect it. One of them is nemo (nmo), which encodes a MAP-like protein kinase that regulates the rate of rotation throughout the entire process, and serves as a link between core planar cell polarity (PCP) factors and the E-cadherin-ß-catenin complex. To determine more precisely the role of nmo in ommatidial rotation, live-imaging analyses in nmo mutant and wild-type early pupal eye discs were performed. We demonstrate that ommatidial rotation is not a continuous process, and that rotating and non-rotating interommatidial cells are very dynamic. Our in vivo analyses also show that nmo regulates the speed of rotation and is required in cone cells for correct ommatidial rotation, and that these cells as well as interommatidial cells are less dynamic in nmo mutants. Furthermore, microarray analyses of nmo and wild-type larval eye discs led us to identify new genes and signaling pathways related to nmo function during this process. One of them, miple, encodes the Drosophila ortholog of the midkine/pleiotrophin secreted cytokines that are involved in cell migration processes. miple is highly up-regulated in nmo mutant discs. Indeed, phenotypic analyses reveal that miple overexpression leads to ommatidial rotation defects. Genetic interaction assays suggest that miple is signaling through Ptp99A, the Drosophila ortholog of the vertebrate midkine/pleiotrophin PTPζ receptor. Accordingly, we propose that one of the roles of Nmo during ommatial rotation is to repress miple expression, which may in turn affect the dynamics in E-cadherin-ß-catenin complexes.


Subject(s)
Body Patterning , Cytokines/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Eye/anatomy & histology , Eye/cytology , Gene Expression Regulation, Developmental , Mitogen-Activated Protein Kinases/metabolism , Animals , Body Patterning/genetics , Cadherins/metabolism , Cytokines/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Eye/metabolism , Eye/ultrastructure , Female , Gene Expression Profiling , Genetic Association Studies , Imaginal Discs/cytology , Imaginal Discs/metabolism , Imaginal Discs/ultrastructure , Imaging, Three-Dimensional , Midkine , Models, Biological , Mutation/genetics , Phenotype , Rotation , beta Catenin/metabolism
14.
Methods Enzymol ; 506: 407-13, 2012.
Article in English | MEDLINE | ID: mdl-22341235

ABSTRACT

Cell competition is a process in which cells with higher fitness ("winners") survive and proliferate at the expense of less fit neighbors ("losers"). It has been suggested that cell competition is involved in a variety of biological processes such as organ size control, tissue homeostasis, cancer progression, and the maintenance of stem cell population. By advent of a genetic mosaic technique, which enables to generate fluorescently marked somatic clones in Drosophila imaginal discs, recent studies have presented some aspects of molecular mechanisms underlying cell competition. Now, with a live-imaging technique using ex vivo-cultured imaginal discs, we can dissect the spatiotemporal nature of competitive cell behaviors within multicellular communities. Here, we describe procedures and tips for live imaging of cell competition in Drosophila imaginal discs.


Subject(s)
Drosophila/ultrastructure , Imaginal Discs/cytology , Imaginal Discs/ultrastructure , Microscopy, Confocal/methods , Animals , Cell Communication , Cell Culture Techniques , Cell Survival , Cells, Cultured , Drosophila/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...