Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Genes Dev ; 33(23-24): 1739-1750, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31753914

ABSTRACT

Proliferating cells, typically considered "nonexcitable," nevertheless, exhibit regulation by bioelectric signals. Notably, voltage-gated sodium channels (VGSC) that are crucial for neuronal excitability are also found in progenitors and up-regulated in cancer. Here, we identify a role for VGSC in proliferation of Drosophila neuroblast (NB) lineages within the central nervous system. Loss of paralytic (para), the sole gene that encodes Drosophila VGSC, reduces neuroblast progeny cell number. The type II neuroblast lineages, featuring a population of transit-amplifying intermediate neural progenitors (INP) similar to that found in the developing human cortex, are particularly sensitive to para manipulation. Following a series of asymmetric divisions, INPs normally exit the cell cycle through a final symmetric division. Our data suggests that loss of Para induces apoptosis in this population, whereas overexpression leads to an increase in INPs and overall neuroblast progeny cell numbers. These effects are cell autonomous and depend on Para channel activity. Reduction of Para expression not only affects normal NB development, but also strongly suppresses brain tumor mass, implicating a role for Para in cancer progression. To our knowledge, our studies are the first to identify a role for VGSC in neural progenitor proliferation. Elucidating the contribution of VGSC in proliferation will advance our understanding of bioelectric signaling within development and disease states.


Subject(s)
Cell Proliferation/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/genetics , Neural Stem Cells/cytology , Sodium Channels/genetics , Sodium Channels/metabolism , Animals , Apoptosis , Cell Count , Cell Lineage/genetics , Gene Expression , Gene Knockdown Techniques
2.
Proc Natl Acad Sci U S A ; 116(11): 5126-5134, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30804200

ABSTRACT

Sensory neurons perceive environmental cues and are important of organismal survival. Peripheral sensory neurons interact intimately with glial cells. While the function of axonal ensheathment by glia is well studied, less is known about the functional significance of glial interaction with the somatodendritic compartment of neurons. Herein, we show that three distinct glia cell types differentially wrap around the axonal and somatodendritic surface of the polymodal dendritic arborization (da) neuron of the Drosophila peripheral nervous system for detection of thermal, mechanical, and light stimuli. We find that glial cell-specific loss of the chromatin modifier gene dATRX in the subperineurial glial layer leads to selective elimination of somatodendritic glial ensheathment, thus allowing us to investigate the function of such ensheathment. We find that somatodendritic glial ensheathment regulates the morphology of the dendritic arbor, as well as the activity of the sensory neuron, in response to sensory stimuli. Additionally, glial ensheathment of the neuronal soma influences dendritic regeneration after injury.


Subject(s)
Dendrites/metabolism , Drosophila melanogaster/metabolism , Neuroglia/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Animals , Axons/metabolism , Axons/radiation effects , Caspases/metabolism , DNA Helicases/metabolism , Dendrites/radiation effects , Drosophila Proteins/metabolism , Enzyme Activation/radiation effects , Light , Neuroglia/radiation effects , Sensory Receptor Cells/radiation effects
3.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36239632

ABSTRACT

Membrane trafficking is essential for sculpting neuronal morphology. The GARP and EARP complexes are conserved tethers that regulate vesicle trafficking in the secretory and endolysosomal pathways, respectively. Both complexes contain the Vps51, Vps52, and Vps53 proteins, and a complex-specific protein: Vps54 in GARP and Vps50 in EARP. In Drosophila, we find that both complexes are required for dendrite morphogenesis during developmental remodeling of multidendritic class IV da (c4da) neurons. Having found that sterol accumulates at the trans-Golgi network (TGN) in Vps54KO/KO neurons, we investigated genes that regulate sterols and related lipids at the TGN. Overexpression of oxysterol binding protein (Osbp) or knockdown of the PI4K four wheel drive (fwd) exacerbates the Vps54KO/KO phenotype, whereas eliminating one allele of Osbp rescues it, suggesting that excess sterol accumulation at the TGN is, in part, responsible for inhibiting dendrite regrowth. These findings distinguish the GARP and EARP complexes in neurodevelopment and implicate vesicle trafficking and lipid transfer pathways in dendrite morphogenesis.


Subject(s)
Dendrites , Multiprotein Complexes , Vesicular Transport Proteins , trans-Golgi Network , Animals , Carrier Proteins , Dendrites/metabolism , Drosophila , Drosophila Proteins , Golgi Apparatus/metabolism , Multiprotein Complexes/metabolism , Receptors, Steroid , Sterols/metabolism , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism
4.
J Neurosci ; 31(9): 3309-19, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368042

ABSTRACT

Dendrites and axons are two major neuronal compartments with differences that are critical for neuronal functions. To learn about the differential regulation of dendritic and axonal development, we conducted a genetic screen in Drosophila and isolated the dendritic arbor reduction 1 (dar1) mutants, which display defects in dendritic but not axonal growth. The dar1 gene encodes a novel transcription regulator in the Krüppel-like factor family. Neurons lacking dar1 function have severely reduced growth of microtubule- but not F-actin-based dendritic branches. In contrast, overexpression of Dar1 dramatically increased the growth of microtubule-based dendritic branches. Our results suggest that Dar1 promotes dendrite growth in part by suppressing the expression of the microtubule-severing protein Spastin. Our study thus uncovers a novel transcriptional program for microtubule regulation that preferentially controls dendrite growth.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Axons/physiology , Dendrites/physiology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/physiology , Kruppel-Like Transcription Factors/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Neuropeptide/physiology , Adenosine Triphosphatases/biosynthesis , Adenosine Triphosphatases/genetics , Animals , Animals, Genetically Modified , Axons/ultrastructure , Dendrites/ultrastructure , Drosophila Proteins/biosynthesis , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Male , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/deficiency , Receptors, Neuropeptide/genetics
5.
Neuron ; 54(3): 403-16, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17481394

ABSTRACT

A neuron's dendrites typically do not cross one another. This intrinsic self-avoidance mechanism ensures unambiguous processing of sensory or synaptic inputs. Moreover, some neurons respect the territory of others of the same type, a phenomenon known as tiling. Different types of neurons, however, often have overlapping dendritic fields. We found that Down's syndrome Cell Adhesion Molecule (Dscam) is required for dendritic self-avoidance of all four classes of Drosophila dendritic arborization (da) neurons. However, neighboring mutant class IV da neurons still exhibited tiling, suggesting that self-avoidance and tiling differ in their recognition and repulsion mechanisms. Introducing 1 of the 38,016 Dscam isoforms to da neurons in Dscam mutants was sufficient to significantly restore self-avoidance. Remarkably, expression of a common Dscam isoform in da neurons of different classes prevented their dendrites from sharing the same territory, suggesting that coexistence of dendritic fields of different neuronal classes requires divergent expression of Dscam isoforms.


Subject(s)
Dendrites/physiology , Drosophila Proteins/physiology , Neurons, Afferent/physiology , Animals , Animals, Genetically Modified , Cell Adhesion Molecules , Cell Shape/physiology , Dendrites/ultrastructure , Drosophila , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Mutation/physiology , Neurons, Afferent/classification , Neurons, Afferent/cytology , Sense Organs/cytology , Staining and Labeling
6.
Neuron ; 51(3): 283-90, 2006 Aug 03.
Article in English | MEDLINE | ID: mdl-16880123

ABSTRACT

Ubiquitin-proteasome system (UPS) is a multistep protein degradation machinery implicated in many diseases. In the nervous system, UPS regulates remodeling and degradation of neuronal processes and is linked to Wallerian axonal degeneration, though the ubiquitin ligases that confer substrate specificity remain unknown. Having shown previously that class IV dendritic arborization (C4da) sensory neurons in Drosophila undergo UPS-mediated dendritic pruning during metamorphosis, we conducted an E2/E3 ubiquitinating enzyme mutant screen, revealing that mutation in ubcD1, an E2 ubiquitin-conjugating enzyme, resulted in retention of C4da neuron dendrites during metamorphosis. Further, we found that UPS activation likely leads to UbcD1-mediated degradation of DIAP1, a caspase-antagonizing E3 ligase. This allows for local activation of the Dronc caspase, thereby preserving C4da neurons while severing their dendrites. Thus, in addition to uncovering E2/E3 ubiquitinating enzymes for dendrite pruning, this study provides a mechanistic link between UPS and the apoptotic machinery in regulating neuronal process remodeling.


Subject(s)
Caspases/metabolism , Dendrites/enzymology , Drosophila Proteins/metabolism , Neurons, Afferent/enzymology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Caspases/genetics , Caspases/physiology , Dendrites/chemistry , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Neurons, Afferent/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics
8.
Neuron ; 89(4): 741-55, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26853303

ABSTRACT

Precise patterning of dendritic arbors is critical for the wiring and function of neural circuits. Dendrite-extracellular matrix (ECM) adhesion ensures that the dendrites of Drosophila dendritic arborization (da) sensory neurons are properly restricted in a 2D space, and thereby facilitates contact-mediated dendritic self-avoidance and tiling. However, the mechanisms regulating dendrite-ECM adhesion in vivo are poorly understood. Here, we show that mutations in the semaphorin ligand sema-2b lead to a dramatic increase in self-crossing of dendrites due to defects in dendrite-ECM adhesion, resulting in a failure to confine dendrites to a 2D plane. Furthermore, we find that Sema-2b is secreted from the epidermis and signals through the Plexin B receptor in neighboring neurons. Importantly, we find that Sema-2b/PlexB genetically and physically interacts with TORC2 complex, Tricornered (Trc) kinase, and integrins. These results reveal a novel role for semaphorins in dendrite patterning and illustrate how epidermal-derived cues regulate neural circuit assembly.


Subject(s)
Dendrites/physiology , Drosophila Proteins/metabolism , Epidermis/physiology , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Developmental/genetics , Semaphorins/metabolism , Sensory Receptor Cells/cytology , Animals , Animals, Genetically Modified , Cell Communication , Drosophila , Drosophila Proteins/genetics , Focal Adhesion Kinase 1/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Larva , Mechanistic Target of Rapamycin Complex 2 , Molecular Biology , Multiprotein Complexes/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Semaphorins/genetics , TOR Serine-Threonine Kinases/metabolism , Transfection
9.
Cancer Res ; 76(16): 4708-19, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325644

ABSTRACT

Proliferating cancer cells are characterized by high rates of glycolysis, lactate production, and altered mitochondrial metabolism. This metabolic reprogramming provides important metabolites for proliferation of tumor cells, including glioblastoma. These biological processes, however, generate oxidative stress that must be balanced through detoxification of reactive oxygen species (ROS). Using an unbiased retroviral loss-of-function screen in nontransformed human astrocytes, we demonstrate that mitochondrial PTEN-induced kinase 1 (PINK1) is a regulator of the Warburg effect and negative regulator of glioblastoma growth. We report that loss of PINK1 contributes to the Warburg effect through ROS-dependent stabilization of hypoxia-inducible factor-1A and reduced pyruvate kinase muscle isozyme 2 activity, both key regulators of aerobic glycolysis. Mechanistically, PINK1 suppresses ROS and tumor growth through FOXO3a, a master regulator of oxidative stress and superoxide dismutase 2. These findings highlight the importance of PINK1 and ROS balance in normal and tumor cells. PINK1 loss was observed in a significant number of human brain tumors including glioblastoma (n > 900) and correlated with poor patient survival. PINK1 overexpression attenuates in vivo glioblastoma growth in orthotopic mouse xenograft models and a transgenic glioblastoma model in Drosophila Cancer Res; 76(16); 4708-19. ©2016 AACR.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Animals , Astrocytes/metabolism , Blotting, Western , Cell Proliferation , Drosophila , Glycolysis/physiology , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Oxidative Stress/physiology
10.
Nat Neurosci ; 18(9): 1236-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258683

ABSTRACT

Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Drug Delivery Systems/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Evolution, Molecular , Thioridazine/administration & dosage , Animals , Brain Neoplasms/diagnosis , COS Cells , Chlorocebus aethiops , Drosophila , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods , Young Adult
11.
Cell Rep ; 9(4): 1446-58, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456135

ABSTRACT

A major gap in our understanding of sensation is how a single sensory neuron can differentially respond to a multitude of different stimuli (polymodality), such as propio- or nocisensation. The prevailing hypothesis is that different stimuli are transduced through ion channels with diverse properties and subunit composition. In a screen for ion channel genes expressed in polymodal nociceptive neurons, we identified Ppk26, a member of the trimeric degenerin/epithelial sodium channel (DEG/ENaC) family, as being necessary for proper locomotion behavior in Drosophila larvae in a mutually dependent fashion with coexpressed Ppk1, another member of the same family. Mutants lacking Ppk1 and Ppk26 were defective in mechanical, but not thermal, nociception behavior. Mutants of Piezo, a channel involved in mechanical nociception in the same neurons, did not show a defect in locomotion, suggesting distinct molecular machinery for mediating locomotor feedback and mechanical nociception.


Subject(s)
Behavior, Animal , Degenerin Sodium Channels/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Epithelial Sodium Channels/metabolism , Locomotion , Sodium Channels/metabolism , Animals , Cell Membrane/metabolism , Dendrites/metabolism , Mutation/genetics , Nociception , Protein Binding , Protein Subunits/metabolism , Temperature
12.
Elife ; 2: e00862, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24192034

ABSTRACT

TMEM16A and TMEM16B are calcium-activated chloride channels (CaCCs) with important functions in mammalian physiology. Whether distant relatives of the vertebrate TMEM16 families also form CaCCs is an intriguing open question. Here we report that a TMEM16 family member from Drosophila melanogaster, Subdued (CG16718), is a CaCC. Amino acid substitutions of Subdued alter the ion selectivity and kinetic properties of the CaCC channels heterologously expressed in HEK 293T cells. This Drosophila channel displays characteristics of classic CaCCs, thereby providing evidence for evolutionarily conserved biophysical properties in the TMEM16 family. Additionally, we show that knockout flies lacking subdued gene activity more readily succumb to death caused by ingesting the pathogenic bacteria Serratia marcescens, suggesting that subdued has novel functions in Drosophila host defense. DOI: http://dx.doi.org/10.7554/eLife.00862.001.


Subject(s)
Calcium/metabolism , Chloride Channels/physiology , Drosophila melanogaster/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calmodulin/physiology , Chloride Channels/chemistry , Chloride Channels/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid
13.
PLoS One ; 7(10): e46724, 2012.
Article in English | MEDLINE | ID: mdl-23056424

ABSTRACT

Neural stem cells (NSCs) are able to self-renew while giving rise to neurons and glia that comprise a functional nervous system. However, how NSC self-renewal is maintained is not well understood. Using the Drosophila larval NSCs called neuroblasts (NBs) as a model, we demonstrate that the Hairy and Enhancer-of-Split (Hes) family protein Deadpan (Dpn) plays important roles in NB self-renewal and specification. The loss of Dpn leads to the premature loss of NBs and truncated NB lineages, a process likely mediated by the homeobox protein Prospero (Pros). Conversely, ectopic/over-expression of Dpn promotes ectopic self-renewing divisions and maintains NB self-renewal into adulthood. In type II NBs, which generate transit amplifying intermediate neural progenitors (INPs) like mammalian NSCs, the loss of Dpn results in ectopic expression of type I NB markers Asense (Ase) and Pros before these type II NBs are lost at early larval stages. Our results also show that knockdown of Notch leads to ectopic Ase expression in type II NBs and the premature loss of type II NBs. Significantly, dpn expression is unchanged in these transformed NBs. Furthermore, the loss of Dpn does not inhibit the over-proliferation of type II NBs and immature INPs caused by over-expression of activated Notch. Our data suggest that Dpn plays important roles in maintaining NB self-renewal and specification of type II NBs in larval brains and that Dpn and Notch function independently in regulating type II NB proliferation and specification.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila Proteins/metabolism , Larva/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptors, Notch/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins , Drosophila , Drosophila Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Notch/genetics
14.
Nat Cell Biol ; 10(10): 1172-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18758451

ABSTRACT

Axons and dendrites differ in both microtubule organization and in the organelles and proteins they contain. Here we show that the microtubule motor dynein has a crucial role in polarized transport and in controlling the orientation of axonal microtubules in Drosophila melanogaster dendritic arborization (da) neurons. Changes in organelle distribution within the dendritic arbors of dynein mutant neurons correlate with a proximal shift in dendritic branch position. Dynein is also necessary for the dendrite-specific localization of Golgi outposts and the ion channel Pickpocket. Axonal microtubules are normally oriented uniformly plus-end-distal; however, without dynein, axons contain both plus- and minus-end distal microtubules. These data suggest that dynein is required for the distinguishing properties of the axon and dendrites: without dynein, dendritic organelles and proteins enter the axon and the axonal microtubules are no longer uniform in polarity.


Subject(s)
Axons/metabolism , Cell Polarity , Dendrites/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Dyneins/metabolism , Microtubules/metabolism , Animals , Biological Transport , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Endosomes/metabolism , Genes, Insect , Golgi Apparatus/metabolism , Green Fluorescent Proteins/metabolism , Male , Mutation/genetics , Recombinant Fusion Proteins/metabolism
15.
Cell ; 130(4): 717-29, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17719548

ABSTRACT

Little is known about how the distinct architectures of dendrites and axons are established. From a genetic screen, we isolated dendritic arbor reduction (dar) mutants with reduced dendritic arbors but normal axons of Drosophila neurons. We identified dar2, dar3, and dar6 genes as the homologs of Sec23, Sar1, and Rab1 of the secretory pathway. In both Drosophila and rodent neurons, defects in Sar1 expression preferentially affected dendritic growth, revealing evolutionarily conserved difference between dendritic and axonal development in the sensitivity to limiting membrane supply from the secretory pathway. Whereas limiting ER-to-Golgi transport resulted in decreased membrane supply from soma to dendrites, membrane supply to axons remained sustained. We also show that dendritic growth is contributed by Golgi outposts, which are found predominantly in dendrites. The distinct dependence between dendritic and axonal growth on the secretory pathway helps to establish different morphology of dendrites and axons.


Subject(s)
Axons/physiology , Dendrites/physiology , Drosophila/embryology , Drosophila/growth & development , Neural Pathways/metabolism , Animals , Cell Membrane/physiology , Cell Polarity , Cells, Cultured , Drosophila/genetics , Embryo, Nonmammalian , Exocytosis , Fluorescence Recovery After Photobleaching , Golgi Apparatus/physiology , Hippocampus/cytology , Mutation , Neurons/cytology , Neurons/metabolism , RNA, Small Interfering/metabolism , Transfection
16.
Development ; 134(1): 55-64, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164414

ABSTRACT

Neurons establish diverse dendritic morphologies during development, and a major challenge is to understand how these distinct developmental programs might relate to, and influence, neuronal function. Drosophila dendritic arborization (da) sensory neurons display class-specific dendritic morphology with extensive coverage of the body wall. To begin to build a basis for linking dendrite structure and function in this genetic system, we analyzed da neuron axon projections in embryonic and larval stages. We found that multiple parameters of axon morphology, including dorsoventral position, midline crossing and collateral branching, correlate with dendritic morphological class. We have identified a class-specific medial-lateral layering of axons in the central nervous system formed during embryonic development, which could allow different classes of da neurons to develop differential connectivity to second-order neurons. We have examined the effect of Robo family members on class-specific axon lamination, and have also taken a forward genetic approach to identify new genes involved in axon and dendrite development. For the latter, we screened the third chromosome at high resolution in vivo for mutations that affect class IV da neuron morphology. Several known loci, as well as putative novel mutations, were identified that contribute to sensory dendrite and/or axon patterning. This collection of mutants, together with anatomical data on dendrites and axons, should begin to permit studies of dendrite diversity in a combined developmental and functional context, and also provide a foundation for understanding shared and distinct mechanisms that control axon and dendrite morphology.


Subject(s)
Central Nervous System/physiology , Dendrites/physiology , Drosophila/growth & development , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Animals , Animals, Genetically Modified , Central Nervous System/embryology , Central Nervous System/metabolism , Dendrites/metabolism , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Embryo, Nonmammalian , Ethyl Methanesulfonate , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Larva/growth & development , Larva/metabolism , Morphogenesis , Mutagens , Mutation , Neurons, Afferent/classification
SELECTION OF CITATIONS
SEARCH DETAIL