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










Publication year range
1.
Elife ; 122023 03 28.
Article in English | MEDLINE | ID: mdl-36975203

ABSTRACT

How does wiring specificity of neural maps emerge during development? Formation of the adult Drosophila olfactory glomerular map begins with the patterning of projection neuron (PN) dendrites at the early pupal stage. To better understand the origin of wiring specificity of this map, we created genetic tools to systematically characterize dendrite patterning across development at PN type-specific resolution. We find that PNs use lineage and birth order combinatorially to build the initial dendritic map. Specifically, birth order directs dendrite targeting in rotating and binary manners for PNs of the anterodorsal and lateral lineages, respectively. Two-photon- and adaptive optical lattice light-sheet microscope-based time-lapse imaging reveals that PN dendrites initiate active targeting with direction-dependent branch stabilization on the timescale of seconds. Moreover, PNs that are used in both the larval and adult olfactory circuits prune their larval-specific dendrites and re-extend new dendrites simultaneously to facilitate timely olfactory map organization. Our work highlights the power and necessity of type-specific neuronal access and time-lapse imaging in identifying wiring mechanisms that underlie complex patterns of functional neural maps.


The brain's ability to sense, act and remember relies on the intricate network of connections between neurons. Organization of these connections into neural maps is critical for processing sensory information. For instance, different odors are represented by specific neurons in a part of the brain known as the olfactory bulb, allowing animals to distinguish between smells. Projection neurons in the olfactory bulb have extensions known as dendrites that receive signals from sensory neurons. Scientists have extensively used the olfactory map in adult fruit flies to study brain wiring because of the specific connections between their sensory and projection neurons. This has led to the discovery of similar wiring strategies in mammals. But how the olfactory map is formed during development is not fully understood. To investigate, Wong et al. built genetic tools to label specific types of olfactory projection neurons during the pupal stage of fruit fly development. This showed that a group of projection neurons directed their dendrites in a clockwise rotation pattern depending on the order in which they were born: the first-born neuron sent dendrites towards the top right of the antennal lobe (the fruit fly equivalent of the olfactory bulb), while the last-born sent dendrites towards the top left. Wong et al. also carried out high-resolution time-lapse imaging of live brains grown in the laboratory to determine how dendrites make wiring decisions. This revealed that projection neurons send dendrites in all directions, but preferentially stabilize those that extend in the direction which the neurons eventually target. Also, live imaging showed neurons could remove old dendrites (used in the larvae) and build new ones (to be used in the adult) simultaneously, allowing them to quickly create new circuits. These experiments demonstrate the value of imaging specific types of neurons to understand the mechanisms that assemble neural maps in the developing brain. Further work could use the genetic tools created by Wong et al. to study how wiring decisions are determined in this and other neural maps by specific genes, potentially yielding insights into neurological disorders associated with wiring defects.


Subject(s)
Drosophila Proteins , Olfactory Receptor Neurons , Animals , Drosophila melanogaster/genetics , Olfactory Pathways , Olfactory Receptor Neurons/physiology , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Time-Lapse Imaging , Drosophila/metabolism
2.
Neuron ; 110(23): 3882-3896.e9, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36220098

ABSTRACT

Cell-surface proteins (CSPs) mediate intercellular communication throughout the lives of multicellular organisms. However, there are no generalizable methods for quantitative CSP profiling in specific cell types in vertebrate tissues. Here, we present in situ cell-surface proteome extraction by extracellular labeling (iPEEL), a proximity labeling method in mice that enables spatiotemporally precise labeling of cell-surface proteomes in a cell-type-specific environment in native tissues for discovery proteomics. Applying iPEEL to developing and mature cerebellar Purkinje cells revealed differential enrichment in CSPs with post-translational protein processing and synaptic functions in the developing and mature cell-surface proteomes, respectively. A proteome-instructed in vivo loss-of-function screen identified a critical, multifaceted role for Armh4 in Purkinje cell dendrite morphogenesis. Armh4 overexpression also disrupts dendrite morphogenesis; this effect requires its conserved cytoplasmic domain and is augmented by disrupting its endocytosis. Our results highlight the utility of CSP profiling in native mammalian tissues for identifying regulators of cell-surface signaling.


Subject(s)
Mammals , Proteomics , Mice , Animals
3.
Neuron ; 110(14): 2299-2314.e8, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35613619

ABSTRACT

Transcription factors specify the fate and connectivity of developing neurons. We investigate how a lineage-specific transcription factor, Acj6, controls the precise dendrite targeting of Drosophila olfactory projection neurons (PNs) by regulating the expression of cell-surface proteins. Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains, and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion molecules and proteins previously not associated with wiring, such as Piezo, whose mechanosensitive ion channel activity is dispensable for its function in PN dendrite targeting. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combined expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, Acj6 controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell-surface executors.


Subject(s)
Drosophila Proteins , Olfactory Receptor Neurons , Animals , Dendrites/physiology , Drosophila/metabolism , Drosophila Proteins/metabolism , Ion Channels/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Olfactory Pathways/physiology , Olfactory Receptor Neurons/metabolism , POU Domain Factors/metabolism , Proteomics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Sci Adv ; 8(7): eabm7723, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35179957

ABSTRACT

Variations in neuronal connectivity occur widely in nervous systems from invertebrates to mammals. Yet, it is unclear how neuronal variability originates, to what extent and at what time scales it exists, and what functional consequences it might carry. To assess inter- and intraindividual neuronal variability, it would be ideal to analyze the same identified neuron across different brain hemispheres and individuals. Here, using genetic labeling and electron microscopy connectomics, we show that an identified inhibitory olfactory local interneuron, TC-LN, exhibits extraordinary variability in its glomerular innervation patterns. Moreover, TC-LN's innervation of the VL2a glomerulus, which processes food signals and modulates mating behavior, is sexually dimorphic, is influenced by female's courtship experience, and correlates with food intake in mated females. Mating also affects output connectivity of TC-LN to specific local interneurons. We propose that mating-associated variability of TC-LNs regulates how food odor is interpreted by an inhibitory network to modulate feeding.

5.
Cell ; 184(20): 5107-5121.e14, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34551316

ABSTRACT

Neural circuit assembly features simultaneous targeting of numerous neuronal processes from constituent neuron types, yet the dynamics is poorly understood. Here, we use the Drosophila olfactory circuit to investigate dynamic cellular processes by which olfactory receptor neurons (ORNs) target axons precisely to specific glomeruli in the ipsi- and contralateral antennal lobes. Time-lapse imaging of individual axons from 30 ORN types revealed a rich diversity in extension speed, innervation timing, and ipsilateral branch locations and identified that ipsilateral targeting occurs via stabilization of transient interstitial branches. Fast imaging using adaptive optics-corrected lattice light-sheet microscopy showed that upon approaching target, many ORN types exhibiting "exploring branches" consisted of parallel microtubule-based terminal branches emanating from an F-actin-rich hub. Antennal nerve ablations uncovered essential roles for bilateral axons in contralateral target selection and for ORN axons to facilitate dendritic refinement of postsynaptic partner neurons. Altogether, these observations provide cellular bases for wiring specificity establishment.


Subject(s)
Olfactory Pathways/cytology , Olfactory Pathways/diagnostic imaging , Time-Lapse Imaging , Animals , Axons/physiology , Cells, Cultured , Dendrites/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Microtubules/metabolism , Olfactory Receptor Neurons/physiology , Time Factors
6.
Elife ; 102021 01 11.
Article in English | MEDLINE | ID: mdl-33427646

ABSTRACT

Neurons undergo substantial morphological and functional changes during development to form precise synaptic connections and acquire specific physiological properties. What are the underlying transcriptomic bases? Here, we obtained the single-cell transcriptomes of Drosophila olfactory projection neurons (PNs) at four developmental stages. We decoded the identity of 21 transcriptomic clusters corresponding to 20 PN types and developed methods to match transcriptomic clusters representing the same PN type across development. We discovered that PN transcriptomes reflect unique biological processes unfolding at each stage-neurite growth and pruning during metamorphosis at an early pupal stage; peaked transcriptomic diversity during olfactory circuit assembly at mid-pupal stages; and neuronal signaling in adults. At early developmental stages, PN types with adjacent birth order share similar transcriptomes. Together, our work reveals principles of cellular diversity during brain development and provides a resource for future studies of neural development in PNs and other neuronal types.


Subject(s)
Drosophila melanogaster/metabolism , Neurites/metabolism , Olfactory Nerve/metabolism , Transcriptome , Animals , Single-Cell Analysis , Time Factors
7.
Neuron ; 109(4): 629-644.e8, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33352118

ABSTRACT

The synaptotrophic hypothesis posits that synapse formation stabilizes dendritic branches, but this hypothesis has not been causally tested in vivo in the mammalian brain. The presynaptic ligand cerebellin-1 (Cbln1) and postsynaptic receptor GluD2 mediate synaptogenesis between granule cells and Purkinje cells in the molecular layer of the cerebellar cortex. Here we show that sparse but not global knockout of GluD2 causes under-elaboration of Purkinje cell dendrites in the deep molecular layer and overelaboration in the superficial molecular layer. Developmental, overexpression, structure-function, and genetic epistasis analyses indicate that these dendrite morphogenesis defects result from a deficit in Cbln1/GluD2-dependent competitive interactions. A generative model of dendrite growth based on competitive synaptogenesis largely recapitulates GluD2 sparse and global knockout phenotypes. Our results support the synaptotrophic hypothesis at initial stages of dendrite development, suggest a second mode in which cumulative synapse formation inhibits further dendrite growth, and highlight the importance of competition in dendrite morphogenesis.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Dendrites/metabolism , Nerve Tissue Proteins/deficiency , Protein Precursors/deficiency , Purkinje Cells/metabolism , Receptors, Glutamate/deficiency , Animals , Dendrites/genetics , Female , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Pregnancy , Protein Binding/physiology , Protein Precursors/genetics , Receptors, Glutamate/genetics
8.
Curr Biol ; 30(7): 1189-1198.e5, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32059767

ABSTRACT

The regulatory mechanisms by which neurons coordinate their physiology and connectivity are not well understood. The Drosophila olfactory receptor neurons (ORNs) provide an excellent system to investigate this question. Each ORN type expresses a unique olfactory receptor, or a combination thereof, and sends their axons to a stereotyped glomerulus. Using single-cell RNA sequencing, we identified 33 transcriptomic clusters for ORNs and mapped 20 to their glomerular types, demonstrating that transcriptomic clusters correspond well with anatomically and physiologically defined ORN types. Each ORN type expresses hundreds of transcription factors. Transcriptome-instructed genetic analyses revealed that (1) one broadly expressed transcription factor (Acj6) only regulates olfactory receptor expression in one ORN type and only wiring specificity in another type, (2) one type-restricted transcription factor (Forkhead) only regulates receptor expression, and (3) another type-restricted transcription factor (Unplugged) regulates both events. Thus, ORNs utilize diverse strategies and complex regulatory networks to coordinate their physiology and connectivity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Forkhead Transcription Factors/genetics , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Olfactory Receptor Neurons/physiology , POU Domain Factors/genetics , Receptors, Odorant/genetics , Transcriptome , Animals , Axons/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Receptors, Odorant/metabolism , Single-Cell Analysis , Smell/physiology
9.
Cell ; 180(2): 373-386.e15, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31955847

ABSTRACT

Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.


Subject(s)
Olfactory Pathways/metabolism , Olfactory Receptor Neurons/metabolism , Proteomics/methods , Animals , Axons/metabolism , Brain/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Neurogenesis/physiology , Olfactory Nerve/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Receptors, Lipoprotein/metabolism , Smell/physiology
10.
Proc Natl Acad Sci U S A ; 116(32): 16068-16073, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31341080

ABSTRACT

Our understanding of the mechanisms of neural circuit assembly is far from complete. Identification of wiring molecules with novel mechanisms of action will provide insights into how complex and heterogeneous neural circuits assemble during development. In the Drosophila olfactory system, 50 classes of olfactory receptor neurons (ORNs) make precise synaptic connections with 50 classes of partner projection neurons (PNs). Here, we performed an RNA interference screen for cell surface molecules and identified the leucine-rich repeat-containing transmembrane protein known as Fish-lips (Fili) as a novel wiring molecule in the assembly of the Drosophila olfactory circuit. Fili contributes to the precise axon and dendrite targeting of a small subset of ORN and PN classes, respectively. Cell-type-specific expression and genetic analyses suggest that Fili sends a transsynaptic repulsive signal to neurites of nonpartner classes that prevents their targeting to inappropriate glomeruli in the antennal lobe.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Olfactory Receptor Neurons/metabolism , Signal Transduction , Synapses/metabolism , Animals , Axons/metabolism , Dendrites/metabolism , Leucine-Rich Repeat Proteins , Mutation/genetics , Phenotype , Proteins/metabolism
11.
Elife ; 82019 06 21.
Article in English | MEDLINE | ID: mdl-31225795

ABSTRACT

Plexins exhibit multitudinous, evolutionarily conserved functions in neural development. How Plexins employ their diverse structural motifs in vivo to perform distinct roles is unclear. We previously reported that Plexin B (PlexB) controls multiple steps during the assembly of the Drosophila olfactory circuit (Li et al., 2018b). Here, we systematically mutagenized structural motifs of PlexB and examined the function of these variants in these multiple steps: axon fasciculation, trajectory choice, and synaptic partner selection. We found that the extracellular Sema domain is essential for all three steps, the catalytic site of the intracellular RapGAP is engaged in none, and the intracellular GTPase-binding motifs are essential for trajectory choice and synaptic partner selection, but are dispensable for fasciculation. Moreover, extracellular PlexB cleavage serves as a regulatory mechanism of PlexB signaling. Thus, the divergent roles of PlexB motifs in distinct steps of neural development contribute to its functional versatility in neural circuit assembly.


Subject(s)
Drosophila Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Receptors, Cell Surface/genetics , Smell/genetics , Animals , Animals, Genetically Modified/genetics , Axons/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Semaphorins/genetics , Signal Transduction/genetics , Smell/physiology
12.
Nat Commun ; 10(1): 2113, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068592

ABSTRACT

Gene editing by CRISPR/Cas9 is commonly used to generate germline mutations or perform in vitro screens, but applicability for in vivo screening has so far been limited. Recently, it was shown that in Drosophila, Cas9 expression could be limited to a desired group of cells, allowing tissue-specific mutagenesis. Here, we thoroughly characterize tissue-specific (ts)CRISPR within the complex neuronal system of the Drosophila mushroom body. We report the generation of a library of gRNA-expressing plasmids and fly lines using optimized tools, which provides a valuable resource to the fly community. We demonstrate the application of our library in a large-scale in vivo screen, which reveals insights into developmental neuronal remodeling.


Subject(s)
Animals, Genetically Modified/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drosophila/genetics , Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , Female , Male , Mushroom Bodies/metabolism , Mutagenesis , Nervous System/growth & development , Neuronal Plasticity/genetics , Neurons/physiology , Plasmids/genetics , RNA, Guide, Kinetoplastida/genetics
13.
Elife ; 72018 08 23.
Article in English | MEDLINE | ID: mdl-30136927

ABSTRACT

The precise assembly of a neural circuit involves many consecutive steps. The conflict between a limited number of wiring molecules and the complexity of the neural network impels each molecule to execute multiple functions at different steps. Here, we examined the cell-type specific distribution of endogenous levels of axon guidance receptor Plexin B (PlexB) in the developing antennal lobe, the first olfactory processing center in Drosophila. We found that different classes of olfactory receptor neurons (ORNs) express PlexB at different levels in two wiring steps - axonal trajectory choice and subsequent target selection. In line with its temporally distinct patterns, the proper levels of PlexB control both steps in succession. Genetic interactions further revealed that the effect of high-level PlexB is antagonized by its canonical partner Sema2b. Thus, PlexB plays a multifaceted role in instructing the assembly of the Drosophila olfactory circuit through temporally-regulated expression patterns and expression level-dependent effects.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Smell/physiology , Animals , Arthropod Antennae/innervation , Arthropod Antennae/physiology , Axons/physiology , Models, Biological , Olfactory Receptor Neurons/physiology
14.
Cell ; 171(5): 1206-1220.e22, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149607

ABSTRACT

The definition of neuronal type and how it relates to the transcriptome are open questions. Drosophila olfactory projection neurons (PNs) are among the best-characterized neuronal types: different PN classes target dendrites to distinct olfactory glomeruli, while PNs of the same class exhibit indistinguishable anatomical and physiological properties. Using single-cell RNA sequencing, we comprehensively characterized the transcriptomes of most PN classes and unequivocally mapped transcriptomes to specific olfactory function for six classes. Transcriptomes of closely related PN classes exhibit the largest differences during circuit assembly but become indistinguishable in adults, suggesting that neuronal subtype diversity peaks during development. Transcription factors and cell-surface molecules are the most differentially expressed genes between classes and are highly informative in encoding cell identity, enabling us to identify a new lineage-specific transcription factor that instructs PN dendrite targeting. These findings establish that neuronal transcriptomic identity corresponds with anatomical and physiological identity defined by connectivity and function.


Subject(s)
Drosophila melanogaster/metabolism , Neurons/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Brain/cytology , Brain/metabolism , Cluster Analysis , Dendrites/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Gene Expression Profiling , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Organ Specificity , Pupa/cytology , Pupa/metabolism , Transcription Factors/metabolism
15.
Proc Natl Acad Sci U S A ; 114(29): 7505-7512, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28674010

ABSTRACT

The formation of complex but highly organized neural circuits requires interactions between neurons and glia. During the assembly of the Drosophila olfactory circuit, 50 olfactory receptor neuron (ORN) classes and 50 projection neuron (PN) classes form synaptic connections in 50 glomerular compartments in the antennal lobe, each of which represents a discrete olfactory information-processing channel. Each compartment is separated from the adjacent compartments by membranous processes from ensheathing glia. Here we show that Thisbe, an FGF released from olfactory neurons, particularly from local interneurons, instructs ensheathing glia to wrap each glomerulus. The Heartless FGF receptor acts cell-autonomously in ensheathing glia to regulate process extension so as to insulate each neuropil compartment. Overexpressing Thisbe in ORNs or PNs causes overwrapping of the glomeruli their axons or dendrites target. Failure to establish the FGF-dependent glia structure disrupts precise ORN axon targeting and discrete glomerular formation.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Fibroblast Growth Factors/physiology , Neuroglia/physiology , Olfactory Bulb/physiology , Signal Transduction , Animals , Animals, Genetically Modified , Axons/physiology , Genotype , Microscopy, Confocal , Neuropil/physiology , Olfactory Pathways/physiology , Olfactory Receptor Neurons/physiology
16.
Elife ; 62017 06 13.
Article in English | MEDLINE | ID: mdl-28606304

ABSTRACT

Precise coordination of synaptic connections ensures proper information flow within circuits. The activity of presynaptic organizing molecules signaling to downstream pathways is essential for such coordination, though such entities remain incompletely known. We show that LRP4, a conserved transmembrane protein known for its postsynaptic roles, functions presynaptically as an organizing molecule. In the Drosophila brain, LRP4 localizes to the nerve terminals at or near active zones. Loss of presynaptic LRP4 reduces excitatory (not inhibitory) synapse number, impairs active zone architecture, and abolishes olfactory attraction - the latter of which can be suppressed by reducing presynaptic GABAB receptors. LRP4 overexpression increases synapse number in excitatory and inhibitory neurons, suggesting an instructive role and a common downstream synapse addition pathway. Mechanistically, LRP4 functions via the conserved kinase SRPK79D to ensure normal synapse number and behavior. This highlights a presynaptic function for LRP4, enabling deeper understanding of how synapse organization is coordinated.


Subject(s)
Brain/physiology , LDL-Receptor Related Proteins/metabolism , Neuronal Plasticity , Neurons/physiology , Presynaptic Terminals/physiology , Receptors, Presynaptic/metabolism , Animals , Drosophila , Gene Expression , Gene Knockout Techniques
17.
Nat Methods ; 12(3): 219-22, 5 p following 222, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25581800

ABSTRACT

The Q system is a repressible binary expression system for transgenic manipulations in living organisms. Through protein engineering and in vivo functional tests, we report here variants of the Q-system transcriptional activator, including QF2, for driving strong and ubiquitous expression in all Drosophila tissues. Our QF2, Gal4QF and LexAQF chimeric transcriptional activators substantially enrich the toolkit available for transgenic regulation in Drosophila melanogaster.


Subject(s)
Drosophila melanogaster/genetics , Genetic Engineering/methods , Trans-Activators/genetics , Transgenes , Animals , Animals, Genetically Modified , Behavior, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian , Female , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Larva , Male , Promoter Regions, Genetic , Sleep/genetics , Transcription Factors/genetics
18.
Dev Cell ; 14(2): 227-38, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18267091

ABSTRACT

Developmental axon pruning is widely used to refine neural circuits. We performed a mosaic screen to identify mutations affecting axon pruning of Drosophila mushroom body gamma neurons. We constructed a modified piggyBac vector with improved mutagenicity and generated insertions in >2000 genes. We identified two cohesin subunits (SMC1 and SA) as being essential for axon pruning. The cohesin complex maintains sister-chromatid cohesion during cell division in eukaryotes. However, we show that the pruning phenotype in SMC1(-/-) clones is rescued by expressing SMC1 in neurons, revealing a postmitotic function. SMC1(-/-) clones exhibit reduced levels of the ecdysone receptor EcR-B1, a key regulator of axon pruning. The pruning phenotype is significantly suppressed by overexpressing EcR-B1 and is enhanced by a reduced dose of EcR, supporting a causal relationship. We also demonstrate a postmitotic role for SMC1 in dendrite targeting of olfactory projection neurons. We suggest that cohesin regulates diverse aspects of neuronal morphogenesis.


Subject(s)
Axons/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Transposable Elements/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Mitosis , Mosaicism , Nuclear Proteins/metabolism , Alleles , Animals , Cell Proliferation , Dendrites/metabolism , Drosophila Proteins/metabolism , Genetic Markers , Mushroom Bodies/cytology , Mutagenesis, Insertional , Mutation/genetics , Olfactory Pathways/metabolism , Phenotype , Receptors, Steroid/metabolism , Transgenes , Cohesins
19.
Nat Neurosci ; 10(7): 828-37, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17529987

ABSTRACT

We identified a mutation in Aats-gly (also known as gars or glycyl-tRNA synthetase), the Drosophila melanogaster ortholog of the human GARS gene that is associated with Charcot-Marie-Tooth neuropathy type 2D (CMT2D), from a mosaic genetic screen. Loss of gars in Drosophila neurons preferentially affects the elaboration and stability of terminal arborization of axons and dendrites. The human and Drosophila genes each encode both a cytoplasmic and a mitochondrial isoform. Using additional mutants that selectively disrupt cytoplasmic or mitochondrial protein translation, we found that cytoplasmic protein translation is required for terminal arborization of both dendrites and axons during development. In contrast, disruption of mitochondrial protein translation preferentially affects the maintenance of dendritic arborization in adults. We also provide evidence that human GARS shows equivalent functions in Drosophila, and that CMT2D causal mutations show loss-of-function properties. Our study highlights different demands of protein translation for the development and maintenance of axons and dendrites.


Subject(s)
Axons/physiology , Cytoplasm/metabolism , Dendrites/physiology , Glycine-tRNA Ligase/genetics , Glycine-tRNA Ligase/physiology , Mitochondria/metabolism , Protein Biosynthesis/physiology , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA/genetics , Drosophila , Genetic Vectors , Humans , Mushroom Bodies/physiology , Mushroom Bodies/ultrastructure , Mutation, Missense/physiology , Neurons/physiology , Neurons/ultrastructure , Phenotype , Point Mutation/genetics , Point Mutation/physiology
20.
Mol Cell Neurosci ; 31(1): 123-30, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16298140

ABSTRACT

Midline glia are a source of cues for neuronal navigation and differentiation in the Drosophila CNS. Despite their importance, how glia and neurons communicate during the development is not fully understood. Here, we examined dynamic morphology of midline glia and assessed their direct cellular interactions with neurons within the embryonic CNS. Midline glia extend filopodia-like "gliopodia" from the onset of axogenesis through the near completion of embryonic neural development. The most abundant and stable within the commissures, gliopodia frequently contact neurites extending from the neuropil on either side of the midline. Misexpression of Rac1N17 in midline glia not only reduces the number of gliopodia but also shifts the position of neuropils towards the midline. Midline-secreted signaling protein Slit accumulates along the surface of gliopodia. Mutant analysis supports the idea that gliopodia contribute to its presentation on neuronal surfaces at both the commissures and neuropils. We propose that gliopodia extend the range of direct glia-neuron communication during CNS development.


Subject(s)
Neuroglia/physiology , Neurons/physiology , Animals , Cell Movement , Drosophila , Nervous System Physiological Phenomena , Neuroglia/cytology , Neurons/cytology , Pseudopodia/physiology , Pseudopodia/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...