Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Neuron ; 112(6): 942-958.e13, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38262414

ABSTRACT

Neurons express various combinations of neurotransmitter receptor (NR) subunits and receive inputs from multiple neuron types expressing different neurotransmitters. Localizing NR subunits to specific synaptic inputs has been challenging. Here, we use epitope-tagged endogenous NR subunits, expansion light-sheet microscopy, and electron microscopy (EM) connectomics to molecularly characterize synapses in Drosophila. We show that in directionally selective motion-sensitive neurons, different multiple NRs elaborated a highly stereotyped molecular topography with NR localized to specific domains receiving cell-type-specific inputs. Developmental studies suggested that NRs or complexes of them with other membrane proteins determine patterns of synaptic inputs. In support of this model, we identify a transmembrane protein selectively associated with a subset of spatially restricted synapses and demonstrate its requirement for synapse formation through genetic analysis. We propose that mechanisms that regulate the precise spatial distribution of NRs provide a molecular cartography specifying the patterns of synaptic connections onto dendrites.


Subject(s)
Connectome , Synapses/physiology , Motor Neurons/metabolism , Microscopy, Electron , Receptors, GABA-A/metabolism
2.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873314

ABSTRACT

Neurons express different combinations of neurotransmitter receptor (NR) subunits and receive inputs from multiple neuron types expressing different neurotransmitters. Localizing NR subunits to specific synaptic inputs has been challenging. Here we use epitope tagged endogenous NR subunits, expansion light-sheet microscopy, and EM connectomics to molecularly characterize synapses in Drosophila. We show that in directionally selective motion sensitive neurons, different multiple NRs elaborated a highly stereotyped molecular topography with NR localized to specific domains receiving cell-type specific inputs. Developmental studies suggested that NRs or complexes of them with other membrane proteins determines patterns of synaptic inputs. In support of this model, we identify a transmembrane protein associated selectively with a subset of spatially restricted synapses and demonstrate through genetic analysis its requirement for synapse formation. We propose that mechanisms which regulate the precise spatial distribution of NRs provide a molecular cartography specifying the patterns of synaptic connections onto dendrites.

3.
Curr Biol ; 33(18): 3998-4005.e6, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37647901

ABSTRACT

Advances in brain connectomics have demonstrated the extraordinary complexity of neural circuits.1,2,3,4,5 Developing neurons encounter the axons and dendrites of many different neuron types and form synapses with only a subset of them. During circuit assembly, neurons express cell-type-specific repertoires comprising many cell adhesion molecules (CAMs) that can mediate interactions between developing neurites.6,7,8 Many CAM families have been shown to contribute to brain wiring in different ways.9,10 It has been challenging, however, to identify receptor-ligand pairs directly matching neurons with their synaptic targets. Here, we integrated the synapse-level connectome of the neural circuit11,12 with the developmental expression patterns7 and binding specificities of CAMs6,13 on pre- and postsynaptic neurons in the Drosophila visual system. To overcome the complexity of neural circuits, we focus on pairs of genetically related neurons that make differential wiring choices. In the motion detection circuit,14 closely related subtypes of T4/T5 neurons choose between alternative synaptic targets in adjacent layers of neuropil.12 This choice correlates with the matching expression in synaptic partners of different receptor-ligand pairs of the Beat and Side families of CAMs. Genetic analysis demonstrated that presynaptic Side-II and postsynaptic Beat-VI restrict synaptic partners to the same layer. Removal of this receptor-ligand pair disrupts layers and leads to inappropriate targeting of presynaptic sites and postsynaptic dendrites. We propose that different Side/Beat receptor-ligand pairs collaborate with other recognition molecules to determine wiring specificities in the fly brain. Combining transcriptomes, connectomes, and protein interactome maps allow unbiased identification of determinants of brain wiring.


Subject(s)
Connectome , Animals , Transcriptome , Ligands , Neurons/physiology , Drosophila/genetics , Drosophila/metabolism , Brain/metabolism , Synapses/physiology , Cell Adhesion Molecules/metabolism
5.
Nature ; 613(7944): 534-542, 2023 01.
Article in English | MEDLINE | ID: mdl-36599984

ABSTRACT

To survive, animals must convert sensory information into appropriate behaviours1,2. Vision is a common sense for locating ethologically relevant stimuli and guiding motor responses3-5. How circuitry converts object location in retinal coordinates to movement direction in body coordinates remains largely unknown. Here we show through behaviour, physiology, anatomy and connectomics in Drosophila that visuomotor transformation occurs by conversion of topographic maps formed by the dendrites of feature-detecting visual projection neurons (VPNs)6,7 into synaptic weight gradients of VPN outputs onto central brain neurons. We demonstrate how this gradient motif transforms the anteroposterior location of a visual looming stimulus into the fly's directional escape. Specifically, we discover that two neurons postsynaptic to a looming-responsive VPN type promote opposite takeoff directions. Opposite synaptic weight gradients onto these neurons from looming VPNs in different visual field regions convert localized looming threats into correctly oriented escapes. For a second looming-responsive VPN type, we demonstrate graded responses along the dorsoventral axis. We show that this synaptic gradient motif generalizes across all 20 primary VPN cell types and most often arises without VPN axon topography. Synaptic gradients may thus be a general mechanism for conveying spatial features of sensory information into directed motor outputs.


Subject(s)
Behavior, Animal , Drosophila , Neurons , Psychomotor Performance , Synapses , Animals , Brain/cytology , Brain/physiology , Drosophila/anatomy & histology , Drosophila/cytology , Drosophila/physiology , Neurons/physiology , Visual Fields/physiology , Synapses/metabolism , Axons , Dendrites , Escape Reaction
6.
bioRxiv ; 2023 Dec 27.
Article in English | MEDLINE | ID: mdl-38234787

ABSTRACT

To visualize the cellular and subcellular localization of neuromodulatory G-protein coupled receptors (GPCRs) in Drosophila , we implement a molecular strategy recently used to add epitope tags to ionotropic receptors at their endogenous loci. Leveraging evolutionary conservation to identify sites more likely to permit insertion of a tag, we generated constitutive and conditional tagged alleles for Drosophila 5-HT1A, 5-HT2A, 5-HT2B, Octß1R, Octß2R, two isoforms of OAMB, and mGluR. The conditional alleles allow for the restricted expression of tagged receptor in specific cell types, an option not available for any previous reagents to label these proteins. We show that 5-HT1A and 5-HT2B localize to the mushroom bodies and central complex respectively, as predicted by their roles in sleep. By contrast, the unexpected enrichment of Octß1R in the central complex and of 5-HT1A and 5-HT2A to nerve terminals in lobular columnar cells in the visual system suggest new hypotheses about their function at these sites. Using an additional tagged allele of the serotonin transporter, a marker of serotonergic tracts, we demonstrate diverse spatial relationships between postsynaptic 5-HT receptors and presynaptic 5-HT neurons, consistent with the importance of both synaptic and volume transmission. Finally, we use the conditional allele of 5-HT1A to show that it localizes to distinct sites within the mushroom bodies as both a postsynaptic receptor in Kenyon cells and a presynaptic autoreceptor. Significance Statement: In Drosophila , despite remarkable advances in both connectomic and genomic studies, antibodies to many aminergic GPCRs are not available. We have overcome this obstacle using evolutionary conservation to identify loci in GPCRs amenable to epitope-tagging, and CRISPR/Cas9 genome editing to generated eight novel lines. This method also may be applied to other GPCRs and allows cell-specific expression of the tagged locus. We have used the tagged alleles we generated to address several questions that remain poorly understood. These include the relationship between pre- and post-synaptic sites that express the same receptor, and the use of relatively distant targets by pre-synaptic release sites that may employ volume transmission as well as standard synaptic signaling.

7.
bioRxiv ; 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38187533

ABSTRACT

We previously reported that vision specifies Layer 2/3 (L2/3) glutamatergic cell-type identity in the primary visual cortex (V1). Using unsupervised clustering of single-nucleus RNA-sequencing data, we identified molecularly distinct L2/3 cell types in normal-reared (NR) and dark-reared (DR) mice, but the two sets exhibited poor correspondence. Here, we show that classification of cell types was confounded in DR by vision-dependent gene programs that are orthogonal to gene programs underlying cell-type identity. A focused clustering analysis successfully matches cell types between DR and NR, suggesting that cell identity-defining gene programs persist under vision deprivation but are overshadowed by vision-dependent transcriptomic variation. Using multi-tasking theory we show that L2/3 cell types form a continuum between three cell-archetypes. Visual deprivation markedly shifts this distribution along the continuum. Thus, dark-rearing markedly influences cell states thereby masking cell-type-identities and changes the distribution of L2/3 types along a transcriptomic continuum.

8.
Cell Rep ; 39(1): 110618, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385751

ABSTRACT

Neurons in the developing brain express many different cell adhesion molecules (CAMs) on their surfaces. CAM-binding affinities can vary by more than 200-fold, but the significance of these variations is unknown. Interactions between the immunoglobulin superfamily CAM DIP-α and its binding partners, Dpr10 and Dpr6, control synaptic targeting and survival of Drosophila optic lobe neurons. We design mutations that systematically change interaction affinity and analyze function in vivo. Reducing affinity causes loss-of-function phenotypes whose severity scales with the magnitude of the change. Synaptic targeting is more sensitive to affinity reduction than is cell survival. Increasing affinity rescues neurons that would normally be culled by apoptosis. By manipulating CAM expression together with affinity, we show that the key parameter controlling circuit assembly is surface avidity, which is the strength of adherence between cell surfaces. We conclude that CAM binding affinities and expression levels are finely tuned for function during development.


Subject(s)
Drosophila Proteins , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Survival , Drosophila/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Neurons/metabolism
9.
Nature ; 603(7899): 112-118, 2022 03.
Article in English | MEDLINE | ID: mdl-35197627

ABSTRACT

The assembly of neural circuits is dependent on precise spatiotemporal expression of cell recognition molecules1-5. Factors controlling cell type specificity have been identified6-8, but how timing is determined remains unknown. Here we describe induction of a cascade of transcription factors by a steroid hormone (ecdysone) in all fly visual system neurons spanning target recognition and synaptogenesis. We demonstrate through single-cell sequencing that the ecdysone pathway regulates the expression of a common set of targets required for synaptic maturation and cell-type-specific targets enriched for cell-surface proteins regulating wiring specificity. Transcription factors in the cascade regulate the expression of the same wiring genes in complex ways, including activation in one cell type and repression in another. We show that disruption of the ecdysone pathway generates specific defects in dendritic and axonal processes and synaptic connectivity, with the order of transcription factor expression correlating with sequential steps in wiring. We also identify shared targets of a cell-type-specific transcription factor and the ecdysone pathway that regulate specificity. We propose that neurons integrate a global temporal transcriptional module with cell-type-specific transcription factors to generate different cell-type-specific patterns of cell recognition molecules regulating wiring.


Subject(s)
Drosophila Proteins , Ecdysone , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Ecdysone/metabolism , Gene Expression Regulation , Synapses/metabolism , Transcription Factors/metabolism
10.
Cell ; 185(2): 311-327.e24, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35063073

ABSTRACT

The role of postnatal experience in sculpting cortical circuitry, while long appreciated, is poorly understood at the level of cell types. We explore this in the mouse primary visual cortex (V1) using single-nucleus RNA sequencing, visual deprivation, genetics, and functional imaging. We find that vision selectively drives the specification of glutamatergic cell types in upper layers (L) (L2/3/4), while deeper-layer glutamatergic, GABAergic, and non-neuronal cell types are established prior to eye opening. L2/3 cell types form an experience-dependent spatial continuum defined by the graded expression of ∼200 genes, including regulators of cell adhesion and synapse formation. One of these genes, Igsf9b, a vision-dependent gene encoding an inhibitory synaptic cell adhesion molecule, is required for the normal development of binocular responses in L2/3. In summary, vision preferentially regulates the development of upper-layer glutamatergic cell types through the regulation of cell-type-specific gene expression programs.


Subject(s)
Vision, Ocular , Visual Cortex/cytology , Visual Cortex/embryology , Animals , Animals, Newborn , Biomarkers/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glutamic Acid/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , RNA-Seq , Transcriptome/genetics , Vision, Binocular/genetics , gamma-Aminobutyric Acid/metabolism
11.
Curr Biol ; 31(19): 4305-4313.e5, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34411526

ABSTRACT

Depth perception emerges from the development of binocular neurons in primary visual cortex. Vision is required for these neurons to acquire their mature responses to visual stimuli. The prevailing view is that vision does not influence binocular circuitry until the onset of the critical period, about a week after eye opening, and that plasticity of visual responses is triggered by increased inhibition. Here, we show that vision is required to form binocular neurons and to improve binocular tuning and matching from eye opening until critical period closure. Enhancing inhibition does not accelerate this process. Vision soon after eye opening improves the tuning properties of binocular neurons by strengthening and sharpening ipsilateral eye cortical responses. This progressively changes the population of neurons in the binocular pool, and this plasticity is sensitive to interocular differences prior to critical period onset. Thus, vision establishes binocular circuitry and guides binocular plasticity from eye opening.


Subject(s)
Visual Cortex , Neurons/physiology , Photic Stimulation , Vision, Binocular/physiology , Vision, Ocular , Visual Cortex/physiology
12.
Neuron ; 108(6): 1045-1057.e6, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33125872

ABSTRACT

Precise patterns of synaptic connections between neurons are encoded in their genetic programs. Here, we use single-cell RNA sequencing to profile neuronal transcriptomes at multiple stages in the developing Drosophila visual system. We devise an efficient strategy for profiling neurons at multiple time points in a single pool, thereby minimizing batch effects and maximizing the reliability of time-course data. A transcriptional atlas spanning multiple stages is generated, including more than 150 distinct neuronal populations; of these, 88 are followed through synaptogenesis. This analysis reveals a common (pan-neuronal) program unfolding in highly coordinated fashion in all neurons, including genes encoding proteins comprising the core synaptic machinery and membrane excitability. This program is overlaid by cell-type-specific programs with diverse cell recognition molecules expressed in different combinations and at different times. We propose that a pan-neuronal program endows neurons with the competence to form synapses and that cell-type-specific programs control synaptic specificity.


Subject(s)
Drosophila/physiology , Neurogenesis/physiology , Neurons/physiology , Visual Pathways/physiology , Animals , Axons/physiology , Synapses/physiology , Transcriptome
13.
Neuron ; 108(4): 735-747.e6, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33091339

ABSTRACT

High acuity stereopsis emerges during an early postnatal critical period when binocular neurons in the primary visual cortex sharpen their receptive field tuning properties. We find that this sharpening is achieved by dismantling the binocular circuit present at critical period onset and building it anew. Longitudinal imaging of receptive field tuning (e.g., orientation selectivity) of thousands of neurons reveals that most binocular neurons present in layer 2/3 at critical period onset are poorly tuned and are rendered monocular. In parallel, new binocular neurons are established by conversion of well-tuned monocular neurons as they gain matched input from the other eye. These improvements in binocular tuning in layer 2/3 are not inherited from layer 4 but are driven by the experience-dependent sharpening of ipsilateral eye responses. Thus, vision builds a new and more sharply tuned binocular circuit in layer 2/3 by cellular exchange and not by refining the original circuit.


Subject(s)
Critical Period, Psychological , Vision, Binocular/physiology , Visual Cortex/physiology , Visual Pathways/physiology , Animals , Female , Male , Mice , Mice, Transgenic , Neurons/physiology , Orientation/physiology , Photic Stimulation , Vision, Monocular/physiology
14.
Neuron ; 108(1): 111-127.e6, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32795398

ABSTRACT

Cajal recognized that the elaborate shape of neurons is fundamental to their function in the brain. However, there are no simple and generalizable genetic methods to study neuronal or glial cell morphology in the mammalian brain. Here, we describe four mouse lines conferring Cre-dependent sparse cell labeling based on mononucleotide repeat frameshift (MORF) as a stochastic translational switch. Notably, the optimized MORF3 mice, with a membrane-bound multivalent immunoreporter, confer Cre-dependent sparse and bright labeling of thousands of neurons, astrocytes, or microglia in each brain, revealing their intricate morphologies. MORF3 mice are compatible with imaging in tissue-cleared thick brain sections and with immuno-EM. An analysis of 151 MORF3-labeled developing retinal horizontal cells reveals novel morphological cell clusters and axonal maturation patterns. Our study demonstrates a conceptually novel, simple, generalizable, and scalable mouse genetic solution to sparsely label and illuminate the morphology of genetically defined neurons and glia in the mammalian brain.


Subject(s)
Astrocytes/ultrastructure , Brain/ultrastructure , Microglia/ultrastructure , Neurons/ultrastructure , Retinal Horizontal Cells/ultrastructure , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Frameshift Mutation/genetics , Green Fluorescent Proteins/genetics , Integrases , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Microsatellite Repeats/genetics , Neurons/metabolism , Neurons/pathology , Retinal Horizontal Cells/metabolism , Retinal Horizontal Cells/pathology
15.
Curr Opin Genet Dev ; 65: 8-13, 2020 12.
Article in English | MEDLINE | ID: mdl-32593792

ABSTRACT

It has long been appreciated that activity sculpts the formation of neuronal circuits in the mammalian brain. By contrast, in Drosophila it was generally thought that genetically hardwired mechanisms of development, mediated by cell recognition molecules, are sufficient to specify the connectome-the complex and stereotyped pattern of connections between neurons. Here, we consider recent findings that activity during development also contributes to circuit assembly in the fly. These observations suggest that activity is a fundamental aspect of brain development in general. They also highlight the opportunities to learn about the role of this phenomenon through the extensive Drosophila toolkit for examining circuit development, structure, and function.


Subject(s)
Brain/growth & development , Connectome , Drosophila/physiology , Neurons/cytology , Synapses/physiology , Animals , Cell Communication
16.
Cell ; 181(6): 1434-1435, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32531247
17.
Cell ; 181(3): 536-556, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32359437

ABSTRACT

Developing neurons connect in specific and stereotyped ways to form the complex circuits that underlie brain function. By comparison to earlier steps in neural development, progress has been slow in identifying the cell surface recognition molecules that mediate these synaptic choices, but new high-throughput imaging, genetic, and molecular methods are accelerating progress. Over the past decade, numerous large and small gene families have been implicated in target recognition, including members of the immunoglobulin, cadherin, and leucine-rich repeat superfamilies. We review these advances and propose ways in which combinatorial use of multifunctional recognition molecules enables the complex neuron-neuron interactions that underlie synaptic specificity.


Subject(s)
Nerve Net/metabolism , Nerve Net/physiology , Synapses/metabolism , Animals , Cadherins/genetics , Cell Communication , Cell Membrane/metabolism , Humans , Neurogenesis , Neurons/metabolism , Neurons/physiology , Synapses/physiology
18.
Elife ; 82019 11 05.
Article in English | MEDLINE | ID: mdl-31687928

ABSTRACT

Patterns of synaptic connectivity are remarkably precise and complex. Single-cell RNA sequencing has revealed a vast transcriptional diversity of neurons. Nevertheless, a clear logic underlying the transcriptional control of neuronal connectivity has yet to emerge. Here, we focused on Drosophila T4/T5 neurons, a class of closely related neuronal subtypes with different wiring patterns. Eight subtypes of T4/T5 neurons are defined by combinations of two patterns of dendritic inputs and four patterns of axonal outputs. Single-cell profiling during development revealed distinct transcriptional programs defining each dendrite and axon wiring pattern. These programs were defined by the expression of a few transcription factors and different combinations of cell surface proteins. Gain and loss of function studies provide evidence for independent control of different wiring features. We propose that modular transcriptional programs for distinct wiring features are assembled in different combinations to generate diverse patterns of neuronal connectivity.


Subject(s)
Axons/physiology , Dendrites/physiology , Neural Conduction , Transcription, Genetic , Animals , Cells, Cultured , Drosophila , Gene Expression Regulation , Single-Cell Analysis
19.
Neuron ; 101(5): 894-904.e5, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30711355

ABSTRACT

Stereotyped synaptic connections define the neural circuits of the brain. In vertebrates, stimulus-independent activity contributes to neural circuit formation. It is unknown whether this type of activity is a general feature of nervous system development. Here, we report patterned, stimulus-independent neural activity in the Drosophila visual system during synaptogenesis. Using in vivo calcium, voltage, and glutamate imaging, we found that all neurons participate in this spontaneous activity, which is characterized by brain-wide periodic active and silent phases. Glia are active in a complementary pattern. Each of the 15 of over 100 specific neuron types in the fly visual system examined exhibited a unique activity signature. The activity of neurons that are synaptic partners in the adult was highly correlated during development. We propose that this cell-type-specific activity coordinates the development of the functional circuitry of the adult brain.


Subject(s)
Action Potentials , Neurogenesis , Photoreceptor Cells, Invertebrate/cytology , Synapses/physiology , Synaptic Potentials , Animals , Calcium/metabolism , Drosophila melanogaster , Glutamic Acid/metabolism , Neuroglia/cytology , Neuroglia/physiology , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/physiology , Visual Pathways/cytology , Visual Pathways/metabolism , Visual Pathways/physiology
20.
Neuron ; 100(6): 1369-1384.e6, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30467079

ABSTRACT

Drosophila Dpr (21 paralogs) and DIP proteins (11 paralogs) are cell recognition molecules of the immunoglobulin superfamily (IgSF) that form a complex protein interaction network. DIP and Dpr proteins are expressed in a synaptic layer-specific fashion in the visual system. How interactions between these proteins regulate layer-specific synaptic circuitry is not known. Here we establish that DIP-α and its interacting partners Dpr6 and Dpr10 regulate multiple processes, including arborization within layers, synapse number, layer specificity, and cell survival. We demonstrate that heterophilic binding between Dpr6/10 and DIP-α and homophilic binding between DIP-α proteins promote interactions between processes in vivo. Knockin mutants disrupting the DIP/Dpr binding interface reveal a role for these proteins during normal development, while ectopic expression studies support an instructive role for interactions between DIPs and Dprs in circuit development. These studies support an important role for the DIP/Dpr protein interaction network in regulating cell-type-specific connectivity patterns.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Neuropil/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Drosophila , Drosophila Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Medulla Oblongata/cytology , Medulla Oblongata/growth & development , Mutation/genetics , Protein Interaction Maps , Surface Plasmon Resonance , Transcription Factors/genetics , Transfection , Visual Pathways/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...