Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
Add more filters

Publication year range
1.
Cell ; 162(2): 351-362, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186189

ABSTRACT

When navigating in their environment, animals use visual motion cues as feedback signals that are elicited by their own motion. Such signals are provided by wide-field neurons sampling motion directions at multiple image points as the animal maneuvers. Each one of these neurons responds selectively to a specific optic flow-field representing the spatial distribution of motion vectors on the retina. Here, we describe the discovery of a group of local, inhibitory interneurons in the fruit fly Drosophila key for filtering these cues. Using anatomy, molecular characterization, activity manipulation, and physiological recordings, we demonstrate that these interneurons convey direction-selective inhibition to wide-field neurons with opposite preferred direction and provide evidence for how their connectivity enables the computation required for integrating opposing motions. Our results indicate that, rather than sharpening directional selectivity per se, these circuit elements reduce noise by eliminating non-specific responses to complex visual information.


Subject(s)
Interneurons/cytology , Motion Perception , Neural Pathways , Optic Lobe, Nonmammalian/physiology , Visual Perception , Animals , Drosophila melanogaster , Interneurons/physiology , Optic Lobe, Nonmammalian/cytology , Synaptic Transmission
2.
Nature ; 629(8014): 1100-1108, 2024 May.
Article in English | MEDLINE | ID: mdl-38778103

ABSTRACT

The rich variety of behaviours observed in animals arises through the interplay between sensory processing and motor control. To understand these sensorimotor transformations, it is useful to build models that predict not only neural responses to sensory input1-5 but also how each neuron causally contributes to behaviour6,7. Here we demonstrate a novel modelling approach to identify a one-to-one mapping between internal units in a deep neural network and real neurons by predicting the behavioural changes that arise from systematic perturbations of more than a dozen neuronal cell types. A key ingredient that we introduce is 'knockout training', which involves perturbing the network during training to match the perturbations of the real neurons during behavioural experiments. We apply this approach to model the sensorimotor transformations of Drosophila melanogaster males during a complex, visually guided social behaviour8-11. The visual projection neurons at the interface between the optic lobe and central brain form a set of discrete channels12, and prior work indicates that each channel encodes a specific visual feature to drive a particular behaviour13,14. Our model reaches a different conclusion: combinations of visual projection neurons, including those involved in non-social behaviours, drive male interactions with the female, forming a rich population code for behaviour. Overall, our framework consolidates behavioural effects elicited from various neural perturbations into a single, unified model, providing a map from stimulus to neuronal cell type to behaviour, and enabling future incorporation of wiring diagrams of the brain15 into the model.


Subject(s)
Brain , Drosophila melanogaster , Models, Neurological , Neurons , Optic Lobe, Nonmammalian , Social Behavior , Visual Perception , Animals , Female , Male , Drosophila melanogaster/physiology , Drosophila melanogaster/cytology , Neurons/classification , Neurons/cytology , Neurons/physiology , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/physiology , Visual Perception/physiology , Nerve Net/cytology , Nerve Net/physiology , Brain/cytology , Brain/physiology
3.
Cell ; 158(5): 1173-1186, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171415

ABSTRACT

Temporal patterning of neural progenitors is one of the core mechanisms generating neuronal diversity in the central nervous system. Here, we show that, in the tips of the outer proliferation center (tOPC) of the developing Drosophila optic lobes, a unique temporal series of transcription factors not only governs the sequential production of distinct neuronal subtypes but also controls the mode of progenitor division, as well as the selective apoptosis of Notch(OFF) or Notch(ON) neurons during binary cell fate decisions. Within a single lineage, intermediate precursors initially do not divide and generate only one neuron; subsequently, precursors divide, but their Notch(ON) progeny systematically die through Reaper activity, whereas later, their Notch(OFF) progeny die through Hid activity. These mechanisms dictate how the tOPC produces neurons for three different optic ganglia. We conclude that temporal patterning generates neuronal diversity by specifying both the identity and survival/death of each unique neuronal subtype.


Subject(s)
Cell Survival , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Neurogenesis , Neuropeptides/metabolism , Optic Lobe, Nonmammalian/cytology , Receptors, Notch/metabolism , Animals , Apoptosis , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Neural Stem Cells , Optic Lobe, Nonmammalian/metabolism
4.
Nature ; 604(7905): 316-322, 2022 04.
Article in English | MEDLINE | ID: mdl-35388222

ABSTRACT

The brain consists of thousands of neuronal types that are generated by stem cells producing different neuronal types as they age. In Drosophila, this temporal patterning is driven by the successive expression of temporal transcription factors (tTFs)1-6. Here we used single-cell mRNA sequencing to identify the complete series of tTFs that specify most Drosophila optic lobe neurons. We verify that tTFs regulate the progression of the series by activating the next tTF(s) and repressing the previous one(s), and also identify more complex mechanisms of regulation. Moreover, we establish the temporal window of origin and birth order of each neuronal type in the medulla and provide evidence that these tTFs are sufficient to explain the generation of all of the neuronal diversity in this brain region. Finally, we describe the first steps of neuronal differentiation and show that these steps are conserved in humans. We find that terminal differentiation genes, such as neurotransmitter-related genes, are present as transcripts, but not as proteins, in immature larval neurons. This comprehensive analysis of a temporal series of tTFs in the optic lobe offers mechanistic insights into how tTF series are regulated, and how they can lead to the generation of a complete set of neurons.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Gene Expression Regulation, Developmental , Optic Lobe, Nonmammalian , Transcription Factors , Vision, Ocular , Visual Perception , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Optic Lobe, Nonmammalian/cytology , RNA-Seq , Single-Cell Analysis , Transcription Factors/metabolism
5.
Nature ; 589(7840): 88-95, 2021 01.
Article in English | MEDLINE | ID: mdl-33149298

ABSTRACT

Deciphering how neuronal diversity is established and maintained requires a detailed knowledge of neuronal gene expression throughout development. In contrast to mammalian brains1,2, the large neuronal diversity of the Drosophila optic lobe3 and its connectome4-6 are almost completely characterized. However, a molecular characterization of this neuronal diversity, particularly during development, has been lacking. Here we present insights into brain development through a nearly complete description of the transcriptomic diversity of the optic lobes of Drosophila. We acquired the transcriptome of 275,000 single cells at adult and at five pupal stages, and built a machine-learning framework to assign them to almost 200 cell types at all time points during development. We discovered two large neuronal populations that wrap neuropils during development but die just before adulthood, as well as neuronal subtypes that partition dorsal and ventral visual circuits by differential Wnt signalling throughout development. Moreover, we show that the transcriptomes of neurons that are of the same type but are produced days apart become synchronized shortly after their production. During synaptogenesis we also resolved neuronal subtypes that, although differing greatly in morphology and connectivity, converge to indistinguishable transcriptomic profiles in adults. Our datasets almost completely account for the known neuronal diversity of the Drosophila optic lobes, and serve as a paradigm to understand brain development across species.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Neurons/classification , Neurons/metabolism , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/growth & development , Anatomy, Artistic , Animals , Apoptosis , Atlases as Topic , Gene Expression Regulation, Developmental , Male , Neurons/cytology , Pupa/cytology , Pupa/growth & development , Single-Cell Analysis , Synapses/metabolism , Transcriptome/genetics , Visual Pathways , Wnt Signaling Pathway
6.
Nature ; 541(7637): 365-370, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28077877

ABSTRACT

In the Drosophila optic lobes, 800 retinotopically organized columns in the medulla act as functional units for processing visual information. The medulla contains over 80 types of neuron, which belong to two classes: uni-columnar neurons have a stoichiometry of one per column, while multi-columnar neurons contact multiple columns. Here we show that combinatorial inputs from temporal and spatial axes generate this neuronal diversity: all neuroblasts switch fates over time to produce different neurons; the neuroepithelium that generates neuroblasts is also subdivided into six compartments by the expression of specific factors. Uni-columnar neurons are produced in all spatial compartments independently of spatial input; they innervate the neuropil where they are generated. Multi-columnar neurons are generated in smaller numbers in restricted compartments and require spatial input; the majority of their cell bodies subsequently move to cover the entire medulla. The selective integration of spatial inputs by a fixed temporal neuroblast cascade thus acts as a powerful mechanism for generating neural diversity, regulating stoichiometry and the formation of retinotopy.


Subject(s)
Body Patterning , Cell Differentiation , Drosophila melanogaster/cytology , Neurogenesis , Neurons/cytology , Optic Lobe, Nonmammalian/cytology , Animals , Body Patterning/genetics , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Movement , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Male , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Neuropil/cytology , Neuropil/metabolism , Optic Lobe, Nonmammalian/growth & development , Optic Lobe, Nonmammalian/metabolism , Pupa/cytology , Pupa/genetics , Pupa/growth & development , Spatio-Temporal Analysis , Time Factors
7.
J Neurosci ; 40(29): 5561-5571, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32499380

ABSTRACT

All animals need information about the direction of motion to be able to track the trajectory of a target (prey, predator, cospecific) or to control the course of navigation. This information is provided by direction selective (DS) neurons, which respond to images moving in a unique direction. DS neurons have been described in numerous species including many arthropods. In these animals, the majority of the studies have focused on DS neurons dedicated to processing the optic flow generated during navigation. In contrast, only a few studies were performed on DS neurons related to object motion processing. The crab Neohelice is an established experimental model for the study of neurons involved in visually-guided behaviors. Here, we describe in male crabs of this species a new group of DS neurons that are highly directionally selective to moving objects. The neurons were physiologically and morphologically characterized by intracellular recording and staining in the optic lobe of intact animals. Because of their arborization in the lobula complex, we called these cells lobula complex directional cells (LCDCs). LCDCs also arborize in a previously undescribed small neuropil of the lateral protocerebrum. LCDCs are responsive only to horizontal motion. This nicely fits in the behavioral adaptations of a crab inhabiting a flat, densely crowded environment, where most object motions are generated by neighboring crabs moving along the horizontal plane.SIGNIFICANCE STATEMENT Direction selective (DS) neurons are key to a variety of visual behaviors including, target tracking (preys, predators, cospecifics) and course control. Here, we describe the physiology and morphology of a new group of remarkably directional neurons exclusively responsive to horizontal motion in crabs. These neurons arborize in the lobula complex and in a previously undescribed small neuropil of the lateral protocerebrum. The strong sensitivity of these cells for horizontal motion represents a clear example of functional neuronal adaptation to the lifestyle of an animal inhabiting a flat environment.


Subject(s)
Adaptation, Physiological , Brachyura/physiology , Motion Perception/physiology , Movement , Neurons/physiology , Action Potentials , Animals , Brachyura/cytology , Male , Neurons/cytology , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/physiology
8.
Dev Biol ; 458(1): 32-42, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31606342

ABSTRACT

The complexity of the nervous system requires the coordination of multiple cellular processes during development. Among them, we find boundary formation, axon guidance, cell migration and cell segregation. Understanding how different cell populations such as glial cells, developing neurons and neural stem cells contribute to the formation of boundaries and morphogenesis in the nervous system is a critical question in neurobiology. Slit is an evolutionary conserved protein essential for the development of the nervous system. For signaling, Slit has to bind to its cognate receptor Robo, a single-pass transmembrane protein. Although the Slit/Robo signaling pathway is well known for its involvement in axon guidance, it has also been associated to boundary formation in the Drosophila visual system. In the optic lobe, Slit is expressed in glial cells, positioned at the boundaries between developing neuropils, and in neurons of the medulla ganglia. Although it has been assumed that glial cells provide Slit to the system, the contribution of the neuronal expression has not been tested. Here, we show that, contrary to what was previously thought, Slit protein provided by medulla neurons is also required for boundary formation and morphogenesis of the optic lobe. Furthermore, tissue specific rescue using modified versions of Slit demonstrates that this protein acts at long range and does not require processing by extracellular proteases. Our data shed new light on our understanding of the cellular mechanisms involved in Slit function in the fly visual system morphogenesis.


Subject(s)
Axon Guidance/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Neuropil/physiology , Optic Lobe, Nonmammalian/growth & development , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Enhancer Elements, Genetic , Gene Knockdown Techniques , Genes, Reporter , Genetic Association Studies , Larva , Morphogenesis , Mutation , Nerve Tissue Proteins/genetics , Neuroglia/physiology , Neuropil/cytology , Optic Lobe, Nonmammalian/cytology , Organ Specificity , Phenotype , Photic Stimulation , Pupa , RNA Interference , Receptors, Immunologic/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Transgenes , Roundabout Proteins
9.
Development ; 145(18)2018 09 25.
Article in English | MEDLINE | ID: mdl-30254066

ABSTRACT

Neural stem cells must balance symmetric and asymmetric cell divisions to generate a functioning brain of the correct size. In both the developing Drosophila visual system and mammalian cerebral cortex, symmetrically dividing neuroepithelial cells transform gradually into asymmetrically dividing progenitors that generate neurons and glia. As a result, it has been widely accepted that stem cells in these tissues switch from a symmetric, expansive phase of cell divisions to a later neurogenic phase of cell divisions. In the Drosophila optic lobe, this switch is thought to occur during larval development. However, we have found that neuroepithelial cells start to produce neuroblasts during embryonic development, demonstrating a much earlier role for neuroblasts in the developing visual system. These neuroblasts undergo neurogenic divisions, enter quiescence and are retained post-embryonically, together with neuroepithelial cells. Later in development, neuroepithelial cells undergo further cell divisions before transforming into larval neuroblasts. Our results demonstrate that the optic lobe neuroepithelium gives rise to neurons and glia over 60 h earlier than was thought previously.


Subject(s)
Drosophila melanogaster/embryology , Neural Stem Cells/cytology , Neuroepithelial Cells/cytology , Neurogenesis/physiology , Optic Lobe, Nonmammalian/cytology , Animals , Cell Division , Neuroglia/cytology , Neurons/cytology
10.
Development ; 144(8): 1510-1517, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28242614

ABSTRACT

The Drosophila larval central nervous system comprises the central brain, ventral nerve cord and optic lobe. In these regions, neuroblasts (NBs) divide asymmetrically to self-renew and generate differentiated neurons or glia. To date, mechanisms of preventing neuron dedifferentiation are still unclear, especially in the optic lobe. Here, we show that the zinc-finger transcription factor Nerfin-1 is expressed in early-stage medulla neurons and is essential for maintaining their differentiation. Loss of Nerfin-1 activates Notch signaling, which promotes neuron-to-NB reversion. Repressing Notch signaling largely rescues dedifferentiation in nerfin-1 mutant clones. Thus, we conclude that Nerfin-1 represses Notch activity in medulla neurons and prevents them from dedifferentiation.


Subject(s)
Cell Differentiation , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Medulla Oblongata/cytology , Neurons/cytology , Neurons/metabolism , Receptors, Notch/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Dedifferentiation , Gene Knockdown Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Optic Lobe, Nonmammalian/anatomy & histology , Optic Lobe, Nonmammalian/cytology , Receptors, Notch/metabolism , Signal Transduction , Up-Regulation , Zinc Fingers
11.
Article in English | MEDLINE | ID: mdl-32088748

ABSTRACT

Praying mantids are the only insects proven to have stereoscopic vision (stereopsis): the ability to perceive depth from the slightly shifted images seen by the two eyes. Recently, the first neurons likely to be involved in mantis stereopsis were described and a speculative neuronal circuit suggested. Here we further investigate classes of neurons in the lobula complex of the praying mantis brain and their tuning to stereoscopically-defined depth. We used sharp electrode recordings with tracer injections to identify visual projection neurons with input in the optic lobe and output in the central brain. In order to measure binocular response fields of the cells the animals watched a vertical bar stimulus in a 3D insect cinema during recordings. We describe the binocular tuning of 19 neurons projecting from the lobula complex and the medulla to central brain areas. The majority of neurons (12/19) were binocular and had receptive fields for both eyes that overlapped in the frontal region. Thus, these neurons could be involved in mantis stereopsis. We also find that neurons preferring different contrast polarity (bright vs dark) tend to be segregated in the mantis lobula complex, reminiscent of the segregation for small targets and widefield motion in mantids and other insects.


Subject(s)
Brain/physiology , Depth Perception , Mantodea/physiology , Neurons/physiology , Optic Lobe, Nonmammalian/physiology , Vision, Binocular , Visual Fields , Animals , Brain/cytology , Evoked Potentials, Visual , Mantodea/cytology , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Visual Pathways/physiology
12.
Article in English | MEDLINE | ID: mdl-31989217

ABSTRACT

Many animals use motion vision information to control dynamic behaviors. For example, flying insects must decide whether to pursue a prey or not, to avoid a predator, to maintain their current flight trajectory, or to land. The neural mechanisms underlying the computation of visual motion have been particularly well investigated in the fly optic lobes. However, the descending neurons, which connect the optic lobes with the motor command centers of the ventral nerve cord, remain less studied. To address this deficiency, we describe motion vision sensitive descending neurons in the hoverfly Eristalis tenax. We describe how the neurons can be identified based on their receptive field properties, and how they respond to moving targets, looming stimuli and to widefield optic flow. We discuss their similarities with previously published visual neurons, in the optic lobes and ventral nerve cord, and suggest that they can be classified as target-selective, looming sensitive and optic flow sensitive, based on these similarities. Our results highlight the importance of using several visual stimuli as the neurons can rarely be identified based on only one response characteristic. In addition, they provide an understanding of the neurophysiology of visual neurons that are likely to affect behavior.


Subject(s)
Brain/physiology , Diptera/physiology , Motion Perception , Neurons/physiology , Optic Lobe, Nonmammalian/physiology , Vision, Ocular , Animals , Brain/cytology , Diptera/cytology , Optic Flow , Optic Lobe, Nonmammalian/cytology , Phenotype , Photic Stimulation , Visual Pathways/physiology
13.
Article in English | MEDLINE | ID: mdl-32020291

ABSTRACT

Color vision is an important sensory capability that enhances the detection of contrast in retinal images. Monochromatic animals exclusively detect temporal and spatial changes in luminance, whereas two or more types of photoreceptors and neuronal circuitries for the comparison of their responses enable animals to differentiate spectral information independent of intensity. Much of what we know about the cellular and physiological mechanisms underlying color vision comes from research on vertebrates including primates. In insects, many important discoveries have been made, but direct insights into the physiology and circuit implementation of color vision are still limited. Recent advances in Drosophila systems neuroscience suggest that a complete insect color vision circuitry, from photoreceptors to behavior, including all elements and computations, can be revealed in future. Here, we review fundamental concepts in color vision alongside our current understanding of the neuronal basis of color vision in Drosophila, including side views to selected other insects.


Subject(s)
Brain/physiology , Color Perception , Color Vision , Compound Eye, Arthropod/physiology , Drosophila melanogaster/physiology , Optic Lobe, Nonmammalian/physiology , Photoreceptor Cells, Invertebrate/physiology , Animals , Behavior, Animal , Brain/cytology , Compound Eye, Arthropod/cytology , Cues , Drosophila melanogaster/cytology , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Visual Pathways/physiology
14.
Article in English | MEDLINE | ID: mdl-31834470

ABSTRACT

The fruit fly Drosophila melanogaster can process chromatic information for true color vision and spectral preference. Spectral information is initially detected by a few distinct photoreceptor channels with different spectral sensitivities and is processed through the visual circuit. The neuroanatomical bases of the circuit are emerging. However, only little information is available in chromatic response properties of higher visual neurons from this important model organism. We used in vivo whole-cell patch-clamp recordings in response to monochromatic light stimuli ranging from 300 to 650 nm with 25-nm steps. We characterized the chromatic response of 33 higher visual neurons, including their general response type and their wavelength tuning. Color-opponent-type responses that had been typically observed in primates and bees were not identified. Instead, the majority of neurons showed excitatory responses to broadband wavelengths. The UV (300-375 nm) and middle wavelength (425-575 nm) ranges could be separated at the population level owing to neurons that preferentially responded to a specific wavelength range. Our results provide a first mapping of chromatic information processing in higher visual neurons of D. melanogaster that is a suitable model for exploring how color-opponent neural mechanisms are implemented in the visual circuits.


Subject(s)
Brain/physiology , Color Perception , Color Vision , Drosophila melanogaster/physiology , Neurons/physiology , Optic Lobe, Nonmammalian/physiology , Animals , Brain/cytology , Drosophila melanogaster/cytology , Evoked Potentials, Visual , Neural Inhibition , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Visual Pathways/physiology
15.
Article in English | MEDLINE | ID: mdl-31811397

ABSTRACT

Stomatopod crustaceans possess tripartite compound eyes; upper and lower hemispheres are separated by an equatorial midband of several ommatidial rows. The organization of stomatopod retinas is well established, but their optic lobes have been studied less. We used histological staining, immunolabeling, and fluorescent tracer injections to compare optic lobes in two 6-row midband species, Neogonodactylus oerstedii and Pseudosquilla ciliata, to those in two 2-row midband species, Squilla empusa and Alima pacifica. Compared to the 6-row species, we found structural differences in all optic neuropils in both 2-row species. Photoreceptor axons from 2-row midband ommatidia supply two sets of lamina cartridges; however, conspicuous spaces lacking lamina cartridges are observed in locations corresponding to where the cartridges of the upper four ommatidial rows of 6-row species would exist. The tripartite arrangement and enlarged projections containing fibers associated with the two rows of midband ommatidia can be traced throughout the entire optic lobe. However, 2-row species lack some features of medullar and lobular neuropils in 6-row species. Our results support the hypothesis that 2-row midband species are derived from a 6-row ancestor, and suggest specializations in the medulla and lobula found solely in 6-row species are important for color and polarization analysis.


Subject(s)
Brain/physiology , Compound Eye, Arthropod/physiology , Crustacea/physiology , Optic Lobe, Nonmammalian/physiology , Photoreceptor Cells, Invertebrate/physiology , Retina/physiology , Vision, Ocular , Visual Perception , Animals , Brain/cytology , Compound Eye, Arthropod/cytology , Crustacea/cytology , Neuroanatomical Tract-Tracing Techniques , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Retina/cytology , Visual Pathways/physiology
16.
Article in English | MEDLINE | ID: mdl-31811399

ABSTRACT

Specialized ommatidia harboring polarization-sensitive photoreceptors exist in the 'dorsal rim area' (DRA) of virtually all insects. Although downstream elements have been described both anatomically and physiologically throughout the optic lobes and the central brain of different species, little is known about their cellular and synaptic adaptations and how these shape their functional role in polarization vision. We have previously shown that in the DRA of Drosophila melanogaster, two distinct types of modality-specific 'distal medulla' cell types (Dm-DRA1 and Dm-DRA2) are post-synaptic to long visual fiber photoreceptors R7 and R8, respectively. Here we describe additional neuronal elements in the medulla neuropil that manifest modality-specific differences in the DRA region, including DRA-specific neuronal morphology, as well as differences in the structure of pre- or post-synaptic membranes. Furthermore, we show that certain cell types (medulla tangential cells and octopaminergic neuromodulatory cells) specifically avoid contacts with polarization-sensitive photoreceptors. Finally, while certain transmedullary cells are specifically absent from DRA medulla columns, other subtypes show specific wiring differences while still connecting the DRA to the lobula complex, as has previously been described in larger insects. This hints towards a complex circuit architecture with more than one pathway connecting polarization-sensitive DRA photoreceptors with the central brain.


Subject(s)
Brain/physiology , Drosophila melanogaster/metabolism , Optic Lobe, Nonmammalian/physiology , Photoreceptor Cells, Invertebrate/physiology , Synapses/physiology , Vision, Ocular , Visual Perception , Adaptation, Physiological , Animals , Animals, Genetically Modified , Brain/cytology , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Visual Pathways/physiology
17.
Article in English | MEDLINE | ID: mdl-31823004

ABSTRACT

The computational organization of sensory systems depends on the diversification of individual cell types with distinct signal-processing capabilities. The Drosophila visual system, for instance, splits information into channels with different temporal properties directly downstream of photoreceptors in the first-order interneurons of the OFF pathway, L2 and L3. However, the biophysical mechanisms that determine this specialization are largely unknown. Here, we show that the voltage-gated Ka channels Shaker and Shal contribute to the response properties of the major OFF pathway input L2. L3 calcium response kinetics postsynaptic to photoreceptors resemble the sustained calcium signals of photoreceptors, whereas L2 neurons decay transiently. Based on a cell-type-specific RNA-seq data set and endogenous protein tagging, we identified Shaker and Shal as the primary candidates to shape L2 responses. Using in vivo two-photon imaging of L2 calcium signals in combination with pharmacological and genetic perturbations of these Ka channels, we show that the wild-type Shaker and Shal function is to enhance L2 responses and cell-autonomously sharpen L2 kinetics. Our results reveal a role for Ka channels in determining the signal-processing characteristics of a specific cell type in the visual system.


Subject(s)
Brain/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Interneurons/metabolism , Optic Lobe, Nonmammalian/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Shaker Superfamily of Potassium Channels/metabolism , Shal Potassium Channels/metabolism , Vision, Ocular , Animals , Animals, Genetically Modified , Brain/cytology , Calcium Channels, L-Type/metabolism , Calcium Signaling , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Evoked Potentials, Visual , Kinetics , Optic Lobe, Nonmammalian/cytology , Photic Stimulation , Shaker Superfamily of Potassium Channels/genetics , Shal Potassium Channels/genetics , Visual Pathways/metabolism , Visual Perception
18.
Dev Biol ; 436(1): 28-41, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29447906

ABSTRACT

Programmed cell death is a conserved strategy for neural development both in vertebrates and invertebrates and is recognized at various developmental stages in the brain from neurogenesis to adulthood. To understand the development of the central nervous system, it is essential to reveal not only molecular mechanisms but also the role of neural cell death (Pinto-Teixeira et al., 2016). To understand the role of cell death in neural development, we investigated the effect of inhibition of cell death on optic lobe development. Our data demonstrate that, in the optic lobe of Drosophila, cell death occurs in neural precursor cells and neurons before neurite formation and functions to prevent various developmental abnormalities. When neuronal cell death was inhibited by an effector caspase inhibitor, p35, multiple abnormal neuropil structures arose during optic lobe development-e.g., enlarged or fused neuropils, misrouted neurons and abnormal neurite lumps. Inhibition of cell death also induced morphogenetic defects in the lamina and medulla development-e.g., failures in the separation of the lamina and medulla cortices and the medulla rotation. These defects were reproduced in the mutant of an initiator caspase, dronc. If cell death was a mechanism for removing the abnormal neuropil structures, we would also expect to observe them in mutants defective for corpse clearance. However, they were not observed in these mutants. When dead cell-membranes were visualized with Apoliner, they were observed only in cortices and not in neuropils. These results suggest that the cell death occurs before mature neurite formation. Moreover, we found that inhibition of cell death induced ectopic neuroepithelial cells, neuroblasts and ganglion mother cells in late pupal stages, at sites where the outer and inner proliferation centers were located at earlier developmental stages. Caspase-3 activation was observed in the neuroepithelial cells and neuroblasts in the proliferation centers. These results indicate that cell death is required for elimination of the precursor cells composing the proliferation centers. This study substantiates an essential role of early neural cell death for ensuring normal development of the central nervous system.


Subject(s)
Apoptosis/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Optic Lobe, Nonmammalian/physiology , Animals , Caspases/metabolism , Drosophila , Drosophila Proteins/metabolism , Immunohistochemistry , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurites/physiology , Neurons/metabolism , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/metabolism , Signal Transduction/physiology
19.
Development ; 143(13): 2431-42, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27381228

ABSTRACT

Differences in neuroepithelial patterning and neurogenesis modes contribute to area-specific diversifications of neural circuits. In the Drosophila visual system, two neuroepithelia, the outer (OPC) and inner (IPC) proliferation centers, generate neuron subtypes for four ganglia in several ways. Whereas neuroepithelial cells in the medial OPC directly convert into neuroblasts, in an IPC subdomain they generate migratory progenitors by epithelial-mesenchymal transition that mature into neuroblasts in a second proliferative zone. The molecular mechanisms that regulate the identity of these neuroepithelia, including their neurogenesis modes, remain poorly understood. Analysis of Polycomblike revealed that loss of Polycomb group-mediated repression of the Hox gene Abdominal-B (Abd-B) caused the transformation of OPC to IPC neuroepithelial identity. This suggests that the neuroepithelial default state is IPC-like, whereas OPC identity is derived. Ectopic Abd-B blocks expression of the highly conserved retinal determination gene network members Eyes absent (Eya), Sine oculis (So) and Homothorax (Hth). These factors are essential for OPC specification and neurogenesis control. Finally, eya and so are also sufficient to confer OPC-like identity, and, in parallel with hth, the OPC-specific neurogenesis mode on the IPC.


Subject(s)
Body Patterning/genetics , Drosophila melanogaster/genetics , Genes, Insect , Neuroepithelial Cells/metabolism , Neurogenesis/genetics , Optic Lobe, Nonmammalian/embryology , Optic Lobe, Nonmammalian/metabolism , Retina/embryology , Animals , Cell Differentiation/genetics , Cell Proliferation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Embryo, Nonmammalian/metabolism , Epithelial-Mesenchymal Transition/genetics , Genetic Testing , Mutation/genetics , Neuroepithelial Cells/cytology , Optic Lobe, Nonmammalian/cytology , Retina/metabolism , Stem Cells/cytology
20.
Dev Biol ; 431(2): 101-110, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28958816

ABSTRACT

Diversification of neuronal types is key to establishing functional variations in neural circuits. The first critical step to generate neuronal diversity is to organize the compartmental domains of developing brains into spatially distinct neural progenitor pools. Neural progenitors in each pool then generate a unique set of diverse neurons through specific spatiotemporal specification processes. In this review article, we focus on an additional mechanism, 'inter-progenitor pool wiring', that further expands the diversity of neural circuits. After diverse types of neurons are generated in one progenitor pool, a fraction of these neurons start migrating toward a remote brain region containing neurons that originate from another progenitor pool. Finally, neurons of different origins are intermingled and eventually form complex but precise neural circuits. The developing cerebral cortex of mammalian brains is one of the best examples of inter-progenitor pool wiring. However, Drosophila visual system development has revealed similar mechanisms in invertebrate brains, suggesting that inter-progenitor pool wiring is an evolutionarily conserved strategy that expands neural circuit diversity. Here, we will discuss how inter-progenitor pool wiring is accomplished in mammalian and fly brain systems.


Subject(s)
Biological Evolution , Neural Stem Cells/cytology , Neurons/cytology , Animals , Cerebral Cortex/cytology , Drosophila/cytology , Optic Lobe, Nonmammalian/cytology
SELECTION OF CITATIONS
SEARCH DETAIL