Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 497
Filter
1.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34183440

ABSTRACT

Like other sensory systems, the visual system is topographically organized: Its sensory neurons, the photoreceptors, and their targets maintain point-to-point correspondence in physical space, forming a retinotopic map. The iterative wiring of circuits in the visual system conveniently facilitates the study of its development. Over the past few decades, experiments in Drosophila have shed light on the principles that guide the specification and connectivity of visual system neurons. In this review, we describe the main findings unearthed by the study of the Drosophila visual system and compare them with similar events in mammals. We focus on how temporal and spatial patterning generates diverse cell types, how guidance molecules distribute the axons and dendrites of neurons within the correct target regions, how vertebrates and invertebrates generate their retinotopic map, and the molecules and mechanisms required for neuronal migration. We suggest that basic principles used to wire the fly visual system are broadly applicable to other systems and highlight its importance as a model to study nervous system development.


Subject(s)
Body Patterning , Drosophila melanogaster/embryology , Nerve Net/embryology , Neurons/physiology , Visual Pathways/embryology , Animals , Cell Adhesion Molecules/metabolism , Cell Movement , Mammals/embryology , Neuroglia/cytology , Receptors, Notch/metabolism , Signal Transduction , Time Factors
2.
Science ; 371(6525)2021 01 08.
Article in English | MEDLINE | ID: mdl-33414193

ABSTRACT

The ability to perceive and interact with the world depends on a diverse array of neural circuits specialized for carrying out specific computations. Each circuit is assembled using a relatively limited number of molecules and common developmental steps, from cell fate specification to activity-dependent synaptic refinement. Given this shared toolkit, how do individual circuits acquire their characteristic properties? We explore this question by comparing development of the circuitry for seeing and hearing, highlighting a few examples where differences in each system's sensory demands necessitate different developmental strategies.


Subject(s)
Auditory Pathways/embryology , Cochlear Nucleus/embryology , Neurogenesis , Retina/embryology , Visual Pathways/embryology , Animals , Hearing/physiology , Mice , Sensory Receptor Cells/ultrastructure , Synapses/ultrastructure , Vision, Ocular/physiology
3.
Elife ; 92020 09 08.
Article in English | MEDLINE | ID: mdl-32896272

ABSTRACT

Ascending visual projections similar to the mammalian thalamocortical pathway are found in a wide range of vertebrate species, but their homology is debated. To get better insights into their evolutionary origin, we examined the developmental origin of a thalamic-like sensory structure of teleosts, the preglomerular complex (PG), focusing on the visual projection neurons. Similarly to the tectofugal thalamic nuclei in amniotes, the lateral nucleus of PG receives tectal information and projects to the pallium. However, our cell lineage study in zebrafish reveals that the majority of PG cells are derived from the midbrain, unlike the amniote thalamus. We also demonstrate that the PG projection neurons develop gradually until late juvenile stages. Our data suggest that teleost PG, as a whole, is not homologous to the amniote thalamus. Thus, the thalamocortical-like projections evolved from a non-forebrain cell population, which indicates a surprising degree of variation in the vertebrate sensory systems.


Subject(s)
Biological Evolution , Cell Lineage , Thalamic Nuclei/embryology , Visual Pathways/embryology , Zebrafish/embryology , Animals , Embryo, Nonmammalian/embryology
4.
Proc Natl Acad Sci U S A ; 116(43): 21812-21820, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591211

ABSTRACT

The developing brain can respond quickly to altered sensory experience by circuit reorganization. During a critical period in early life, neurons in the primary visual cortex rapidly lose responsiveness to an occluded eye and come to respond better to the open eye. While physiological and some of the molecular mechanisms of this process have been characterized, its structural basis, except for the well-known changes in the thalamocortical projection, remains obscure. To elucidate the relationship between synaptic remodeling and functional changes during this experience-dependent process, we used 2-photon microscopy to image synaptic structures of sparsely labeled layer 2/3 neurons in the binocular zone of mouse primary visual cortex. Anatomical changes at presynaptic and postsynaptic sites in mice undergoing monocular visual deprivation (MD) were compared to those in control mice with normal visual experience. We found that postsynaptic spines remodeled quickly in response to MD, with neurons more strongly dominated by the deprived eye losing more spines. These postsynaptic changes parallel changes in visual responses during MD and their recovery after restoration of binocular vision. In control animals with normal visual experience, the formation of presynaptic boutons increased during the critical period and then declined. MD affected bouton formation, but with a delay, blocking it after 3 d. These findings reveal intracortical anatomical changes in cellular layers of the cortex that can account for rapid activity-dependent plasticity.


Subject(s)
Amblyopia/physiopathology , Neuronal Plasticity/physiology , Visual Cortex/embryology , Visual Pathways/embryology , Animals , Mice , Mice, Inbred C57BL , Presynaptic Terminals/physiology , Sensory Deprivation/physiology , Vision, Binocular/physiology , Vision, Monocular/physiology , Visual Cortex/physiology
5.
J Comp Neurol ; 527(3): 508-521, 2019 02 15.
Article in English | MEDLINE | ID: mdl-29744881

ABSTRACT

In the developing mouse optic tract, retinal ganglion cell (RGC) axon position is organized by topography and laterality (i.e., eye-specific or ipsi- and contralateral segregation). Our lab previously showed that ipsilaterally projecting RGCs are segregated to the lateral aspect of the developing optic tract and found that ipsilateral axons self-fasciculate to a greater extent than contralaterally projecting RGC axons in vitro. However, the full complement of axon-intrinsic and -extrinsic factors mediating eye-specific segregation in the tract remain poorly understood. Glia, which are known to express several guidance cues in the visual system and regulate the navigation of ipsilateral and contralateral RGC axons at the optic chiasm, are natural candidates for contributing to eye-specific pre-target axon organization. Here, we investigate the spatiotemporal expression patterns of both putative astrocytes (Aldh1l1+ cells) and microglia (Iba1+ cells) in the embryonic and neonatal optic tract. We quantified the localization of ipsilateral RGC axons to the lateral two-thirds of the optic tract and analyzed glia position and distribution relative to eye-specific axon organization. While our results indicate that glial segregation patterns do not strictly align with eye-specific RGC axon segregation in the tract, we identify distinct spatiotemporal organization of both Aldh1l1+ cells and microglia in and around the developing optic tract. These findings inform future research into molecular mechanisms of glial involvement in RGC axon growth and organization in the developing retinogeniculate pathway.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Neuroglia/metabolism , Optic Tract/embryology , Optic Tract/metabolism , Retinal Dehydrogenase/metabolism , Retinal Ganglion Cells/metabolism , Age Factors , Aldehyde Dehydrogenase 1 Family/analysis , Animals , Axons/metabolism , Mice , Mice, Inbred C57BL , Optic Tract/cytology , Retinal Dehydrogenase/analysis , Visual Pathways/cytology , Visual Pathways/embryology , Visual Pathways/metabolism
6.
J Neurosci ; 38(26): 5854-5871, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29793976

ABSTRACT

NADPH oxidase (Nox)-derived reactive oxygen species (ROS) have been linked to neuronal polarity, axonal outgrowth, cerebellar development, regeneration of sensory axons, and neuroplasticity. However, the specific roles that individual Nox isoforms play during nervous system development in vivo remain unclear. To address this problem, we investigated the role of Nox activity in the development of retinotectal connections in zebrafish embryos. Zebrafish broadly express four nox genes (nox1, nox2/cybb, nox5, and duox) throughout the CNS during early development. Application of a pan-Nox inhibitor, celastrol, during the time of optic nerve (ON) outgrowth resulted in significant expansion of the ganglion cell layer (GCL), thinning of the ON, and a decrease in retinal axons reaching the optic tectum (OT). With the exception of GCL expansion, these effects were partially ameliorated by the addition of H2O2, a key ROS involved in Nox signaling. To address isoform-specific Nox functions, we used CRISPR/Cas9 to generate mutations in each zebrafish nox gene. We found that nox2/cybb chimeric mutants displayed ON thinning and decreased OT innervation. Furthermore, nox2/cybb homozygous mutants (nox2/cybb-/-) showed significant GCL expansion and mistargeted retinal axons in the OT. Neurite outgrowth from cultured zebrafish retinal ganglion cells was reduced by Nox inhibitors, suggesting a cell-autonomous role for Nox in these neurons. Collectively, our results show that Nox2/Cybb is important for retinotectal development in zebrafish.SIGNIFICANCE STATEMENT Most isoforms of NADPH oxidase (Nox) only produce reactive oxygen species (ROS) when activated by an upstream signal, making them ideal candidates for ROS signaling. Nox enzymes are present in neurons and their activity has been shown to be important for neuronal development and function largely by in vitro studies. However, whether Nox is involved in the development of axons and formation of neuronal connections in vivo has remained unclear. Using mutant zebrafish embryos, this study shows that a specific Nox isoform, Nox2/Cybb, is important for the establishment of axonal connections between retinal ganglion cells and the optic tectum.


Subject(s)
NADPH Oxidase 2/metabolism , Neurogenesis/physiology , Optic Lobe, Nonmammalian/embryology , Retina/embryology , Visual Pathways/embryology , Animals , Embryo, Nonmammalian , Optic Lobe, Nonmammalian/metabolism , Retina/metabolism , Visual Pathways/metabolism , Zebrafish
7.
J Comp Neurol ; 526(7): 1077-1096, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29322522

ABSTRACT

Prior to forming and refining synaptic connections, axons of projection neurons navigate long distances to their targets. While much is known about guidance cues for axon navigation through intermediate choice points, whether and how axons are organized within tracts is less clear. Here we analyze the organization of retinal ganglion cell (RGC) axons in the developing mouse retinogeniculate pathway. RGC axons are organized by both eye-specificity and topography in the optic nerve and tract: ipsilateral RGC axons are segregated from contralateral axons and are offset laterally in the tract relative to contralateral axon topographic position. To identify potential cell-autonomous factors contributing to the segregation of ipsilateral and contralateral RGC axons in the visual pathway, we assessed their fasciculation behavior in a retinal explant assay. Ipsilateral RGC neurites self-fasciculate more than contralateral neurites in vitro and maintain this difference in the presence of extrinsic chiasm cues. To further probe the role of axon self-association in circuit formation in vivo, we examined RGC axon organization and fasciculation in an EphB1-/- mutant, in which a subset of ipsilateral RGC axons aberrantly crosses the midline but targets the ipsilateral zone in the dorsal lateral geniculate nucleus on the opposite side. Aberrantly crossing axons retain their association with ipsilateral axons in the contralateral tract, indicating that cohort-specific axon affinity is maintained independently of guidance signals present at the midline. Our results provide a comprehensive assessment of RGC axon organization in the retinogeniculate pathway and suggest that axon self-association contributes to pre-target axon organization.


Subject(s)
Axons/physiology , Optic Nerve/physiology , Retinal Ganglion Cells/cytology , Visual Pathways , Amino Acids/metabolism , Animals , Animals, Newborn , Embryo, Mammalian , Eye/cytology , Eye/innervation , Fasciculation , Functional Laterality , In Vitro Techniques , Intermediate Filaments/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve/embryology , Optic Nerve/growth & development , Receptor, EphB1/genetics , Receptor, EphB1/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Visual Pathways/anatomy & histology , Visual Pathways/embryology , Visual Pathways/growth & development
8.
Methods Mol Biol ; 1650: 167-176, 2017.
Article in English | MEDLINE | ID: mdl-28809020

ABSTRACT

To elucidate a gene function, in vivo analysis is indispensable. We can carry out gain and loss of function experiment of a gene of interest by electroporation in ovo and ex ovo culture system on early-stage and advanced-stage chick embryos, respectively. In this section, we introduce in/ex ovo electroporation methods for the development of the chick central nervous system and visual system investigation.


Subject(s)
Central Nervous System/metabolism , Chick Embryo , Chickens/genetics , Electroporation/methods , Gene Transfer Techniques , Animals , Central Nervous System/embryology , DNA Transposable Elements , Visual Pathways/embryology , Visual Pathways/metabolism
9.
Cell Rep ; 20(6): 1255-1261, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28793250

ABSTRACT

The 40,000 neurons of the medulla, the largest visual processing center of the Drosophila brain, derive from a sheet of neuroepithelial cells. During larval development, a wave of differentiation sweeps across the neuroepithelium, converting neuroepithelial cells into neuroblasts that sequentially express transcription factors specifying different neuronal cell fates. The switch from neuroepithelial cells to neuroblasts is controlled by a complex gene regulatory network and is marked by the expression of the proneural gene l'sc. We discovered that microRNA miR-7 is expressed at the transition between neuroepithelial cells and neuroblasts. We showed that miR-7 promotes neuroepithelial cell-to-neuroblast transition by targeting downstream Notch effectors to limit Notch signaling. miR-7 acts as a buffer to ensure that a precise and stereotypical pattern of transition is maintained, even under conditions of environmental stress, echoing the role that miR-7 plays in the eye imaginal disc. This common mechanism reflects the importance of robust visual system development.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Neurogenesis , Visual Pathways/embryology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Visual Pathways/metabolism
10.
J Comp Neurol ; 525(3): 459-477, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27410778

ABSTRACT

Information from the retina is carried along the visual pathway with accuracy and spatial conservation as a result of topographically mapped axonal connections. The optic tectum in the midbrain is the primary region to which retinal ganglion cells project their axons in the chick. The two primary axes of the retina project independently onto the tectum using different sets of guidance cues to give rise to the retinotectal map. Specificity of the map is determined by attractive or repulsive interactions between molecular tags that are distributed in gradients in the retina and the tectum. Despite several studies, knowledge of the retinotectal guidance molecules is far from being complete. We screened for all molecules that are expressed differentially along the anterior-posterior and medial-lateral axes of the chick tectum using microarray based transcriptional profiling and identified several novel candidate retinotectal guidance molecules. Two such genes, encoding Wnt5a and Raldh2, the synthesizing enzymes for retinoic acid, were further analyzed for their function as putative regulators of retinotectal map formation. Wnt5a and retinoic acid were found to exhibit differential effects on the growth of axons from retinal explants derived from different quadrants of the retina. This screen also yielded a large number of genes expressed in a lamina-specific manner in the tectum, which may have other roles in tectal development. J. Comp. Neurol. 525:459-477, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Developmental , Retina/embryology , Retina/metabolism , Superior Colliculi/embryology , Superior Colliculi/metabolism , Transcriptome , Animals , Avian Proteins/metabolism , Chick Embryo , Gene Expression Profiling , In Situ Hybridization , Microarray Analysis , Neuronal Outgrowth/physiology , Real-Time Polymerase Chain Reaction , Retina/cytology , Superior Colliculi/cytology , Tissue Culture Techniques , Visual Pathways/cytology , Visual Pathways/embryology , Visual Pathways/metabolism
11.
J Neurosci ; 36(24): 6503-13, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27307238

ABSTRACT

UNLABELLED: During brain development, various types of neuronal populations are produced from different progenitor pools to produce neuronal diversity that is sufficient to establish functional neuronal circuits. However, the molecular mechanisms that specify the identity of each progenitor pool remain obscure. Here, we show that Wnt signaling is essential for the specification of the identity of posterior progenitor pools in the Drosophila visual center. In the medulla, the largest component of the visual center, different types of neurons are produced from two progenitor pools: the outer proliferation center (OPC) and glial precursor cells (GPCs; also known as tips of the OPC). We found that OPC-type neurons are produced from the GPCs at the expense of GPC-type neurons when Wnt signaling is suppressed in the GPCs. In contrast, GPC-type neurons are ectopically induced when Wnt signaling is ectopically activated in the OPC. These results suggest that Wnt signaling is necessary and sufficient for the specification of the progenitor pool identity. We also found that Homothorax (Hth), which is temporally expressed in the OPC, is ectopically induced in the GPCs by suppression of Wnt signaling and that ectopic induction of Hth phenocopies the suppression of Wnt signaling in the GPCs. Thus, Wnt signaling is involved in regionalization of the fly visual center through the specification of the progenitor pool located posterior to the medulla by suppressing Hth expression. SIGNIFICANCE STATEMENT: Brain consists of considerably diverse neurons of different origins. In mammalian brain, excitatory and inhibitory neurons derive from the dorsal and ventral telencephalon, respectively. Multiple progenitor pools also contribute to the neuronal diversity in fly brain. However, it has been unclear how differences between these progenitor pools are established. Here, we show that Wnt signaling, an evolutionarily conserved signaling, is involved in the process that establishes the differences between these progenitor pools. Because ß-catenin signaling, which is under the control of Wnt ligands, specifies progenitor pool identity in the developing mammalian thalamus, Wnt signaling-mediated specification of progenitor pool identity may be conserved in insect and mammalian brains.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental/genetics , Neurons/physiology , Signal Transduction/physiology , Visual Pathways/physiology , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified , Brain/cytology , Brain/embryology , CD8 Antigens/genetics , CD8 Antigens/metabolism , Drosophila , Drosophila Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Visual Pathways/embryology , Visual Pathways/growth & development , Wnt Proteins/genetics
12.
Elife ; 5: e13715, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26987017

ABSTRACT

The precise recognition of appropriate synaptic partner neurons is a critical step during neural circuit assembly. However, little is known about the developmental context in which recognition specificity is important to establish synaptic contacts. We show that in the Drosophila visual system, sequential segregation of photoreceptor afferents, reflecting their birth order, lead to differential positioning of their growth cones in the early target region. By combining loss- and gain-of-function analyses we demonstrate that relative differences in the expression of the transcription factor Sequoia regulate R cell growth cone segregation. This initial growth cone positioning is consolidated via cell-adhesion molecule Capricious in R8 axons. Further, we show that the initial growth cone positioning determines synaptic layer selection through proximity-based axon-target interactions. Taken together, we demonstrate that birth order dependent pre-patterning of afferent growth cones is an essential pre-requisite for the identification of synaptic partner neurons during visual map formation in Drosophila.


Subject(s)
Drosophila/embryology , Growth Cones/physiology , Synapses/physiology , Animals , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/metabolism , Visual Pathways/embryology
13.
Development ; 143(7): 1134-48, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26903505

ABSTRACT

The transcript encoding translationally controlled tumor protein (Tctp), a molecule associated with aggressive breast cancers, was identified among the most abundant in genome-wide screens of axons, suggesting that Tctp is important in neurons. Here, we tested the role of Tctp in retinal axon development in Xenopus laevis We report that Tctp deficiency results in stunted and splayed retinotectal projections that fail to innervate the optic tectum at the normal developmental time owing to impaired axon extension. Tctp-deficient axons exhibit defects associated with mitochondrial dysfunction and we show that Tctp interacts in the axonal compartment with myeloid cell leukemia 1 (Mcl1), a pro-survival member of the Bcl2 family. Mcl1 knockdown gives rise to similar axon misprojection phenotypes, and we provide evidence that the anti-apoptotic activity of Tctp is necessary for the normal development of the retinotectal projection. These findings suggest that Tctp supports the development of the retinotectal projection via its regulation of pro-survival signalling and axonal mitochondrial homeostasis, and establish a novel and fundamental role for Tctp in vertebrate neural circuitry assembly.


Subject(s)
Axons/metabolism , Biomarkers, Tumor/genetics , Optic Lobe, Nonmammalian/embryology , Retina/embryology , Retinal Ganglion Cells/cytology , Visual Pathways/embryology , Animals , Blastomeres/cytology , Cells, Cultured , Embryo, Nonmammalian/embryology , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/physiology , Mitochondria/physiology , Mitochondrial Dynamics/genetics , Morpholinos/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neurogenesis/physiology , Rats , Rats, Inbred F344 , Retinal Ganglion Cells/metabolism , Tumor Protein, Translationally-Controlled 1 , Visual Pathways/metabolism , Xenopus laevis
14.
Eur J Neurosci ; 43(2): 162-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26535920

ABSTRACT

The visual system of adult pigeons shows a lateralization of object discrimination with a left hemispheric dominance on the behavioural, physiological and anatomical levels. The crucial trigger for the establishment of this asymmetry is the position of the embryo inside the egg, which exposes the right eye to light falling through the egg shell. As a result, the right-sided retina is more strongly stimulated with light during embryonic development. However, it is unknown how this embryonic light stimulation is transduced to the brain as rods and cones are not yet functional. A possible solution could be the blue-light-sensitive molecule cryptochrome 1 (Cry1), which is expressed in retinal ganglion cells (RGCs) of several mammalian and avian species. RGCs have been shown to be functional during the time of induction of asymmetry and possess projections to primary visual areas. Therefore, Cry1-containing RGCs could be responsible for induction of asymmetry. The aim of this study was to identify the expression pattern of the Cry1 subtype Cry1b in the retina of embryonic, post-hatch and adult pigeons by immunohistochemical staining and to show whether Cry1b-containing RGCs project to the optic tectum. Cry1b-positive cells were indeed mainly found in the RGC layer and to lesser extent in the inner nuclear layer at all ages, including the embryonic stage. Tracing in adult animals revealed that at least a subset of Cry1b-containing RGCs project to the optic tectum. Thus, Cry1b-containing RGCs within the embryonic retina could be involved in the induction of asymmetries in the visual system of pigeons.


Subject(s)
Avian Proteins/metabolism , Cryptochromes/metabolism , Functional Laterality , Retina/metabolism , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Animals , Columbidae , Retina/embryology , Superior Colliculi/embryology , Visual Pathways/embryology , Visual Pathways/metabolism
15.
Elife ; 42015 Nov 14.
Article in English | MEDLINE | ID: mdl-26568314

ABSTRACT

Biophysical properties of neurons become increasingly diverse over development, but mechanisms underlying and constraining this diversity are not fully understood. Here we investigate electrophysiological characteristics of Xenopus tadpole midbrain neurons across development and during homeostatic plasticity induced by patterned visual stimulation. We show that in development tectal neuron properties not only change on average, but also become increasingly diverse. After sensory stimulation, both electrophysiological diversity and functional differentiation of cells are reduced. At the same time, the amount of cross-correlations between cell properties increase after patterned stimulation as a result of homeostatic plasticity. We show that tectal neurons with similar spiking profiles often have strikingly different electrophysiological properties, and demonstrate that changes in intrinsic excitability during development and in response to sensory stimulation are mediated by different underlying mechanisms. Overall, this analysis and the accompanying dataset provide a unique framework for further studies of network maturation in Xenopus tadpoles.


Subject(s)
Electrophysiological Phenomena , Mesencephalon/embryology , Neuronal Plasticity , Neurons/physiology , Visual Pathways/cytology , Visual Pathways/embryology , Xenopus/embryology , Action Potentials , Animals , Photic Stimulation
16.
Neural Dev ; 10: 23, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26492970

ABSTRACT

BACKGROUND: In the visual system of most binocular vertebrates, the axons of retinal ganglion cells (RGCs) diverge at the diencephalic midline and extend to targets on both ipsi- and contralateral sides of the brain. While a molecular mechanism explaining ipsilateral guidance decisions has been characterized, less is known of how RGC axons cross the midline. RESULTS: Here, we took advantage of the zebrafish, in which all RGC axons project contralaterally at the optic chiasm, to characterize Islr2 as an RGC receptor required for complete retinal axon midline crossing. We used a systematic extracellular protein-protein interaction screening assay to identify two Vasorin paralogs, Vasna and Vasnb, as specific Islr2 ligands. Antibodies against Vasna and Vasnb reveal cellular populations surrounding the retinal axon pathway, suggesting the involvement of these proteins in guidance decisions made by axons of the optic nerve. Specifically, Vasnb marks the membranes of a cellular barricade located anteriorly to the optic chiasm, a structure termed the "glial knot" in higher vertebrates. Loss of function mutations in either vasorin paralog, individually or combined, however, do not exhibit an overt retinal axon projection phenotype, suggesting that additional midline factors, acting either independently or redundantly, compensate for their loss. Analysis of Islr2 knockout mice supports a scenario in which Islr2 controls the coherence of RGC axons through the ventral midline and optic tract. CONCLUSIONS: Although stereotypic guidance of RGC axons at the vertebrate optic chiasm is controlled by multiple, redundant mechanisms, and despite the differences in ventral diencephalic tissue architecture, we identify a novel role for the LRR receptor Islr2 in ensuring proper axon navigation at the optic chiasm of both zebrafish and mouse.


Subject(s)
Axons/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Optic Chiasm/embryology , Retina/embryology , Animals , Body Patterning/physiology , Image Processing, Computer-Assisted , In Situ Hybridization , Mice , Mice, Inbred C57BL , Visual Pathways/embryology , Zebrafish
17.
Elife ; 42015 Oct 29.
Article in English | MEDLINE | ID: mdl-26512889

ABSTRACT

Filopodial dynamics are thought to control growth cone guidance, but the types and roles of growth cone dynamics underlying neural circuit assembly in a living brain are largely unknown. To address this issue, we have developed long-term, continuous, fast and high-resolution imaging of growth cone dynamics from axon growth to synapse formation in cultured Drosophila brains. Using R7 photoreceptor neurons as a model we show that >90% of the growth cone filopodia exhibit fast, stochastic dynamics that persist despite ongoing stepwise layer formation. Correspondingly, R7 growth cones stabilize early and change their final position by passive dislocation. N-Cadherin controls both fast filopodial dynamics and growth cone stabilization. Surprisingly, loss of N-Cadherin causes no primary targeting defects, but destabilizes R7 growth cones to jump between correct and incorrect layers. Hence, growth cone dynamics can influence wiring specificity without a direct role in target recognition and implement simple rules during circuit assembly.


Subject(s)
Drosophila/embryology , Growth Cones/physiology , Pseudopodia/physiology , Visual Pathways/embryology , Animals , Cadherins/metabolism , Drosophila Proteins/metabolism , Optical Imaging
18.
Brain Res ; 1625: 324-36, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26363092

ABSTRACT

Ocular albinism (OA) is characterized by inadequate L-3, 4-dihydroxyphenylalanine (L-DOPA) and dopamine (DA) in the eyes. This study investigated DA-related signaling pathways in mouse chiasm projection patterns and the potential role of ocular albinism type 1 (OA1) and dopamine 1A (D1A) receptors in the optic pathway. In embryonic day (E) E13-E15 retina, most L-DOPA and OA1-positive cells were distributed among Müller glial cells on E13 and retinal ganglion cells (RGC) on E14. In the ventral diencephalon, OA1 and L-DOPA were strongly expressed on the optic chiasm (OC) and optic tract (OT), respectively, but weak on the optic stalk (OS). At E13-E15, DA and D1A staining was predominately expressed in radially arranged cells with a neuronal expression pattern. In the ventral diencephalon, DA and D1A were strongly expressed on the OC, OT and OS. Furthermore, L-DOPA significantly inhibited retinal axon outgrowth in both the dorsal nasal (DN) and ventral temporal (VT) groups. DA inhibited retinal axon outgrowth, which was abolished by the D1A antagonist SCH23390. Brain slice cultures indicated that L-DOPA inhibited axons that crossed at the OC of E13 embryos, which was not abolished by DA. L-DOPA also inhibited axons that crossed at the OC of albino mice. Albino mice exhibited reduced ipsilateral retinal projections compared with C57 pigmented mice. No significant difference was identified in the uncrossed projections of albino mice following L-DOPA and DA expression. Furthermore, transcription factor Zic family member 2 (Zic2) upregulated OA1 mRNA expression. Our findings provide critical insights into DA-related signaling in retinal development.


Subject(s)
Dopamine/metabolism , Optic Chiasm/metabolism , Retina/metabolism , Signal Transduction/physiology , Visual Pathways/metabolism , Animals , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/physiology , In Vitro Techniques , Levodopa/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/metabolism , Optic Chiasm/embryology , Organ Culture Techniques , Pregnancy , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Retina/cytology , Signal Transduction/genetics , Transcription Factors/metabolism , Visual Pathways/embryology
19.
J Neurosci ; 35(11): 4729-40, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25788689

ABSTRACT

Retina ganglion cell (RGC) axons grow along a stereotyped pathway undergoing coordinated rounds of fasciculation and defasciculation, which are critical to establishing proper eye-brain connections. How this coordination is achieved is poorly understood, but shedding of guidance cues by metalloproteinases is emerging as a relevant mechanism. Secreted Frizzled Related Proteins (Sfrps) are multifunctional proteins, which, among others, reorient RGC growth cones by regulating intracellular second messengers, and interact with Tolloid and ADAM metalloproteinases, thereby repressing their activity. Here, we show that the combination of these two functions well explain the axon guidance phenotype observed in Sfrp1 and Sfrp2 single and compound mouse mutant embryos, in which RGC axons make subtle but significant mistakes during their intraretinal growth and inappropriately defasciculate along their pathway. The distribution of Sfrp1 and Sfrp2 in the eye is consistent with the idea that Sfrp1/2 normally constrain axon growth into the fiber layer and the optic disc. Disheveled axon growth instead seems linked to Sfrp-mediated modulation of metalloproteinase activity. Indeed, retinal explants from embryos with different Sfrp-null alleles or explants overexpressing ADAM10 extend axons with a disheveled appearance, which is reverted by the addition of Sfrp1 or an ADAM10-specific inhibitor. This mode of growth is associated with an abnormal proteolytic processing of L1 and N-cadherin, two ADAM10 substrates previously implicated in axon guidance. We thus propose that Sfrps contribute to coordinate visual axon growth with a dual mechanism: by directly signaling at the growth cone and by regulating the processing of other relevant cues.


Subject(s)
Axons/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Retinal Ganglion Cells/physiology , Visual Pathways/embryology , Visual Pathways/growth & development , Animals , Female , Frizzled Receptors/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Dev Neurobiol ; 75(6): 621-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25787992

ABSTRACT

Spontaneous activity during early development is necessary for the formation of precise neural connections, but it remains uncertain whether activity plays an instructive or permissive role in brain wiring. In the visual system, retinal ganglion cell (RGC) projections to the brain form two prominent sensory maps, one reflecting eye of origin and the other retinotopic location. Recent studies provide compelling evidence supporting an instructive role for spontaneous retinal activity in the development of eye-specific projections, but evidence for a similarly instructive role in the development of retinotopy is more equivocal. Here, we report on experiments in which we knocked down the expression of ß2-containing nicotinic acetylcholine receptors (ß2-nAChRs) specifically in the retina through a Cre-loxP recombination strategy. Overall levels of spontaneous retinal activity in retina-specific ß2-nAChR mutant mice (Rx-ß2cKO), examined in vitro and in vivo, were reduced to a degree comparable to that observed in whole animal ß2-nAChR mouse mutants (ß2KO). However, many residual spontaneous waves in Rx-ß2cKO mice displayed local propagating features with strong correlations between nearby but not distant RGCs typical of waves observed in wild-type (WT) but not ß2KO mice. We further observed that eye-specific segregation was disrupted in Rx-ß2cKO mice, but retinotopy was spared in a competition-dependent manner. These results suggest that propagating patterns of spontaneous retinal waves are essential for normal development of the retinotopic map, even while overall activity levels are significantly reduced, and support an instructive role for spontaneous retinal activity in both eye-specific segregation and retinotopic refinement.


Subject(s)
Brain Mapping , Retina/cytology , Retinal Ganglion Cells/physiology , Visual Pathways , Action Potentials/physiology , Amino Acids/metabolism , Animals , Calcium Signaling/physiology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Superior Colliculi , Visual Pathways/cytology , Visual Pathways/embryology , Visual Pathways/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL