Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(3): e2216537120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36630455

ABSTRACT

Protein degradation is critical for brain function through processes that remain incompletely understood. Here, we investigated the in vivo function of the 20S neuronal membrane proteasome (NMP) in the brain of Xenopus laevis tadpoles. With biochemistry, immunohistochemistry, and electron microscopy, we demonstrated that NMPs are conserved in the tadpole brain and preferentially degrade neuronal activity-induced newly synthesized proteins in vivo. Using in vivo calcium imaging in the optic tectum, we showed that acute NMP inhibition rapidly increased spontaneous neuronal activity, resulting in hypersynchronization across tectal neurons. At the circuit level, inhibiting NMPs abolished learning-dependent improvement in visuomotor behavior in live animals and caused a significant deterioration in basal behavioral performance following visual training with enhanced visual experience. Our data provide in vivo characterization of NMP functions in the vertebrate nervous system and suggest that NMP-mediated degradation of activity-induced nascent proteins may serve as a homeostatic modulatory mechanism in neurons that is critical for regulating neuronal activity and experience-dependent circuit plasticity.


Subject(s)
Neurons , Proteasome Endopeptidase Complex , Animals , Proteasome Endopeptidase Complex/metabolism , Neurons/metabolism , Superior Colliculi/physiology , Tectum Mesencephali , Xenopus laevis/metabolism , Avoidance Learning/physiology , Larva/metabolism , Neuronal Plasticity/physiology
2.
eNeuro ; 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34099488

ABSTRACT

The Xenopus laevis experimental system has provided significant insight into the development and plasticity of neural circuits. Xenopus neuroscience research would be enhanced by additional tools to study neural circuit structure and function. Rabies viruses are powerful tools to label and manipulate neural circuits and have been widely used to study mesoscale connectomics. Whether rabies virus can be used to transduce neurons and express transgenes in Xenopus has not been systematically investigated. Glycoprotein-deleted rabies virus transduces neurons at the axon terminal and retrogradely labels their cell bodies. We show that glycoprotein-deleted rabies virus infects local and projection neurons in the Xenopus tadpole when directly injected into brain tissue. Pseudotyping glycoprotein-deleted rabies with EnvA restricts infection to cells with exogenous expression of the EnvA receptor, TVA. EnvA pseudotyped virus specifically infects tadpole neurons with promoter-driven expression of TVA, demonstrating its utility to label targeted neuronal populations. Neuronal cell types are defined by a combination of features including anatomical location, expression of genetic markers, axon projection sites, morphology, and physiological properties. We show that driving TVA expression in one hemisphere and injecting EnvA pseudotyped virus into the contralateral hemisphere, retrogradely labels neurons defined by cell body location and axon projection site. Using this approach, rabies can be used to identify cell types in Xenopus brain and simultaneously to express transgenes which enable monitoring or manipulation of neuronal activity. This makes rabies a valuable tool to study the structure and function of neural circuits in Xenopus.Significance StatementStudies in Xenopus have contributed a great deal to our understanding of brain circuit development and plasticity, regeneration, and hormonal regulation of behavior and metamorphosis. Here, we show that recombinant rabies virus transduces neurons in the Xenopus tadpole, enlarging the toolbox that can be applied to studying Xenopus brain. Rabies can be used for retrograde labeling and expression of a broad range of transgenes including fluorescent proteins for anatomical tracing and studying neuronal morphology, voltage or calcium indicators to visualize neuronal activity, and photo- or chemosensitive channels to control neuronal activity. The versatility of these tools enables diverse experiments to analyze and manipulate Xenopus brain structure and function, including mesoscale connectivity.

3.
J Neurophysiol ; 121(1): 306-320, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30517041

ABSTRACT

Traumatic brain injuries introduce functional and structural circuit deficits that must be repaired for an organism to regain function. We developed an injury model in which Xenopus laevis tadpoles are given a penetrating stab wound that damages the optic tectal circuit and impairs visuomotor behavior. In tadpoles, as in other systems, injury induces neurogenesis. The newly generated neurons are thought to integrate into the existing circuit; however, whether they integrate via the same mechanisms that govern normal neuronal maturation during development is not understood. Development of the functional visuomotor circuit in Xenopus is driven by sensory activity. We hypothesized that enhanced visual experience would improve recovery from injury by facilitating integration of newly generated neurons into the tectal circuit. We labeled newly generated neurons in the injured tectum by green fluorescent protein expression and examined their circuit integration using electrophysiology and in vivo imaging. Providing animals with brief bouts of enhanced visual experience starting 24 h after injury increased synaptogenesis and circuit integration of new neurons and facilitated behavioral recovery. To investigate mechanisms of neuronal integration and behavioral recovery after injury, we interfered with N-methyl-d-aspartate (NMDA) receptor function. Ifenprodil, which blocks GluN2B-containing NMDA receptors, impaired dendritic arbor elaboration. GluN2B blockade inhibited functional integration of neurons generated in response to injury and prevented behavioral recovery. Furthermore, tectal GluN2B knockdown blocked the beneficial effects of enhanced visual experience on functional plasticity and behavioral recovery. We conclude that visual experience-mediated rehabilitation of the injured tectal circuit occurs by GluN2B-containing NMDA receptor-dependent integration of newly generated neurons. NEW & NOTEWORTHY Recovery from brain injury is difficult in most systems. The study of regenerative animal models that are capable of injury repair can provide insight into cellular and circuit mechanisms underlying repair. Using Xenopus tadpoles, we show enhanced sensory experience rehabilitates the injured visual circuit and that this experience-dependent recovery depends on N-methyl-d-aspartate receptor function. Understanding the mechanisms of rehabilitation in this system may facilitate recovery in brain regions and systems where repair is currently impossible.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/rehabilitation , Receptors, N-Methyl-D-Aspartate/metabolism , Visual Perception/physiology , Animals , Brain Injuries/pathology , Excitatory Amino Acid Antagonists/pharmacology , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Larva , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/metabolism , Neural Pathways/pathology , Neurogenesis , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Photic Stimulation/methods , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Recovery of Function/drug effects , Recovery of Function/physiology , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Visual Perception/drug effects , Xenopus laevis
4.
J Neurophysiol ; 119(5): 1947-1961, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29442555

ABSTRACT

The circuit controlling visually guided behavior in nonmammalian vertebrates, such as Xenopus tadpoles, includes retinal projections to the contralateral optic tectum, where visual information is processed, and tectal motor outputs projecting ipsilaterally to hindbrain and spinal cord. Tadpoles have an intertectal commissure whose function is unknown, but it might transfer information between the tectal lobes. Differences in visual experience between the two eyes have profound effects on the development and function of visual circuits in animals with binocular vision, but the effects on animals with fully crossed retinal projections are not clear. We tested the effect of monocular visual experience on the visuomotor circuit in Xenopus tadpoles. We show that cutting the intertectal commissure or providing visual experience to one eye (monocular visual experience) is sufficient to disrupt tectally mediated visual avoidance behavior. Monocular visual experience induces asymmetry in tectal circuit activity across the midline. Repeated exposure to monocular visual experience drives maturation of the stimulated retinotectal synapses, seen as increased AMPA-to-NMDA ratios, induces synaptic plasticity in intertectal synaptic connections, and induces bilaterally asymmetric changes in the tectal excitation-to-inhibition ratio (E/I). We show that unilateral expression of peptides that interfere with AMPA or GABAA receptor trafficking alters E/I in the transfected tectum and is sufficient to degrade visuomotor behavior. Our study demonstrates that monocular visual experience in animals with fully crossed visual systems produces asymmetric circuit function across the midline and degrades visuomotor behavior. The data further suggest that intertectal inputs are an integral component of a bilateral visuomotor circuit critical for behavior. NEW & NOTEWORTHY The developing optic tectum of Xenopus tadpoles represents a unique circuit in which laterally positioned eyes provide sensory input to a circuit that is transiently monocular, but which will be binocular in the animal's adulthood. We challenge the idea that the two lobes of tadpole optic tectum function independently by testing the requirement of interhemispheric communication and demonstrate that unbalanced sensory input can induce structural and functional plasticity in the tectum sufficient to disrupt function.


Subject(s)
Behavior, Animal/physiology , Neuronal Plasticity/physiology , Psychomotor Performance/physiology , Retina/physiology , Tectum Mesencephali/physiology , Vision, Binocular/physiology , Vision, Monocular/physiology , Visual Pathways/physiology , Xenopus laevis/physiology , Animals , Larva/physiology , Superior Colliculi/physiology
5.
J Neurophysiol ; 116(5): 2281-2297, 2016 11.
Article in English | MEDLINE | ID: mdl-27582296

ABSTRACT

Communication between optic tecta/superior colliculi is thought to be required for sensorimotor behaviors by comparing inputs across the midline, however the development of and the role of visual experience in the function and plasticity of intertectal connections are unclear. We combined neuronal tracing, in vivo time-lapse imaging, and electrophysiology to characterize the structural and functional development of intertectal axons and synapses in Xenopus tadpole optic tectum. We find that intertectal connections are established early during optic tectal circuit development. We determined the neurotransmitter identity of intertectal neurons using both rabies virus-mediated tracing combined with post-hoc immunohistochemistry, and electrophysiology. Excitatory and inhibitory intertectal neuronal somata are similarly distributed throughout the tectum. Excitatory and inhibitory intertectal axons are structurally similar and elaborate broadly in the contralateral tectum. We demonstrate that intertectal and retinotectal axons converge onto tectal neurons by recording postsynaptic currents after stimulating intertectal and retinotectal inputs. Cutting the intertectal commissure removes synaptic responses to contralateral tectal stimulation. In vivo time-lapse imaging demonstrated that visual experience drives plasticity in intertectal bouton size and dynamics. Finally, visual experience coordinately drives the maturation of excitatory and inhibitory intertectal inputs by increasing AMPA- and GABA-receptor mediated currents, comparable to experience-dependent maturation of retinotectal inputs. These data indicate that visual experience regulates plasticity of excitatory and inhibitory intertectal inputs, maintaining the excitatory: inhibitory ratio of intertectal input. These studies place intertectal inputs as key players in tectal circuit development and suggest that they may play a role in sensory information processing critical to sensorimotor behaviors.

6.
eNeuro ; 2(1): e0055, 2015.
Article in English | MEDLINE | ID: mdl-25844398

ABSTRACT

Fragile X Syndrome (FXS) is the leading known monogenic form of autism and the most common form of inherited intellectual disability. FXS results from silencing the FMR1 gene during embryonic development, leading to loss of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein that regulates mRNA transport, stability, and translation. FXS is commonly thought of as a disease of synaptic dysfunction, however, FMRP expression is lost early in embryonic development, well before most synaptogenesis occurs. Recent studies suggest that loss of FMRP results in aberrant neurogenesis, but neurogenic defects have been variable. We investigated whether FMRP affects neurogenesis in Xenopus laevis tadpoles which express a homolog of FMR1. We used in vivo time-lapse imaging of neural progenitor cells and their neuronal progeny to evaluate the effect of acute loss or over-expression of FMRP on neurogenesis in the developing optic tectum. We complimented the time-lapse studies with SYTOX labeling to quantify apoptosis and CldU labeling to measure cell proliferation. Animals with increased or decreased levels of FMRP have significantly decreased neuronal proliferation and survival. They also have increased neuronal differentiation, but deficient dendritic arbor elaboration. The presence and severity of these defects was highly sensitive to FMRP levels. These data demonstrate that FMRP plays an important role in neurogenesis and suggest that endogenous FMRP levels are carefully regulated. These studies show promise in using Xenopus as an experimental system to study fundamental deficits in brain development with loss of FMRP and give new insight into the pathophysiology of FXS.

7.
J Neurosci ; 30(48): 16376-82, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123583

ABSTRACT

Throughout the nervous system, neurons restrict their connections to specific depths or "layers" of their targets to constrain the type and number of synapses they make. Despite the importance of lamina-specific synaptic connectivity, the mechanisms that give rise to this feature in mammals remain poorly understood. Here we examined the cellular events underlying the formation of lamina-specific retinal ganglion cell (RGC) axonal projections to the superior colliculus (SC) of the mouse. By combining a genetically encoded marker of a defined RGC subtype (OFF-αRGCs) with serial immunoelectron microscopy, we resolved the ultrastructure of axon terminals fated for laminar stabilization versus those fated for removal. We found that OFF-αRGCs form synapses across the full depth of the retinorecipient SC before undergoing lamina-specific arbor retraction and synapse elimination to arrive at their mature, restricted pattern of connectivity. Interestingly, we did not observe evidence of axon degeneration or glia-induced synapse engulfment during this process. These findings indicate that lamina-specific visual connections are generated through the selective stabilization of correctly targeted axon arbors and suggest that the decision to maintain or eliminate an axonal projection reflects the molecular compatibility of presynaptic and postsynaptic neurons at a given laminar depth.


Subject(s)
Axons/physiology , Basement Membrane/physiology , Retinal Ganglion Cells/physiology , Synapses/physiology , Visual Pathways/physiology , Animals , Animals, Newborn , Axons/ultrastructure , Basement Membrane/ultrastructure , Mice , Mice, Knockout , Neural Pathways/physiology , Neural Pathways/ultrastructure , Retinal Ganglion Cells/ultrastructure , Synapses/ultrastructure , Visual Pathways/ultrastructure
8.
Proc Natl Acad Sci U S A ; 105(37): 14157-62, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18780780

ABSTRACT

New neurons are continuously generated in restricted regions of the adult mammalian brain. Although these adult-born neurons have been shown to receive synaptic inputs, little is known about their synaptic outputs. Using retrovirus-mediated birth-dating and labeling in combination with serial section electron microscopic reconstruction, we report that mossy fiber en passant boutons of adult-born dentate granule cells form initial synaptic contacts with CA3 pyramidal cells within 2 weeks after their birth and reach morphologic maturity within 8 weeks in the adult hippocampus. Knockdown of Disrupted-in-Schizophrenia-1 (DISC1) in newborn granule cells leads to defects in axonal targeting and development of synaptic outputs in the adult brain. Together with previous reports of synaptic inputs, these results demonstrate that adult-born neurons are fully integrated into the existing neuronal circuitry. Our results also indicate a role for DISC1 in presynaptic development and may have implications for the etiology of schizophrenia and related mental disorders.


Subject(s)
Aging/physiology , Mossy Fibers, Hippocampal/growth & development , Neurons/cytology , Synapses/physiology , Animals , Animals, Newborn , Female , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mossy Fibers, Hippocampal/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Interference , Synapses/ultrastructure
9.
Neural Dev ; 3: 21, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18727829

ABSTRACT

BACKGROUND: The development of the corticospinal tract (CST) in higher vertebrates relies on a series of axon guidance decisions along its long projection pathway. Several guidance molecules are known to be involved at various decision points to regulate the projection of CST axons. However, previous analyses of the CST guidance defects in mutant mice lacking these molecules have suggested that there are other molecules involved in CST axon guidance that are yet to be identified. In this study, we investigate the role of plexin signaling in the guidance of motor CST axons in vivo. RESULTS: Expression pattern studies show that plexin-A3, plexin-A4, and neuropilin-1 are expressed in the developing cerebral cortex when the motor CST axons originating from layer V cortical neurons are guided down to the spinal cord. By analyzing mutant mice, we show that motor CST axons that turn dorsally to cross the midline at the pyramidal decussation require plexin-A3 and plexin-A4 signaling. Although other CST guidance defects are found in neuropilin-1 mutants, this dorsal turning defect is not observed in either neuropilin-1 or neuropilin-2 mutants, suggesting that the local cues that activate plexin signaling at the dorsal turning point are membrane-bound semaphorins. Further expression pattern study and mutant analysis indicate that Sema6A is one of the local cues for motor CST axon turning at the pyramidal decussation. CONCLUSION: Dorsal turning and midline crossing at the pyramidal decussation is a crucial step to properly direct CST axons into the dorsal spinal cord. We show that the signaling of plexin-A3, plexin-A4, and Sema6A is at least partially required for dorsal turning of the CST axons, while neuropilin-1 is required for proper fasciculation of the tract at midline crossing. Together with previous reports, these results demonstrate that several guidance cues are specifically utilized to regulate the dorsal turning and midline crossing of developing CST axons.


Subject(s)
Axons/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Pyramidal Tracts , Receptors, Cell Surface/metabolism , Age Factors , Animals , Cytoskeletal Proteins , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Motor Neurons/ultrastructure , Nerve Tissue Proteins/genetics , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Pyramidal Tracts/cytology , Pyramidal Tracts/growth & development , Pyramidal Tracts/metabolism , Receptors, Cell Surface/genetics , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction/physiology
10.
Proc Natl Acad Sci U S A ; 105(23): 8136-41, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18523013

ABSTRACT

Neurons in the developing CNS tend to send out long axon collaterals to multiple target areas. For these neurons to attain specific connections, some of their axon collaterals are subsequently pruned-a process called stereotyped axon pruning. One of the most striking examples of stereotyped pruning in the CNS is the pruning of corticospinal tract (CST) axons. The long CST collaterals from layer V neurons of the visual and motor cortices are differentially pruned during development. Here we demonstrate that select plexins and neuropilins, which serve as coreceptors for semaphorins, are expressed in visual cortical neurons at the time when CST axon collaterals are stereotypically pruned. By analyzing mutant mice, we find that the pruning of visual, but not motor, CST axon collaterals depends on plexin-A3, plexin-A4, and neuropilin-2. Expression pattern study suggests that Sema3F is a candidate local cue for the pruning of visual CST axons. Using electron microscopic analysis, we also show that visual CST axon collaterals form synaptic contacts in the spinal cord before pruning and that the unpruned collaterals in adult mutant mice are unmyelinated and maintain their synaptic contacts. Our results indicate that the stereotyped pruning of the visual and motor CST axon collaterals is differentially regulated and that this specificity arises from the differential expression of plexin receptors in the cortex.


Subject(s)
Axons/metabolism , Nerve Tissue Proteins/metabolism , Pyramidal Tracts/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Visual Cortex/metabolism , Animals , Axons/ultrastructure , Mice , Motor Neurons/metabolism , Myelin Sheath/metabolism , Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/deficiency , Neuropilin-2/metabolism , Neuropilins/metabolism , Pyramidal Tracts/cytology , Pyramidal Tracts/ultrastructure , Receptors, Cell Surface/deficiency , Semaphorins/metabolism , Superior Colliculi/cytology , Superior Colliculi/metabolism , Synapses
11.
Cell ; 130(6): 1146-58, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17825401

ABSTRACT

Adult neurogenesis occurs throughout life in discrete regions of the adult mammalian brain. Little is known about the mechanism governing the sequential developmental process that leads to integration of new neurons from adult neural stem cells into the existing circuitry. Here, we investigated roles of Disrupted-In-Schizophrenia 1 (DISC1), a schizophrenia susceptibility gene, in adult hippocampal neurogenesis. Unexpectedly, downregulation of DISC1 leads to accelerated neuronal integration, resulting in aberrant morphological development and mispositioning of new dentate granule cells in a cell-autonomous fashion. Functionally, newborn neurons with DISC1 knockdown exhibit enhanced excitability and accelerated dendritic development and synapse formation. Furthermore, DISC1 cooperates with its binding partner NDEL1 in regulating adult neurogenesis. Taken together, our study identifies DISC1 as a key regulator that orchestrates the tempo of functional neuronal integration in the adult brain and demonstrates essential roles of a susceptibility gene for major mental illness in neuronal development, including adult neurogenesis.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Schizophrenia/metabolism , Stem Cells/metabolism , Synapses/metabolism , Action Potentials , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cell Size , Dendrites/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Genetic Vectors , Genotype , Hippocampus/embryology , Hippocampus/growth & development , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/pathology , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Schizophrenia/genetics , Schizophrenia/physiopathology , Stem Cells/pathology , Synapses/pathology , Synaptic Transmission , Time Factors
12.
Dev Neurosci ; 29(1-2): 6-13, 2007.
Article in English | MEDLINE | ID: mdl-17148945

ABSTRACT

During early development of the central nervous system (CNS), there is an exuberant outgrowth of projections which later need to be refined to achieve precise connectivity. One widely used strategy for this refinement is axon pruning. Axon pruning has also been suggested to be involved in creating more diverse connection patterns between different species. An understanding of the mechanism of pruning, however, has been elusive in the CNS. Recent studies have focused on a stereotyped pruning event that occurs within the mossy fibers of the developing vertebrate hippocampus. In the following discussion, we will review the cellular and molecular factors that are known to regulate pruning in the hippocampus and highlight some advantages this system presents for future studies on pruning in the developing CNS.


Subject(s)
Axons/metabolism , Axons/ultrastructure , Cell Differentiation/physiology , Hippocampus/cytology , Hippocampus/embryology , Semaphorins/metabolism , Animals , Hippocampus/metabolism , Humans , Mossy Fibers, Hippocampal/embryology , Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Signal Transduction/physiology , Vertebrates/embryology , Vertebrates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...