Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 26(12): 2158-2170, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919424

ABSTRACT

Neuronal homeostasis prevents hyperactivity and hypoactivity. Age-related hyperactivity suggests homeostasis may be dysregulated in later life. However, plasticity mechanisms preventing age-related hyperactivity and their efficacy in later life are unclear. We identify the adult cortical plasticity response to elevated activity driven by sensory overstimulation, then test how plasticity changes with age. We use in vivo two-photon imaging of calcium-mediated cellular/synaptic activity, electrophysiology and c-Fos-activity tagging to show control of neuronal activity is dysregulated in the visual cortex in late adulthood. Specifically, in young adult cortex, mGluR5-dependent population-wide excitatory synaptic weakening and inhibitory synaptogenesis reduce cortical activity following overstimulation. In later life, these mechanisms are downregulated, so that overstimulation results in synaptic strengthening and elevated activity. We also find overstimulation disrupts cognition in older but not younger animals. We propose that specific plasticity mechanisms fail in later life dysregulating neuronal microcircuit homeostasis and that the age-related response to overstimulation can impact cognitive performance.


Subject(s)
Neurons , Visual Cortex , Animals , Neurons/physiology , Homeostasis/physiology , Visual Cortex/physiology , Neuronal Plasticity/physiology
2.
Elife ; 102021 03 16.
Article in English | MEDLINE | ID: mdl-33722342

ABSTRACT

Spontaneous activity drives the establishment of appropriate connectivity in different circuits during brain development. In the mouse primary visual cortex, two distinct patterns of spontaneous activity occur before vision onset: local low-synchronicity events originating in the retina and global high-synchronicity events originating in the cortex. We sought to determine the contribution of these activity patterns to jointly organize network connectivity through different activity-dependent plasticity rules. We postulated that local events shape cortical input selectivity and topography, while global events homeostatically regulate connection strength. However, to generate robust selectivity, we found that global events should adapt their amplitude to the history of preceding cortical activation. We confirmed this prediction by analyzing in vivo spontaneous cortical activity. The predicted adaptation leads to the sparsification of spontaneous activity on a slower timescale during development, demonstrating the remarkable capacity of the developing sensory cortex to acquire sensitivity to visual inputs after eye-opening.


Subject(s)
Adaptation, Physiological , Visual Cortex/physiology , Animals , Brain Mapping/methods , Mice , Models, Neurological , Neuronal Plasticity/physiology , Neurons/physiology , Retina/physiology , Synapses/physiology , Vision, Ocular/physiology , Visual Cortex/growth & development
3.
Cell Calcium ; 95: 102365, 2021 05.
Article in English | MEDLINE | ID: mdl-33610083

ABSTRACT

The adult neocortex is not hard-wired but instead retains the capacity to reorganise across multiple spatial scales long into adulthood. Plastic reorganisation occurs at the level of mesoscopic sensory maps, functional neuronal assemblies and synaptic ensembles and is thought to be a critical feature of neuronal network function. Here, we describe a series of approaches that use calcium imaging to measure network reorganisation across multiple spatial scales in vivo. At the mesoscopic level, we demonstrate that sensory activity can be measured in animals undergoing longitudinal behavioural assessment involving automated touchscreen tasks. At the cellular level, we show that network dynamics can be longitudinally measured at both stable and transient functional assemblies. At the level of single synapses, we show that functional subcellular calcium imaging approaches can be used to measure synaptic ensembles of dendritic spines in vivo. Finally, we demonstrate that all three levels of imaging can be spatially related to local pathology in a preclinical rodent model of amyloidosis. We propose that multi-scale in vivo calcium imaging can be used to measure parallel plasticity processes operating across multiple spatial scales in both the healthy brain and preclinical models of disease.


Subject(s)
Amyloidosis/metabolism , Calcium/metabolism , Microscopy, Fluorescence, Multiphoton/methods , Neocortex/metabolism , Nerve Net/metabolism , Amyloidosis/diagnostic imaging , Animals , Disease Models, Animal , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/diagnostic imaging , Nerve Net/diagnostic imaging
4.
J Neurosci ; 40(28): 5495-5509, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32527982

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common monogenic neurodevelopmental disorder associated with physical and cognitive problems. The cognitive issues are thought to arise from increased release of the neurotransmitter GABA. Modulating the signaling pathways causing increased GABA release in a mouse model of NF1 reverts deficits in hippocampal learning. However, clinical trials based on these approaches have so far been unsuccessful. We therefore used a combination of slice electrophysiology, in vivo two-photon calcium imaging, and optical imaging of intrinsic signal in a mouse model of NF1 to investigate whether cortical development is affected in NF1, possibly causing lifelong consequences that cannot be rescued by reducing inhibition later in life. We find that, in NF1 mice of both sexes, inhibition increases strongly during the development of the visual cortex and remains high. While this increase in cortical inhibition does not affect spontaneous cortical activity patterns during early cortical development, the critical period for ocular dominance plasticity is shortened in NF1 mice due to its early closure but unaltered onset. Notably, after environmental enrichment, differences in inhibitory innervation and ocular dominance plasticity between NF1 mice and WT littermates disappear. These results provide the first evidence for critical period dysregulation in NF1 and suggest that treatments aimed at normalizing levels of inhibition will need to start at early stages of development.SIGNIFICANCE STATEMENT Neurofibromatosis type 1 is associated with cognitive problems for which no treatment is currently available. This study shows that, in a mouse model of neurofibromatosis type 1, cortical inhibition is increased during development and critical period regulation is disturbed. Rearing the mice in an environment that stimulates cognitive function overcomes these deficits. These results uncover critical period dysregulation as a novel mechanism in the pathogenesis of neurofibromatosis type 1. This suggests that targeting the affected signaling pathways in neurofibromatosis type 1 for the treatment of cognitive disabilities may have to start at a much younger age than has so far been tested in clinical trials.


Subject(s)
Cerebral Cortex/physiopathology , Neurofibromatosis 1/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Animals , Critical Period, Psychological , Disease Models, Animal , Female , Male , Mice , Optical Imaging , Visual Cortex/physiopathology
5.
Nat Commun ; 10(1): 5684, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831751

ABSTRACT

We experience the world through multiple senses simultaneously. To better understand mechanisms of multisensory processing we ask whether inputs from two senses (auditory and visual) can interact and drive plasticity in neural-circuits of the primary visual cortex (V1). Using genetically-encoded voltage and calcium indicators, we find coincident audio-visual experience modifies both the supra and subthreshold response properties of neurons in L2/3 of mouse V1. Specifically, we find that after audio-visual pairing, a subset of multimodal neurons develops enhanced auditory responses to the paired auditory stimulus. This cross-modal plasticity persists over days and is reflected in the strengthening of small functional networks of L2/3 neurons. We find V1 processes coincident auditory and visual events by strengthening functional associations between feature specific assemblies of multimodal neurons during bouts of sensory driven co-activity, leaving a trace of multisensory experience in the cortical network.


Subject(s)
Auditory Cortex/physiology , Auditory Perception/physiology , Nerve Net/physiology , Visual Cortex/physiology , Visual Perception/physiology , Acoustic Stimulation , Animals , Mice , Models, Animal , Models, Biological , Neuronal Plasticity , Neurons/physiology , Photic Stimulation , Sensory Deprivation/physiology
6.
Front Neural Circuits ; 13: 57, 2019.
Article in English | MEDLINE | ID: mdl-31616256

ABSTRACT

Fragile X syndrome (FXS) is the most prevalent inherited cause of autism and is accompanied by behavioral and sensory deficits. Errors in the wiring of the brain during early development likely contribute to these deficits, but the underlying mechanisms are unclear. Spontaneous activity patterns, which are required for fine-tuning neuronal networks before the senses become active, are perturbed in rodent models of FXS. Here, we investigated spontaneous network activity patterns in the developing visual cortex of the Fmr1 knockout mouse using in vivo calcium imaging during the second postnatal week, before eye opening. We found that while the frequency, mean amplitude and duration of spontaneous network events were unchanged in the knockout mouse, pair-wise correlations between neurons were increased compared to wild type littermate controls. Further analysis revealed that interneuronal correlations were not generally increased, rather that low-synchronization events occurred relatively less frequently than high-synchronization events. Low-, but not high-, synchronization events have been associated with retinal inputs previously. Since we found that spontaneous retinal waves were normal in the knockout, our results suggest that peripherally driven activity is underrepresented in the Fmr1 KO visual cortex. Therefore, we propose that central gating of retinal inputs may be affected in FXS and that peripherally and centrally driven activity patterns are already unbalanced before eye opening in this disorder.


Subject(s)
Calcium/metabolism , Fragile X Mental Retardation Protein/genetics , Neurons/physiology , Visual Cortex/physiology , Animals , Disease Models, Animal , Fragile X Syndrome/physiopathology , Mice , Mice, Knockout , Visual Cortex/growth & development
8.
Hum Mol Genet ; 23(6): 1538-50, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24163243

ABSTRACT

Mutations in CACNA1F encoding the α1-subunit of the retinal Cav1.4 L-type calcium channel have been linked to Cav1.4 channelopathies including incomplete congenital stationary night blindness type 2A (CSNB2), Åland Island eye disease (AIED) and cone-rod dystrophy type 3 (CORDX3). Since CACNA1F is located on the X chromosome, Cav1.4 channelopathies are typically affecting male patients via X-chromosomal recessive inheritance. Occasionally, clinical symptoms have been observed in female carriers, too. It is currently unknown how these mutations lead to symptoms in carriers and how the retinal network in these females is affected. To investigate these clinically important issues, we compared retinal phenotypes in Cav1.4-deficient and Cav1.4 heterozygous mice and in human female carrier patients. Heterozygous Cacna1f carrier mice have a retinal mosaic consistent with differential X-chromosomal inactivation, characterized by adjacent vertical columns of affected and non-affected wild-type-like retinal network. Vertical columns in heterozygous mice are well comparable to either the wild-type retinal network of normal mice or to the retina of homozygous mice. Affected retinal columns display pronounced rod and cone photoreceptor synaptopathy and cone degeneration. These changes lead to vastly impaired vision-guided navigation under dark and normal light conditions and reduced retinal electroretinography (ERG) responses in Cacna1f carrier mice. Similar abnormal ERG responses were found in five human CACNA1F carriers, four of which had novel mutations. In conclusion, our data on Cav1.4 deficient mice and human female carriers of mutations in CACNA1F are consistent with a phenotype of mosaic CSNB2.


Subject(s)
Calcium Channels/genetics , Eye Diseases, Hereditary/pathology , Genetic Diseases, X-Linked/pathology , Myopia/pathology , Night Blindness/pathology , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Animals , Calcium Channels/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Electroretinography , Eye Diseases, Hereditary/genetics , Female , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Myopia/genetics , Night Blindness/genetics , Phenotype , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , X Chromosome , X Chromosome Inactivation
9.
Invest Ophthalmol Vis Sci ; 54(13): 8079-90, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24255040

ABSTRACT

PURPOSE: Endocannabinoids are important modulators of synaptic transmission and plasticity throughout the central nervous system. The cannabinoid receptor type 1 (CB1R) is extensively expressed in the adult retina of rodents, while CB2R mRNA and protein expression have been only recently demonstrated in retinal tissue. The activation of cannabinoid receptors modulates neurotransmitter release from photoreceptors and could also affect bipolar cell synaptic release. However, the impact of CB1R and CB2R on the retinal function as a whole is currently unknown. METHODS: In the present study, we investigated the function of cannabinoid receptors in the retina by recording electroretinographic responses (ERGs) from mice lacking either CB1 or CB2 receptors (cnr1(-/-) and cnr2(-/-), respectively). We also documented the precise distribution of CB2R by immunohistochemistry. RESULTS: Our results showed that CB2R is localized in cone and rod photoreceptors, horizontal cells, some amacrine cells, and bipolar and ganglion cells. In scotopic conditions, the amplitudes of the a-wave of the ERG were increased in cnr2(-/-) mice, while they remained unchanged in cnr1(-/-) mice. The analysis of the velocity-time profile of the a-wave revealed that the increased amplitude was due to a slower deceleration rather than an increase in acceleration of the waveform. Under photopic conditions, b-wave amplitudes of cnr2(-/-) mice required more light adaptation time to reach stable values. No effects were observed in cnr1(-/-) mice. CONCLUSIONS: The data indicated that CB2R is likely to be involved in shaping retinal responses to light and suggest that CB1 and CB2 receptors could have different roles in visual processing.


Subject(s)
Photoreceptor Cells, Vertebrate/metabolism , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Retinal Neurons/metabolism , Animals , Blotting, Western , Dark Adaptation , Electroretinography , Endocannabinoids/physiology , Female , Fluorescent Antibody Technique, Indirect , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal
10.
PLoS One ; 8(8): e70849, 2013.
Article in English | MEDLINE | ID: mdl-23951024

ABSTRACT

Navigation of retinal projections towards their targets is regulated by guidance molecules and growth cone transduction mechanisms. Here, we present in vitro and in vivo evidences that the cannabinoid receptor 2 (CB2R) is expressed along the retino-thalamic pathway and exerts a modulatory action on axon guidance. These effects are specific to CB2R since no changes were observed in mice where the gene coding for this receptor was altered (cnr2 (-/-)). The CB2R induced morphological changes observed at the growth cone are PKA dependent and require the presence of the netrin-1 receptor, Deleted in Colorectal Cancer. Interfering with endogenous CB2R signalling using pharmacological agents increased retinal axon length and induced aberrant projections. Additionally, cnr2 (-/-) mice showed abnormal eye-specific segregation of retinal projections in the dorsal lateral geniculate nucleus (dLGN) indicating CB2R's implication in retinothalamic development. Overall, this study demonstrates that the contribution of endocannabinoids to brain development is not solely mediated by CB1R, but also involves CB2R.


Subject(s)
Axons/metabolism , Endocannabinoids/metabolism , Geniculate Bodies/metabolism , Receptor, Cannabinoid, CB2/genetics , Retinal Ganglion Cells/metabolism , Animals , Axons/ultrastructure , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental , Geniculate Bodies/cytology , Geniculate Bodies/growth & development , Mice , Mice, Knockout , Netrin Receptors , Neurogenesis/physiology , Primary Cell Culture , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Retinal Ganglion Cells/cytology , Visual Pathways/physiology
11.
PLoS One ; 8(5): e63853, 2013.
Article in English | MEDLINE | ID: mdl-23675510

ABSTRACT

Several studies have shown the importance of calcium channels in the development and/or maturation of synapses. The Ca(V)1.4(α(1F)) knockout mouse is a unique model to study the role of calcium channels in photoreceptor synapse formation. It features abnormal ribbon synapses and aberrant cone morphology. We investigated the expression and targeting of several key elements of ribbon synapses and analyzed the cone morphology in the Ca(V)1.4(α(1F)) knockout retina. Our data demonstrate that most abnormalities occur after eye opening. Indeed, scaffolding proteins such as Bassoon and RIM2 are properly targeted at first, but their expression and localization are not maintained in adulthood. This indicates that either calcium or the Ca(V)1.4 channel, or both are necessary for the maintenance of their normal expression and distribution in photoreceptors. Other proteins, such as Veli3 and PSD-95, also display abnormal expression in rods prior to eye opening. Conversely, vesicle related proteins appear normal. Our data demonstrate that the Ca(V)1.4 channel is important for maintaining scaffolding proteins in the ribbon synapse but less vital for proteins related to vesicular release. This study also confirms that in adult retinae, cones show developmental features such as sprouting and synaptogenesis. Overall we present evidence that in the absence of the Ca(V)1.4 channel, photoreceptor synapses remain immature and are unable to stabilize.


Subject(s)
Aging/genetics , Calcium Channels/metabolism , Gene Expression Regulation, Developmental , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Synapses/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Calcium Channels/genetics , Calcium Channels, L-Type , Disks Large Homolog 4 Protein , Gene Expression Profiling , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Rod Photoreceptor Cells/ultrastructure , Synapses/genetics , Synapses/ultrastructure , rab3 GTP-Binding Proteins/genetics , rab3 GTP-Binding Proteins/metabolism
12.
Cell Mol Life Sci ; 70(10): 1831-47, 2013 May.
Article in English | MEDLINE | ID: mdl-23269435

ABSTRACT

In the mammalian retina, light signals generated in photoreceptors are passed to bipolar and horizontal cells via synaptic contacts. In various pathological conditions, these second-order neurons extend neurites into the outer nuclear layer (ONL). However, the molecular events associated with this neurite outgrowth are not known. Here, we characterized the morphological synaptic changes in the CNGA3/CNGB1 double-knockout (A3B1) mouse, a model of retinitis pigmentosa. In these mice, horizontal cells looked normal until postnatal day (p) 11, but started growing neurites into the ONL 1 day later. At p28, the number of sprouting processes decreased, but the remaining sprouts developed synapse-like contacts at rod cell bodies, with an ultrastructural appearance reminiscent of ribbon synapses. Hence, neurite outgrowth and ectopic synaptogenesis in the A3B1 retina were precisely timed events starting at p12 and p28, respectively. We therefore performed microarray analysis of retinal gene expression in A3B1 and wild-type mice at those ages to evaluate the genomic response underlying these two events. This analysis identified 163 differentially regulated genes in the A3B1 retina related to neurite outgrowth or plasticity of synapses. The global changes in gene expression in the A3B1 retina were consistent with activation of signaling pathways related to Tp53, Smad, and Stat3. Moreover, key molecules of these signaling pathways could be localized at or in close proximity to outgrowing neurites. We therefore propose that Tp53, Smad, and Stat3 signaling pathways contribute to the synaptic plasticity in the A3B1 retina.


Subject(s)
Neurites/metabolism , Retinal Bipolar Cells/metabolism , Retinal Horizontal Cells/metabolism , Synapses/metabolism , Animals , Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Retina/metabolism , Retinal Bipolar Cells/pathology , Retinal Horizontal Cells/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Synapses/ultrastructure , Tumor Suppressor Protein p53/metabolism
13.
PLoS One ; 7(4): e35109, 2012.
Article in English | MEDLINE | ID: mdl-22506070

ABSTRACT

Far from being a simple sensor, the retina actively participates in processing visual signals. One of the best understood aspects of this processing is the detection of motion direction. Direction-selective (DS) retinal circuits include several subtypes of ganglion cells (GCs) and inhibitory interneurons, such as starburst amacrine cells (SACs). Recent studies demonstrated a surprising complexity in the arrangement of synapses in the DS circuit, i.e. between SACs and DS ganglion cells. Thus, to fully understand retinal DS mechanisms, detailed knowledge of all synaptic elements involved, particularly the nature and localization of neurotransmitter receptors, is needed. Since inhibition from SACs onto DSGCs is crucial for generating retinal direction selectivity, we investigate here the nature of the GABA receptors mediating this interaction. We found that in the inner plexiform layer (IPL) of mouse and rabbit retina, GABA(A) receptor subunit α2 (GABA(A)R α2) aggregated in synaptic clusters along two bands overlapping the dendritic plexuses of both ON and OFF SACs. On distal dendrites of individually labeled SACs in rabbit, GABA(A)R α2 was aligned with the majority of varicosities, the cell's output structures, and found postsynaptically on DSGC dendrites, both in the ON and OFF portion of the IPL. In GABA(A)R α2 knock-out (KO) mice, light responses of retinal GCs recorded with two-photon calcium imaging revealed a significant impairment of DS responses compared to their wild-type littermates. We observed a dramatic drop in the proportion of cells exhibiting DS phenotype in both the ON and ON-OFF populations, which strongly supports our anatomical findings that α2-containing GABA(A)Rs are critical for mediating retinal DS inhibition. Our study reveals for the first time, to the best of our knowledge, the precise functional localization of a specific receptor subunit in the retinal DS circuit.


Subject(s)
Motion Perception/physiology , Receptors, GABA-A/metabolism , Retina/physiology , Amacrine Cells/metabolism , Amacrine Cells/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Rabbits , Retina/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Synapses/metabolism
14.
J Neurosci ; 31(4): 1489-99, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273433

ABSTRACT

Endocannabinoids (eCBs) are retrograde neurotransmitters that modulate the function of many types of synapses. The presence of eCBs, their CB1 receptor (CB1R), and metabolizing enzymes at embryonic and early postnatal periods have been linked to developmental processes such as neuronal proliferation, differentiation, and migration, axon guidance, and synaptogenesis. Here, we demonstrate the presence of a functional eCB system in the developing visual system and the role of CB1R during axon growth and retinothalamic development. Pharmacological treatment of retinal explants and primary cortical neuron cultures with ACEA, a selective CB1R agonist, induced a collapse of the growth cone (GC). Furthermore the application of AM251, a CB1R inverse agonist, to the neuronal cultures increased the surface area of GC. In vivo, intraocular injection of ACEA diminished retinal projection growth, while AM251 promoted growth and caused aberrant projections. In addition, compared with their wild-type littermates, CB1R-deficient adult mice revealed a lower level of eye-specific segregation of retinal projections in the dorsal lateral geniculate nucleus. Finally, we found that pharmacological modulation of CB1R affected the trafficking of Deleted in colorectal cancer (DCC) receptor to the plasma membrane in a PKA-dependent manner. Moreover, pharmacological inhibition or genetic inactivation of DCC abolished the CB1R-induced reorganization of the GC. Overall, these findings establish a mechanism by which the CB1R influences GC behavior and nervous system development in concerted action with DCC.


Subject(s)
Axons/physiology , Neurons/metabolism , Receptor, Cannabinoid, CB1/physiology , Receptors, Cell Surface/physiology , Retina/metabolism , Tumor Suppressor Proteins/physiology , Animals , Cannabinoid Receptor Modulators/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , DCC Receptor , Drug Inverse Agonism , Growth Cones/physiology , In Vitro Techniques , Mice , Mice, Knockout , Neurotransmitter Agents/metabolism , Protein Transport , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Retina/embryology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/ultrastructure , Thalamus/embryology , Thalamus/metabolism
15.
Neurosci Lett ; 329(3): 257-60, 2002 Sep 06.
Article in English | MEDLINE | ID: mdl-12183025

ABSTRACT

The Ca(2+)-independent protein kinase C (PKC) Apl II, but not the Ca(2+)-activated PKC Apl I, becomes autonomously active during intermediate periods of facilitation in Aplysia neurons. We examined the ability of superoxide formed by the enzymatic reaction of xanthine with xanthine oxidase (X/XO) to induce autonomous activity of PKCs in Aplysia. X/XO stimulated autonomous PKC activity in Aplysia nervous system homogenates, but this activity resulted solely from activation of PKC Apl I. PKC Apl I is also more sensitive to activation by X/XO when expressed in insect cells. Our results suggest that oxidation can autonomously activate PKC Apl I in the Aplysia nervous system, but that the activation of PKC Apl II during synaptic facilitation is not due to oxidation of the enzyme.


Subject(s)
Isoenzymes/metabolism , Neurons/enzymology , Protein Kinase C/metabolism , Animals , Aplysia , Conditioning, Psychological/physiology , Memory/physiology , Neuronal Plasticity/physiology , Oxidation-Reduction , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...