Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
RSC Adv ; 13(18): 12035-12049, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37077270

ABSTRACT

A series of acylsulfenyl iodides (RCOSI) were synthesized by the reactions of carbothioic acid group 11-16 element derivatives with iodine or N-iodosuccinimides in moderate to good yields. The structure of the PhCOSI was nearly square planar based on the X-ray analysis, where the C[double bond, length as m-dash]O⋯I distance (3.153(5) Å) was significantly shorter than the sum of the van der Waals radii of the atoms (Σr vdW), indicating close contact within the molecule. The distances between an iodine atom and the neighbouring two iodine atoms were also less than Σr vdW, perhaps due to the energy lowering effect of the interactions. The acylsulfenyl iodides readily reacted with alkenes and alkynes to give the expected addition products in moderate to good yields at approximately 0 °C. A new synthesis of acylated sulfines, sulfenamides and sulfenochalcogenides using acylsulfenyl iodides is also described. Theoretical calculations were performed on PhCOSI with the Sapporo-TZP(+1s1p) basis sets at the MP2 level, which perfectly reproduced the observed structures. Similar calculations were performed on the reactions, exemplified by those of MeCOSI and CH2[double bond, length as m-dash]CH2, together with those of MeSI and CH2[double bond, length as m-dash]CH2. Mechanisms for both reactions were proposed, which were very similar. The proposed mechanism for the former was understood based on that of the latter. For both mechanisms, the episulfuranes and episulfonium ions played an important role. The dynamic and static nature of the bonds in the COSI group of PhCOSI and MeCOSI were elucidated based on QTAIM dual functional analysis.

2.
Sci Rep ; 9(1): 4387, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867513

ABSTRACT

Keratan sulfate (KS) is a carbohydrate side chain covalently attached to extracellular proteoglycans. KS is composed of disaccharide units of 6-sulfated N-acetylglucosamine (GlcNAc) and galactose. We have previously shown that GlcNAc-6-O-sulfotransferase (GlcNAc6ST) 1 encoded by Chst2 is an enzyme necessary for the synthesis of GlcNAc-6-sulfated KS chains that are required for neuronal plasticity in the visual cortex of the mouse brain during the critical period, but not in adulthood. Here, we show that GlcNAc-6-sulfated KS recognized by the R-10G anti-KS antibody, of which the minimum epitope structure is Galß1-4GlcNAc(6S)ß1-3Galß1-4GlcNAc(6S), distributes diffusely in neuropils and presents densely in close proximity to the perineuronal region of the perineuronal net (PNN)-positive neurons in the adult visual cortex. Surprisingly, GlcNAc6ST3, which was discovered as an intestinal GlcNAc6ST encoded by Chst5, is a major brain KS sulfotransferase expressed in oligodendrocytes in adulthood. Moreover, we identified an isoform of the protein-tyrosine phosphatase PTPRZ as a R-10G-reactive KS proteoglycan. These results indicate that GlcNAc6ST3 may play a role in synthesis of a component of PNN in the adult brain, and that the KS-modified isoform of PTPRZ encoded by Ptprz1 could be an extracellular molecule associated with PNNs.


Subject(s)
Brain/enzymology , Brain/metabolism , Keratan Sulfate/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Sulfotransferases/metabolism , Animals , Female , Immunoprecipitation , Male , Mice , Microscopy, Confocal , Protein Tyrosine Phosphatases , Proteoglycans/metabolism , Synaptophysin/metabolism , Carbohydrate Sulfotransferases
3.
J Neurosci ; 38(36): 7852-7869, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30064994

ABSTRACT

Early visual experience is essential for the maturation of visual functions in which the primary visual cortex plays crucial roles. The extraction of visual features based on response selectivity of individual neurons, a fundamental process in the cortex, is basically established by eye opening in rodents, suggesting that visual experience is required for the development of neural functions other than feature extraction. Here, we show that synchronized firing, which is important for visual information processing, occurs selectively in adjacent neurons sharing similar orientation or spatial frequency preferences in layers 2-4 (upper layer) of rat visual cortex. This feature-selective spike synchrony was rudimentary when the eyes opened and became prominent during the first few weeks after eye opening only in the presence of pattern vision. In contrast, synchronization in layers 5-6 (lower layer) was almost independent of orientation similarity and more weakly dependent on spatial frequency similarity compared with upper layer synchrony. Lower layer synchronization was strengthened during development after eye opening independently of visual experience as a whole. However, the feature selectivity of synchronization was regulated by visual inputs, whereas the inputs without contours were sufficient for this regulation. Therefore, we speculate that feature-selective synchronization in the upper layer may convey detailed information on visual objects to the higher-order cortex, whereas weakly feature-selective synchronization in the lower layer may covey rather rough visual information to the subcortical areas or higher-order cortex. A major role of visual experience may be to establish the specific neural circuits underlying highly feature-selective synchronization.SIGNIFICANCE STATEMENT The neuronal mechanisms underlying experience-dependent improvement of visual functions still remain unresolved. In this study, we investigated whether early visual experience contributes to the development of synchronized neural firing in the primary visual cortex, which plays important roles in visual information processing. We found that synchronized firing depends more remarkably on the similarity of preferred visual stimuli in the upper than lower layer neurons. Pattern vision during development was required for the establishment of spike synchrony in the upper but not the lower layer. These findings provide a new view regarding the role of sensory experience in the functional development of the cortex and the differences in the modes of information processing in the upper and lower cortical layers.


Subject(s)
Neurons/physiology , Sensory Deprivation/physiology , Visual Cortex/physiology , Visual Perception/physiology , Action Potentials/physiology , Animals , Female , Male , Orientation, Spatial/physiology , Photic Stimulation , Rats , Rats, Long-Evans , Visual Pathways/physiology
4.
Neurobiol Learn Mem ; 152: 61-70, 2018 07.
Article in English | MEDLINE | ID: mdl-29783061

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays an important role in synaptic plasticity related to learning and memory. We previously reported that SPARC-related protein containing immunoglobulin domains 1 (SPIG1, also known as Follistatin-like protein 4, FSTL4) binds to pro-BDNF and negatively regulates BDNF maturation; however, its neurological functions, particularly in learning and memory, have not yet been elucidated. We herein examined the electrophysiological and behavioral phenotypes of Spig1-knockout (Spig1-KO) mice. Adult Spig1-KO mice exhibited greater excitability and facilitated long-term potentiation (LTP) in the CA1 region of hippocampal slices than age- and sex-matched wild-type (WT) mice. Facilitated LTP was reduced to the level of WT by the bath application of an anti-BDNF antibody to hippocampal slices. A step-through inhibitory avoidance learning paradigm revealed that the extinction of aversive memories was significantly enhanced in adult Spig1-KO mice, while they showed the normal acquisition of aversive memories; besides, spatial reference memory formation was also normal in the standard Morris water maze task. An intracerebroventricular (icv) injection of anti-BDNF in the process of extinction learning transiently induced the recurrence of aversive memories in Spig1-KO mice, but exerted no effects in WT mice. These results indicate a critical role for SPIG1 in BDNF-mediated synaptic plasticity in extinction of inhibitory avoidance memory.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Extinction, Psychological/physiology , Follistatin-Related Proteins/physiology , Long-Term Potentiation , Animals , Conditioning, Classical , Electroshock , Follistatin-Related Proteins/genetics , Hippocampus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Synaptic Transmission
5.
Neurosci Res ; 120: 36-44, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28284708

ABSTRACT

Manipulation of visual experience can considerably modify visual responses of visual cortical neurons even in adulthood in the mouse, although the modification is less profound than that observed during the critical period. Our previous studies demonstrated that low-frequency (2Hz) stimulation for 15min applied to layer 4 induces T-type Ca2+ channel-dependent long-term potentiation (LTP) at excitatory synapses in layer 2/3 neurons of visual cortex during the critical period. In this study, we investigated whether low-frequency stimulation could induce synaptic plasticity in adult mice. We found that 2Hz stimulation induced LTP of extracellular field potentials evoked by stimulation of layer 4 in layer 2/3 in adulthood as during the critical period. LTP in adulthood was blocked by L-type, but not T-type, Ca2+ channel antagonists, whereas LTP during the critical period was blocked by T-type, but not L-type, Ca2+ channel antagonists. This developmental change in LTP was prevented by dark rearing. Under pharmacological blockade of GABAA receptors, T-type Ca2+ channel-dependent LTP occurred, whereas L-type Ca2+ channel-dependent LTP did not occur. These results suggest that different forms of synaptic plasticity can contribute separately to experience-dependent modification of visual responses during the critical period and in adulthood.


Subject(s)
Long-Term Potentiation , Visual Cortex/physiology , Animals , Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/physiology , Electric Stimulation , Mice, Inbred C57BL , Sensory Deprivation , Visual Cortex/growth & development
6.
J Am Soc Nephrol ; 27(3): 766-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26376860

ABSTRACT

Hyponatremia is the most common clinical electrolyte disorder. Once thought to be asymptomatic in response to adaptation by the brain, recent evidence suggests that chronic hyponatremia may be linked to attention deficits, gait disturbances, risk of falls, and cognitive impairments. Such neurologic defects are associated with a reduction in quality of life and may be a significant cause of mortality. However, because underlying diseases such as adrenal insufficiency, heart failure, liver cirrhosis, and cancer may also affect brain function, the contribution of hyponatremia alone to neurologic manifestations and the underlying mechanisms remain unclear. Using a syndrome of inappropriate secretion of antidiuretic hormone rat model, we show here that sustained reduction of serum sodium ion concentration induced gait disturbances; facilitated the extinction of a contextual fear memory; caused cognitive impairment in a novel object recognition test; and impaired long-term potentiation at hippocampal CA3-CA1 synapses. In vivo microdialysis revealed an elevated extracellular glutamate concentration in the hippocampus of chronically hyponatremic rats. A sustained low extracellular sodium ion concentration also decreased glutamate uptake by primary astrocyte cultures, suggesting an underlying mechanism of impaired long-term potentiation. Furthermore, gait and memory performances of corrected hyponatremic rats were equivalent to those of control rats. Thus, these results suggest chronic hyponatremia in humans may cause gait disturbance and cognitive impairment, but these abnormalities are reversible and careful correction of this condition may improve quality of life and reduce mortality.


Subject(s)
Gait Disorders, Neurologic/etiology , Hyponatremia/complications , Inappropriate ADH Syndrome/physiopathology , Memory Disorders/etiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/physiopathology , Cells, Cultured , Chronic Disease , Cognition Disorders/blood , Cognition Disorders/etiology , Disease Models, Animal , Fear/physiology , Gait Disorders, Neurologic/blood , Glutamic Acid/metabolism , Hyponatremia/blood , Hyponatremia/psychology , Inappropriate ADH Syndrome/complications , Inappropriate ADH Syndrome/psychology , Male , Memory Disorders/blood , Microdialysis , Neuronal Plasticity , Rats , Rats, Sprague-Dawley , Sodium/blood , Sodium/pharmacology , Synapses/physiology
7.
Cereb Cortex ; 26(2): 669-682, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25316339

ABSTRACT

To understand the functions of the neocortex, it is essential to characterize the properties of neurons constituting cortical circuits. Here, we focused on a distinct group of GABAergic neurons that are defined by a specific colocalization of intense labeling for both neuronal nitric oxide synthase (nNOS) and substance P (SP) receptor [neurokinin 1 (NK1) receptors]. We investigated the mechanisms of the SP actions on these neurons in visual cortical slices obtained from young glutamate decarboxylase 67-green fluorescent protein knock-in mice. Bath application of SP induced a nonselective cation current leading to depolarization that was inhibited by the NK1 antagonists in nNOS-immunopositive neurons. Ruthenium red and La(3+), transient receptor potential (TRP) channel blockers, suppressed the SP-induced current. The SP-induced current was mediated by G proteins and suppressed by D609, an inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC), but not by inhibitors of phosphatidylinositol-specific PLC, adenylate cyclase or Src tyrosine kinases. Ca(2+) imaging experiments under voltage clamp showed that SP induced a rise in intracellular Ca(2+) that was abolished by removal of extracellular Ca(2+) but not by depletion of intracellular Ca(2+) stores. These results suggest that SP regulates nNOS neurons by activating TRP-like Ca(2+)-permeable nonselective cation channels through a PC-PLC-dependent signaling pathway.


Subject(s)
Calcium/metabolism , GABAergic Neurons/drug effects , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/physiology , Substance P/pharmacology , Type C Phospholipases/metabolism , Visual Cortex/cytology , Animals , Anthracenes/pharmacology , Boron Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , GABAergic Neurons/physiology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Imidazoles/pharmacology , In Vitro Techniques , Isoindoles/pharmacology , Mice , Mice, Transgenic , Neurokinin-1 Receptor Antagonists/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Quinolines/pharmacology , Quinuclidines/pharmacology , Signal Transduction/drug effects
8.
Exp Neurol ; 274(Pt B): 145-55, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26277687

ABSTRACT

Proteoglycans play important roles in regulating the development and functions of the brain. They consist of a core protein and glycosaminoglycans, which are long sugar chains of repeating disaccharide units with sulfation. A recent study demonstrated that the sulfation pattern of chondroitin sulfate on proteoglycans contributes to regulation of the critical period of experience-dependent plasticity in the mouse visual cortex. In the present study, we investigated the role of keratan sulfate (KS), another glycosaminoglycan, in critical period plasticity in the mouse visual cortex. Immunohistochemical analyses demonstrated the presence of KS containing disaccharide units of N-acetylglucosamine (GlcNAc)-6-sulfate and nonsulfated galactose during the critical period, although KS containing disaccharide units of GlcNAc-6-sulfate and galactose-6-sulfate was already known to disappear before that period. The KS chains were distributed diffusely in the extracellular space and densely around the soma of a large population of excitatory and inhibitory neurons. Electron microscopic analysis revealed that the KS was localized within the perisynaptic spaces and dendrites but not in presynaptic sites. KS was mainly located on phosphacan. In mice deficient in GlcNAc-6-O-sulfotransferase 1, which is one of the enzymes necessary for the synthesis of KS chains, the expression of KS was one half that in wild-type mice. In the knockout mice, monocular deprivation during the critical period resulted in a depression of deprived-eye responses but failed to produce potentiation of nondeprived-eye responses. In addition, T-type Ca(2+) channel-dependent long-term potentiation (LTP), which occurs only during the critical period, was not observed. These results suggest that regulation by KS-phosphacan with a specific sulfation pattern is necessary for the generation of LTP and hence the potentiation of nondeprived-eye responses after monocular deprivation.


Subject(s)
Neuronal Plasticity/physiology , Neurons/physiology , Proteoglycans/metabolism , Sulfotransferases/genetics , Visual Cortex/growth & development , Age Factors , Animals , Animals, Newborn , Epitopes/metabolism , Evoked Potentials, Visual/genetics , Gene Expression Regulation/genetics , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/genetics , Neurons/ultrastructure , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Sensory Deprivation/physiology , Sulfotransferases/deficiency , Synaptophysin/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Visual Cortex/cytology , Visual Cortex/metabolism , Carbohydrate Sulfotransferases
9.
Neurosci Res ; 96: 37-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25701075

ABSTRACT

Monocular deprivation produces depression and potentiation of visual responses evoked in visual cortical neurons by stimulation of deprived and nondeprived eyes, respectively, during the critical period of ocular dominance plasticity. Our previous studies suggested that T-type Ca(2+) channel-dependent long-term potentiation (LTP), induced by 2 Hz stimulation, mediates the potentiation of visual responses. However, it was proposed that the experience-dependent response potentiation is mediated by tumor necrosis factor-α (TNFα)-dependent homeostatic synaptic scaling but not by Hebbian synaptic plasticity, because the potentiation was absent in TNFα knockout (TNFα-KO) mice. In this study, we investigated whether TNFα is required for LTP induced by 2 Hz stimulation using visual cortical slices prepared from critical period mice and rats. The production of LTP was prevented by pharmacological blockade of TNFα in rats and mice. LTP production was also prevented by an inhibitor of TNFα-converting enzyme that converts membrane-bound TNFα to soluble TNFα. In TNFα-KO mice, LTP did not occur and was rescued by exogenous soluble TNFα. Soluble TNFα was required for LTP production only during a restricted time window soon after 2 Hz stimulation. These results strengthen the view that T-type Ca(2+) channel-dependent LTP contributes to the potentiation of nondeprived eye responses following monocular deprivation.


Subject(s)
Calcium Channels, T-Type/physiology , Long-Term Potentiation , Tumor Necrosis Factor-alpha/physiology , Visual Cortex/physiology , Animals , Critical Period, Psychological , Dominance, Ocular , Electric Stimulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Long-Evans , Tumor Necrosis Factor-alpha/genetics
10.
J Neurosci ; 34(37): 12576-86, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25209295

ABSTRACT

Visual cortical neurons selectively respond to particular features of visual stimuli and this selective responsiveness emerges from specific connectivity in the cortex. Most visual response properties are basically established by eye opening and are thereafter modified or refined by visual experience based on activity-dependent synaptic modifications during an early postnatal period. Visual deprivation during this period impairs development of visual functions, such as visual acuity. We previously demonstrated that fine-scale networks composed of a population of interconnected layer 2/3 (L2/3) pyramidal neurons receiving common inputs from adjacent neurons are embedded in a small area in rat visual cortex. We suggested that this network could be a functional unit for visual information processing. In this study, we investigated the effects of early visual experience on the development of fine-scale networks and individual synaptic connections in rat visual cortical slices. We used two kinds of deprivation, binocular deprivation and dark rearing, which allowed visual inputs with only diffuse light and no visual input, respectively. The probability and strength of excitatory connections to L2/3 pyramidal cells increased during the 2 weeks after eye opening, and these changes were prevented by dark rearing, but not binocular deprivation. Fine-scale networks were absent just after eye opening and established during the following 2 weeks in rats reared with normal visual experience, but not with either type of deprivation. These results indicate that patterned vision is required for the emergence of the fine-scale network, whereas diffuse light stimulation is sufficient for the maturation of individual synapses.


Subject(s)
Aging/physiology , Learning/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Visual Cortex/physiology , Visual Perception/physiology , Animals , Animals, Newborn , Cells, Cultured , Executive Function/physiology , Female , Male , Rats , Rats, Long-Evans
11.
Neurosci Res ; 87: 33-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25017998

ABSTRACT

Visual cortical neurons undergo depression and potentiation of their visual responses to stimulation of the deprived and non-deprived eyes, respectively, after monocular deprivation. This modification occurs predominantly during an early postnatal period in normal development, and this critical period is postponed until adulthood in animals reared in darkness from birth. We have proposed that Ni(2+)-sensitive T-type Ca(2+) channel-dependent long-term potentiation (T-LTP) mediates the potentiation of non-deprived eye responses. In this study, to investigate the development of Ni(2+)-sensitive T-type Ca(2+) channels, presumed CaV3.2 channels, we performed whole-cell recordings from layer 2/3 pyramidal neurons in rat visual cortical slices. T-type Ca(2+) channel currents were activated by voltage steps from -100mV to -40mV under a pharmacological blockade of Na(+) and K(+) channels. We estimated presumed CaV3.2 currents from the currents obtained after subtraction of the currents in the presence of Ni(2+) (50µM) from those in control solution. The estimated currents were very small before eye opening, peaked during the critical period and then returned to a small value by adulthood. Dark rearing prevented the developmental decline in these currents until adulthood. These results suggest that the regulation of CaV3.2 currents underlies the developmental changes in T-LTP and ocular dominance plasticity.


Subject(s)
Calcium Channels, T-Type/metabolism , Dominance, Ocular , Long-Term Potentiation , Neurons/physiology , Visual Cortex/physiology , Age Factors , Animals , Calcium/metabolism , Rats , Rats, Long-Evans , Sensory Deprivation/physiology , Visual Cortex/cytology
12.
J Neurophysiol ; 109(8): 2064-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23343903

ABSTRACT

Immature excitatory synapses often have NMDA receptors but not AMPA receptors in central neurons, including visual cortical pyramidal neurons. These synapses, called silent synapses, are converted to functional synapses with AMPA receptors by NMDA receptor activation during early development. It is likely that this process underlies the activity-dependent refinement of neuronal circuits and brain functions. In the present study, we investigated postnatal development of excitatory synapses, focusing on the role of visual inputs in the conversion of silent to functional synapses in mouse visual cortex. We analyzed presumably unitary excitatory postsynaptic currents (EPSCs) between a pair of layer 2/3 pyramidal neurons, using minimal stimulation with a patch pipette attached to the soma of one of the pair. The proportion of silent synapses was estimated by the difference in the failure rate between AMPA- and NMDA-EPSCs. In normal development, silent synapses were present abundantly before eye opening, decreased considerably by the critical period of ocular dominance plasticity, and almost absent in adulthood. This decline in silent synapses was prevented by dark rearing. The amplitude of presumably unitary AMPA-EPSCs increased with age, but this increase was suppressed by dark rearing. The quantal amplitude of AMPA-EPSCs and paired-pulse ratio of NMDA-EPSCs both remained unchanged during development, independent of visual experience. These results indicate that visual inputs are required for the conversion of silent to functional synapses and this conversion largely contributes to developmental increases in the amplitude of presumably unitary AMPA-EPSCs.


Subject(s)
Darkness , Pyramidal Cells/physiology , Synapses/physiology , Visual Cortex/physiology , Animals , Excitatory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , N-Methylaspartate/metabolism , Photic Stimulation , Pyramidal Cells/metabolism , Visual Cortex/cytology , Visual Cortex/growth & development , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
13.
Nat Neurosci ; 15(3): 414-22, S1-2, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22246436

ABSTRACT

Cortical plasticity is most evident during a critical period in early life, but the mechanisms that restrict plasticity after the critical period are poorly understood. We found that a developmental increase in the 4-sulfation/6-sulfation (4S/6S) ratio of chondroitin sulfate proteoglycans (CSPGs), which are components of the brain extracellular matrix, leads to the termination of the critical period for ocular dominance plasticity in the mouse visual cortex. Condensation of CSPGs into perineuronal nets that enwrapped synaptic contacts on parvalbumin-expressing interneurons was prevented by cell-autonomous overexpression of chondroitin 6-sulfation, which maintains a low 4S/6S ratio. Furthermore, the increase in the 4S/6S ratio was required for the accumulation of Otx2, a homeoprotein that activates the development of parvalbumin-expressing interneurons, and for functional maturation of the electrophysiological properties of these cells. Our results indicate that the critical period for cortical plasticity is regulated by the 4S/6S ratio of CSPGs, which determines the maturation of parvalbumin-expressing interneurons.


Subject(s)
Chondroitin Sulfates/metabolism , Gene Expression Regulation, Developmental/physiology , Neuronal Plasticity/physiology , Up-Regulation/physiology , Visual Cortex/cytology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Aggrecans/metabolism , Amphetamines/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/genetics , Electroporation/methods , Functional Laterality/physiology , Gene Expression Regulation, Developmental/genetics , Humans , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/genetics , Otx Transcription Factors/metabolism , Parvalbumins/metabolism , Patch-Clamp Techniques , Plant Lectins/metabolism , Receptors, N-Acetylglucosamine/metabolism , Sensory Deprivation/physiology , Sulfotransferases/genetics , Time Factors , Up-Regulation/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Visual Cortex/drug effects , Visual Cortex/physiology , Visual Pathways/metabolism , Carbohydrate Sulfotransferases
14.
J Neuropathol Exp Neurol ; 68(11): 1207-18, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19816196

ABSTRACT

The neurological mouse mutation shambling (shm) exhibits ataxia and hindlimb paresis. Positional cloning of shm showed that it encodes contactin-associated protein (Caspr), which is required for formation of the paranodal junction in myelinated nerves. The shm mutation is a TT insertion in the Caspr gene that results in a frame shift and a premature stop codon at the COOH-terminus. The truncated Caspr protein that is generated lacks the transmembrane and cytoplasmic domains. Here, we found that the nodal/paranodal axoplasm of shm mice lack paranodal junctions and contain large mitochondria and abnormal accumulations of cytoplasmic organelles that indicate altered axonal transport. Immunohistochemical analysis of mutant mice showed reduced expression of Caspr, contactin, and neurofascin 155, which are thought to form a protein complex in the paranodal region; protein 4.1B, however, was normally distributed. The mutant mice had aberrant localization of voltage-gated ion channels on the axolemma of nodal/paranodal regions. Electrophysiological analysis demonstrated that the velocity of saltatory conduction was reduced in sciatic nerves and that the visual response was attenuated in the primary visual cortex. These abnormalities likely contribute to the neurological phenotype of the mutant mice.


Subject(s)
Axons/pathology , Cell Adhesion Molecules, Neuronal/genetics , Mutation , Myelin Sheath/pathology , Nerve Fibers, Myelinated/pathology , Neuroglia/pathology , Phenotype , Amino Acid Sequence , Animals , Axons/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Molecular Sequence Data , Myelin Sheath/chemistry , Myelin Sheath/genetics , Nerve Fibers, Myelinated/chemistry , Neuroglia/chemistry
15.
Neurosci Res ; 64(2): 191-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428700

ABSTRACT

T-type Ca2+ channel-dependent long-term potentiation (LTP) occurs predominantly during the critical period of ocular dominance plasticity in rat and cat visual cortex. Noradrenaline and serotonin are known to facilitate ocular dominance plasticity. In this study using rat visual cortical slices, we tested whether this LTP is modulated by these neuromodulators in the same way as ocular dominance plasticity. Extracellular field potentials evoked by layer 4 stimulation were recorded from layer 2/3 and LTP was induced by low-frequency (2 Hz) stimulation continued for 15 min. The induction of LTP was suppressed by beta, but not alpha, adrenergic receptor antagonists. LTP induction was also inhibited by selective antagonists for 5-HT(1A) or 5-HT(2) receptors. In slices prepared from rats in which noradrenaline or serotonin was depleted by selective neurotoxins, the magnitude of LTP was significantly smaller compared with control slices. These results indicate that the same types of adrenergic and serotonergic receptors facilitate both LTP and ocular dominance plasticity, supporting our hypothesis that T-type Ca2+ channel-dependent LTP mediates experience-dependent enhancement of visual responses of cortical neurons during the critical period.


Subject(s)
Long-Term Potentiation , Norepinephrine/physiology , Serotonin/physiology , Visual Cortex/physiology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Antagonists , Animals , Calcium Channels, T-Type/physiology , Electric Stimulation , In Vitro Techniques , Rats , Rats, Long-Evans , Receptor, Serotonin, 5-HT1A/physiology , Receptors, Serotonin, 5-HT2/physiology , Visual Cortex/growth & development
16.
Eur J Neurosci ; 28(4): 730-43, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18657180

ABSTRACT

Neocortical neuronal circuits are refined by experience during the critical period of early postnatal life. The shift of ocular dominance in the visual cortex following monocular deprivation has been intensively studied to unravel the mechanisms underlying the experience-dependent modification. Synaptic plasticity is considered to be involved in this process. We previously showed in layer 2/3 pyramidal neurons of rat visual cortex that low-frequency stimulation-induced long-term potentiation (LTP) at excitatory synapses, which requires the activation of Ni(2+)-sensitive (R-type or T-type) voltage-gated Ca(2+) channels (VGCCs) for induction, shared a similar age and experience dependence with ocular dominance plasticity. In this study, we examined whether this LTP is involved in ocular dominance plasticity. In visual cortical slices, LTP was blocked by mibefradil, kurtoxin and R-(-)-efonidipine, T-type VGCC blockers, but not by SNX-482, an R-type VGCC blocker, indicating that LTP induction requires T-type VGCC activation. Mibefradil did not affect synaptic transmission even at a dose about 30 times higher than that required for LTP blockade. Therefore, this drug was used to test the effect of T-type VGCC blockade on ocular dominance shift produced by 6 days of monocular deprivation during the critical period using visual evoked potentials (VEPs). Although this monocular deprivation commonly produced both depression of deprived eye responses and potentiation of nondeprived eye responses, only the former change occurred when mibefradil was infused into the visual cortex during monocular deprivation. Mibefradil infusion produced no acute effects on VEPs. These results suggest that T-type VGCC-dependent LTP contributes to the experience-dependent enhancement of visual responses.


Subject(s)
Calcium Channels, T-Type/metabolism , Critical Period, Psychological , Sensory Deprivation/physiology , Synapses/physiology , Vision, Monocular/physiology , Visual Cortex/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/genetics , Dominance, Ocular , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Mibefradil/pharmacology , Neuronal Plasticity/physiology , Rats , Rats, Long-Evans , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Visual Cortex/cytology
17.
Neurosci Res ; 61(2): 192-200, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18395922

ABSTRACT

High-frequency stimulation (HFS) induces long-term potentiation (LTP) at inhibitory synapses of layer 5 pyramidal neurons in developing rat visual cortex. This LTP requires postsynaptic Ca2+ rise for induction, while the maintenance mechanism is present at the presynaptic site, suggesting presynaptic LTP expression and the necessity of retrograde signaling. We investigated whether the supposed signal is mediated by brain-derived neurotrophic factor (BDNF), which is expressed in pyramidal neurons but not inhibitory interneurons. LTP did not occur when HFS was applied in the presence of the Trk receptor tyrosine kinase inhibitor K252a in the perfusion medium. HFS produced LTP when bath application of K252a was started after HFS or when K252a was loaded into postsynaptic cells. LTP did not occur in the presence of TrkB-IgG scavenging BDNF or function-blocking anti-BDNF antibody in the medium. In cells loaded with the Ca2+ chelator BAPTA, the addition of BDNF to the medium enabled HFS to induce LTP without affecting baseline synaptic transmission. These results suggest that BDNF released from postsynaptic cells activates presynaptic TrkB, leading to LTP. Because BDNF, expressed activity dependently, regulates the maturation of cortical inhibition, inhibitory LTP may contribute to this developmental process, and hence experience-dependent functional maturation of visual cortex.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Long-Term Potentiation/physiology , Pyramidal Cells/physiology , Signal Transduction/physiology , Synapses/physiology , Visual Cortex/physiology , Animals , Brain-Derived Neurotrophic Factor/drug effects , Carbazoles/pharmacology , Electric Stimulation , Enzyme Inhibitors/pharmacology , Indole Alkaloids/pharmacology , Long-Term Potentiation/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Rats , Rats, Long-Evans , Receptor, trkB/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Visual Cortex/drug effects
18.
Neuron ; 57(6): 905-16, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18367091

ABSTRACT

Cortical pyramidal neurons alter their responses to input signals depending on behavioral state. We investigated whether changes in somatic inhibition contribute to these alterations. In layer 5 pyramidal neurons of rat visual cortex, repetitive firing from a depolarized membrane potential, which typically occurs during arousal, produced long-lasting depression of somatic inhibition. In contrast, slow membrane oscillations with firing in the depolarized phase, which typically occurs during slow-wave sleep, produced long-lasting potentiation. The depression is mediated by L-type Ca2+ channels and GABA(A) receptor endocytosis, whereas potentiation is mediated by R-type Ca2+ channels and receptor exocytosis. It is likely that the direction of modification is mainly dependent on the ratio of R- and L-type Ca2+ channel activation. Furthermore, somatic inhibition was stronger in slices prepared from rats during slow-wave sleep than arousal. This bidirectional modification of somatic inhibition may alter pyramidal neuron responsiveness in accordance with behavioral state.


Subject(s)
Dendrites/physiology , Neural Inhibition/physiology , Pyramidal Cells/cytology , Visual Cortex/cytology , 2-Amino-5-phosphonovalerate/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Animals, Newborn , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Dendrites/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/radiation effects , Patch-Clamp Techniques/methods , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Spider Venoms/pharmacology , gamma-Aminobutyric Acid/pharmacology
19.
FASEB J ; 22(6): 1797-806, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18198214

ABSTRACT

Interferon-gamma (IFN-gamma) is a proinflammatory cytokine that plays a pivotal role in pathology of diseases in the central nervous system (CNS), such as multiple sclerosis. However, the direct effect of IFN-gamma on neuronal cells has yet to be elucidated. We show here that IFN-gamma directly induces neuronal dysfunction, which appears as dendritic bead formation in mouse cortical neurons and enhances glutamate neurotoxicity mediated via alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic (AMPA) receptors but not N-methyl-D-aspartate receptors. In the CNS, IFN-gamma receptor forms a unique, neuron-specific, calcium-permeable receptor complex with AMPA receptor subunit GluR1. Through this receptor complex, IFN-gamma phosphorylates GluR1 at serine 845 position by JAK1.2/STAT1 pathway, increases Ca(2+) influx and following nitric oxide production, and subsequently decreases ATP production, leading to the dendritic bead formation. These findings provide novel mechanisms of neuronal excitotoxicity, which may occur in both inflammatory and neurodegenerative diseases in the CNS.


Subject(s)
Interferon-gamma/pharmacology , Multiprotein Complexes/metabolism , Neurons/pathology , Neurotoxicity Syndromes/etiology , Receptors, AMPA/metabolism , Receptors, Interferon/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Mice , Phosphorylation , Interferon gamma Receptor
20.
Science ; 316(5825): 758-61, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17478724

ABSTRACT

We found that, in the mouse visual cortex, action potentials generated in a single layer-2/3 pyramidal (excitatory) neuron can reliably evoke large, constant-latency inhibitory postsynaptic currents in other nearby pyramidal cells. This effect is mediated by axo-axonic ionotropic glutamate receptor-mediated excitation of the nerve terminals of inhibitory interneurons, which connect to the target pyramidal cells. Therefore, individual cortical excitatory neurons can generate inhibition independently from the somatic firing of inhibitory interneurons.


Subject(s)
Inhibitory Postsynaptic Potentials , Pyramidal Cells/physiology , Synapses/physiology , Visual Cortex/physiology , Action Potentials , Animals , Axons/metabolism , Electric Stimulation , Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Interneurons/physiology , Mice , Mice, Inbred C57BL , Neural Inhibition , Patch-Clamp Techniques , Presynaptic Terminals/physiology , Receptors, AMPA/physiology , Receptors, Kainic Acid/physiology , Synaptic Transmission , Visual Cortex/cytology , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...