Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Intellect Disabil Res ; 67(4): 362-374, 2023 04.
Article in English | MEDLINE | ID: mdl-36625000

ABSTRACT

BACKGROUND: Neurofibromatosis type 1 (NF1) is a neurodevelopmental genetic disorder associated with visual-spatial and visuomotor deficits, which have not been studied well in adults with NF1. METHODS: In 22 adults with NF1 and 31 controls, visuomotor functioning was assessed by measuring eye latency, hand latency and hand accuracy during visuomotor tasks. Visual-spatial functioning was assessed by measuring eye movement responses during the Visual Threshold Task. RESULTS: The NF1 group had a significantly shorter eye latency than the control group and was less accurate in their hand movements during specific visuomotor tasks. The groups showed no differences in eye movement responses during the Visual Threshold Task and in hand latency during the visuomotor tasks. CONCLUSIONS: In contrast to studies in children with NF1, we found no alterations in visual-spatial information processing in adults. Impairments in eye latency and hand accuracy during specific visuomotor tasks may indicate deficits in visuomotor functioning in adults with NF1.


Subject(s)
Neurofibromatosis 1 , Child , Humans , Adult , Neurofibromatosis 1/complications , Eye Movements , Visual Perception/physiology , Hand , Psychomotor Performance/physiology
2.
Sci Rep ; 12(1): 19041, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36351971

ABSTRACT

Individuals with Neurofibromatosis type 1 (NF1) experience a high degree of motor problems. The cerebellum plays a pivotal role in motor functioning and the NF1 gene is highly expressed in cerebellar Purkinje cells. However, it is not well understood to what extent NF1 affects cerebellar functioning and how this relates to NF1 motor functioning. Therefore, we subjected global Nf1+/- mice to a cerebellum-dependent associative learning task, called Pavlovian eyeblink conditioning. Additionally, we assessed general motor function and muscle strength in Nf1+/- mice. To our surprise, we found that Nf1+/- mice showed a moderately increased learning rate of conditioned eyeblink responses, as well as improved accuracy in the adaptive timing of the eyeblink responses. Locomotion, balance, general motor function, and muscle strength were not affected in Nf1+/- mice. Together, our results support the view that cerebellar function in Nf1+/- mice is unimpaired.


Subject(s)
Neurofibromatosis 1 , Mice , Animals , Neurofibromatosis 1/genetics , Cerebellum/physiology , Conditioning, Classical/physiology , Purkinje Cells/physiology , Blinking
3.
Clin Neurophysiol ; 142: 220-227, 2022 10.
Article in English | MEDLINE | ID: mdl-36081238

ABSTRACT

OBJECTIVE: The inability to properly process visual information has been frequently associated with neurofibromatosis type 1 (NF1). Based on animal studies, the cause of cognitive disabilities in NF1 is hypothesized to arise from decreased synaptic plasticity. Visual cortical plasticity in humans can be investigated by studying visual evoked potentials (VEPs) in response to visual stimulation. METHODS: VEP plasticity was assessed by measuring the increase of the peak amplitudes C1, P1, and N1 induced by 10-min modulation of checkerboard reversals in 22 adult NF1 patients and 30 controls. VEP signals were recorded pre-modulation, during modulation, and at 2, 7, 12, 17, 22, 27 min post-modulation. RESULTS: The P1 amplitude increased significantly comparing post-modulation to pre-modulation in the control group. This potentiation was not observed in the NF1 group. CONCLUSIONS: Visual cortical plasticity could be measured using VEPs in response to visual stimulation in the control group. Individuals with NF1 may have reduced visual cortical plasticity, as indicated by their non-potentiated response to VEP induction. These findings should be interpreted with caution due to high inter-subject variability. SIGNIFICANCE: The present study contributes to an improved assessment of the feasibility for using neurophysiological outcome measures in intervention studies of cognitive deficits among patients with NF1.


Subject(s)
Neurofibromatosis 1 , Visual Cortex , Adult , Animals , Evoked Potentials, Visual , Humans , Neuronal Plasticity/physiology , Photic Stimulation
4.
J Autism Dev Disord ; 48(7): 2278-2285, 2018 07.
Article in English | MEDLINE | ID: mdl-29423604

ABSTRACT

In a non-selected sample of children with Neurofibromatosis type 1 (NF1) the prevalence rate of autism spectrum disorder (ASD) and predictive value of an observational (ADOS)-and questionnaire-based screening instrument were assessed. Complete data was available for 128 children. The prevalence rate for clinical ASD was 10.9%, which is clearly higher than in the general population. This prevalence rate is presumably more accurate than in previous studies that examined children with NF1 with an ASD presumption or solely based on screening instruments. The combined observational- and screening based classifications demonstrated the highest positive predictive value for DSM-IV diagnosis, highlighting the importance of using both instruments in children with NF1.


Subject(s)
Autism Spectrum Disorder/epidemiology , Neurofibromatosis 1/complications , Child , Cohort Studies , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , Male , Neurofibromatosis 1/epidemiology , Prevalence
5.
Nat Commun ; 8(1): 1052, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29051493

ABSTRACT

De novo mutations in specific mTOR pathway genes cause brain overgrowth in the context of intellectual disability (ID). By analyzing 101 mMTOR-related genes in a large ID patient cohort and two independent population cohorts, we show that these genes modulate brain growth in health and disease. We report the mTOR activator gene RHEB as an ID gene that is associated with megalencephaly when mutated. Functional testing of mutant RHEB in vertebrate animal models indicates pathway hyperactivation with a concomitant increase in cell and head size, aberrant neuronal migration, and induction of seizures, concordant with the human phenotype. This study reveals that tight control of brain volume is exerted through a large community of mTOR-related genes. Human brain volume can be altered, by either rare disruptive events causing hyperactivation of the pathway, or through the collective effects of common alleles.


Subject(s)
Brain/anatomy & histology , Intellectual Disability/genetics , Megalencephaly/genetics , Mutation , Ras Homolog Enriched in Brain Protein/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Movement , Cell Size , Cells, Cultured , Humans , Intellectual Disability/pathology , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Organ Size , Seizures/genetics , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Zebrafish/genetics
6.
J Neurol ; 264(1): 161-167, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878438

ABSTRACT

Cognitive development in patients with tuberous sclerosis complex is highly variable. Predictors in the infant years would be valuable to counsel parents and to support development. The aim of this study was to confirm factors that have been reported to be independently correlated with cognitive development. 102 patients included in this study were treated at the ENCORE-TSC expertise center of the Erasmus Medical Center-Sophia Children's Hospital. Data from the first 24 months of life were used, including details on epilepsy, motor development and mutation status. Outcome was defined as cognitive development (intellectual equivalent, IE) as measured using tests appropriate to the patients age and cognitive abilities (median age at testing 8.2 years, IQR 4.7-12.0). Univariable and multivariable regression analyses were used. In a univariable analysis, predictors of lower IE were: the presence of infantile spasms (ß = -18.3, p = 0.000), a larger number of antiepileptic drugs used (ß = -6.3, p = 0.000), vigabatrin not used as first drug (ß = -14.6, p = 0.020), corticosteroid treatment (ß = -33.2, p = 0.005), and a later age at which the child could walk independently (ß = -2.1, p = 0.000). An older age at seizure onset predicted higher IE (ß = 1.7, p = 0.000). In a multivariable analysis, only age at seizure onset was significantly correlated to IE (ß = 1.2, p = 0.005), contributing to 28% of the variation in IE. In our cohort, age at seizure onset was the only variable that independently predicted IE. Factors predicting cognitive development could aid parents and physicians in finding the appropriate support and schooling for these patients.


Subject(s)
Cognition , Intelligence , Tuberous Sclerosis/diagnosis , Tuberous Sclerosis/psychology , Age of Onset , Child , Child Development , Child, Preschool , Epilepsy/diagnosis , Epilepsy/epidemiology , Epilepsy/psychology , Epilepsy/therapy , Female , Follow-Up Studies , Humans , Intellectual Disability/diagnosis , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Intelligence Tests , Male , Multivariate Analysis , Prognosis , Psychology, Child , Regression Analysis , Retrospective Studies , Tuberous Sclerosis/epidemiology , Tuberous Sclerosis/therapy
8.
Adv Healthc Mater ; 5(7): 850-62, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-26845073

ABSTRACT

Recent discoveries indicate that during neuronal development the signaling processes that regulate extracellular sensing (e.g., adhesion, cytoskeletal dynamics) are important targets for ubiquitination-dependent regulation, in particular through E3 ubiquitin ligases. Among these, Ubiquitin E3a ligase (UBE3A) has a key role in brain functioning, but its function and how its deficiency results in the neurodevelopmental disorder Angelman syndrome is still unclear. Here, the role of UBE3A is investigated in neurite contact guidance during neuronal development, in vitro. The microtopography sensing of wild-type and Ube3a-deficient hippocampal neurons is studied by exploiting gratings with different topographical characteristics, with the aim to compare their capabilities to read and follow physical directional stimuli. It is shown that neuronal contact guidance is defective in Ube3a-deficient neurons, and this behavior is linked to an impaired activation of the focal adhesion signaling pathway. Taken together, the results suggest that the neuronal contact sensing machinery might be affected in Angelman syndrome.


Subject(s)
Hippocampus/cytology , Nanostructures/chemistry , Neurites/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin/metabolism , Animals , Cell Nucleus/metabolism , Cell Nucleus Shape , Cell Shape , Female , Focal Adhesions/metabolism , Male , Mice , Nanostructures/ultrastructure , Tubulin/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Mol Psychiatry ; 21(3): 364-75, 2016 03.
Article in English | MEDLINE | ID: mdl-25802982

ABSTRACT

Memories are encoded within sparsely distributed neuronal ensembles. However, the defining cellular properties of neurons within a memory trace remain incompletely understood. Using a fluorescence-based Arc reporter, we were able to visually identify the distinct subset of lateral amygdala (LA) neurons activated during auditory fear conditioning. We found that Arc-expressing neurons have enhanced intrinsic excitability and are preferentially recruited into newly encoded memory traces. Furthermore, synaptic potentiation of thalamic inputs to the LA during fear conditioning is learning-specific, postsynaptically mediated and highly localized to Arc-expressing neurons. Taken together, our findings validate the immediate-early gene Arc as a molecular marker for the LA neuronal ensemble recruited during fear learning. Moreover, these results establish a model of fear memory formation in which intrinsic excitability determines neuronal selection, whereas learning-related encoding is governed by synaptic plasticity.


Subject(s)
Basolateral Nuclear Complex/metabolism , Conditioning, Classical/physiology , Cytoskeletal Proteins/metabolism , Fear/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Acoustic Stimulation/adverse effects , Action Potentials/drug effects , Action Potentials/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basolateral Nuclear Complex/cytology , Central Nervous System Stimulants/pharmacology , Choline O-Acetyltransferase/metabolism , Cytoskeletal Proteins/genetics , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurons/physiology , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , Picrotoxin/pharmacology , Proto-Oncogene Proteins c-fos/metabolism
11.
Mol Psychiatry ; 20(11): 1311-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25917366

ABSTRACT

Cognitive impairments are a major clinical feature of the common neurogenetic disease neurofibromatosis type 1 (NF1). Previous studies have demonstrated that increased neuronal inhibition underlies the learning deficits in NF1, however, the molecular mechanism underlying this cell-type specificity has remained unknown. Here, we identify an interneuron-specific attenuation of hyperpolarization-activated cyclic nucleotide-gated (HCN) current as the cause for increased inhibition in Nf1 mutants. Mechanistically, we demonstrate that HCN1 is a novel NF1-interacting protein for which loss of NF1 results in a concomitant increase of interneuron excitability. Furthermore, the HCN channel agonist lamotrigine rescued the electrophysiological and cognitive deficits in two independent Nf1 mouse models, thereby establishing the importance of HCN channel dysfunction in NF1. Together, our results provide detailed mechanistic insights into the pathophysiology of NF1-associated cognitive defects, and identify a novel target for clinical drug development.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neurofibromatosis 1/complications , Potassium Channels/metabolism , Animals , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Hippocampus/cytology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Lamotrigine , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Potassium Channels/genetics , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Triazines/therapeutic use
12.
Genes Brain Behav ; 10(4): 404-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21352477

ABSTRACT

Neurofibromatosis type 1 (NF1) is an autosomal dominantly inherited disease, characterized by various neurocutaneous symptoms, cognitive impairments and problems in fine and gross motor performance. Although cognitive deficits in NF1 have been attributed to increased release of the inhibitory neurotransmitter γ-amino butyric acid (GABA) in the hippocampus, the origin of the motor deficits is unknown. Cerebellar Purkinje cells, the sole output neurons of the cerebellar cortex, are GABAergic neurons and express neurofibromin at high levels, suggesting an important role for the cerebellum in the observed motor deficits in NF1. To test this, we determined the cerebellar contribution to motor problems in Nf1(+/-) mice, a validated mouse model for NF1. Using the Rotarod, a non-specific motor performance test, we confirmed that, like NF1 patients, Nf1(+/-) mice have motor deficits. Next, to evaluate the role of the cerebellum in these deficits, mice were subjected to cerebellum-specific motor performance and learning tests. Nf1(+/-) mice showed no impairment on the Erasmus ladder, as step time and number of missteps were not different. Furthermore, when compensatory eye movements were tested, no performance deficits were found in the optokinetic reflex and vestibulo-ocular reflex in the dark (VOR) or in the light (VVOR). Finally, Nf1(+/-) mice successfully completed short- and long-term VOR adaptation paradigms, tests that both depend on cerebellar function. Thus, despite the confirmed presence of motor performance problems in Nf1(+/-) mice, we found no indication of a cerebellar component. These results, combined with recent clinical data, suggest that cerebellar function is not overtly affected in NF1 patients.


Subject(s)
Cerebellum/physiopathology , Motor Activity/genetics , Motor Skills Disorders/etiology , Neurofibromatosis 1/genetics , Purkinje Cells/physiology , Animals , Eye Movements/genetics , Genes, Neurofibromatosis 1 , Hand Strength/physiology , Heterozygote , Learning/physiology , Mice , Mice, Neurologic Mutants , Motor Skills Disorders/genetics , Motor Skills Disorders/physiopathology , Neurofibromin 1/genetics , Rotarod Performance Test
13.
Neuron ; 67(4): 618-28, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20797538

ABSTRACT

Cerebellar motor learning is required to obtain procedural skills. Studies have provided supportive evidence for a potential role of kinase-mediated long-term depression (LTD) at the parallel fiber to Purkinje cell synapse in cerebellar learning. Recently, phosphatases have been implicated in the induction of potentiation of Purkinje cell activities in vitro, but it remains to be shown whether and how phosphatase-mediated potentiation contributes to motor learning. Here, we investigated its possible role by creating and testing a Purkinje cell-specific knockout of calcium/calmodulin-activated protein-phosphatase-2B (L7-PP2B). The selective deletion of PP2B indeed abolished postsynaptic long-term potentiation in Purkinje cells and their ability to increase their excitability, whereas LTD was unaffected. The mutants showed impaired "gain-decrease" and "gain-increase" adaptation of their vestibulo-ocular reflex (VOR) as well as impaired acquisition of classical delay conditioning of their eyeblink response. Thus, our data indicate that PP2B may indeed mediate potentiation in Purkinje cells and contribute prominently to cerebellar motor learning.


Subject(s)
Calcineurin/metabolism , Learning/physiology , Long-Term Potentiation/physiology , Motor Activity/physiology , Purkinje Cells/physiology , Action Potentials/physiology , Adaptation, Psychological/physiology , Animals , Calcineurin/genetics , Cerebellum/cytology , Cerebellum/physiology , Conditioning, Classical/physiology , Conditioning, Eyelid/physiology , Long-Term Synaptic Depression/physiology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/physiology , Purkinje Cells/cytology , Reflex, Vestibulo-Ocular/physiology , Time Factors
14.
Eur J Neurosci ; 27(10): 2542-50, 2008 May.
Article in English | MEDLINE | ID: mdl-18547242

ABSTRACT

Corticosterone (100 nm) rapidly increases the frequency of miniature excitatory postsynaptic currents in mouse CA1 pyramidal neurons via membrane-located mineralocorticoid receptors (MRs). We now show that a presynaptic ERK1/2 signalling pathway mediates the nongenomic effect, as it was blocked by the MEK inhibitors U0126 (10 microm) and PD098059 (40 microm) and occluded in H-Ras(G12V)-mutant mice with constitutive activation of the ERK1/2 presynaptic pathway. Notably, the increase in mEPSC frequency was not mediated by retrograde signalling through endocannabinoids or nitric oxide, supporting presynaptic localization of the signalling pathway. Unexpectedly, corticosterone was also found to have a direct postsynaptic effect, rapidly decreasing the peak amplitude of I(A) currents. This effect takes place via postsynaptic membrane MRs coupled to a G protein-mediated pathway, as the effect of corticosterone on I(A) was effectively blocked by 0.5 mm GDP-beta-S administered via the recording pipette into the postsynaptic cell. Taken together, these results indicate that membrane MRs mediate rapid, nongenomic effects via pre- as well as postsynaptic pathways. Through these dual pathways, high corticosterone concentrations such as occur after stress could contribute to enhanced CA1 pyramidal excitability.


Subject(s)
Adrenal Cortex Hormones/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Synaptic Membranes/metabolism , Synaptic Transmission/physiology , Animals , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Hippocampus/ultrastructure , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Mineralocorticoid/drug effects , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Synaptic Membranes/drug effects , Synaptic Transmission/drug effects , Thionucleotides/pharmacology
15.
AJNR Am J Neuroradiol ; 29(4): 816-22, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18339726

ABSTRACT

BACKGROUND AND PURPOSE: Hyperintensities on T2-weighted images are seen in the brains of most patients with neurofibromatosis type I (NF-1), but the origin of these unidentified bright objects (UBOs) remains obscure. In the current study, we examined the diffusion characteristics of brain tissue in children with NF-1 to test the hypothesis that a microstructural abnormality is present in NF-1. MATERIALS AND METHODS: Diffusion tensor imaging (DTI) was performed in 50 children with NF-1 and 8 controls. Circular regions of interest were manually placed in 7 standardized locations in both hemispheres, including UBO sites. Apparent diffusion coefficients (ADC), fractional anisotropy (FA), and axial anisotropy (A(m)) were used to differentiate quantitatively between healthy and disordered brain matter. Differences in eigenvalues (lambda(1), lambda(2), lambda(3)) were determined to examine parenchymal integrity. RESULTS: We found higher ADC values for UBOs than for normal-appearing sites (P < .01) and higher ADC values for normal-appearing sites than for controls (P < .04 in 5 of 7 regions). In most regions, we found no differences in FA or A(m). Eigenvalues lambda(2) and lambda(3) were higher at UBO sites than in normal-appearing sites (P < .04). CONCLUSION: With ADC, it was possible to differentiate quantitatively between normal- and abnormal-appearing brain matter in NF-1 and also between normal-appearing brain matter in NF-1 and healthy brain matter in controls, indicating subtle pathologic damage disrupting the tissue microstructure in the NF-1 brain. Higher diffusivity for lambda(1), lambda(2), and lambda(3) indicates that this disturbance of microstructure is caused by accumulation of fluid or vacuolation.


Subject(s)
Brain/pathology , Diffusion Magnetic Resonance Imaging , Neurofibromatosis 1/pathology , Adolescent , Child , Female , Humans , Male , Observer Variation
16.
Neuroscience ; 151(1): 43-55, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18082335

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are critical determinants of bidirectional synaptic plasticity, however, studies of NMDAR function have been based primarily on pharmacological and electrophysiological manipulations, and it is still debated whether there are subunit-selective forms of long-term potentiation (LTP) and long-term depression (LTD). Here we provide ultrastructural analyses of axospinous synapses in cornu ammonis field 1 of hippocampus (CA1) stratum radiatum of transgenic mice with mutations to two key underlying postsynaptic density (PSD) proteins, postsynaptic density protein 95 (PSD-95) and the alpha-isoform of calcium-calmodulin-dependent protein kinase II (alphaCaMKII). Distribution profiles of synaptic proteins in these mice reveal very different patterns of subunit-specific NMDAR localization, which may be related to the divergent phenotypes of the two mutants. In PSD-95, Dlg, ZO-1/Dlg-homologous region (PDZ) 3-truncated mutant mice in which LTD could not be induced but LTP was found to be enhanced, we found a subtle, yet preferential displacement of synaptic N-methyl-d-aspartate receptor subunit 2B (NR2B) subunits in lateral regions of the synapse without affecting changes in the localization of N-methyl-d-aspartate receptor subunit 2A (NR2A) subunits. In persistent inhibitory alphaCaMKII Thr305 substituted with Asp in alpha-isoform of calcium-calmodulin kinase II (T305D) mutant mice with severely impaired LTP but stable LTD expression, we found a selective reduction of NR2A subunits at both the synapse and throughout the cytoplasm of the spine without any effect on the NR2B subunit. In an experiment of mutual exclusivity, neither PSD-95 nor alphaCaMKII localization was found to be affected by mutations to the corresponding PSD protein suggesting that they are functionally independent of the other in the regulation of NR2A- and NR2B-containing NMDARs preceding synaptic activity. Consequently, there may exist at least two distinct PSD-95 and alphaCaMKII-specific NMDAR complexes involved in mediating LTP and LTD through opposing signal transduction pathways in synapses of the hippocampus. The contrasting phenotypes of the PSD-95 and alphaCaMKII mutant mice further establish the prospect of an independent and, possibly, competing mechanism for the regulation of NMDAR-dependent bidirectional synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Receptors, N-Methyl-D-Aspartate/biosynthesis , Synapses/metabolism , Animals , Antibody Specificity , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disks Large Homolog 4 Protein , Guanylate Kinases , Image Processing, Computer-Assisted , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Isomerism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation/physiology , Neuronal Plasticity/physiology , Phenotype
17.
Science ; 301(5640): 1736-9, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-14500987

ABSTRACT

Mammals can be trained to make a conditioned movement at a precise time, which is correlated to the interval between the conditioned stimulus and unconditioned stimulus during the learning. This learning-dependent timing has been shown to depend on an intact cerebellar cortex, but which cellular process is responsible for this form of learning remains to be demonstrated. Here, we show that protein kinase C-dependent long-term depression in Purkinje cells is necessary for learning-dependent timing of Pavlovian-conditioned eyeblink responses.


Subject(s)
Blinking , Cerebellum/physiology , Conditioning, Eyelid , Learning , Long-Term Synaptic Depression , Purkinje Cells/physiology , Animals , Electroshock , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , N-Methylaspartate/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Time Factors
18.
Ann N Y Acad Sci ; 978: 391-404, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12582068

ABSTRACT

Electrotonic coupling by gap junctions between neurons in the inferior olive has been claimed to underly complex spike (CS) synchrony of Purkinje cells in the cerebellar cortex and thereby to play a role in the coordination of movements. Here, we investigated the motor performance of mice that lack connexin36 (Cx36), which appears necessary for functional olivary gap junctions. Cx36 null-mutants are not ataxic, they show a normal performance on the accelerating rotorod, and they have a regular walking pattern. In addition, they show normal compensatory eye movements during sinusoidal visual and/or vestibular stimulation. To find out whether the normal motor performance in mutants reflects normal CS activity or some compensatory mechanism downstream of the cerebellar cortex, we determined the CS firing rate, climbing-fiber pause, and degree of CS synchrony. None of these parameters in the mutants differed from those in wildtype littermates. Finally, we investigated whether the role of coupling becomes apparent under challenging conditions, such as during application of the tremorgenic drug harmaline, which specifically turns olivary neurons into an oscillatory state at a high frequency. In both the mutants and wildtypes this application induced tremors of a similar duration with similar peak frequencies and amplitudes. Thus surprisingly, the present data does not support the notion that electrotonic coupling by gap junctions underlies synchronization of olivary spike activity and that these gap junctions are essential for normal motor performance.


Subject(s)
Action Potentials/physiology , Connexins/deficiency , Gap Junctions/physiology , Olivary Nucleus/physiology , Psychomotor Performance/physiology , Action Potentials/drug effects , Animals , Connexins/genetics , Eye Proteins/genetics , Gap Junctions/drug effects , Mice , Mice, Knockout , Mice, Neurologic Mutants , Olivary Nucleus/drug effects , Psychomotor Performance/drug effects , Gap Junction delta-2 Protein
20.
Biol Psychiatry ; 47(3): 200-9, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10682217

ABSTRACT

Recent studies on the molecular and cellular basis of learning and memory have brought us closer than ever to understanding the mechanisms of synaptic plasticity and their relevance to memory formation. Genetic approaches have played a central role in these new findings because the same mutant mice can be studied with molecular, cellular, circuit, and behavioral tools. Therefore, the results can be used to construct models that cut across levels of analytical complexity, forging connections from the biochemistry of the modified protein to the behavior of the mutant mice. These findings are not only improving our understanding of learning and memory, they are also enriching our understanding of cognitive disorders, such as neurofibromatosis type I. Mechanisms underlying long-term changes in synaptic function are likely to be at the heart of many cognitive and emotional processes in humans. Therefore, molecular and cellular insights into learning and memory undoubtedly will have a profound impact on the understanding and treatment of psychiatric disorders.


Subject(s)
Cognition Disorders/genetics , Depressive Disorder/genetics , Schizophrenia/genetics , Animals , Brain/physiopathology , Cognition Disorders/physiopathology , Depressive Disorder/physiopathology , Humans , Mental Recall/physiology , Mice , Mice, Mutant Strains , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Schizophrenia/physiopathology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL