Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 58
1.
Clin Genet ; 97(1): 125-137, 2020 01.
Article En | MEDLINE | ID: mdl-30873608

Neurexins are presynaptic cell adhesion molecules critically involved in synaptogenesis and vesicular neurotransmitter release. They are encoded by three genes (NRXN1-3), each yielding a longer alpha (α) and a shorter beta (ß) transcript. Deletions spanning the promoter and the initial exons of the NRXN1 gene, located in chromosome 2p16.3, are associated with a variety of neurodevelopmental, psychiatric, neurological and neuropsychological phenotypes. We have performed a systematic review to define (a) the clinical phenotypes most associated with mono-allelic exonic NRXN1 deletions, and (b) the phenotypic features of NRXN1 bi-allelic deficiency due to compound heterozygous deletions/mutations. Clinically, three major conclusions can be drawn: (a) incomplete penetrance and pleiotropy do not allow reliable predictions of clinical outcome following prenatal detection of mono-allelic exonic NRXN1 deletions. Newborn carriers should undergo periodic neuro-behavioral observations for the timely detection of warning signs and the prescription of early behavioral intervention; (b) the presence of additional independent genetic risk factors should always be sought, as they may influence prognosis; (c) children with exonic NRXN1 deletions displaying early-onset, severe psychomotor delay in the context of a Pitt-Hopkins-like syndrome 2 phenotype, should undergo DNA sequencing of the spared NRXN1 allele in search for mutations or very small insertions/deletions.


Calcium-Binding Proteins/genetics , Cell Adhesion Molecules, Neuronal/genetics , Genetic Predisposition to Disease , Neural Cell Adhesion Molecules/genetics , Neurodevelopmental Disorders/genetics , Calcium-Binding Proteins/deficiency , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Mental Disorders/genetics , Mental Disorders/pathology , Mutation , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Neural Cell Adhesion Molecules/deficiency , Neurodevelopmental Disorders/pathology , Phenotype
2.
Proc Natl Acad Sci U S A ; 115(23): E5382-E5389, 2018 06 05.
Article En | MEDLINE | ID: mdl-29784826

Leucine-rich repeat transmembrane (LRRTM) proteins are synaptic cell adhesion molecules that influence synapse formation and function. They are genetically associated with neuropsychiatric disorders, and via their synaptic actions likely regulate the establishment and function of neural circuits in the mammalian brain. Here, we take advantage of the generation of a LRRTM1 and LRRTM2 double conditional knockout mouse (LRRTM1,2 cKO) to examine the role of LRRTM1,2 at mature excitatory synapses in hippocampal CA1 pyramidal neurons. Genetic deletion of LRRTM1,2 in vivo in CA1 neurons using Cre recombinase-expressing lentiviruses dramatically impaired long-term potentiation (LTP), an impairment that was rescued by simultaneous expression of LRRTM2, but not LRRTM4. Mutation or deletion of the intracellular tail of LRRTM2 did not affect its ability to rescue LTP, while point mutations designed to impair its binding to presynaptic neurexins prevented rescue of LTP. In contrast to previous work using shRNA-mediated knockdown of LRRTM1,2, KO of these proteins at mature synapses also caused a decrease in AMPA receptor-mediated, but not NMDA receptor-mediated, synaptic transmission and had no detectable effect on presynaptic function. Imaging of recombinant photoactivatable AMPA receptor subunit GluA1 in the dendritic spines of cultured neurons revealed that it was less stable in the absence of LRRTM1,2. These results illustrate the advantages of conditional genetic deletion experiments for elucidating the function of endogenous synaptic proteins and suggest that LRRTM1,2 proteins help stabilize synaptic AMPA receptors at mature spines during basal synaptic transmission and LTP.


CA1 Region, Hippocampal/physiology , Long-Term Potentiation/physiology , Neural Cell Adhesion Molecules/deficiency , Pyramidal Cells/physiology , Receptors, AMPA/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials/physiology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
3.
Cell Rep ; 21(13): 3637-3645, 2017 12 26.
Article En | MEDLINE | ID: mdl-29281813

Synaptopathies contributing to neurodevelopmental disorders are linked to mutations in synaptic organizing molecules, including postsynaptic neuroligins, presynaptic neurexins, and MDGAs, which regulate their interaction. The role of MDGA1 in suppressing inhibitory versus excitatory synapses is controversial based on in vitro studies. We show that genetic deletion of MDGA1 in vivo elevates hippocampal CA1 inhibitory, but not excitatory, synapse density and transmission. Furthermore, MDGA1 is selectively expressed by pyramidal neurons and regulates perisomatic, but not distal dendritic, inhibitory synapses. Mdga1-/- hippocampal networks demonstrate muted responses to neural excitation, and Mdga1-/- mice are resistant to induced seizures. Mdga1-/- mice further demonstrate compromised hippocampal long-term potentiation, consistent with observed deficits in spatial and context-dependent learning and memory. These results suggest that mutations in MDGA1 may contribute to cognitive deficits through altered synaptic transmission and plasticity by loss of suppression of inhibitory synapse development in a subcellular domain- and cell-type-selective manner.


Cognition , Nerve Net/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Inhibition , Synapses/metabolism , Animals , CA1 Region, Hippocampal/pathology , Gene Deletion , Long-Term Potentiation , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Synapses/ultrastructure , Synaptic Transmission
4.
Sci Rep ; 7(1): 13631, 2017 10 19.
Article En | MEDLINE | ID: mdl-29051583

The neural cell adhesion molecule (NCAM) is the major carrier of polysialic acid (PSA) which modulates NCAM functions of neural cells at the cell surface. In previous studies, we have shown that stimulation of cultured neurons with surrogate NCAM ligands leads to the generation and nuclear import of PSA-lacking and -carrying NCAM fragments. Here, we show that the nuclear import of the PSA-carrying NCAM fragment is mediated by positive cofactor 4 and cofilin, which we identified as novel PSA-binding proteins. In the nucleus, the PSA-carrying NCAM fragment interacts via PSA with PC4 and cofilin, which are involved in RNA polymerase II-dependent transcription. Microarray analysis revealed that the nuclear PSA-carrying and -lacking NCAM fragments affect expression of different genes. By qPCR and immunoblot analysis we verified that the nuclear PSA-carrying NCAM fragment increases mRNA and protein expression of nuclear receptor subfamily 2 group F member 6, whereas the PSA-lacking NCAM fragment increases mRNA and protein expression of low density lipoprotein receptor-related protein 2 and α-synuclein. Differential gene expression evoked by nuclear NCAM fragments without and with PSA indicates that PSA-carrying and -lacking NCAM play different functional roles in the nervous system.


Neural Cell Adhesion Molecules/metabolism , Sialic Acids/metabolism , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
5.
Behav Brain Res ; 317: 311-318, 2017 01 15.
Article En | MEDLINE | ID: mdl-27693610

Neurotrimin (Ntm) belongs to the IgLON family of cell adhesion molecules with Lsamp, Obcam and kilon that regulate the outgrowth of neurites mostly by forming heterodimers. IgLONs have been associated with psychiatric disorders, intelligence, body weight, heart disease and tumours. This study provides an initial behavioural and pharmacological characterization of the phenotype of Ntm-deficient mice. We expected to see at least some overlap with the phenotype of Lsamp-deficient mice as Ntm and Lsamp are the main interaction partners in the IgLON family and are colocalized in some brain regions. However, Ntm-deficient mice displayed none of the deviations in behaviour that we have previously shown in Lsamp-deficient mice, but differently from Lsamp-deficient mice, had a deficit in emotional learning in the active avoidance task. The only overlap was decreased sensitivity to the locomotor stimulating effect of amphetamine in both knockout models. Thus, despite being interaction partners, on the behavioural level Lsamp seems to play a much more central role than Ntm and the roles of these two proteins seem to be complementary rather than overlapping.


Emotions/physiology , Learning Disabilities/genetics , Neural Cell Adhesion Molecules/deficiency , Amphetamine/pharmacology , Animals , Avoidance Learning/physiology , Body Weight/drug effects , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Central Nervous System Stimulants/pharmacology , Conditioning, Psychological/drug effects , Dark Adaptation/drug effects , Dark Adaptation/genetics , Disease Models, Animal , Exploratory Behavior/drug effects , Fasting/physiology , Fear/drug effects , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , Locomotion/drug effects , Locomotion/genetics , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/genetics , Reflex/genetics
6.
Glia ; 64(8): 1314-30, 2016 08.
Article En | MEDLINE | ID: mdl-27159043

Microglia are tissue macrophages and mediators of innate immune responses in the brain. The protein-modifying glycan polysialic acid (polySia) is implicated in modulating microglia activity. Cultured murine microglia maintain a pool of Golgi-confined polySia, which is depleted in response to lipopolysaccharide (LPS)-induced activation. Polysialylated neuropilin-2 (polySia-NRP2) contributes to this pool but further polySia protein carriers have remained elusive. Here, we use organotypic brain slice cultures to demonstrate that injury-induced activation of microglia initiates Golgi-confined polySia expression in situ. An unbiased glycoproteomic approach with stem cell-derived microglia identifies E-selectin ligand-1 (ESL-1) as a novel polySia acceptor. Together with polySia-NRP2, polySia-ESL-1 is also detected in primary cultured microglia, in brain slice cultures and in phorbol ester-induced THP-1 macrophages. Induction of stem cell-derived microglia, activated microglia in brain slice cultures and THP-1 macrophages by LPS, but not interleukin-4, causes polySia depletion and, as shown for stem cell-derived microglia, a metalloproteinase-dependent release of polySia-ESL-1 and polySia-NRP2. Moreover, soluble polySia attenuates LPS-induced production of nitric oxide and proinflammatory cytokines. Thus, shedding of polySia-ESL-1 and polySia-NRP2 after LPS-induced activation of microglia and THP-1 macrophages may constitute a mechanism for negative feedback regulation. GLIA 2016 GLIA 2016;64:1314-1330.


Lipopolysaccharides/toxicity , Macrophages/immunology , Microglia/immunology , Neuropilin-2/metabolism , Polysaccharides/toxicity , THP-1 Cells/immunology , Animals , Brain/immunology , Brain/pathology , Cells, Cultured , Cytokines/metabolism , Escherichia coli , Humans , Macrophages/pathology , Mice, Knockout , Microglia/pathology , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Nitric Oxide/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sialoglycoproteins/metabolism , Sialyltransferases/deficiency , Sialyltransferases/genetics , THP-1 Cells/pathology , Tissue Culture Techniques
7.
Cell Rep ; 14(3): 560-571, 2016 Jan 26.
Article En | MEDLINE | ID: mdl-26776515

The subventricular zone (SVZ) provides a specialized neurogenic microenvironment for proliferation and aggregation of basal progenitors (BPs). Our study reveals a mechanism for the aggregation of BPs within the SVZ required for their proliferation and generation of cortical layer neurons. The autism-related IgCAM, MDGA1, is locally expressed in the BP cell membrane where it co-localizes and complexes with the gap junction protein Connexin43. To address MDGA1 function, we created a floxed allele of MDGA1 and deleted it from BPs. MDGA1 deletion results in reduced BP proliferation and size of the SVZ, with an aberrant population of BPs ectopically positioned in the cortical plate. These defects are manifested in diminished production of cortical layer neurons and a significant reduction of the cortical layers. We conclude that MDGA1 functions to aggregate and maintain BPs within the SVZ providing the neurogenic niche required for their proliferation and generation of cortical layer neurons.


Lateral Ventricles/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Cell Differentiation , Cell Proliferation , Connexin 43/genetics , Connexin 43/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Stem Cells/cytology , Stem Cells/metabolism
8.
Behav Neurosci ; 129(6): 765-76, 2015 Dec.
Article En | MEDLINE | ID: mdl-26595880

The neurexins are a family of presynaptic cell adhesion molecules. Human genetic studies have found heterozygous deletions affecting NRXN1 and NRXN2, encoding α-neurexin I (Nrxn1α) and α-neurexin II (Nrxn2α), in individuals with autism spectrum disorders and schizophrenia. However, the link between α-neurexin deficiency and the manifestation of psychiatric disorders remain unclear. To assess whether the heterozygous loss of neurexins results in behaviors relevant to autism or schizophrenia, we used mice with heterozygous (HET) deletion of Nrxn1α or Nrxn2α. We found that in a test of social approach, Nrxn1α HET mice show no social memory for familiar versus novel conspecifics. In a passive avoidance test, female Nrxn1α HET mice cross to the conditioned chamber sooner than female wild-type and Nrxn2α HET mice. Nrxn2α HET mice also express a lack of long-term object discrimination, indicating a deficit in cognition. The observed Nrxn1α and Nrxn2α genotypic effects were specific, as neither HET deletion had effects on a wide range of other behavioral measures, including several measures of anxiety. Our findings demonstrate that the heterozygous loss of α-neurexin I and α-neurexin II in mice leads to phenotypes relevant to autism and schizophrenia.


Discrimination, Psychological/physiology , Learning/physiology , Memory/physiology , Nerve Tissue Proteins/deficiency , Neural Cell Adhesion Molecules/deficiency , Social Behavior , Animals , Anxiety/metabolism , Autistic Disorder , Calcium-Binding Proteins , Cognition/physiology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/genetics , Phenotype , Schizophrenic Psychology , Sequence Deletion
9.
Eur Neuropsychopharmacol ; 25(12): 2394-403, 2015 Dec.
Article En | MEDLINE | ID: mdl-26499173

Mood disorders are associated with alterations in serotonergic system, deficient BDNF (brain-derived neurotrophic factor) signaling and abnormal synaptic plasticity. Increased degradation and reduced functions of NCAM (neural cell adhesion molecule) have recently been associated with depression and NCAM deficient mice show depression-related behavior and impaired learning. The aim of the present study was to investigate potential changes in serotonergic and BDNF systems in NCAM knock-out mice. Serotonergic nerve fiber density and SERT (serotonin transporter) protein levels were robustly reduced in the hippocampus, prefrontal cortex and basolateral amygdala of adult NCAM(-)(/-) mice. This SERT reduction was already evident during early postnatal development. [(3)H]MADAM binding experiments further demonstrated reduced availability of SERT in cell membranes of NCAM(-)(/-) mice. Moreover, the levels of serotonin and its major metabolite 5-HIAA were down regulated in the brains of NCAM(-)(/-) mice. NCAM(-)(/-) mice also showed a dramatic reduction in the BDNF protein levels in the hippocampus and prefrontal cortex. This BDNF deficiency was associated with reduced phosphorylation of its receptor TrkB. Importantly, chronic administration of antidepressant amitriptyline partially or completely restored these changes in serotonergic and BDNF systems, respectively. In conclusion, NCAM deficiency lead to prominent and persistent abnormalities in brain serotonergic and BDNF systems, which likely contributes to the behavioral and neurobiological phenotype of NCAM(-/-) mice.


Adrenergic Uptake Inhibitors/therapeutic use , Amitriptyline/therapeutic use , Brain Diseases, Metabolic , Brain-Derived Neurotrophic Factor/metabolism , Neural Cell Adhesion Molecules/deficiency , Serotonin/metabolism , Animals , Brain/metabolism , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Disease Models, Animal , Electrochemical Techniques , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Cell Adhesion Molecules/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Binding/drug effects , Protein Binding/genetics , Receptor, trkB/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism
10.
Genes Brain Behav ; 13(5): 468-77, 2014 Jun.
Article En | MEDLINE | ID: mdl-24751161

The neural cell adhesion molecule (NCAM) has been implicated in the development and plasticity of neural circuits and the control of hippocampus- and amygdala-dependent learning and behaviour. Previous studies in constitutive NCAM null mutants identified emotional behaviour deficits related to disturbances of hippocampal and amygdala functions. Here, we studied these behaviours in mice conditionally deficient in NCAM in the postmigratory forebrain neurons. We report deficits in both innate and learned avoidance behaviours, as observed in elevated plus maze and passive avoidance tasks. In contrast, general locomotor activity, trait anxiety or neophobia were unaffected by the mutation. Altered avoidance behaviour of the conditional NCAM mutants was associated with a deficit in serotonergic signalling, as indicated by their reduced responsiveness to (±)-8-hydroxy-2-(dipropylamino)-tetralin-induced hypothermia. Another serotonin-dependent behaviour, namely intermale aggression that is massively increased in constitutively NCAM-deficient mice, was not affected in the forebrain-specific mutants. Our data suggest that genetically or environmentally induced changes of NCAM expression in the late postnatal and mature forebrain determine avoidance behaviour and serotonin (5-HT)1A receptor signalling.


Avoidance Learning , Neural Cell Adhesion Molecules/genetics , Prosencephalon/metabolism , Aggression , Animals , Dopamine Agonists/pharmacology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mutation , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/metabolism , Prosencephalon/drug effects , Prosencephalon/physiology
11.
Stress ; 16(6): 638-46, 2013 Nov.
Article En | MEDLINE | ID: mdl-24000815

The neural cell adhesion molecule (NCAM) plays a crucial role in stress-related brain function, emotional behavior and memory formation. In this study, we investigated the functions of the glucocorticoid and serotonergic systems in mice constitutively deficient for NCAM (NCAM-/- mice). Our data provide evidence for a hyperfunction of the hypothalamic-pituitary-adrenal axis, with enlarged adrenal glands and increased stress-induced corticosterone release, but reduced hippocampal glucocorticoid receptor expression in NCAM-/- mice when compared to NCAM+/+ mice. We also obtained evidence for a hypofunction of 5-HT1A autoreceptors as indicated by increased 8-0H-DPAT-induced hypothermia. These findings suggest a disturbance of both humoral and neural stress systems in NCAM-/- mice. Accordingly, we not only confirmed previously observed hyperarousal of NCAM-/- mice in various anxiety tests, but also observed an increased response to novelty exposure in these animals. Spatial learning deficits of the NCAM-/- mice in a Morris Water maze persisted, even when mice were pretrained to prevent effects of novelty or stress. We suggest that NCAM-mediated processes are involved in both novelty/stress-related emotional behavior and in cognitive function during spatial learning.


Hippocampus/metabolism , Maze Learning/physiology , Neural Cell Adhesion Molecules/deficiency , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Anxiety/chemically induced , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Hypothermia/chemically induced , Memory, Short-Term/physiology , Mice , Motor Activity/physiology , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/biosynthesis , Stress, Psychological/physiopathology
12.
Stress ; 16(6): 647-54, 2013 Nov.
Article En | MEDLINE | ID: mdl-24010949

The neural cell adhesion molecule (NCAM) is a key regulator of brain plasticity. Substantial evidence indicates that NCAM is down-regulated by exposure to sustained stress and chronic stress seems to lead to increased aggression. In addition, constitutional NCAM deletion in mice has been shown to lead to increased intermale aggression and altered emotionality Forebrain-specific postnatal NCAM knockout was previously shown to impair cognitive function, particularly when animals were exposed to subchronic stress, but the effects on emotional and social behavior remain unclear. In this study, we investigated the potential interplay of a forebrain-specific postnatal NCAM deletion and exposure to different lengths of repeated stress (i.e. subchronic: 14 days; chronic: 29 days) on aggressive and emotional behavior. Our results show that postnatal deletion of NCAM in the forebrain leads to increased aggression and altered emotionality depending on the duration of stress, whereas conditional NCAM knockout has no basal impact on these behaviors. These findings support the involvement of NCAM in the regulation of emotional and aggressive behaviors, suggesting that diminished NCAM expression might be a critical vulnerability factor for the development of these behavioral alterations under repeated exposure to stress.


Aggression/physiology , Neural Cell Adhesion Molecules/physiology , Prosencephalon/metabolism , Stress, Psychological/physiopathology , Animals , Anxiety/metabolism , Exploratory Behavior/physiology , Male , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency
13.
Learn Mem ; 20(4): 183-93, 2013 Mar 15.
Article En | MEDLINE | ID: mdl-23504516

Most of the mechanisms involved in neural plasticity support cognition, and aging has a considerable effect on some of these processes. The neural cell adhesion molecule (NCAM) of the immunoglobulin superfamily plays a pivotal role in structural and functional plasticity and is required to modulate cognitive and emotional behaviors. However, whether aging is associated with NCAM alterations that might contribute to age-related cognitive decline is not currently known. In this study, we determined whether conditional NCAM-deficient mice display increased vulnerability to age-related cognitive and emotional alterations. We assessed the NCAM expression levels in the hippocampus and medial prefrontal cortex (mPFC) and characterized the performance of adult and aged conditional NCAM-deficient mice and their age-matched wild-type littermates in a delayed matching-to-place test in the Morris water maze and a delayed reinforced alternation test in the T-maze. Although aging in wild-type mice is associated with an isoform-specific reduction of NCAM expression levels in the hippocampus and mPFC, these mice exhibited only mild impairments in working/episodic-like memory performance. However, aged conditional NCAM-deficient mice displayed pronounced impairments in both the delayed matching-to-place and the delayed reinforced alternation tests. Importantly, the deficits of aged NCAM-deficient mice in these working/episodic-like memory tasks could not be attributed to increased anxiety-like behaviors or to differences in locomotor activity. Taken together, these data indicate that reduced NCAM expression in the forebrain might be a critical factor for the occurrence of cognitive impairments during aging.


Aging/physiology , Cognition Disorders/genetics , Neural Cell Adhesion Molecules/deficiency , Analysis of Variance , Animals , Anxiety/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dark Adaptation/genetics , Exploratory Behavior/physiology , Female , Gene Expression Regulation/genetics , Hippocampus/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Cell Adhesion Molecules/genetics , Prefrontal Cortex/metabolism , Protein Isoforms/metabolism , Reinforcement, Psychology , Swimming/physiology , Swimming/psychology
14.
J Neurosci ; 32(7): 2263-75, 2012 Feb 15.
Article En | MEDLINE | ID: mdl-22396402

Neural cell adhesion molecule (NCAM) is the predominant carrier of the unusual glycan polysialic acid (PSA). Deficits in PSA and/or NCAM expression cause impairments in hippocampal long-term potentiation and depression (LTP and LTD) and are associated with schizophrenia and aging. In this study, we show that impaired LTP in adult NCAM-deficient (NCAM(-/-)) mice is restored by increasing the activity of the NMDA subtype of glutamate receptor (GluN) through either reducing the extracellular Mg2+ concentration or applying d-cycloserine (DCS), a partial agonist of the GluN glycine binding site. Pharmacological inhibition of the GluN2A subtype reduced LTP to the same level in NCAM(-/-) and wild-type (NCAM(+/+)) littermate mice and abolished the rescue by DCS in NCAM(-/-) mice, suggesting that the effects of DCS are mainly mediated by GluN2A. The insufficient contribution of GluN to LTD in NCAM(-/-) mice was also compensated for by DCS. Furthermore, impaired contextual and cued fear conditioning levels were restored in NCAM(-/-) mice by administration of DCS before conditioning. In 12-month-old NCAM(-/-), but not NCAM(+/+) mice, there was a decline in LTP compared with 3-month-old mice that could be rescued by DCS. In 24-month-old mice of both genotypes, there was a reduction in LTP that could be fully restored by DCS in NCAM(+/+) mice but only partially restored in NCAM(-/-) mice. Thus, several deficiencies of NCAM(-/-) mice can be ameliorated by enhancing GluN2A-mediated neurotransmission with DCS.


Aging/physiology , Learning/physiology , Neural Cell Adhesion Molecules/deficiency , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Age Factors , Aging/genetics , Animals , Cycloserine/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Knockout , Neural Inhibition/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Transmission/drug effects
15.
Brain Res ; 1447: 106-18, 2012 Apr 04.
Article En | MEDLINE | ID: mdl-22361116

The neural cell adhesion molecule (NCAM) plays an important role in brain plasticity. Using mice deficient in all isoforms of NCAM we have previously demonstrated that constitutive deficiency in the NCAM gene (NCAM-/-) resulted in cognitive impairment, anhedonic behaviour and a reduced ability to cope with stress. This was accompanied by reduced basal phosphorylation of the fibroblast growth factor receptor 1 (FGFR1) and reduced phosphorylation of calcium-calmodulin kinase (CaMK) II and IV and cAMP response element binding protein (CREB). The present study was aimed to investigate how partial deficiency in NCAM in mice (NCAM+/-) affected phenotype. We found that NCAM+/- mice showed a longer period of immobility in the tail suspension test, increased latency to feed in the novelty-suppressed feeding test and reduced preference for sucrose in sucrose preference test. Both NCAM+/- and NCAM-/- mice showed reduced extinction of contextual fear. In contrast to NCAM-/- mice, NCAM+/- mice did not demonstrate memory impairment in either object recognition or contextual fear conditioning tests. Levels of phosphorylated FGFR1 in the hippocampus and prefrontal/frontal cortex of NCAM+/- mice were partially reduced and no changes in the phosphorylation of CaMKII, CaMKIV or CREB in the hippocampus were found. We conclude that a constitutive partial reduction in NCAM proteins results in a behavioural phenotype related to depression without impairment in cognitive functions, also affecting the level of FGFR1 phosphorylation without major alterations in CaMKII and CaMKIV intracellular signalling. Partial reduction in FGFR1 phosphorylation might explain the observed behavioural phenotype in NCAM+/- mice.


Cognition/physiology , Depression/metabolism , Genetic Carrier Screening , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/physiology , Animals , Depression/genetics , Depression/psychology , Fear/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neural Cell Adhesion Molecules/genetics , Neural Inhibition/physiology
16.
Stress ; 15(2): 195-206, 2012 Mar.
Article En | MEDLINE | ID: mdl-21939373

Previous studies in rodents showed that chronic stress induces structural and functional alterations in several brain regions, including shrinkage of the hippocampus and the prefrontal cortex, which are accompanied by cognitive and emotional disturbances. Reduced expression of the neural cell adhesion molecule (NCAM) following chronic stress has been proposed to be crucially involved in neuronal retraction and behavioral alterations. Since NCAM gene polymorphisms and altered expression of alternatively spliced NCAM isoforms have been associated with bipolar depression and schizophrenia in humans, we hypothesized that reduced expression of NCAM renders individuals more vulnerable to the deleterious effects of stress on behavior. Here, we specifically questioned whether mice in which the NCAM gene is inactivated in the forebrain by cre-recombinase under the control of the calcium-calmodulin-dependent kinase II promoter (conditional NCAM-deficient mice), display increased vulnerability to stress. We assessed the evolving of depressive-like behaviors and spatial learning and memory impairments following a subchronic stress protocol (2 weeks) that does not result in behavioral dysfunction, nor in altered NCAM expression, in wild-type mice. Indeed, while no behavioral alterations were detected in wild-type littermates after subchronic stress, conditional NCAM-deficient mice showed increased immobility in the tail suspension test and deficits in reversal spatial learning in the water maze. These findings indicate that diminished NCAM expression might be a critical vulnerability factor for the development of behavioral alterations by stress and further support a functional involvement of NCAM in stress-induced cognitive and emotional disturbances.


Behavior, Animal/physiology , Neural Cell Adhesion Molecules/genetics , Stress, Psychological/physiopathology , Animals , Hippocampus/metabolism , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , Prosencephalon/metabolism , Prosencephalon/physiology
17.
PLoS One ; 6(7): e22716, 2011.
Article En | MEDLINE | ID: mdl-21818371

Recent genetic linkage analysis has shown that LRRTM1 (Leucine rich repeat transmembrane neuronal 1) is associated with schizophrenia. Here, we characterized Lrrtm1 knockout mice behaviorally and morphologically. Systematic behavioral analysis revealed reduced locomotor activity in the early dark phase, altered behavioral responses to novel environments (open-field box, light-dark box, elevated plus maze, and hole board), avoidance of approach to large inanimate objects, social discrimination deficit, and spatial memory deficit. Upon administration of the NMDA receptor antagonist MK-801, Lrrtm1 knockout mice showed both locomotive activities in the open-field box and responses to the inanimate object that were distinct from those of wild-type mice, suggesting that altered glutamatergic transmission underlay the behavioral abnormalities. Furthermore, administration of a selective serotonin reuptake inhibitor (fluoxetine) rescued the abnormality in the elevated plus maze. Morphologically, the brains of Lrrtm1 knockout mice showed reduction in total hippocampus size and reduced synaptic density. The hippocampal synapses were characterized by elongated spines and diffusely distributed synaptic vesicles, indicating the role of Lrrtm1 in maintaining synaptic integrity. Although the pharmacobehavioral phenotype was not entirely characteristic of those of schizophrenia model animals, the impaired cognitive function may warrant the further study of LRRTM1 in relevance to schizophrenia.


Cognition/physiology , Genetic Predisposition to Disease , Hippocampus/physiopathology , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Schizophrenia/genetics , Synapses/pathology , Adaptation, Psychological/drug effects , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Clozapine/administration & dosage , Clozapine/pharmacology , Clozapine/therapeutic use , Cognition/drug effects , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Environment , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Gene Targeting , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/ultrastructure , Membrane Proteins , Memory/drug effects , Memory/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/metabolism , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Synapses/drug effects , Synapses/ultrastructure
18.
J Biol Chem ; 286(29): 26127-37, 2011 Jul 22.
Article En | MEDLINE | ID: mdl-21628472

The neural cell adhesion molecule (NCAM) was recently shown to be involved in the progression of various tumors with diverse effects. We previously demonstrated that NCAM potentiates the cellular invasion and metastasis of melanoma. Here we further report that the growth of melanoma is obviously retarded when the expression of NCAM is silenced. We found that the proliferation of murine B16F0 melanoma cells, their colony formation on soft agar, and growth of transplanted melanoma in vivo are clearly inhibited by the introduction of NCAM siRNA. Interestingly, change of NCAM expression level is shown to regulate the activity of Wnt signaling molecule, ß-catenin, markedly. This novel machinery requires the function of FGF receptor and glycogen synthase kinase-3ß but is independent of the Wnt receptors, MAPK-Erk and PI3K/Akt pathways. In addition, NCAM is found to form a functional complex with ß-catenin, FGF receptor, and glycogen synthase kinase-3ß. Moreover, up-regulation of NCAM140 and NCAM180 appears more potent than NCAM120 in activation of ß-catenin, suggesting that the intracellular domain of NCAM is required for facilitating the ß-catenin signaling. Furthermore, the melanoma cells also exhibit distinct differentiation phenotypes with the NCAM silencing. Our findings reveal a novel regulatory role of NCAM in the progression of melanoma that might serve as a new therapeutic target for the treatment of melanoma.


Glycogen Synthase Kinase 3/metabolism , Melanoma, Experimental/pathology , Neural Cell Adhesion Molecules/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Silencing , Glycogen Synthase Kinase 3 beta , Melanoma, Experimental/genetics , Mice , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Protein Binding , Signal Transduction/genetics
19.
Brain Res ; 1404: 55-62, 2011 Aug 02.
Article En | MEDLINE | ID: mdl-21704981

Neurons that express high levels of polysialylated neural cell adhesion molecule (PSA-NCAM) in adult spinal substantia gelatinosa also express the µ-opioid receptor. While PSA removal from NCAM by spinal intrathecal injection of endoneuraminidase-N (endo-N) did not detectably change opioid receptor expression, morphine-induced analgesia was significantly increased. This analgesic strengthening was detected as early as 15 min after endo-N treatment and persisted for at least 7 days. In addition, the tolerance that develops with chronic morphine treatment was overcome in the absence of PSA. Interestingly, the same effects on analgesia and tolerance were also produced by selective deletion of the NCAM-180 isoform.


Analgesics, Opioid/pharmacology , Drug Tolerance/physiology , Gene Expression Regulation/drug effects , Morphine/pharmacology , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Animals , Behavior, Animal , Drug Interactions , Glycoside Hydrolases/pharmacology , Male , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Pain Measurement/methods , Receptors, Opioid, mu/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Substantia Gelatinosa/drug effects , Substantia Gelatinosa/metabolism
20.
J Neurosci ; 31(20): 7275-90, 2011 May 18.
Article En | MEDLINE | ID: mdl-21593312

Oligomannosidic glycans play important roles in nervous system development and function. By performing a phage display screening with oligomannose-specific antibodies, we identified an oligomannose-mimicking peptide that was functionally active in modulating neurite outgrowth and neuron-astrocyte adhesion. Using the oligomannose-mimicking peptide in crosslinking experiments, synapsin I was identified as a novel oligomannose-binding protein in mouse brain. Further analyses not only verified that synapsin I is an oligomannose-binding lectin, but also indicated that it is a glycoprotein carrying oligomannose and Lewis(x). We also found that synapsin I is expressed in glia-enriched cultures and is released from glial cells via exosomes. Incubation of glial-derived exosomes in the presence of high KCl concentrations or subjecting glial cell cultures to either oxygen/glucose deprivation or hydrogen peroxide resulted in release of synapsin I from exosomes. Application of synapsin I promoted neurite outgrowth from hippocampal neurons and increased survival of cortical neurons upon hydrogen peroxide treatment or oxygen/glucose deprivation. Coculture experiments using wild-type hippocampal neurons and wild-type or synapsin-deficient glial cells showed enhanced neurite outgrowth when synapsin was expressed by glial cells. Synapsin-induced neurite outgrowth was dependent on oligomannose on synapsin I and the neural cell adhesion molecule NCAM at the neuronal cell surface. The data indicate that, under conditions of high neuronal activity and/or oxidative stress, synapsin can be released from glial-derived exosomes and promotes neurite outgrowth and neuronal survival by modulating the interactions between glia and neurons.


Exosomes/metabolism , Lectins/metabolism , Neurites/metabolism , Neuroglia/metabolism , Neurons/metabolism , Oligosaccharides/metabolism , Synapsins/metabolism , Animals , Cell Communication/genetics , Cell Communication/physiology , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Female , Glycoproteins/metabolism , Male , Mannose/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Pregnancy , Protein Binding/physiology
...