Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Curr Alzheimer Res ; 17(4): 382-392, 2020.
Article in English | MEDLINE | ID: mdl-32321402

ABSTRACT

BACKGROUND: Cognitive capacities in Alzheimer's Disease (AD) are impaired by an epigenetic blockade mediated by histone deacetylase 2 (HDAC2), which prevents the transcription of genes that are important for synaptic plasticity. OBJECTIVE: Investigation of the functional relationship between cell adhesion molecule L1 and HDAC2 in AD. METHODS: Cultures of dissociated cortical and hippocampal neurons from wild-type or L1-deficient mice were treated with Aß1-42 for 24 h. After removal of Aß1-42 cells were treated with the recombinant L1 extracellular domain (rL1) for 24 h followed by immunohistochemistry, western blotting, and reverse transcription PCR to evaluate the interaction between L1 and HDAC2. RESULTS: Aß and HDAC2 protein levels were increased in APPSWE/L1+/- mutant brains compared to APPSWE mutant brains. Administration of the recombinant extracellular domain of L1 to cultured cortical and hippocampal neurons reduced HDAC2 mRNA and protein levels. In parallel, reduced phosphorylation levels of glucocorticoid receptor 1 (GR1), which is implicated in regulating HDAC2 levels, was observed in response to L1 administration. Application of a glucocorticoid receptor inhibitor reduced Aß-induced GR1 phosphorylation and prevented the increase in HDAC2 levels. HDAC2 protein levels were increased in cultured cortical neurons from L1-deficient mice. This change could be reversed by the administration of the recombinant extracellular domain of L1. CONCLUSION: Our results suggest that some functionally interdependent activities of L1 and HDAC2 contribute to ameliorating the phenotype of AD by GR1 dephosphorylation, which leads to reduced HDAC2 expression. The combined findings encourage further investigations on the beneficial effects of L1 in the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation, Enzymologic , Histone Deacetylase 2/biosynthesis , Neural Cell Adhesion Molecule L1/administration & dosage , Neural Cell Adhesion Molecule L1/deficiency , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Female , Hippocampus/metabolism , Hippocampus/pathology , Histone Deacetylase 2/antagonists & inhibitors , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/toxicity
2.
Neuropsychopharmacology ; 41(6): 1670-80, 2016 May.
Article in English | MEDLINE | ID: mdl-26530284

ABSTRACT

Antidepressant drugs are too often ineffective, the exact mechanism of efficacy is still ambiguous, and there has been a paucity of novel targets for pharmacotherapy. In an attempt to understand the pathogenesis of depression and subsequently develop more efficacious antidepressant drugs, multiple theories have been proposed, including the modulation of neurotransmission, the upregulation of neurogenesis and neurotrophic factors, normalizing hypothalamic-pituitary-adrenal reactivity, and the reduction of neuroinflammation; all of which have supporting lines of evidence. Therefore, an ideal molecular target for novel pharmaceutical intervention would function at the confluence of these theories. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) functions broadly, serving to mediate synaptic plasticity, neurogenesis, neurotrophic factor signaling, and inflammatory signaling throughout the brain; all of which are associated with the pathophysiology and treatment of depression. Moreover, the expression of PSA-NCAM is reduced by depression, and conversely enhanced by antidepressant treatment, particularly within the hippocampus. Here we demonstrate that selectively cleaving the polysialic acid moiety, using the bacteriophage-derived enzyme endoneuraminidase N, completely inhibits the antidepressant efficacy of the selective-serotonin reuptake inhibitor fluoxetine (FLX) in a chronic unpredictable stress model of depression. We also observe a corresponding attenuation of FLX-induced hippocampal neuroplasticity, including decreased hippocampal neurogenesis, synaptic density, and neural activation. These data indicate that PSA-NCAM-mediated neuroplasticity is necessary for antidepressant action; therefore PSA-NCAM represents an interesting, and novel, target for pharmacotherapy.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluoxetine/pharmacology , Neural Cell Adhesion Molecule L1/physiology , Sialic Acids/physiology , Animals , Corticosterone/blood , Depression/drug therapy , Disease Models, Animal , Glycoside Hydrolases/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Sialic Acids/deficiency , Sialic Acids/metabolism , Testosterone/blood , Treatment Outcome
3.
Cereb Cortex ; 21(2): 401-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20576928

ABSTRACT

Neural cell adhesion molecule close homolog of L1 (CHL1) is a regulator of topographic targeting of thalamic axons to the somatosensory cortex (S1) but little is known about its cooperation with other L1 class molecules. To investigate this, CHL1(-/-)/L1(-/y) double mutant mice were generated and analyzed for thalamocortical axon topography. Double mutants exhibited a striking posterior shift of axons from motor thalamic nuclei to the visual cortex (V1), which was not observed in single mutants. In wild-type (WT) embryos, L1 and CHL1 were coexpressed in the dorsal thalamus (DT) and on fibers along the thalamocortical projection in the ventral telencephalon and cortex. L1 and CHL1 colocalized on growth cones and neurites of cortical and thalamic neurons in culture. Growth cone collapse assays with WT and mutant neurons demonstrated a requirement for L1 and CHL1 in repellent responses to EphrinA5, a guidance factor for thalamic axons. L1 coimmunoprecipitated with the principal EphrinA5 receptors expressed in the DT (EphA3, EphA4, and EphA7), whereas CHL1 associated selectively with EphA7. These results implicate a novel mechanism in which L1 and CHL1 interact with individual EphA receptors and cooperate to guide subpopulations of thalamic axons to distinct neocortical areas essential for thalamocortical connectivity.


Subject(s)
Axons/physiology , Cell Adhesion Molecules/metabolism , Cerebral Cortex/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Pathways/physiology , Thalamus/metabolism , Amino Acids/metabolism , Animals , Axons/drug effects , Axons/ultrastructure , Cell Adhesion Molecules/deficiency , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Ephrin-A5/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Growth Cones/physiology , Humans , Immunoprecipitation/methods , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Cell Adhesion Molecule L1/deficiency , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Thalamus/cytology , Transfection/methods
4.
Brain ; 132(Pt 8): 2180-95, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19541848

ABSTRACT

The adhesion molecule L1 is one of the few adhesion molecules known to be beneficial for repair processes in the adult central nervous system of vertebrates by promoting axonal growth and neuronal survival. In the peripheral nervous system, L1 is up-regulated by myelination-competent Schwann cells and regenerating axons after nerve damage but its functional role has remained unknown. Here we tested the hypothesis that L1 is, as in the central nervous system, beneficial for nerve regeneration in the peripheral nervous system by performing combined functional and histological analyses of adult L1-deficient mice (L1y/-) and wild-type (L1y/+) littermates. Contrary to our hypothesis, quantitative video-based motion analysis revealed better locomotor recovery in L1y/- than in L1y/+ mice at 4-12 weeks after transection and surgical repair of the femoral nerve. Motoneuron regeneration in L1y/- mice was also enhanced as indicated by attenuated post-traumatic loss of motoneurons, enhanced precision of motor reinnervation, larger cell bodies of regenerated motoneurons and diminished loss of inhibitory synaptic input to motoneurons. In search of mechanisms underlying the observed effects, we analysed peripheral nerves at short time-periods (3-14 days) after transection and found that Schwann cell proliferation is strongly augmented in L1y/- versus L1y/+ mice. L1-deficient Schwann cells showed increased proliferation than wild-type Schwann cells, both in vivo and in vitro. These findings suggest a novel role for L1 in nerve regeneration. We propose that L1 negatively regulates Schwann cell proliferation after nerve damage, which in turn restricts functional recovery by limiting the trophic support for regenerating motoneurons.


Subject(s)
Neural Cell Adhesion Molecule L1/physiology , Peripheral Nervous System/injuries , Schwann Cells/pathology , Animals , Axons/pathology , Cell Proliferation , Cell Survival , Femoral Nerve/injuries , Femoral Nerve/physiology , Femoral Nerve/surgery , Lameness, Animal/pathology , Lameness, Animal/physiopathology , Lameness, Animal/surgery , Male , Mice , Mice, Knockout , Motor Activity/physiology , Motor Neurons/pathology , Motor Neurons/physiology , Nerve Regeneration/physiology , Neural Cell Adhesion Molecule L1/deficiency , Peripheral Nervous System/physiology , Presynaptic Terminals/pathology , Recovery of Function , Synapses/physiology
5.
Dev Dyn ; 238(3): 708-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19235728

ABSTRACT

The enteric nervous system arises predominantly from vagal level neural crest cells that migrate into and along the developing gut. As the neural crest-derived cells migrate within the gut, a subpopulation begins to differentiate into enteric neurons. Here, we show that the differentiation of neural crest-derived cells into enteric neurons is delayed in L1-deficient mice, compared with littermate controls. However, glial cell differentiation is not affected in L1-deficient mice. These mice also show a delay in the differentiation of a neurotransmitter-specific subtype of enteric neuron within the gastrointestinal tract. Together, these results suggest a role for the cell adhesion molecule, L1, in the differentiation of neural crest-derived cells into enteric neurons within the developing enteric nervous system.


Subject(s)
Cell Differentiation , Digestive System/embryology , Digestive System/metabolism , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/cytology , Animals , Cell Death , Cell Proliferation , Digestive System/cytology , Enteric Nervous System/cytology , Gene Expression Regulation, Developmental , Mice , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/genetics , Neurons/metabolism
6.
EMBO J ; 27(1): 188-200, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18046458

ABSTRACT

Apical dendrites of pyramidal neurons in the neocortex have a stereotypic orientation that is important for neuronal function. Neural recognition molecule Close Homolog of L1 (CHL1) has been shown to regulate oriented growth of apical dendrites in the mouse caudal cortex. Here we show that CHL1 directly associates with NB-3, a member of the F3/contactin family of neural recognition molecules, and enhances its cell surface expression. Similar to CHL1, NB-3 exhibits high-caudal to low-rostral expression in the deep layer neurons of the neocortex. NB-3-deficient mice show abnormal apical dendrite projections of deep layer pyramidal neurons in the visual cortex. Both CHL1 and NB-3 interact with protein tyrosine phosphatase alpha (PTPalpha) and regulate its activity. Moreover, deep layer pyramidal neurons of PTPalpha-deficient mice develop misoriented, even inverted, apical dendrites. We propose a signaling complex in which PTPalpha mediates CHL1 and NB-3-regulated apical dendrite projection in the developing caudal cortex.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cell Adhesion Molecules/physiology , Dendrites/enzymology , Neocortex/cytology , Neural Cell Adhesion Molecule L1/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/physiology , Animals , COS Cells , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Chlorocebus aethiops , Dendrites/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neocortex/enzymology , Neocortex/metabolism , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/genetics , Prefrontal Cortex/cytology , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics
7.
Neurosci Lett ; 420(3): 277-81, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17540505

ABSTRACT

Repair strategies for spinal cord injury often focus on promoting regeneration of injured axons and stimulating subsequent functional recovery. Although many of these strategies have proven their merits, less is known about potential unwanted side-effects, such as sprouting of nociceptive CGRP immunoreactive axons, which may bring about pain-related behavior. Sprouting of CGRP axons into lesion sites spontaneously occurs after spinal cord injury (SCI). Using L1-deficient mice we show a reduction of such CGRP growth response. This reduction was specific for CGRP axons since the overall neurofilament positive fibre in-growth into the spinal lesion site was not affected. Our results may have important implications on the development and assessment of repair strategies that should not only stimulate functional recovery, but also prevent the development of pain or autonomic dysreflexia.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Nerve Fibers/physiology , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Animals , Axons/pathology , Axons/physiology , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers/pathology , Neurons, Afferent/pathology , Neurons, Afferent/physiology
8.
Neuroscience ; 144(3): 788-96, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17140740

ABSTRACT

The neural cell adhesion molecule (NCAM) has been implicated in regulating synaptic plasticity mechanisms as well as memory consolidation processes. Attachment of polysialic acid to NCAM (PSA-NCAM) has been reported to down-regulate its adhesive forces, a process hypothesized to be implicated in synapse selection after learning experiences. PSA-NCAM has been critically implicated in hippocampus-related synaptic plasticity and memory storage, but information about its functional role in other brain areas remains scarce. Here, we studied mice deficient for polysialyltransferase-1 (ST8SialV/PST-1), an enzyme which attaches PSA to NCAM during postnatal development and adulthood, and whose deficiency results in a drastic reduction of PSA-NCAM expression throughout the brain in adulthood. Mice were tested for their performance in the water maze and auditory fear conditioning (AFC). We report that ST8SiaIV knockout mice were impaired in spatial as well as reversal learning in the water maze. On the other hand, AFC was intact and ST8SiaIV mice exhibited no impairments in the acquisition or retention of cued fear memories. Spatial orientation learning and reversal learning require complex integration of spatial information and response selection involving the hippocampus and prefrontal cortex, whereas cued fear conditioning is an associative type of emotional memory that highly depends on amygdala function. Therefore, our results indicate that PSA-NCAM contributes differentially to learning processes that differ in the nature of the neural computations involved, which probably reflects a differential role of this molecule in different brain regions.


Subject(s)
Brain/metabolism , Learning Disabilities/metabolism , Memory Disorders/metabolism , Neural Cell Adhesion Molecule L1/deficiency , Sialic Acids/deficiency , Amygdala/metabolism , Amygdala/physiopathology , Animals , Avoidance Learning/physiology , Brain/physiopathology , Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Learning Disabilities/genetics , Learning Disabilities/physiopathology , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Knockout , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Sialyltransferases/genetics
9.
J Neurosci ; 26(42): 10888-109898, 2006 Oct 18.
Article in English | MEDLINE | ID: mdl-17050727

ABSTRACT

Polysialic acid (PSA) regulates functions of the neural cell adhesion molecule (NCAM) during development and in neuroplasticity in the adult; the underlying mechanisms at different phases of learning and memory consolidation are, however, unknown. To investigate the contributions of PSA versus the extracellular domain of the NCAM glycoprotein backbone to synaptic plasticity, we applied NCAM, PSA-NCAM, and PSA to acute slices of the hippocampal CA1 region of NCAM-deficient mice and measured their effects on long-term potentiation (LTP). Remarkably, only PSA and PSA-NCAM, but not NCAM restored normal LTP. Application of these molecules to the dorsal hippocampus of wild-type mice showed that PSA-NCAM and PSA, but not NCAM, injected before fear conditioning, impaired formation of hippocampus-dependent contextual memory. Consolidation of contextual memory was affected by PSA-NCAM only when injected during its late, but not early phases. None of the tested compounds disturbed extrahippocampal-cued memory. Mice lacking the polysialyltransferase (ST8SialV/PST) responsible for attachment of PSA to NCAM in adulthood showed a mild deficit only in hippocampal contextual learning, when compared with NCAM-deficient mice that were disturbed in both contextual and cued memories. Contextual and tone memory in NCAM-deficient mice could be partially restored by injection of PSA-NCAM, but not of NCAM, into the hippocampus, suggesting that the impact of PSA-NCAM in synaptic plasticity and learning is not mediated by modulation of NCAM-NCAM homophilic interactions. In conclusion, our data support the view that polysialylated NCAM is involved in both formation and late consolidation of contextual memory.


Subject(s)
Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/physiology , Memory/physiology , Neural Cell Adhesion Molecule L1/physiology , Sialic Acids/physiology , Animals , Conditioning, Psychological/drug effects , Fear/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/pharmacology , Sialic Acids/deficiency , Sialic Acids/pharmacology
10.
Gastroenterology ; 130(4): 1221-32, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16618414

ABSTRACT

BACKGROUND & AIMS: During development, the enteric nervous system is derived from neural crest cells that emigrate from the hindbrain, enter the foregut, and colonize the gut. Defects in neural crest migration can result in intestinal aganglionosis. Hirschsprung's disease (congenital aganglionosis) is a human condition in which enteric neurons are absent from the distal bowel. A number of clinical studies have implicated the cell adhesion molecule L1 in Hirschsprung's disease. We examined the role of L1 in the migration of neural crest cells through the developing mouse gut. METHODS: A variety of in vitro and in vivo assays were used to examine: (1) the effect of L1 blocking antibodies or exogenous soluble L1 protein known to compromise L1 function on the rate of crest cell migration, (2) the effect of blocking L1 activity on the dynamic behavior of crest cells using time-lapse microscopy, and (3) whether the colonization of the gut by crest cells in L1-deficient mice differs from control mice. RESULTS: We show that L1 is expressed by neural crest cells as they colonize the gut. Perturbation studies show that disrupting L1 activity retards neural crest migration and increases the number of solitary neural crest cells. L1-deficient mice show a small but significant reduction in neural crest cell migration at early developmental stages, but the entire gastrointestinal tract is colonized. CONCLUSIONS: L1 is important for the migration of neural crest cells through the developing gut and is likely to be involved in the etiology of Hirschsprung's disease.


Subject(s)
Cell Movement/physiology , Intestines/embryology , Neural Cell Adhesion Molecule L1/physiology , Neural Crest/physiology , Animals , Antibodies/pharmacology , Embryo, Mammalian/cytology , Embryonic Development , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy/methods , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Time Factors
11.
Eur J Neurosci ; 23(8): 1997-2011, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16630048

ABSTRACT

L1 is a member of the immunoglobulin superfamily of cell adhesion molecules that is associated with axonal growth, including formation of the corticospinal tract (CST). The present study describes the effects of L1 deletion on hindlimb function in locomotion, and examines the role of L1 in recovery and remodeling after contusive spinal cord injury (SCI) in mice. Uninjured adult L1 knockout (Y/-) mice had impaired performance on locomotor tests compared with their wild-type littermates (Y/+). Anterograde tracing demonstrated that CST axons project to thoracic, but not lumbar, levels of the spinal cord of Y/- mice, and revealed a diversion of these fibers from their position in the base of the dorsal columns. Retrograde tracing also revealed reduced numbers of descending projections from paraventricular hypothalamus and red nuclei to the lumbar spinal cord in Y/- mice. SCI at the mid-thoracic level produced a lesion encompassing the center of the spinal cord, including the site of the dorsal CST and surrounding gray matter (GM). The injury caused lasting deficits in fine aspects of locomotion. There was no effect of genotype on final lesion size or the growth of axons into the lesion area. However, injured Y/- mice demonstrated a robust expansion of CST projections throughout the GM of the cervical and thoracic spinal cord rostral to the lesion compared with Y/+ littermates. Thus, L1 is important for the development of multiple spinal projections and also contributes to the restriction of CST sprouting rostral to the site of a SCI in adults.


Subject(s)
Locomotion/genetics , Neural Cell Adhesion Molecule L1/deficiency , Pyramidal Tracts/physiopathology , Recovery of Function/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Biotin/analogs & derivatives , Brain/metabolism , Brain/pathology , Cell Count/methods , Dextrans , Female , Functional Laterality/genetics , Hindlimb/physiopathology , Immunohistochemistry/methods , Male , Mice , Mice, Knockout , Nerve Regeneration , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Psychomotor Performance/physiology , Pyramidal Tracts/pathology , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord Injuries/genetics , Stilbamidines , Time Factors
12.
Brain Res ; 1029(1): 131-4, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15533325

ABSTRACT

Mutations in the gene encoding the cell adhesion molecule L1 or its close homologue, CHL1 (close homologue of L1), cause brain dysfunction in both humans and mice. Here we report that prepulse inhibition (PPI) of the acoustic startle response is impaired in mice deficient in either L1 or CHL1. This newly identified feature may provide a basis for using these mice as models for sensorimotor gating impairment found in some neuropsychiatric disorders such as schizophrenia.


Subject(s)
Ion Channel Gating/physiology , Neural Cell Adhesion Molecule L1/physiology , Neural Inhibition/physiology , Reflex, Startle/physiology , Acoustic Stimulation , Animals , Cell Adhesion Molecules , Female , Ion Channel Gating/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Animal , Neural Cell Adhesion Molecule L1/deficiency , Neural Inhibition/genetics , Proteins/physiology , Reflex, Startle/genetics
13.
Behav Brain Res ; 152(2): 327-34, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15196800

ABSTRACT

Polysialylated neural cell adhesion molecule (PSA-NCAM) is predominantly expressed during development where it regulates biological functions including axon targeting and neuronal precursor cell migration. Although dramatically down regulated after birth in most regions of the nervous system, PSA-NCAM remains highly expressed into adulthood in areas that have ongoing regeneration and plasticity such as in the olfactory bulb and hippocampus. Consequently, lack of PSA-NCAM in NCAM null mice results in distinct morphological changes to these areas. The functional correlates of these changes are not well defined although there have been reports that learning is impaired in NCAM null mice. In the present study, we assessed the ability of old and young NCAM null mice to learn an odour discrimination task. We tested male and female experimental and control animals of two different ages: 30-60 days and 12-15 months. During 4 days of training, NCAM null and C57BL/6J received trials where one odour (CS+) was paired with sugar while another odour (CS-) was not. In a subsequent preference test, conducted in the absence of sugar, all animals, regardless of strain or age, spent significantly more time digging in the CS+ odour than in the CS- odour. In addition, there was no significant difference in digging behaviour in the CS+ between the NCAM null and the control animals. These data indicate that deletion of the NCAM gene may change the morphology of the olfactory bulb but does not interfere with the ability to learn an odour discrimination task.


Subject(s)
Discrimination Learning/physiology , Mice, Knockout/physiology , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/physiology , Sialic Acids/deficiency , Sialic Acids/physiology , Age Factors , Animals , Behavior, Animal , Conditioning, Psychological/physiology , Female , Histology , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Odorants , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Sex Factors
14.
Mol Cell Neurosci ; 26(1): 191-203, 2004 May.
Article in English | MEDLINE | ID: mdl-15121190

ABSTRACT

Cell adhesion molecules have been implicated in neural development and hippocampal synaptic plasticity. Here, we investigated the role of the neural cell adhesion molecule L1 in regulation of basal synaptic transmission and plasticity in the CA1 area of the hippocampus of juvenile mice. We show that theta-burst stimulation (TBS) and pairing of low-frequency presynaptic stimulation with depolarization of postsynaptic CA1 pyramidal cells induced similar levels of LTP in L1-deficient and wild-type mice. The basal excitatory synaptic transmission and density of asymmetric excitatory synapses in the stratum radiatum were also normal in L1-deficient mice. Since L1 is expressed not only by principal cells but also by inhibitory interneurons, we recorded inhibitory postsynaptic currents (IPSCs) evoked in CA1 pyramidal cells by minimal stimulation of perisomatic interneurons. L1-deficient mice showed a reduction in the mean amplitude of putative unitary IPSCs, higher values of the coefficient of amplitude variation, higher number of failures in transmitter release, and a reduction in frequency but not amplitude of miniature IPSCs. The use-dependent modulation of inhibitory transmission by paired-pulse or short tetanic stimulation was, however, normal in L1-deficient mice. The physiological abnormalities correlated with a strong reduction in the density of inhibitory active zones, indicating that L1 is involved in establishing inhibitory perisomatic synapses in the hippocampus.


Subject(s)
Hippocampus/metabolism , Neural Cell Adhesion Molecule L1/deficiency , Neural Inhibition/genetics , Synapses/metabolism , Synaptic Transmission/genetics , gamma-Aminobutyric Acid/metabolism , Animals , Cell Differentiation/genetics , Down-Regulation/genetics , Electric Stimulation , Gene Expression Regulation, Developmental/genetics , Hippocampus/growth & development , Hippocampus/ultrastructure , In Vitro Techniques , Interneurons/metabolism , Interneurons/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron , Neural Cell Adhesion Molecule L1/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Synapses/ultrastructure , Theta Rhythm
15.
J Cell Biol ; 165(1): 145-54, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15067019

ABSTRACT

A new mouse line has been produced in which the sixth Ig domain of the L1 cell adhesion molecule has been deleted. Despite the rather large deletion, L1 expression is preserved at normal levels. In vitro experiments showed that L1-L1 homophilic binding was lost, along with L1-alpha5beta1 integrin binding. However, L1-neurocan and L1-neuropilin binding were preserved and sema3a responses were intact. Surprisingly, many of the axon guidance defects present in the L1 knockout mice, such as abnormal corticospinal tract and corpus callosum, were not observed. Nonetheless, when backcrossed on the C57BL/6 strain, a severe hydrocephalus was observed and after several generations, became an embryonic lethal. These results imply that L1 binding to L1, TAG-1, or F3, and L1-alpha5beta1 integrin binding are not essential for normal development of a variety of axon pathways, and suggest that L1-L1 homophilic binding is important in the production of X-linked hydrocephalus.


Subject(s)
Brain/abnormalities , Hydrocephalus/genetics , Nervous System Malformations/metabolism , Neural Cell Adhesion Molecule L1/deficiency , Animals , Brain/cytology , Brain/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/metabolism , Contactin 2 , Contactins , Female , Genes, Lethal/genetics , Hydrocephalus/metabolism , Hydrocephalus/physiopathology , Inbreeding , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Lectins, C-Type , Male , Mice , Mice, Knockout , Mice, Neurologic Mutants , Nerve Tissue Proteins/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Neural Cell Adhesion Molecule L1/genetics , Neural Pathways/abnormalities , Neural Pathways/cytology , Neural Pathways/metabolism , Neurocan , Neuropilins/metabolism , Protein Binding/genetics
16.
Neurosci Res ; 48(4): 471-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15041201

ABSTRACT

To examine the role of neural cell adhesion molecule L1 in thalamocortical projections, we analysed L1 deficient (L1-/y) mice. Immunohistochemistry of pleiotrophin/HB-GAM, a marker for thalamocortical axons and axonal tracing experiments showed that thalamocortical axons were abnormally and highly fasciculated when they pass through the developing internal capsule. Within the cortex, however, their course was more diffuse. The corticofugal fibres immunoreactive for TAG-1 were also more strongly fasciculated and their number was decreased in L1-/y mice. Furthermore, no TAG-1-positive corticofugal axons reached the dorsal thalamus. These data suggest that L1 plays an important role in the fasciculation and routing of axons connecting between the thalamus and the cortex.


Subject(s)
Axons/physiology , Neocortex/anatomy & histology , Neural Cell Adhesion Molecule L1/metabolism , Neural Pathways/anatomy & histology , Thalamus/anatomy & histology , Animals , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Contactin 2 , Cytokines/metabolism , Immunohistochemistry , Mice , Neocortex/growth & development , Neural Cell Adhesion Molecule L1/deficiency , Neural Pathways/metabolism , Thalamus/growth & development
17.
Cereb Cortex ; 14(2): 121-31, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14704209

ABSTRACT

The neural cell adhesion molecule L1 has been found to play important roles in axon growth and fasciculation. Our main objective was to determine the role of L1 during the development of connections between thalamus and cortex. We find that thalamocortical and corticothalamic axons in mice lacking L1 are hyperfasciculated, a subset of thalamocortical axons make pathfinding errors and thalamocortical axon growth cones are abnormally long in the subplate. These defects occur despite formation of six cortical layers and formation of topographically appropriate thalamocortical connections. The loss of L1 is accompanied by loss of expression of ankyrin-B, an intracellular L1 binding partner, suggesting that L1 is involved in the regulation of Ank2 stability. We postulate that the pathfinding errors, growth cone abnormalities and hyperfasciculation of axons following loss of L1 reflect both a shift in binding partners among axons and different substrates and a loss of appropriate interactions with the cytoskeleton.


Subject(s)
Axons/physiology , Neural Cell Adhesion Molecule L1/physiology , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecules , Neural Pathways/growth & development , Neural Pathways/physiology , Thalamus/growth & development , Thalamus/physiology
18.
J Neurosci ; 23(32): 10419-32, 2003 Nov 12.
Article in English | MEDLINE | ID: mdl-14614101

ABSTRACT

L1, a neural cell adhesion molecule of the immunoglobulin superfamily, is involved in neuronal migration and differentiation and axon outgrowth and guidance. Mutations in the human and mouse L1 gene result in similarly severe neurological abnormalities. To dissociate the functional roles of L1 in the adult brain from developmental abnormalities, we have generated a mutant in which the L1 gene is inactivated by cre-recombinase under the control of the calcium/calmodulin-dependent kinase II promoter. This mutant (L1fy+) did not show the overt morphological and behavioral abnormalities observed previously in constitutive L1-deficient (L1-/-) mice; however, there was an increase in basal excitatory synaptic transmission that was not apparent in L1-/- mice. Similar to L1-/- mice, no defects in short- and long-term potentiation in the CA1 region of the hippocampus were observed. Interestingly, L1fy+ mice showed decreased anxiety in the open field and elevated plus-maze, contrary to L1-/- mice, and altered place learning in the water maze, similar to L1-/- mice. Thus, mice conditionally deficient in L1 expression in the adult brain share some abnormalities, but also display different ones, as compared with L1-/- mice, highlighting the role of L1 in the regulation of synaptic transmission and behavior in adulthood.


Subject(s)
Anxiety/genetics , Hippocampus/physiology , Learning/physiology , Neural Cell Adhesion Molecule L1/genetics , Spatial Behavior/physiology , Synaptic Transmission/genetics , Animals , Behavior, Animal/physiology , Chimera , Electrophysiology , Gene Targeting/methods , Hippocampus/cytology , Immunohistochemistry , Integrases/biosynthesis , Integrases/genetics , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Motor Activity/genetics , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/physiology , Prosencephalon/cytology , Prosencephalon/metabolism , Viral Proteins/biosynthesis , Viral Proteins/genetics
19.
J Neurobiol ; 57(1): 67-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12973829

ABSTRACT

In humans, loss or alteration of the CHL1/CALL gene may contribute to mental impairment associated with the 3p-syndrome, caused by distal deletions of the short (p) arm of chromosome 3, and schizophrenia. Mice deficient for the Close Homologue of L1 (CHL1) show aberrant connectivity of hippocampal mossy fibers and olfactory sensory axons, suggesting participation of CHL1 in the establishment of neuronal networks. Furthermore, behavioral studies showed that CHL1-deficient mice react differently towards novel experimental environments. These data raise the hypothesis that processing of information, possibly novel versus familiar, may be altered in the absence of CHL1. To test this hypothesis, brain activities were investigated after presentation of a novel, familiar, or neutral gustatory stimulus using metabolic mapping with ((14)C)-2-deoxyglucose (2-DG) and analysis of mRNA expression of the immediate early genes (IEGs) c-fos and arg 3.1/arc by in situ hybridization. 2-DG labeling revealed only small differences between CHL1-deficient and wild-type littermate mice. In contrast, while the specific novelty-induced increase in c-fos expression was maintained in most of the brain areas analyzed, c-fos mRNA expression was similar after the novel and familiar taste in several brain areas of the CHL1-deficient mice. Furthermore, in these mutants, arg 3.1/arc expression was slightly reduced after the novel taste and increased after the familiar taste, leading to a similar arg 3.1/arc mRNA expression after both stimuli. Our results indicate that, in contrast to controls, CHL1-deficient mice might process novel and familiar information similarly and suggest that the altered neuronal connectivity in these mutants disturbs information processing at the molecular level.


Subject(s)
Brain Mapping , Brain/physiology , Genes, Immediate-Early/physiology , Mental Processes/physiology , Neural Cell Adhesion Molecule L1/deficiency , Proteins , Animals , Cell Adhesion Molecules , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Genes, fos/physiology , In Situ Hybridization , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/genetics , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...