Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neurosci ; 34(34): 11274-87, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143608

ABSTRACT

Neuron-glial related cell adhesion molecule (NrCAM) is a regulator of axon growth and repellent guidance, and has been implicated in autism spectrum disorders. Here a novel postsynaptic role for NrCAM in Semaphorin3F (Sema3F)-induced dendritic spine remodeling was identified in pyramidal neurons of the primary visual cortex (V1). NrCAM localized to dendritic spines of star pyramidal cells in postnatal V1, where it was coexpressed with Sema3F. NrCAM deletion in mice resulted in elevated spine densities on apical dendrites of star pyramidal cells at both postnatal and adult stages, and electron microscopy revealed increased numbers of asymmetric synapses in layer 4 of V1. Whole-cell recordings in cortical slices from NrCAM-null mice revealed increased frequency of mEPSCs in star pyramidal neurons. Recombinant Sema3F-Fc protein induced spine retraction on apical dendrites of wild-type, but not NrCAM-null cortical neurons in culture, while re-expression of NrCAM rescued the spine retraction response. NrCAM formed a complex in brain with Sema3F receptor subunits Neuropilin-2 (Npn-2) and PlexinA3 (PlexA3) through an Npn-2-binding sequence (TARNER) in the extracellular Ig1 domain. A trans heterozygous genetic interaction test demonstrated that Sema3F and NrCAM pathways interacted in vivo to regulate spine density in star pyramidal neurons. These findings reveal NrCAM as a novel postnatal regulator of dendritic spine density in cortical pyramidal neurons, and an integral component of the Sema3F receptor complex. The results implicate NrCAM as a contributor to excitatory/inhibitory balance in neocortical circuits.


Subject(s)
Brain/cytology , Cell Adhesion Molecules/physiology , Dendritic Spines/physiology , Gene Expression Regulation, Developmental/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Age Factors , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Cell Adhesion Molecules/deficiency , Cells, Cultured , Chlorocebus aethiops , Dendritic Spines/ultrastructure , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Antagonists/pharmacology , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/deficiency , Sodium Channel Blockers/pharmacology , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
2.
J Neurochem ; 128(2): 267-79, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117969

ABSTRACT

EphrinA/EphA-dependent axon repulsion is crucial for synaptic targeting in developing neurons but downstream molecular mechanisms remain obscure. Here, it is shown that ephrinA5/EphA3 triggers proteolysis of the neural cell adhesion molecule (NCAM) by the metalloprotease a disintegrin and metalloprotease (ADAM)10 to promote growth cone collapse in neurons from mouse neocortex. EphrinA5 induced ADAM10 activity to promote ectodomain shedding of polysialic acid-NCAM in cortical neuron cultures, releasing a ~ 250 kDa soluble fragment consisting of most of its extracellular region. NCAM shedding was dependent on ADAM10 and EphA3 kinase activity as shown in HEK293T cells transfected with dominant negative ADAM10 and kinase-inactive EphA3 (K653R) mutants. Purified ADAM10 cleaved NCAM at a sequence within the E-F loop of the second fibronectin type III domain (Leu(671) -Lys(672) /Ser(673) -Leu(674) ) identified by mass spectrometry. Mutations of NCAM within the ADAM10 cleavage sequence prevented EphA3-induced shedding of NCAM in HEK293T cells. EphrinA5-induced growth cone collapse was dependent on ADAM10 activity, was inhibited in cortical cultures from NCAM null mice, and was rescued by WT but not ADAM10 cleavage site mutants of NCAM. Regulated proteolysis of NCAM through the ephrin5/EphA3/ADAM10 mechanism likely impacts synapse development, and may lead to excess NCAM shedding when disrupted, as implicated in neurodevelopmental disorders such as schizophrenia. PSA-NCAM and ephrinA/EphA3 coordinately regulate inhibitory synapse development. Here, we have found that ephrinA5 stimulates EphA3 kinase and ADAM10 activity to promote PSA-NCAM cleavage at a site in its second FNIII repeat, which regulates ephrinA5-induced growth cone collapse in GABAergic and non-GABAergic neurons. These findings identify a new regulatory mechanism which may contribute to inhibitory connectivity.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Growth Cones/physiology , Membrane Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Receptor, EphA3/metabolism , Receptor, EphA5/metabolism , ADAM10 Protein , Animals , Cells, Cultured , Cerebral Cortex/cytology , Fibronectins/metabolism , Growth Cones/ultrastructure , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neural Cell Adhesion Molecules/genetics , Protein Structure, Tertiary
3.
PLoS One ; 8(9): e73000, 2013.
Article in English | MEDLINE | ID: mdl-24023801

ABSTRACT

NrCAM (Neuron-glial related cell adhesion molecule), a member of the L1 family of cell adhesion molecules, reversibly binds ankyrin and regulates axon growth, but it has not been studied for a role in retinotopic mapping. During development of retino-collicular topography, NrCAM was expressed uniformly in retinal ganglion cells (RGCs) along both mediolateral and anteroposterior retinal axes, and was localized on RGC axons within the optic tract and superior colliculus (SC). Anterograde tracing of RGC axons in NrCAM null mutant mice at P10, when the map resembles its mature form, revealed laterally displaced ectopic termination zones (eTZs) of axons from the temporal retina, indicating defective mediolateral topography, which is governed by ephrinB/EphBs. Axon tracing at P2 revealed that interstitial branch orientation of ventral-temporal RGC axons in NrCAM null mice was compromised in the medial direction, likely accounting for displacement of eTZs. A similar retinocollicular targeting defect in EphB mutant mice suggested that NrCAM and EphB interact to regulate mediolateral retino-collicular targeting. We found that EphB2 tyrosine kinase but not an EphB2 kinase dead mutant, phosphorylated NrCAM at a conserved tyrosine residue in the FIGQY ankyrin binding motif, perturbing ankyrin recruitment in NrCAM transfected HEK293 cells. Furthermore, the phosphorylation of NrCAM at FIGQY in SC was decreased in EphB1/3 and EphB1/2/3 null mice compared to WT, while phospho-FIGQY of NrCAM in SC was increased in EphB2 constitutively active (F620D/F620D) mice. These results demonstrate that NrCAM contributes to mediolateral retinocollicular axon targeting by regulating RGC branch orientation through a likely mechanism in which ephrinB/EphB phosphorylates NrCAM to modulate linkage to the actin cytoskeleton.


Subject(s)
Cell Adhesion Molecules/metabolism , Retina/metabolism , Superior Colliculi/metabolism , Animals , Axons/metabolism , Cell Adhesion Molecules/genetics , Cell Line , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Knockout , Retinal Ganglion Cells/metabolism
4.
Cereb Cortex ; 23(1): 162-77, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22275477

ABSTRACT

A novel function for the neural cell adhesion molecule (NCAM) was identified in ephrinA/EphA-mediated repulsion as an important regulatory mechanism for development of GABAergic inhibitory synaptic connections in mouse prefrontal cortex. Deletion of NCAM, EphA3, or ephrinA2/3/5 in null mutant mice increased the numbers and size of perisomatic synapses between GABAergic basket interneurons and pyramidal cells in the developing cingulate cortex (layers II/III). A functional consequence of NCAM loss was increased amplitudes and faster kinetics of miniature inhibitory postsynaptic currents in NCAM null cingulate cortex. NCAM and EphA3 formed a molecular complex and colocalized with the inhibitory presynaptic marker vesicular GABA transporter (VGAT) in perisomatic puncta and neuropil in the cingulate cortex. EphrinA5 treatment promoted axon remodeling of enhanced green fluorescent protein-labeled basket interneurons in cortical slice cultures and induced growth cone collapse in wild-type but not NCAM null mutant neurons. NCAM modified with polysialic acid (PSA) was required to promote ephrinA5-induced axon remodeling of basket interneurons in cortical slices, likely by providing a permissive environment for ephrinA5/EphA3 signaling. These results reveal a new mechanism in which NCAM and ephrinAs/EphA3 coordinate to constrain GABAergic interneuronal arborization and perisomatic innervation, potentially contributing to excitatory/inhibitory balance in prefrontal cortical circuitry.


Subject(s)
Ephrins/metabolism , GABAergic Neurons/physiology , Interneurons/physiology , Neural Cell Adhesion Molecules/metabolism , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Synapses/physiology , Animals , Cells, Cultured , Female , Male , Mice , Mice, Transgenic , gamma-Aminobutyric Acid/metabolism
5.
Front Mol Neurosci ; 5: 76, 2012.
Article in English | MEDLINE | ID: mdl-22723769

ABSTRACT

To study the potential role of neural cell adhesion molecule (NCAM) in the development of thalamocortical (TC) axon topography, wild type, and NCAM null mutant mice were analyzed for NCAM expression, projection, and targeting of TC afferents within the somatosensory area of the neocortex. Here we report that NCAM and its α-2,8-linked polysialic acid (PSA) are expressed in developing TC axons during projection to the neocortex. Pathfinding of TC axons in wild type and null mutant mice was mapped using anterograde DiI labeling. At embryonic day E16.5, null mutant mice displayed misguided TC axons in the dorsal telencephalon, but not in the ventral telencephalon, an intermediate target that initially sorts TC axons toward correct neocortical areas. During the early postnatal period, rostrolateral TC axons within the internal capsule along the ventral telencephalon adopted distorted trajectories in the ventral telencephalon and failed to reach the neocortex in NCAM null mutant animals. NCAM null mutants showed abnormal segregation of layer IV barrels in a restricted portion of the somatosensory cortex. As shown by Nissl and cytochrome oxidase staining, barrels of the anterolateral barrel subfield (ALBSF) and the most distal barrels of the posteromedial barrel subfield (PMBSF) did not segregate properly in null mutant mice. These results indicate a novel role for NCAM in axonal pathfinding and topographic sorting of TC axons, which may be important for the function of specific territories of sensory representation in the somatosensory cortex.

6.
Neurobiol Dis ; 43(2): 372-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21515372

ABSTRACT

The neural cell adhesion molecule, NCAM, is a pivotal regulator of neural development, with key roles in axonal and dendritic growth and synaptic plasticity. Alterations in NCAM expression or proteolytic cleavage have been linked to human neuropsychiatric disorders such as schizophrenia, bipolar disorder and Alzheimer's disease, and may contribute to cognitive dysfunction. We have generated mice overexpressing the NCAM extracellular (EC) proteolytic cleavage fragment which has been reported to be increased in schizophrenic versus normal brains. These mice show impaired GABAergic innervation and reduced number of apical dendritic spines on pyramidal neurons in the prefrontal cortex (PFC). Here, these NCAM-EC transgenic mice were subjected to behavioral tasks and electrophysiological measurements to determine the impact of structural abnormalities in the PFC on synaptic and cognitive functions. NCAM-EC mice exhibited impaired working memory in a delayed non-match-to-sample task, which requires PFC function, but showed no differences in anxiety, olfactory abilities, or sociability. Transgenic mice displayed impaired long- and short-term potentiation in the PFC but normal synaptic plasticity in the hippocampus, suggesting that the abnormal synaptic innervation in NCAM-EC mice impairs PFC plasticity and alters working memory. These findings may have implications for cognitive dysfunctions observed in neuropsychiatric disorders.


Subject(s)
Cerebral Cortex/pathology , Memory, Short-Term/physiology , Neural Cell Adhesion Molecules/genetics , Neuronal Plasticity/genetics , Neurons/pathology , Animals , Cerebral Cortex/physiology , Disease Models, Animal , Extracellular Space/genetics , Extracellular Space/metabolism , Female , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Cell Adhesion Molecules/biosynthesis , Neurons/metabolism , Organ Culture Techniques , Protein Structure, Tertiary/genetics , Proteolysis
7.
J Neurosci ; 31(4): 1545-58, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273439

ABSTRACT

NrCAM is a neural cell adhesion molecule of the L1 family that has been linked to autism spectrum disorders, a disease spectrum in which abnormal thalamocortical connectivity may contribute to visual processing defects. Here we show that NrCAM interaction with neuropilin-2 (Npn-2) is critical for semaphorin 3F (Sema3F)-induced guidance of thalamocortical axon subpopulations at the ventral telencephalon (VTe), an intermediate target for thalamic axon sorting. Genetic deletion of NrCAM or Npn-2 caused contingents of embryonic thalamic axons to misproject caudally in the VTe. The resultant thalamocortical map of NrCAM-null mutants showed striking mistargeting of motor and somatosensory thalamic axon contingents to the primary visual cortex, but retinogeniculate targeting and segregation were normal. NrCAM formed a molecular complex with Npn-2 in brain and neural cells, and was required for Sema3F-induced growth cone collapse in thalamic neuron cultures, consistent with a vital function for NrCAM in Sema3F-induced axon repulsion. NrCAM-null mice displayed reduced responses to visual evoked potentials recorded from layer IV in the binocular zone of primary visual cortex (V1), particularly when evoked from the ipsilateral eye, indicating abnormal visual acuity and ocularity. These results demonstrate that NrCAM is required for normal maturation of cortical visual acuity, and suggest that the aberrant projection of thalamic motor and somatosensory axons to the visual cortex in NrCAM-null mutant mice impairs cortical functions.


Subject(s)
Axons/physiology , Cell Adhesion Molecules/physiology , Motor Cortex/ultrastructure , Somatosensory Cortex/ultrastructure , Thalamus/ultrastructure , Visual Acuity , Visual Cortex/ultrastructure , Animals , Cell Adhesion Molecules/genetics , Evoked Potentials, Visual , Female , Growth Cones/physiology , Male , Membrane Proteins/physiology , Mice , Mice, Knockout , Motor Cortex/embryology , Motor Cortex/growth & development , Nerve Tissue Proteins/physiology , Neuropilin-2/genetics , Neuropilin-2/physiology , Somatosensory Cortex/embryology , Somatosensory Cortex/growth & development , Thalamus/embryology , Thalamus/growth & development , Visual Cortex/embryology , Visual Cortex/growth & development
8.
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
10.
Mol Biol Cell ; 20(24): 5236-49, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19846666

ABSTRACT

Epidermal growth factor receptor (EGFR) activation by GPCRs regulates many important biological processes. ADAM metalloprotease activity has been implicated as a key step in transactivation, yet the regulatory mechanisms are not fully understood. Here, we investigate the regulation of transforming growth factor-alpha (TGF-alpha) shedding by reactive oxygen species (ROS) through the ATP-dependent activation of the P2Y family of GPCRs. We report that ATP stimulates TGF-alpha proteolysis with concomitant EGFR activation and that this process requires TACE/ADAM17 activity in both murine fibroblasts and CHO cells. ATP-induced TGF-alpha shedding required calcium and was independent of Src family kinases and PKC and MAPK signaling. Moreover, ATP-induced TGF-alpha shedding was completely inhibited by scavengers of ROS, whereas calcium-stimulated shedding was partially inhibited by ROS scavenging. Hydrogen peroxide restored TGF-alpha shedding after calcium chelation. Importantly, we also found that ATP-induced shedding was independent of the cytoplasmic NADPH oxidase complex. Instead, mitochondrial ROS production increased in response to ATP and mitochondrial oxidative complex activity was required to activate TACE-dependent shedding. These results reveal an essential role for mitochondrial ROS in regulating GPCR-induced growth factor shedding.


Subject(s)
ADAM Proteins/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Transforming Growth Factor alpha/metabolism , ADAM17 Protein , Adenosine Triphosphate/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , ErbB Receptors/genetics , ErbB Receptors/metabolism , HL-60 Cells , Humans , Mice , Mitochondria/drug effects , Mitochondria/enzymology , NADPH Oxidases/metabolism , Receptors, Purinergic P2/metabolism , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...