Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Neuroscience ; 165(1): 107-15, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-19819308

ABSTRACT

The cell adhesion molecule close homologue of L1 (CHL1) is important for apical dendritic projection and laminar positioning of pyramidal neurons in caudal regions of the cerebral cortex. The p21-activated kinase (PAK1-3) subfamily of serine/threonine kinases has also been implicated in regulating cell adhesion, migration, and morphology. Immunofluorescence staining in mouse embryonic brain showed that PAK1-3 was expressed in embryonic cortex and colocalized with CHL1 during neuronal migration and differentiation. To investigate a cooperative function for CHL1 and PAK in pyramidal cell differentiation or migration, a dominant-negative PAK mutant (PAK1 AID) that inhibits PAK1-3 kinase activity while coexpressing a green fluorescent protein (GFP) reporter was electroporated into the lateral ventricles of wild type (WT) and CHL1 null mutant mouse embryos (E14.5), then brain slices were cultured and neurons analyzed for laminar positioning and morphology by confocal microscopy after 3 days in vitro. Expression of PAK1 AID in CHL1 mutant cortex inactivated PAK and caused embryonic cortical neurons to branch profusely in the intermediate zone (IZ) and cortical plate (CP). The number of nodes, terminals and length of leading processes/apical dendrites of CHL1 mutant embryos expressing PAK1 AID increased dramatically, compared to CHL1 mutants without PAK1 AID, or WT embryos with or without PAK1 AID. These findings suggest that CHL1 and PAK1-3 kinase cooperate, most likely in independent pathways, in regulating morphological development of the leading process/apical dendrite of embryonic cortical neurons.


Subject(s)
Cell Adhesion Molecules/metabolism , Cerebral Cortex/cytology , Neurons/physiology , p21-Activated Kinases/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Differentiation , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Mice, Knockout , p21-Activated Kinases/antagonists & inhibitors
2.
Neuroscience ; 162(1): 96-105, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19393299

ABSTRACT

Neural cell adhesion molecule (NCAM) is a membrane-bound cell recognition molecule that exerts important functions in normal neurodevelopment including cell migration, neurite outgrowth, axon fasciculation, and synaptic plasticity. Alternative splicing of NCAM mRNA generates three main protein isoforms: NCAM-180, -140, and -120. Ectodomain shedding of NCAM isoforms can produce an extracellular 105-115 kilodalton soluble neural cell adhesion molecule fragment (NCAM-EC) and a smaller intracellular cytoplasmic fragment (NCAM-IC). NCAM also undergoes a unique post-translational modification in brain by the addition of polysialic acid (PSA)-NCAM. Interestingly, both PSA-NCAM and NCAM-EC have been implicated in the pathophysiology of schizophrenia. The developmental expression patterns of the main NCAM isoforms and PSA-NCAM have been described in rodent brain, but no studies have examined NCAM expression across human cortical development. Western blotting was used to quantify NCAM in human postmortem prefrontal cortex in 42 individuals ranging in age from mid-gestation to early adulthood. Each NCAM isoform (NCAM-180, -140, and -120), post-translational modification (PSA-NCAM) and cleavage fragment (NCAM-EC and NCAM-IC) demonstrated developmental regulation in frontal cortex. NCAM-180, -140, and -120, as well as PSA-NCAM, and NCAM-IC all showed strong developmental regulation during fetal and early postnatal ages, consistent with their identified roles in axon growth and plasticity. NCAM-EC demonstrated a more gradual increase from the early postnatal period to reach a plateau by early adolescence, potentially implicating involvement in later developmental processes. In summary, this study implicates the major NCAM isoforms, PSA-NCAM and proteolytically cleaved NCAM in pre- and postnatal development of the human prefrontal cortex. These data provide new insights on human cortical development and also provide a basis for how altered NCAM signaling during specific developmental intervals could affect synaptic connectivity and circuit formation, and thereby contribute to neurodevelopmental disorders.


Subject(s)
Gene Expression Regulation, Developmental , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Adolescent , Adult , Aging/genetics , Aging/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Prefrontal Cortex/embryology , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Rats , Rats, Sprague-Dawley , Sialic Acids/genetics , Sialic Acids/metabolism , Young Adult
3.
Brain Res Dev Brain Res ; 126(1): 21-30, 2001 Jan 31.
Article in English | MEDLINE | ID: mdl-11172883

ABSTRACT

Dopaminergic neurons of the mouse mesencephalon originate in the ventricular zone and migrate radially along radial glia then tangentially along nerve fibers that express the neural cell adhesion molecule L1 to form the substantia nigra (A9 group) and ventral tegmental area (VTA) (A10 group). The role of L1 in migration of dopaminergic neuronal precursors was investigated in L1 knockout mice by tyrosine hydroxylase (TH) immunostaining. An altered rostrocaudal distribution of dopaminergic neurons was observed within the substantia nigra and VTA of L1-minus mice. In L1-minus mice at postnatal day 0, TH-positive cells were present abnormally in the dorsomedial mesencephalon, suggesting impaired migration. Axons projecting from the substantia nigra to the caudate putamen also exhibited an abnormal targeting pattern. There was no evidence of dopaminergic cell loss in the mutant SN. Abnormal localization of dopaminergic neurons in L1-minus mice was also evident in the zona incerta of the thalamus (A13 group), and the arcuate (A12) and periventricular nucleus (A14) of the hypothalamus. Cell bodies and axons in the substantia nigra, VTA, and hypothalamus of wild type mouse embryos expressed L1. These results suggested that L1 plays an important developmental role in the organization of dopaminergic neuronal cell groups in the mesencephalon and diencephalon.


Subject(s)
Dopamine/physiology , Membrane Glycoproteins/genetics , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Animals , Cell Movement/physiology , Female , Hypothalamus/cytology , Hypothalamus/immunology , In Situ Nick-End Labeling , Leukocyte L1 Antigen Complex , Male , Mice , Mice, Knockout , Neurons/enzymology , Substantia Nigra/cytology , Substantia Nigra/embryology , Tyrosine 3-Monooxygenase/analysis , Ventral Tegmental Area/cytology , Ventral Tegmental Area/embryology
4.
J Neurosci ; 21(5): 1490-500, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222639

ABSTRACT

The neural adhesion molecule L1 mediates the axon outgrowth, adhesion, and fasciculation that are necessary for proper development of synaptic connections. L1 gene mutations are present in humans with the X-linked mental retardation syndrome CRASH (corpus callosum hypoplasia, retardation, aphasia, spastic paraplegia, hydrocephalus). Three missense mutations associated with CRASH syndrome reside in the cytoplasmic domain of L1, which contains a highly conserved binding region for the cytoskeletal protein ankyrin. In a cellular ankyrin recruitment assay that uses transfected human embryonic kidney (HEK) 293 cells, two of the pathologic mutations located within the conserved SFIGQY sequence (S1224L and Y1229H) strikingly reduced the ability of L1 to recruit 270 kDa ankyrinG protein that was tagged with green fluorescent protein (ankyrin-GFP) to the plasma membrane. In contrast, the L1 missense mutation S1194L and an L1 isoform lacking the neuron-specific sequence RSLE in the cytoplasmic domain were as effective as RSLE-containing neuronal L1 in the recruitment of ankyrin-GFP. Ankyrin binding by L1 was independent of cell-cell interactions. Receptor-mediated endocytosis of L1 regulates intracellular signal transduction, which is necessary for neurite outgrowth. In rat B35 neuroblastoma cell lines stably expressing L1 missense mutants, antibody-induced endocytosis was unaffected by S1224L or S1194L mutations but appeared to be enhanced by the Y1229H mutation. These results suggested a critical role for tyrosine residue 1229 in the regulation of L1 endocytosis. In conclusion, specific mutations within key residues of the cytoplasmic domain of L1 (Ser(1224), Tyr(1229)) destabilize normal L1-ankyrin interactions and may influence L1 endocytosis to contribute to the mechanism of neuronal dysfunction in human X-linked mental retardation.


Subject(s)
Ankyrins/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Intellectual Disability/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Animals , Cell Line , Conserved Sequence/genetics , Cytoplasm/metabolism , Endocytosis/genetics , Green Fluorescent Proteins , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Intellectual Disability/metabolism , Leukocyte L1 Antigen Complex , Luminescent Proteins/genetics , Mutation, Missense , Neuroblastoma/metabolism , Neurons/cytology , Neurons/metabolism , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Signal Transduction/genetics , Syndrome , Transfection , X Chromosome/genetics
5.
J Neurosci ; 20(11): 4177-88, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10818153

ABSTRACT

The neural cell adhesion molecule L1 mediates the axon outgrowth, adhesion, and fasciculation necessary for proper development of synaptic connections. Mutations of human L1 cause an X-linked mental retardation syndrome termed CRASH (corpus callosum hypoplasia, retardation, aphasia, spastic paraplegia, and hydrocephalus), and L1 knock-out mice display defects in neuronal process extension resembling the CRASH phenotype. Little is known about the biochemical or cellular mechanism by which L1 performs neuronal functions. Here it is demonstrated that clustering of L1 with antibodies or L1 protein in rodent B35 neuroblastoma and cerebellar neuron cultures induced the phosphorylation/activation of the mitogen-activated protein kinases (MAPKs) and extracellular signal-regulated kinases 1 and 2. MAPK activation was essential for L1-dependent neurite outgrowth, because chemical inhibitors [2-(2'-amino-3'-methoxyphenyl)-oxanaphthalen-4-one and 1,4-diamino-2, 3-dicyano-1,4-bis(2-aminophenylthio)butadiene] of the MAPK kinase MEK strongly suppressed neurite outgrowth by cerebellar neurons on L1. The nonreceptor tyrosine kinase pp60(c-src) was required for L1-triggered MAPK phosphorylation, as shown in src-minus cerebellar neurons and by expression of the kinase-inactive mutant Src(K295M) in B35 neuroblastoma cells. Phosphatidylinositol 3-kinase (PI3-kinase) and the small GTPase p21(rac) were identified as signaling intermediates to MAPK by phosphoinositide and Rac-GTP assays and expression of inhibitory mutants. Antibody-induced endocytosis of L1, visualized by immunofluorescence staining and confocal microscopy of B35 cells, was blocked by expression of kinase-inactive Src(K295M) and dominant-negative dynamin(K44A) but not by inhibitors of MEK or PI3-kinase. Dynamin(K44A) also inhibited L1 antibody-triggered MAPK phosphorylation. This study supports a model in which pp60(c-src) regulates dynamin-mediated endocytosis of L1 as an essential step in MAPK-dependent neurite outgrowth on an L1 substrate.


Subject(s)
Cerebellum/physiology , Intercellular Adhesion Molecule-1/physiology , MAP Kinase Signaling System/physiology , Neurites/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Animals , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Cells, Cultured , Cerebellum/cytology , Cytoskeleton/genetics , Cytoskeleton/physiology , Endocytosis/physiology , Growth Cones/physiology , Growth Cones/ultrastructure , Humans , Mice , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/physiology , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/metabolism , p21-Activated Kinases
6.
Curr Biol ; 10(24): 1607-10, 2000.
Article in English | MEDLINE | ID: mdl-11137015

ABSTRACT

Genetic evidence indicates that cell adhesion molecules of the immunoglobulin superfamily (IgCAMs) are critical for activity-dependent synapse formation at the neuromuscular junction in Drosophila and have also been implicated in synaptic remodelling during learning in Aplysia (see [1] for review). In mammals, a widely adopted model for the process of learning at the cellular level is long-term potentiation (LTP) in the hippocampal formation. Studies in vitro have shown that antibodies to the IgCAMs L1 and NCAM reduce LTP in CA1 neurons of rat hippocampus, suggesting a role for these molecules in the modulation of synaptic efficacy, perhaps by regulating synaptic remodelling [2]. A role for NCAM in LTP has been confirmed in mice lacking NCAM [3] (but see [4]), but similar studies have not been reported for L1. Here we examine LTP in the hippocampus of mice lacking L1 [5,6], using different experimental protocols in three different laboratories. In tests of LTP in vitro and in vivo we found no significant differences between mutant animals and controls. Thus, contrary to expectation, our data suggest that L1 function is not necessary for the establishment or maintenance of LTP in the hippocampus. Impaired performance in spatial learning exhibited by L1 mutants may therefore not be due to hippocampal dysfunction [6].


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Membrane Glycoproteins/physiology , Neural Cell Adhesion Molecules/physiology , Neurons/physiology , Animals , Electrophysiology , Hippocampus/cytology , Immunoglobulins , Leukocyte L1 Antigen Complex , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mutation , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/immunology , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurosci ; 19(12): 4907-20, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10366625

ABSTRACT

In humans, mutations in the L1 cell adhesion molecule are associated with a neurological syndrome termed CRASH, which includes corpus callosum agenesis, mental retardation, adducted thumbs, spasticity, and hydrocephalus. A mouse model with a null mutation in the L1 gene (Cohen et al., 1997) was analyzed for brain abnormalities by Nissl and Golgi staining and immunocytochemistry. In the motor, somatosensory, and visual cortex, many pyramidal neurons in layer V exhibited undulating apical dendrites that did not reach layer I. The hippocampus of L1 mutant mice was smaller than normal, with fewer pyramidal and granule cells. The corpus callosum of L1-minus mice was reduced in size because of the failure of many callosal axons to cross the midline. Enlarged ventricles and septal abnormalities were also features of the mutant mouse brain. Immunoperoxidase staining showed that L1 was abundant in developing neurons at embryonic day 18 (E18) in wild-type cerebral cortex, hippocampus, and corpus callosum and then declined to low levels with maturation. In the E18 cortex, L1 colocalized with microtubule-associated protein 2, a marker of dendrites and somata. These new findings suggest new roles for L1 in the mechanism of cortical dendrite differentiation, as well as in guidance of callosal axons and regulation of hippocampal development. The phenotype of the L1 mutant mouse indicates that it is a potentially valuable model for the human CRASH syndrome.


Subject(s)
Cerebral Ventricles/abnormalities , Hippocampus/abnormalities , Membrane Glycoproteins/genetics , Neural Cell Adhesion Molecules/genetics , Pyramidal Cells/pathology , Agenesis of Corpus Callosum , Animals , Antigens, Surface/genetics , Axons/pathology , Axons/physiology , Brain Chemistry/genetics , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Cerebral Ventricles/embryology , Cerebral Ventricles/pathology , Corpus Callosum/embryology , Corpus Callosum/pathology , DNA Nucleotidylexotransferase/analysis , Dendrites/pathology , Dendrites/physiology , Female , Genotype , Hippocampus/cytology , Hippocampus/embryology , Immunoenzyme Techniques , In Situ Nick-End Labeling , Intellectual Disability/genetics , Intellectual Disability/pathology , Leukocyte L1 Antigen Complex , Male , Mice , Mice, Knockout , Pyramidal Cells/enzymology , Pyramidal Cells/ultrastructure , Septal Nuclei/abnormalities , Septal Nuclei/embryology , Septal Nuclei/pathology
8.
J Neurobiol ; 38(4): 542-58, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10084688

ABSTRACT

The neural cell adhesion molecule NCAM plays an important role in axonal growth, learning, and memory. A signaling pathway has been elucidated in which clustering of the NCAM140 isoform in the neural plasma membrane stimulated the activating phosphorylation of mitogen-activated protein kinases (MAPKs) and the transcription factor cyclic AMP response-element binding protein (CREB). NCAM clustering transiently induced dual phosphorylation (activation) of the MAPKs ERK1 and ERK2 (extracellular signal-regulated kinases) by a pathway regulated by the focal adhesion kinase p125fak, p59fyn, Ras, and MAPK kinase. CREB phosphorylation at serine 133 induced by NCAM was dependent in part on an intact MAPK pathway. c-Jun N-terminal kinase, which is associated with apoptosis and cellular stress, was not activated by NCAM. Inhibition of the MAPK pathway in rat cerebellar neuron cultures selectively reduced NCAM-stimulated neurite outgrowth. These results define an NCAM signal transduction mechanism with the potential for modulating the expression of genes needed for axonal growth, survival, and synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Mitogen-Activated Protein Kinases , Neural Cell Adhesion Molecules/physiology , Neurites/physiology , Neurons/metabolism , ras Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , JNK Mitogen-Activated Protein Kinases , L Cells , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/immunology , Neuroblastoma , Phosphorylation , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
9.
Exp Eye Res ; 68(2): 201-10, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10068485

ABSTRACT

Rek is a receptor-class tyrosine kinase of the Axl/Tyro3 family that has transforming capability and is expressed in the avian nervous system. To gain insight into its normal cellular function, the spatial and temporal expression of Rek was analyzed in the developing chick retina by immunoperoxidase staining and Western blotting using Rek-specific antibodies. Rek was developmentally regulated in the retina with maximal expression of the 140 kD full-length kinase at embryonic stage 34 (E8), when retinal glia and neurons begin to differentiate. Rek immunoreactivity was most prominent in the processes of developing and mature retinal Müller glia, and appeared transiently in the bodies of differentiating ganglion and amacrine neurons. In the developing optic tract Rek was found in oligodendroglial-type cells but not in ganglion cell axons. Rek antibodies also stained brain ependymal cells and some cerebellar neuronal cell types (Purkinje, basket and stellate cells). This expression pattern suggests that the Rek tyrosine kinase participates in an aspect of Müller glial function, and may also contribute to the development of restricted populations of glia and neurons in the brain and retina.


Subject(s)
Neuroglia/enzymology , Neurons/enzymology , Retina/embryology , Animals , Blotting, Western , Brain/embryology , Brain/enzymology , Cell Differentiation , Chick Embryo , Immunoenzyme Techniques , Neurons/cytology , Oncogene Proteins/metabolism , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/metabolism , Retina/enzymology , Axl Receptor Tyrosine Kinase
10.
J Neurobiol ; 36(1): 53-63, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658338

ABSTRACT

The Src-family tyrosine kinases p59fyn and pp60c-src are localized on axons of the mouse olfactory nerve during the initial stages of axonal growth, but their functional roles remain to be defined. To study the role of these kinases, we analyzed the trajectory of the olfactory nerve in E11.5 homozygous null mutant mice lacking single src or fyn gens and double mutants lacking both genes. Primary olfactory axons of single and double mutants exited the olfactory epithelium and projected toward the telencephalon, but displayed differences in fasciculation. The fyn-minus olfactory nerve had significantly more fascicles than than src-minus nerve. Most strikingly, the primary olfactory nerve of src/fyn double mutants showed the greatest degree of defasciculation. These defects, identified by NCAM labeling, were not due to apparent changes in the size of the olfactory epithelium. With the exception of the src-minus mice, which had fever fascicles than the wild type, no obvious differences were observed in coalescence of vomeronasal axons from mutant mice. The mesenchyme of the double and single mutants exhibited only subtle changes in laminin and fibronectin staining, indicating that the adhesive environment of the mesenchyme may contribute in part to defects in fasciculation. The results suggest that signaling pathways mediated by p59fyn and pp60c-src contribute to the appropriate fasciculation of axons in the nascent olfactory system, and comprise partially compensatory mechanisms for axonal adhesion and guidance.


Subject(s)
Axons/physiology , Olfactory Pathways/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Proto-Oncogene Proteins/physiology , Animals , Embryonic and Fetal Development/physiology , Extracellular Matrix Proteins/metabolism , Mice , Mutation , Olfactory Pathways/embryology , Olfactory Pathways/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , Proto-Oncogene Proteins pp60(c-src)/genetics , Vomeronasal Organ/embryology , Vomeronasal Organ/innervation
11.
J Neurocytol ; 27(2): 119-26, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9609402

ABSTRACT

Brain microtubule dynamics were studied by video-enhanced differential interference contrast microscopy in a cytosolic extract from fetal rat brain, prepared under conditions designed to produce minimal alterations in microtubule stability. With urchin sperm axoneme fragments as nucleation seeds, the extract was shown to support cellular-like microtubule dynamics. Microtubules elongated from one end of the axonemes, and did not spontaneously self-assemble in the absence of axonemes. The following microtubule kinetic parameters were measured in the extract: velocity of elongation (8.1 mm/min), velocity of rapid shortening (5.8 mm/min), catastrophe frequency (0.17 min(-1)), and rescue frequency (1 min(-1)). These parameters were in close agreement with reported values for growth cones of living neurons. Microtubule properties in the fetal brain extract were shown to be affected by agents with known effects on the cytoskeleton. pp60c-src, a tyrosine kinase important in cell adhesion molecule-dependent axon growth, caused small increases in the frequency of microtubule catastrophe (0.31 min(-1)) and rescue (2 min(-1)) without changing the velocities of elongation or rapid-shortening. Although pp60c-src phosphorylated purified porcine brain tubulin in vitro, it did not elicit significant changes in its polymerization properties, suggesting that other cytoskeletal proteins in the brain extract are involved in modulating microtubule dynamics. The cytosolic extract of fetal rat brain provides a useful system for studying regulation of microtubule assembly in neuronal growth cones.


Subject(s)
Brain Chemistry , Cytosol/metabolism , Microtubules/metabolism , Animals , Cytosol/chemistry , Cytosol/enzymology , Fetus , Microtubules/chemistry , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Sprague-Dawley , Tubulin/metabolism , Tyrosine/metabolism
12.
J Biol Chem ; 272(13): 8310-9, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9079653

ABSTRACT

Axonal growth cones respond to adhesion molecules and extracellular matrix components by rapid morphological changes and growth rate modification. Neurite outgrowth mediated by the neural cell adhesion molecule (NCAM) requires the src family tyrosine kinase p59(fyn) in nerve growth cones, but the molecular basis for this interaction has not been defined. The NCAM140 isoform, which is found in migrating growth cones, selectively co-immunoprecipitated with p59(fyn) from nonionic detergent (Brij 96) extracts of early postnatal mouse cerebellum and transfected rat B35 neuroblastoma and COS-7 cells. p59(fyn) did not associate significantly with the NCAM180 isoform, which is found at sites of stable neural cell contacts, or with the glycophosphatidylinositol-linked NCAM120 isoform. pp60(c-)src, a tyrosine kinase that promotes neurite growth on the neuronal cell adhesion molecule L1, did not interact with any NCAM isoform. Whereas p59(fyn) was constitutively associated with NCAM140, the focal adhesion kinase p125(fak), a nonreceptor tyrosine kinase known to mediate integrin-dependent signaling, became recruited to the NCAM140-p59(fyn) complex when cells were reacted with antibodies against the extracellular region of NCAM. Treatment of cells with a soluble NCAM fusion protein or with NCAM antibodies caused a rapid and transient increase in tyrosine phosphorylation of p125(fak) and p59(fyn). These results suggest that NCAM140 binding interactions at the cell surface induce the assembly of a molecular complex of NCAM140, p125(fak), and p59(fyn) and activate the catalytic function of these tyrosine kinases, initiating a signaling cascade that may modulate growth cone migration.


Subject(s)
Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Insulin/metabolism , Animals , COS Cells , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Molecular Weight , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-fyn , Rats , Transfection
13.
J Biol Chem ; 271(46): 29049-59, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8910558

ABSTRACT

Rek (retina-expressed kinase) has been identified as a putative novel receptor-type tyrosine kinase of the Axl/Tyro3 family with a potential role in neural cell development. rek clones were isolated from a chick embryonic brain cDNA library with a DNA probe obtained by reverse transcriptase-polymerase chain reaction of mRNA from Müller glia-like cells cultured from chick embryonic retina. Sequence analysis indicated that Rek is a protein of 873 amino acids with an extracellular region composed of two immunoglobulin-like domains followed by two fibronectin type III domains with eight predicted N-glycosylation sites. Two consensus src homology 2 domain binding sites are present in the cytoplasmic domain, suggesting that Rek activates several signal transduction pathways. Northern analysis of rek mRNA revealed a 5.5-kilobase transcript in chick brain, retina, and kidney and in primary cultures of retinal Müller glia-like cells. Rek protein was identified by immunoprecipitation and immunoblotting as a 140-kDa protein expressed in the chick retina at embryonic days 6-13, which corresponded to the major period of neuronal and glial differentiation. Transfection of rek cDNA into COS cells resulted in transient expression of a putative precursor of 106 kDa that autophosphorylated in immune complex protein kinase assays. Overexpression of rek cDNA in mouse NIH3T3 fibroblasts resulted in activation of the 140-kDa rek kinase and induction of morphologically transformed foci. These properties indicated that Rek has oncogenic potential when overexpressed, but its normal function is likely to be related to cell-cell recognition events governing the differentiation or proliferation of neural cells.


Subject(s)
Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line, Transformed , Chick Embryo , Cloning, Molecular , DNA, Complementary , Eye Proteins/genetics , Eye Proteins/metabolism , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Sequence Homology, Amino Acid , Axl Receptor Tyrosine Kinase
14.
Perspect Dev Neurobiol ; 4(2-3): 169-81, 1996.
Article in English | MEDLINE | ID: mdl-9168199

ABSTRACT

Nerve growth cone guidance is a highly complex feat, involving coordination of cell adhesion molecules, trophic factor gradients, and extracellular matrix proteins. While navigating through the developing nervous system, the growth cone must integrate diverse environmental signals into a singular response. The repertoire of growth cone responses to these extracellular cues includes axonal growth, fasciculation, and synaptic stabilization, which are achieved through dynamic changes in the cytoskeleton and modulation of gene expression. It has become evident that interactions between cell adhesion molecules can activate intracellular signaling pathways in neurons. Such signaling pathways are just beginning to be defined for the axonal growth promoting molecules L1 and NCAM which are members of the immunoglobulin (Ig) superfamily. Recent findings have revealed that L1 and NCAM induce neurite outgrowth by activating intracellular signaling pathways in the growth cone mediated by two different members of the src family of nonreceptor protein tyrosine kinases (PTKs), pp60(c-src) and p59(fyn5,6). Growth cones display diverse morphologies and variable motility on these different cell adhesion molecules, which are likely to be generated by src kinases. In this review we will address novel features of nonreceptor PTKs of the src family which dictate their distinctive molecular interactions with cell adhesion molecules and signaling components.


Subject(s)
Neural Cell Adhesion Molecules/physiology , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/physiology , Signal Transduction , src-Family Kinases/physiology , Animals , Humans , Neurites/physiology , Structure-Activity Relationship , src-Family Kinases/genetics
15.
J Neurochem ; 65(1): 84-95, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7790898

ABSTRACT

Triggering of the cell adhesion molecules L1 or N-CAM in a nerve growth cone membrane fraction from fetal rat brain with purified L1 or N-CAM or specific antibodies decreases the steady-state levels of protein tyrosine phosphorylation in the membranes. Here we report that triggering of L1 and N-CAM in the growth cone-enriched membrane fraction with a subset of antibodies directed against the extracellular region of L1 and N-CAM elicited dephosphorylation of endogenous protein substrates, indicating the presence of a cell adhesion molecule-activated phosphatase. The most prominent substrates were a membrane-associated 200-kDa protein and tubulin, both of which were dephosphorylated on tyrosine and serine/threonine residues in response to L1 or N-CAM triggering. The antibody-induced phosphatase was inhibited by agents that blocked tyrosine and serine/threonine phosphatases, including sodium orthovanadate, vanadyl sulfate, zinc cations, heparin, and sodium pyrophosphate. Purified L1 and N-CAM fragments and other antibodies reacting with the extracellular region of these adhesion molecules did not activate the phosphatase but did inhibit tyrosine phosphorylation. These properties suggested that triggering of L1 and N-CAM can lead to either phosphatase activation or tyrosine kinase inhibition in growth cone membranes. These findings implicate protein phosphatases in addition to tyrosine kinases as components of L1 and N-CAM intracellular signaling pathways in growth cones.


Subject(s)
Antibodies/physiology , Axons/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/immunology , Phosphoprotein Phosphatases/metabolism , Amino Acids/analysis , Animals , Cells, Cultured , Enzyme Activation , Leukocyte L1 Antigen Complex , Membrane Proteins/chemistry , Membranes/metabolism , Molecular Weight , Phosphoprotein Phosphatases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Tubulin/chemistry
16.
Brain Res Mol Brain Res ; 28(1): 110-6, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7707863

ABSTRACT

Regulation of protein function through tyrosine phosphorylation is critical to many developmental processes involving cell-cell communication. A number of protein tyrosine phosphatases (PTPs) have been identified in the early postnatal and mature central nervous system (CNS), but the PTPs expressed during its development have not been well characterized. Using a polymerase chain reaction with degenerate primers, we analyzed PTPs expressed in fetal (E18) rat brain and Müller glia cultures from embryonic chick retina, two systems in which cell-to-cell contacts are numerous. Fetal rat brain expressed four known receptor-like PTPs (PTP delta, LAR, LAR-PTP2, LRP (PTP alpha)) and the non-receptor phosphatase PTP1B. Müller glia exhibited a distinct but overlapping pattern of expression: four known receptor PTPs (PTP alpha, PTP gamma, PTP delta, PTP zeta) and PTP1B. In addition, two novel PTPs, termed MG-PTP1 and 2 (Müller glia PTP 1 and 2) were identified in Müller glia cDNA. MG-PTP1 was related to, but distinct from PTP delta, while MG-PTP2 was related to, but distinct from the cytosolic T-cell phosphatase. These results demonstrate that a distinct but overlapping set of PTPs is expressed in the developing brain and retinal Müller glia, including two novel PTPs that may participate in neural cell communication.


Subject(s)
Brain/metabolism , Neuroglia/metabolism , Protein Tyrosine Phosphatases/biosynthesis , Retina/metabolism , Animals , Chick Embryo , DNA, Complementary , Gene Expression , Molecular Sequence Data , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Sequence Homology, Amino Acid
17.
J Cell Biol ; 127(3): 825-33, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7962063

ABSTRACT

Src-related nonreceptor protein tyrosine kinases in nerve growth cones (p59fyn, pp60c-src, and pp62c-yes) are potential intracellular signaling molecules for cell adhesion molecule-directed axonal growth. To determine whether src-related tyrosine kinases mediate NCAM-dependent neurite outgrowth, cultures of cerebellar and sensory neurons from fyn-, src-, and yes- minus mice were analyzed for neurite outgrowth on monolayers of NCAM140-transfected L fibroblasts. NCAM-dependent neurite outgrowth was selectively inhibited in cultures of cerebellar and dorsal root ganglion neurons from fyn-, but not src- or yes- mice. Neurite outgrowth by fyn-, src-, or yes- neurons on untransfected fibroblast monolayers was unaffected, indicating that these kinases do not contribute significantly to axon growth on at least some integrins or other adhesive substrates present on fibroblasts. This study demonstrates that p59fyn is an essential component of the NCAM signaling pathway leading to axonal growth.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cerebellum/physiology , Neurites/physiology , Neurons/physiology , Proto-Oncogene Proteins/genetics , Animals , Base Sequence , Cell Adhesion Molecules, Neuronal/biosynthesis , Cells, Cultured , DNA Primers , Enzyme-Linked Immunosorbent Assay , Kinetics , L Cells , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Molecular Sequence Data , Neurites/ultrastructure , Neurons, Afferent/physiology , Polymerase Chain Reaction , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn , Transfection
18.
Neuron ; 12(4): 873-84, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7512817

ABSTRACT

The nonreceptor tyrosine protein kinases pp60c-src, p59fyn, and pp62c-yes are localized in growth cones of developing neurons, but their function is undefined. To determine whether these tyrosine kinases were capable of regulating substrate-dependent axon growth, cultures of cerebellar neurons from wild-type, src-, fyn-, and yes- mice were analyzed for neurite outgrowth on the neural cell adhesion molecule L1 or the extracellular matrix protein laminin. The rate of neurite extension on L1 was reduced in src-, but not in fyn- or yes- neurons. Neurite extension on laminin was unaltered in src-, fyn-, or yes- neurons, indicating that pp60c-src, p59fyn, or pp62c-yes is not likely to participate in integrin-dependent axon growth. These results demonstrate that pp60c-src is a component of the intracellular signaling pathway in L1-mediated axonal growth and suggest that Src-related nonreceptor tyrosine kinases may have distinct, nonredundant functions in the nervous system.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cerebellum/cytology , Neurites/physiology , Neurons/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src)/physiology , Animals , Cells, Cultured , Immunoblotting , Laminin/physiology , Leukocyte L1 Antigen Complex , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Phosphotyrosine , Tyrosine/analogs & derivatives , Tyrosine/metabolism
19.
Oncogene ; 8(6): 1429-36, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502471

ABSTRACT

Expression of the c-fyn proto-oncogene was analysed in the developing and adult rat brain by subcellular fractionation and immunocytochemistry using polyclonal antibodies specific for its protein product (p59fyn). Immunoperoxidase staining revealed widespread localization of p59fyn in developing axonal tracts throughout the fetal (E18) rat brain. fyn immunoreactivity was not observed in most axon-rich regions of the adult brain, but continued to be expressed at elevated levels in the adult olfactory and vomeronasal systems. At other sites in the adult rat brain, fyn immunoreactivity was restricted to cell bodies of neuronal subpopulations, especially those in brain stem and hypothalamic nuclei, and to subpopulations of glial cells along axonal tracts in the medulla, optic nerve and white matter of the spinal cord. In the peripheral nervous system, fyn staining was also prominent in Schwann cells. Subcellular fractionation of fetal and adult rat brain confirmed the immunocytochemical localization, demonstrating an enrichment of p59fyn in membranes from a fetal brain fraction containing nerve growth cones, and lower levels in adult brain where there was only a small enrichment in synaptosomal membranes. The developmental regulation of p59fyn suggests that the fyn tyrosine kinase may serve separate cellular roles in axonal growth and specialized functions of mature neurons and glia.


Subject(s)
Axons/ultrastructure , Brain/cytology , Neurons/cytology , Optic Nerve/cytology , Proto-Oncogene Proteins/analysis , Sciatic Nerve/cytology , Spinal Cord/cytology , Trigeminal Nerve/cytology , Animals , Axons/physiology , Brain/embryology , Chickens , Fetus , Immunoblotting , Immunohistochemistry , Neuroglia/cytology , Neuroglia/physiology , Neurons/physiology , Optic Nerve/embryology , Organ Specificity , Protein-Tyrosine Kinases/analysis , Proto-Oncogene Proteins c-fyn , Rats , Rats, Sprague-Dawley , Sciatic Nerve/embryology , Spinal Cord/embryology , Subcellular Fractions/chemistry , Trigeminal Nerve/embryology
20.
J Neurochem ; 60(6): 2134-44, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7684068

ABSTRACT

Genistein and other inhibitors of protein tyrosine kinases were examined for effects on neurite elongation and growth cone morphology in the rat PC12 pheochromocytoma cell line. Genistein increased the rate of neurite elongation in PC12 cells grown on a collagen/polylysine substratum after priming with nerve growth factor (NGF), but had no effect on undifferentiated cells. Steady-state levels of phosphotyrosine-modified proteins (105, 59, 52, and 46 kDa) were reduced in NGF-primed cells by genistein treatment. The target of genistein action did not appear to be the NGF receptor/trk tyrosine kinase because the presence of NGF in cultures of NGF-primed cells was not necessary for genistein-stimulated neurite outgrowth. The tyrosine kinase inhibitors tyrphostin RG508964 and herbimycin A also increased the rate of neurite elongation in NGF-primed PC12 cells. Video-enhanced differential interference contrast microscopy revealed that growth cones of genistein-treated cells had less complex morphologies and were less dynamic than untreated cells, with short filopodia restricted to the leading edge, unlike untreated cells whose growth cones exhibited longer, more numerous filopodia and lamellipodia, which remodeled continuously. These results suggest that protein tyrosine kinase activity in PC12 cells negatively regulates neurite outgrowth and directly or indirectly affects growth cone morphology.


Subject(s)
Catechols/pharmacology , Isoflavones/pharmacology , Nerve Growth Factors/pharmacology , Neurites/physiology , Nitriles/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrphostins , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation , Cell Division/drug effects , Electrophoresis, Polyacrylamide Gel , Genistein , Kinetics , Molecular Weight , Neurites/drug effects , Neurites/ultrastructure , PC12 Cells , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphotyrosine , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, trkA , Tyrosine/analogs & derivatives , Tyrosine/analysis , Video Recording
SELECTION OF CITATIONS
SEARCH DETAIL
...