Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Neurochem ; 137(5): 744-55, 2016 06.
Article in English | MEDLINE | ID: mdl-26991250

ABSTRACT

Axonal growth cone motility requires precise regulation of adhesion to navigate the complex environment of the nervous system and reach its target. Myristoylated alanine-rich C kinase substrate (MARCKS) protein is enriched in the developing brain and plays an important, phosphorylation-dependent role in the modulation of axonal growth cone adhesion. The ratio of phospho-MARCKS (MARCKS-P) to total MARCKS controls adhesion modulation and spreading of the axonal growth cone. Pin1, a peptidyl-prolyl cis/trans isomerase (PPIase) that recognizes and binds to phosphorylated serine/threonine residues preceded by a proline (pSer/Thr-Pro) is also expressed in the developing brain. Here, we show that Pin1 is present in the growth cone, interacts with MARCKS-P, and regulates its dephosphorylation. We also described morphological alterations in the corpus callosum and cerebral cortex fibers of the Pin1 knockout mouse brain that may be caused by the misregulation of MARCKS-P and alterations of neuronal adhesion. We have shown that MARCKS, a critical protein in the movement of neuronal growth cones, is in turn regulated by both phosphorylation and cis-trans peptidyl isomerization mediated by Pin1. In the absence of Pin1, MARCKS is hyperphosphorylated, leading to loss of adhesions, and collapse of the growth cone. The Pin1 KO mice exhibited disturbed neuronal projections from the cerebral cortex and reduced white matter tracks such as the corpus callosum. This study highlights a novel function of Pin1 in neurodevelopment.


Subject(s)
Axons/physiology , Cell Adhesion/physiology , Growth Cones/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Female , Intracellular Signaling Peptides and Proteins/chemistry , Isomerism , Membrane Proteins/chemistry , Mice , Mice, Knockout , Myristoylated Alanine-Rich C Kinase Substrate , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley
2.
Nat Cell Biol ; 16(5): 434-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24705552

ABSTRACT

Development of the nervous system requires extensive axonal and dendritic growth during which neurons massively increase their surface area. Here we report that the endoplasmic reticulum (ER)-resident SNARE Sec22b has a conserved non-fusogenic function in plasma membrane expansion. Sec22b is closely apposed to the plasma membrane SNARE syntaxin1. Sec22b forms a trans-SNARE complex with syntaxin1 that does not include SNAP23/25/29, and does not mediate fusion. Insertion of a long rigid linker between the SNARE and transmembrane domains of Sec22b extends the distance between the ER and plasma membrane, and impairs neurite growth but not the secretion of VSV-G. In yeast, Sec22 interacts with lipid transfer proteins, and inhibition of Sec22 leads to defects in lipid metabolism at contact sites between the ER and plasma membrane. These results suggest that close apposition of the ER and plasma membrane mediated by Sec22 and plasma membrane syntaxins generates a non-fusogenic SNARE bridge contributing to plasma membrane expansion, probably through non-vesicular lipid transfer.


Subject(s)
Cell Membrane/metabolism , Cerebral Cortex/metabolism , Endoplasmic Reticulum/metabolism , Neurons/metabolism , R-SNARE Proteins/metabolism , Animals , Animals, Newborn , COS Cells , Carrier Proteins/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Chlorocebus aethiops , Gestational Age , HeLa Cells , Humans , Lipid Metabolism , Mice , R-SNARE Proteins/genetics , RNA Interference , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Syntaxin 1/genetics , Syntaxin 1/metabolism , Time Factors , Transfection
3.
FASEB J ; 28(1): 195-205, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24036883

ABSTRACT

Amyloid precursor protein (APP), encoded on Hsa21, functions as a cell adhesion molecule (CAM) in axonal growth cones (GCs) of the developing brain. We show here that axonal GCs of human fetal Down syndrome (DS) neurons (and of a DS mouse model) overexpress APP protein relative to euploid controls. We investigated whether DS neurons generate an abnormal, APP-dependent GC phenotype in vitro. On laminin, which binds APP and ß1 integrins (Itgb1), DS neurons formed enlarged and faster-advancing GCs compared to controls. On peptide matrices that bind APP only, but not on those binding exclusively Itgb1 or L1CAM, DS GCs were significantly enlarged (2.0-fold), formed increased close adhesions (1.8-fold), and advanced faster (1.4-fold). In assays involving alternating stripes of monospecific matrices, human control GCs exhibited no preference for any of the substrates, whereas DS GCs preferred the APP-binding matrix (cross-over decreased significantly from 48.2 to 27.2%). Reducing APP expression in DS GCs with siRNA normalized most measures of the phenotype, including substrate choice. These experiments show that human DS neurons exhibit an APP-dependent, abnormal GC phenotype characterized by increased adhesion and altered contact guidance. The results suggest that APP overexpression may perturb axonal pathfinding and circuit formation in developing DS brain.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Down Syndrome/metabolism , Animals , Brain/metabolism , Cell Adhesion Molecules/metabolism , Growth Cones/metabolism , Hippocampus/metabolism , Humans , In Vitro Techniques , Laminin/metabolism , Mice
4.
FASEB J ; 28(2): 692-704, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174428

ABSTRACT

The aim of this study was to identify the molecular signals produced in human endothelial cells (ECs) by the interaction of α5ß1 integrin with soluble vascular endothelial growth factor receptor-1 (sVEGFR-1) present in the extracellular matrix. We generated a gene expression profile of ECs adhering to sVEGFR-1 or to fibronectin, the classic extracellular matrix ligand for α5ß1 integrin or in a nonadhering condition. Several biological pathways were differently modulated, 3 protein kinase C substrates [adducin, myristoylated alanine-rich protein kinase C substrate (MARCKS), and radixin] were differently expressed and phosphorylated when cells adhering to sVEGFR-1 were compared with those adhering to fibronectin. Rac1 activation and Gα13 protein involvement through the interaction with radixin were also detected after attachment to sVEGFR-1, and these responses depended on active VEGFR-2 signaling. On sVEGFR-1, ECs exhibited a motile phenotype that was consistent with the abundant presence of MARCKS, a stabilizer of dynamic adhesions. Moreover, ECs silenced for radixin expression no longer responded to the proangiogenic VEGFR-1-derived peptide 12. We propose that the presence of sVEGFR-1 in the EC microenvironment directs α5ß1 integrin signaling to generate a dynamic, motile phenotype. Our findings also provide new insights into the mechanism of action of proangiogenic peptide 12, relevant to a therapeutic perspective.


Subject(s)
Cell Adhesion/physiology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Blotting, Western , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Fibronectins/metabolism , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
5.
PLoS One ; 8(5): e64521, 2013.
Article in English | MEDLINE | ID: mdl-23691241

ABSTRACT

Amyloid precursor protein (APP), a transmembrane glycoprotein, is well known for its involvement in the pathogenesis of Alzheimer disease of the aging brain, but its normal function is unclear. APP is a prominent component of the adult as well as the developing brain. It is enriched in axonal growth cones (GCs) and has been implicated in cell adhesion and motility. We tested the hypothesis that APP is an extracellular matrix adhesion molecule in experiments that isolated the function of APP from that of well-established adhesion molecules. To this end we plated wild-type, APP-, or ß1-integrin (Itgb1)- misexpressing mouse hippocampal neurons on matrices of either laminin, recombinant L1, or synthetic peptides binding specifically to Itgb1 s or APP. We measured GC adhesion, initial axonal outgrowth, and substrate preference on alternating matrix stripes and made the following observations: Substrates of APP-binding peptide alone sustain neurite outgrowth; APP dosage controls GC adhesion to laminin and APP-binding peptide as well as axonal outgrowth in Itgb1- independent manner; and APP directs GCs in contact guidance assays. It follows that APP is an independently operating cell adhesion molecule that affects the GC's phenotype on APP-binding matrices including laminin, and that it is likely to affect axon pathfinding in vivo.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Adhesion Molecules/metabolism , Growth Cones/metabolism , Animals , Axons/metabolism , Brain/cytology , Cell Adhesion , Gene Expression Regulation , Laminin/metabolism , Mice , Pyramidal Cells/cytology , Rats
6.
PLoS One ; 7(2): e31858, 2012.
Article in English | MEDLINE | ID: mdl-22384089

ABSTRACT

The growth cone, the tip of the emerging neurite, plays a crucial role in establishing the wiring of the developing nervous system. We performed an extensive proteomic analysis of axonal growth cones isolated from the brains of fetal Sprague-Dawley rats. Approximately 2000 proteins were identified at ≥ 99% confidence level. Using informatics, including functional annotation cluster and KEGG pathway analysis, we found great diversity of proteins involved in axonal pathfinding, cytoskeletal remodeling, vesicular traffic and carbohydrate metabolism, as expected. We also found a large and complex array of proteins involved in translation, protein folding, posttranslational processing, and proteasome/ubiquitination-dependent degradation. Immunofluorescence studies performed on hippocampal neurons in culture confirmed the presence in the axonal growth cone of proteins representative of these processes. These analyses also provide evidence for rough endoplasmic reticulum and reveal a reticular structure equipped with Golgi-like functions in the axonal growth cone. Furthermore, Western blot revealed the growth cone enrichment, relative to fetal brain homogenate, of some of the proteins involved in protein synthesis, folding and catabolism. Our study provides a resource for further research and amplifies the relatively recently developed concept that the axonal growth cone is equipped with proteins capable of performing a highly diverse range of functions.


Subject(s)
Axons/metabolism , Growth Cones/metabolism , Proteomics/methods , Animals , Axons/physiology , Brain/embryology , Brain/metabolism , Carbohydrates/chemistry , Cluster Analysis , Computational Biology/methods , Hippocampus/metabolism , Microscopy, Fluorescence/methods , Protein Folding , Protein Processing, Post-Translational , Proteome , Rats , Rats, Sprague-Dawley
7.
J Neurochem ; 120(6): 974-84, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22220903

ABSTRACT

The repellent semaphorin 3A (Sema3A) causes growth cone turning or collapse by triggering cytoskeletal rearrangements and detachment of adhesion sites. Growth cone detachment is dependent on eicosanoid activation of protein kinase C epsilon (PKCε), but the characterization of the phospholipase A(2) (PLA(2) ) that releases arachidonic acid (AA) for eicosanoid synthesis has remained elusive. Here, we show, in rat dorsal root ganglion (DRG) neurons, that Sema3A stimulates PLA(2) activity, that Sema3A-induced growth cone turning and collapse are dependent on the release of AA, and that the primary PLA(2) involved is the group IV α isoform (GIVA). Silencing GIVA expression renders growth cones resistant to Sema3A-induced collapse, and GIVA inhibition reverses Sema3A-induced repulsion into attraction. These studies identify a novel, early step in Sema3A-signaling and a PLA(2) necessary for growth cone repulsion and collapse.


Subject(s)
Group IV Phospholipases A2/metabolism , Growth Cones/metabolism , Neurons/cytology , Animals , Cell Movement/physiology , Cells, Cultured , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Group IV Phospholipases A2/genetics , Growth Cones/drug effects , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/metabolism , RNA, Small Interfering/pharmacology , Rats , Semaphorin-3A/metabolism , Semaphorin-3A/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors , Transfection
8.
Mol Genet Metab ; 104(1-2): 13-22, 2011.
Article in English | MEDLINE | ID: mdl-21835664

ABSTRACT

A December 2010 meeting, "Down Syndrome: National Conference on Patient Registries, Research Databases, and Biobanks," was jointly sponsored by the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) at the National Institutes of Health (NIH) in Bethesda, MD, and the Global Down Syndrome Foundation (GDSF)/Linda Crnic Institute for Down Syndrome based in Denver, CO. Approximately 70 attendees and organizers from various advocacy groups, federal agencies (Centers for Disease Control and Prevention, and various NIH Institutes, Centers, and Offices), members of industry, clinicians, and researchers from various academic institutions were greeted by Drs. Yvonne Maddox, Deputy Director of NICHD, and Edward McCabe, Executive Director of the Linda Crnic Institute for Down Syndrome. They charged the participants to focus on the separate issues of contact registries, research databases, and biobanks through both podium presentations and breakout session discussions. Among the breakout groups for each of the major sessions, participants were asked to generate responses to questions posed by the organizers concerning these three research resources as they related to Down syndrome and then to report back to the group at large with a summary of their discussions. This report represents a synthesis of the discussions and suggested approaches formulated by the group as a whole.


Subject(s)
Biological Specimen Banks/statistics & numerical data , Biomedical Research/statistics & numerical data , Databases as Topic/statistics & numerical data , Down Syndrome/epidemiology , Registries/statistics & numerical data , Humans , United States/epidemiology
9.
J Neurosci ; 29(42): 13292-301, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19846717

ABSTRACT

Axonal elongation is one of the hallmarks of neuronal polarization. This phenomenon requires axonal membrane growth by exocytosis of plasmalemmal precursor vesicles (PPVs) at the nerve growth cone, a process regulated by IGF-1 activation of the PI3K (phosphatidylinositol-3 kinase) pathway. Few details are known, however, about the targeting mechanisms for PPVs. Here, we show, in cultured hippocampal pyramidal neurons and growth cones isolated from fetal rat brain, that IGF-1 activates the GTP-binding protein TC10, which triggers translocation to the plasma membrane of the exocyst component exo70 in the distal axon and growth cone. We also show that TC10 and exo70 function are necessary for addition of new membrane and, thus, axon elongation stimulated by IGF-1. Moreover, expression silencing of either TC10 or exo70 inhibit the establishment of neuronal polarity by hindering the insertion of IGF-1 receptor in one of the undifferentiated neurites. We conclude that, in hippocampal pyramidal neurons in culture, (1) membrane expansion at the axonal growth cone is regulated by IGF-1 via a cascade involving TC10 and the exocyst complex, (2) TC10 and exo70 are essential for the polarized externalization of IGF-1 receptor, and (3) this process is necessary for axon specification.


Subject(s)
Axons/physiology , Axons/ultrastructure , Insulin-Like Growth Factor I/pharmacology , Pyramidal Cells/cytology , Vesicular Transport Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Axons/drug effects , Cells, Cultured , Cellular Structures/drug effects , Cellular Structures/metabolism , Chromones/pharmacology , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Morpholines/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptor, IGF Type 1/physiology , Time Factors , Transfection/methods
10.
J Cell Sci ; 122(Pt 13): 2300-10, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509053

ABSTRACT

Cell motility necessitates the rapid formation and disassembly of cell adhesions. We have studied adhesions in a highly motile melanoma cell line using various biochemical approaches and microscopic techniques to image close adhesions. We report that WM-1617 melanoma cells contain at least two types of close adhesion: classic focal adhesions and more extensive, irregularly shaped adhesions that tend to occur along lamellipodial edges. In contrast to focal adhesions, these latter adhesions are highly dynamic and can be disassembled rapidly via protein kinase C (PKC) activation (e.g. by eicosanoid) and MARCKS phosphorylation. MARCKS overexpression, however, greatly increases the area of close adhesions and renders them largely refractory to PKC stimulation. This indicates that nonphosphorylated MARCKS is an adhesion stabilizer. Unlike focal adhesions, the dynamic adhesions contain alpha3 integrin and MARCKS, but they do not contain the focal adhesion marker vinculin. Overall, these results begin to define the molecular and functional properties of dynamic close adhesions involved in cell motility.


Subject(s)
Cell Adhesion , Intercellular Junctions/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma, Experimental/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Humans , Integrin alpha3/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Melanoma/pathology , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Mice , Myristoylated Alanine-Rich C Kinase Substrate , Paxillin/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/metabolism , Vinculin/metabolism
11.
Nat Rev Neurosci ; 10(4): 251-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19259102

ABSTRACT

The formation of axons and dendrites and maintenance of the neuron's vastly expanded surface require the continuous addition of new membrane. This is achieved by membrane synthesis through the secretory pathway followed by regulated vesicle fusion with the plasma membrane, typically in the distal neurite. However, it is far from simple: multiple distinct membrane carriers are used to target specific membrane domains, dendrites seem to operate semi-autonomously from the rest of the neuron, and exocytosis for membrane expansion is different from that for release of synaptic vesicles. Current knowledge of this process and its implications for neuronal development, function and repair are reviewed.


Subject(s)
Axons/physiology , Cell Membrane/physiology , Exocytosis/physiology , Neurites/physiology , Neurons/physiology , Animals , Humans , Membrane Proteins/metabolism , Neurons/cytology
12.
Nutrition ; 24(9): 860-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18725084

ABSTRACT

OBJECTIVES: The role of neuropeptides in nervous system function is still in many cases undefined. In the present study we examined a possible role of the 36-amino acid neuropeptide Y (NPY) with regard to three functions: axon guidance and attraction/repulsion, adult neurogenesis, and control of food intake. METHODS: Growth cones from embryonic dorsal root ganglion neurons were studied in culture during asymmetrical gradient application of NPY. Growth cones were monitored over a 60-min period, and final turning angle and growth rate were recorded. In the second part the NPY Y(1) and Y(2) receptors were studied in the subventricular zone, the rostral migratory stream, and the olfactory bulb in normal mice and mice with genetically deleted NPY Y(1) or Y(2) receptors. In the third part an anorectic mouse was analyzed with immunohistochemistry. RESULTS: 1) NPY elicited an attractive turning response and an increase in growth rate, effects exerted via the NPY Y(1) receptor. 2) The NPY Y(1) receptor was expressed in neuroblasts in the anterior rostral migratory stream. Mice deficient in the Y(1) or Y(2) receptor had fewer proliferating precursor cells and neuroblasts in the subventricular zone and rostral migratory stream and fewer neurons in the olfactory bulb expressing calbindin, calretinin or tyrosine hydroxylase. 3) In the anorectic mouse markers for microglia were strongly upregulated in the arcuate nucleus and in projection areas of the NPY/agouti gene-related protein arcuate system. CONCLUSION: NPY participates in several mechanisms involved in the development of the nervous system and is of importance in the control of food intake.


Subject(s)
Anorexia/physiopathology , Axons/physiology , Feeding Behavior , Neurogenesis/physiology , Neuropeptide Y/physiology , Animals , Anorexia/genetics , Disease Models, Animal , Eating , Mice
13.
Eur J Neurosci ; 28(2): 268-78, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18702698

ABSTRACT

During nervous system development axons reach their target areas under the influence of numerous guidance cues that affect rate and direction of growth. This report addresses the unsettled question of whether and to what extent growth velocity and turning responses (attraction, repulsion) are interdependent. We exposed individual growth cones of fetal rat dorsal root ganglion neurons in culture asymmetrically to gradients of seven different factors and recorded their growth rates and turning angles. Growth cones exhibited divergent patterns of turning and growth responses. For example, hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1) and thrombin all promoted growth, but HGF was a powerful attractant, thrombin a potent repellent and IGF-1 did not elicit turning. Galanin and neuropeptide Y also affected growth and/or turning differentially. Finally, nerve growth factor in the culture medium not only inhibited the turning responses to HGF, but also converted growth promotion of HGF and IGF-1 into inhibition. Overall, our studies indicate that: (i) turning and advance are regulated independently, except that strong attractive or repulsive responses generally are accompanied by growth promotion; (ii) asymmetric growth factor application per se does not elicit attraction; (iii) regulation of the two parameters may occur through a single receptor; and (iv) the effects of combined growth factors may not be additive and can be inhibitory.


Subject(s)
Ganglia, Spinal/embryology , Growth Cones/physiology , Intercellular Signaling Peptides and Proteins/physiology , Amides/pharmacology , Animals , Cells, Cultured , Drug Synergism , Embryonic Development/drug effects , Embryonic Development/physiology , Fetus , Galanin/pharmacology , Ganglia, Spinal/drug effects , Growth/drug effects , Growth Cones/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Neuropeptide Y/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Thrombin/pharmacology
14.
Mol Biol Cell ; 17(12): 5115-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987960

ABSTRACT

Repellents evoke growth cone turning by eliciting asymmetric, localized loss of actin cytoskeleton together with changes in substratum attachment. We have demonstrated that semaphorin-3A (Sema3A)-induced growth cone detachment and collapse require eicosanoid-mediated activation of protein kinase C epsilon (PKC epsilon) and that the major PKC epsilon target is the myristoylated, alanine-rich C-kinase substrate (MARCKS). Here, we show that PKC activation is necessary for growth cone turning and that MARCKS, while at the membrane, colocalizes with alpha3-integrin in a peripheral adhesive zone of the growth cone. Phosphorylation of MARCKS causes its translocation from the membrane to the cytosol. Silencing MARCKS expression dramatically reduces growth cone spread, whereas overexpression of wild-type MARCKS inhibits growth cone collapse triggered by PKC activation. Expression of phosphorylation-deficient, mutant MARCKS greatly expands growth cone adhesion, and this is characterized by extensive colocalization of MARCKS and alpha3-integrin, resistance to eicosanoid-triggered detachment and collapse, and reversal of Sema3A-induced repulsion into attraction. We conclude that MARCKS is involved in regulating growth cone adhesion as follows: its nonphosphorylated form stabilizes integrin-mediated adhesions, and its phosphorylation-triggered release from adhesions causes localized growth cone detachment critical for turning and collapse.


Subject(s)
Growth Cones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Actins/metabolism , Animals , Cell Adhesion/drug effects , Cytoskeleton/drug effects , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Gene Expression/drug effects , Gene Silencing , Growth Cones/drug effects , Humans , Integrin alpha3/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Semaphorin-3A/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
15.
Nat Neurosci ; 9(8): 993-5, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16845384

ABSTRACT

How a neuron becomes polarized remains largely unknown. Results obtained with a function-blocking antibody and an siRNA targeting the insulin-like growth factor-1 (IGF-1) receptor suggest that an essential step in the establishment of hippocampal neuronal polarity and the initiation of axonal outgrowth is the activation of the phosphatidylinositol 3-kinase (PI3k)-Cdc42 pathway by the IGF-1 receptor, but not by the TrkA or TrkB receptors.


Subject(s)
Cell Polarity , Hippocampus/cytology , Neurons/cytology , Receptor, IGF Type 1/metabolism , Animals , Cells, Cultured , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, trkA/metabolism , Receptor, trkB/metabolism , cdc42 GTP-Binding Protein/metabolism
16.
J Immunol ; 176(7): 4419-30, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547280

ABSTRACT

In this study, we describe the identification and in vitro functional activity of a novel multiple domain complement regulatory protein discovered based on its homology to short consensus repeat (SCR)-containing proteins of the regulators of complement activation (RCA) gene family. The rat cDNA encodes a predicted 388-kDa protein consisting of 14 N-terminal CUB domains that are separated from each other by a SCR followed by 15 tandem SCR domains, a transmembrane domain, and a short cytoplasmic tail. This protein is the homolog of the human protein of unknown function called the CUB and sushi multiple domains 1 (CSMD1) protein. A cloning strategy that incorporates the two C-terminal CUB-SCR domains and 12 of the tandem SCR repeats was used to produce a soluble rat CSMD1 protein. This protein blocked classical complement pathway activation in a comparable fashion with rat Crry but did not block alternative pathway activation. Analysis of CSMD1 mRNA expression by in situ hybridization and immunolabeling of neurons indicates that the primary sites of synthesis are the developing CNS and epithelial tissues. Of particular significance is the enrichment of CSMD1 in the nerve growth cone, the amoeboid-leading edge of the growing neuron. These results suggest that CSMD1 may be an important regulator of complement activation and inflammation in the developing CNS, and that it may also play a role in the context of growth cone function.


Subject(s)
Central Nervous System/metabolism , Epithelium/metabolism , Membrane Proteins/metabolism , Aging/physiology , Animals , Cell Line , Central Nervous System/cytology , Cloning, Molecular , Complement Pathway, Classical , Erythrocytes/drug effects , Female , Gene Expression Regulation , Growth Cones/metabolism , Hemolysis/drug effects , Humans , In Situ Hybridization , Male , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Membrane Proteins/pharmacology , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Sheep , Solubility
17.
J Cell Sci ; 119(Pt 1): 172-83, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16371655

ABSTRACT

The 'signal regulatory protein' SIRPalpha is an Ig superfamily, transmembrane glycoprotein with a pair of cytoplasmic domains that can bind the phosphatase SHP-2 when phosphorylated on tyrosine. SIRPalpha is prominent in growth cones of rat cortical neurons and located, together with the tetraspanin CD81, in the growth cone periphery. SIRPalpha is dynamically associated with Triton-X-100-sensitive, but Brij-98-resistant, lipid microdomains, which also contain CD81. Challenge of growth cones with the integrin-binding extracellular-matrix (ECM) protein, laminin, or with the growth factors, IGF-1 or BDNF, increases SIRPalpha phosphorylation and SHP-2 binding rapidly and transiently, via Src family kinase activation; phosphorylated SIRPalpha dissociates from the lipid microdomains. A cytoplasmic tail fragment of SIRPalpha (cSIRPalpha), when expressed in primary cortical neurons, also is phosphorylated and binds SHP-2. Expression of wild-type cSIRPalpha, but not of a phosphorylation-deficient mutant, substantially decreases IGF-1-stimulated axonal growth on laminin. On poly-D-lysine and in control conditions, axonal growth is slower than on laminin, but there is no further reduction in growth rate induced by the expression of cSIRPalpha. Thus, the effect of cSIRPalpha on axon growth is dependent upon integrin activation by laminin. These results suggest that SIRPalpha functions in the modulation of axonal growth by ECM molecules, such as laminin.


Subject(s)
Growth Cones/metabolism , Neurons , Receptors, Immunologic/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Female , Growth Cones/ultrastructure , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Laminin/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Neurons/cytology , Neurons/metabolism , Pregnancy , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
J Cell Sci ; 118(Pt 16): 3653-62, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16046480

ABSTRACT

Exocytotic incorporation of plasmalemmal precursor vesicles (PPVs) into the cell surface is necessary for axonal outgrowth and is known to occur mainly at the nerve growth cone. We have demonstrated recently that plasmalemmal expansion is regulated at the growth cone by IGF-1, but not by BDNF, in a manner that is quasi independent of the neuron's perikaryon. To begin elucidating the signaling pathway by which exocytosis of the plasmalemmal precursor is regulated, we studied activation of the IRS/PI3K/Akt pathway in isolated growth cones and hippocampal neurons in culture stimulated with IGF-1 or BDNF. Our results show that IGF-1, but not BDNF, significantly and rapidly stimulates IRS/PI3K/Akt and membrane expansion. Inhibition of PI3K with Wortmannin or LY294002 blocked IGF-1-stimulated plasmalemmal expansion at the growth cones of cultured neurons. Finally, our results show that, upon stimulation with IGF-1, most active PI3K becomes associated with distal microtubules in the proximal or central domain of the growth cone. Taken together, our results suggest a critical role for IGF-1 and the IRS/PI3K/Akt pathway in the process of membrane assembly at the axonal growth cone.


Subject(s)
Cell Membrane/metabolism , Central Nervous System/embryology , Exocytosis/physiology , Growth Cones/metabolism , Insulin-Like Growth Factor I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/drug effects , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/growth & development , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors , Exocytosis/drug effects , Growth Cones/drug effects , Growth Cones/ultrastructure , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/growth & development , Insulin-Like Growth Factor I/pharmacology , Membrane Fusion/drug effects , Membrane Fusion/physiology , Microtubules/drug effects , Microtubules/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Transport/drug effects , Protein Transport/physiology , Rats , Signal Transduction/physiology , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure
19.
J Neurochem ; 86(1): 55-60, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12807424

ABSTRACT

Growth cone gp93 is a highly heterogeneous membrane glycoprotein with an Mr of about 93 kDa. It was purified from adult rat brain and microsequenced. The sequences of four different peptide fragments of gp93 matched those of the 'signal regulatory protein' SIRPalpha (also known as SHPS-1, BIT or P84), an Ig superfamily member. SIRPalpha contains a cytoplasmic tail that is a tyrosine kinase substrate and binds the protein tyrosine phosphatase SHP-2. SIRPalpha and gp93 also were immunochemically cross-reactive. A PCR strategy was used to determine whether gp93/SIRPalpha heterogeneity in the brain depended upon the presence of different transcripts and, thus, sequence heterogeneity. However, we observed only a single full-length transcript. A short splice variant also was detected. These data identify gp93 as the Ig superfamily member SIRPalpha. Together with our previous results, the data also demonstrate that, in rat brain, gp93/SIRPalpha heterogeneity is the result of differential glycosylation (plus phosphorylation), rather than sequence heterogeneity.


Subject(s)
Antigens, Differentiation , Brain Chemistry , Growth Cones/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/metabolism , Receptors, Immunologic , Signal Transduction/physiology , Amino Acid Sequence , Animals , Glycosylation , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neural Cell Adhesion Molecule L1/genetics , Phosphorylation , Polymerase Chain Reaction , Precipitin Tests , Rats , Sequence Analysis, Protein
20.
J Biol Chem ; 278(23): 21168-77, 2003 Jun 06.
Article in English | MEDLINE | ID: mdl-12665507

ABSTRACT

Exposure of growing neurons to thrombin or semaphorin 3A stimulates a receptor-mediated signaling cascade that results in collapse of their growth cones. This collapse response necessitates eicosanoid production, as we have shown earlier. The present report investigates whether and which protein kinase C (PKC) isoforms may be activated by such eicosanoids. To examine these questions, we isolated growth cones from fetal rat brain and tested whether thrombin or the eicosanoid, 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE), could activate endogenous growth cone PKC. We show that both thrombin and 12(S)-HETE stimulate the phosphorylation of the myristoylated alanine-rich protein kinase C substrate, an 87-kDa adhesion site protein. Furthermore, we show both with immunoprecipitated and with recombinant PKC that 12(S)-HETE activation is selective for the epsilon isoform and does not require accessory proteins. Last, we demonstrate that PKC activation is necessary for thrombin-induced growth cone collapse. These data indicate that eicosanoid-mediated repellent effects result from the direct and selective activation of PKCepsilon and suggest the involvement of myristoylated alanine-rich protein kinase C substrate phosphorylation in growth cone collapse.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Growth Cones/drug effects , Growth Cones/enzymology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Protein Kinase C/metabolism , Animals , Cell Adhesion/physiology , Enzyme Activation/drug effects , Hemostatics/pharmacology , Myristoylated Alanine-Rich C Kinase Substrate , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Precipitin Tests , Protein Kinase C/genetics , Protein Kinase C/isolation & purification , Protein Kinase C-epsilon , Proteins/metabolism , Rats , Recombinant Proteins/metabolism , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...