Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neurochem ; 121(6): 915-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22469063

ABSTRACT

Apart from its hematopoietic activity, erythropoietin (EPO) is also known as a tissue-protective cytokine. In the brain, EPO and its receptor are up-regulated in response to insult and exert pro-survival effects. EPO binds to its receptor (EPOR) via high- and low-affinity binding sites (Sites 1 and 2, respectively), inducing conformational changes in the receptor, followed by the activation of downstream signaling cascades. Based on the crystal structure of the EPO:EPOR(2) complex, we designed a peptide, termed Epobis, whose sequence encompassed amino acids from binding Site 1. The present study shows that the Epobis peptide specifically binds to EPOR and induces neurite outgrowth from primary neurons in an EPOR-expression dependent manner. Furthermore, Epobis promoted the survival of hippocampal and cerebellar neuronal cultures after kainate treatment and KCl deprivation, respectively. Thus, we identified a new functional agonist of EPOR with the potential to promote neuroregeneration and neuroprotection.


Subject(s)
Neurites/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Erythropoietin/chemistry , Erythropoietin/metabolism , Gene Knockdown Techniques , Humans , Models, Molecular , Neuroprotective Agents/metabolism , Peptides/metabolism , Protein Binding , Protein Structure, Quaternary , Rats , Rats, Wistar , Signal Transduction/physiology , Surface Plasmon Resonance , Transfection
2.
PLoS One ; 6(8): e23433, 2011.
Article in English | MEDLINE | ID: mdl-21887252

ABSTRACT

The key roles played by the neural cell adhesion molecule (NCAM) in plasticity and cognition underscore this membrane protein as a relevant target to develop cognitive-enhancing drugs. However, NCAM is a structurally and functionally complex molecule with multiple domains engaged in a variety of actions, which raise the question as to which NCAM fragment should be targeted. Synthetic NCAM mimetic peptides that mimic NCAM sequences relevant to specific interactions allow identification of the most promising targets within NCAM. Recently, a decapeptide ligand of NCAM--plannexin, which mimics a homophilic trans-binding site in Ig2 and binds to Ig3--was developed as a tool for studying NCAM's trans-interactions. In this study, we investigated plannexin's ability to affect neural plasticity and memory formation. We found that plannexin facilitates neurite outgrowth in primary hippocampal neuronal cultures and improves spatial learning in rats, both under basal conditions and under conditions involving a deficit in a key plasticity-promoting posttranslational modification of NCAM, its polysialylation. We also found that plannexin enhances excitatory synaptic transmission in hippocampal area CA1, where it also increases the number of mushroom spines and the synaptic expression of the AMPAR subunits GluA1 and GluA2. Altogether, these findings provide compelling evidence that plannexin is an important facilitator of synaptic functional, structural and molecular plasticity in the hippocampal CA1 region, highlighting the fragment in NCAM's Ig3 module where plannexin binds as a novel target for the development of cognition-enhancing drugs.


Subject(s)
Hippocampus/drug effects , Hippocampus/physiology , Learning/drug effects , Neural Cell Adhesion Molecules/metabolism , Neuronal Plasticity/drug effects , Oligopeptides/pharmacology , Animals , Binding Sites , Biomarkers/metabolism , Cell Survival/drug effects , Cells, Cultured , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Glycoside Hydrolases/metabolism , Hippocampus/metabolism , Hippocampus/ultrastructure , Male , Maze Learning/drug effects , Mice , Models, Molecular , Neurites/drug effects , Neurites/metabolism , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Glutamate/metabolism , Sialic Acids/metabolism , Synaptic Transmission/drug effects
3.
Eur J Cell Biol ; 89(11): 817-27, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20692716

ABSTRACT

Neural cell adhesion molecule (NCAM)-mediated cell adhesion results in activation of intracellular signaling cascades that lead to cellular responses such as neurite outgrowth, neuronal survival, and modulation of synaptic activity associated with cognitive processes. The crystal structure of the immunoglobulin (Ig) 1-2-3 fragment of the NCAM ectodomain has revealed novel mechanisms for NCAM homophilic adhesion. The present study addressed the biological significance of the so called dense zipper formation of NCAM. Two peptides, termed dennexinA and dennexinB, were modeled after the contact interfaces between Ig1 and Ig3 and between Ig2 and Ig2, respectively, observed in the crystal structure. Although the two dennexin peptides differed in amino acid sequence, they both modulated cell adhesion, reflected by inhibition of NCAM-mediated neurite outgrowth. Both dennexins also promoted neuronal survival, and the effect of dennexinA was independent of polysialic acid expression. Consistent with the effect of dennexinA on NCAM-mediated adhesion in vitro, the peptide impaired long-term memory retention in rats in the Morris water maze test. Thus, dennexins are novel site-specific pharmacological tools for elucidation of the role of NCAM in a variety of biological processes under normal and pathological conditions.


Subject(s)
Maze Learning/drug effects , Neural Cell Adhesion Molecules/physiology , Neurons/physiology , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line , Male , Mice , Models, Molecular , Molecular Sequence Data , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction
4.
Brain ; 133(Pt 8): 2281-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20435631

ABSTRACT

Erythropoietin, a member of the type 1 cytokine superfamily, controls proliferation and differentiation of erythroid progenitor cells through binding to and dimerization of the erythropoietin receptor. Both erythropoietin and its receptor are also expressed in the central nervous system, where they are involved in tissue protection. However, the use of erythropoietin as a neuroprotective agent may be hampered by its erythropoietic activity. Therefore, developing non-haematopoietic erythropoietin mimetics is important. Based on the crystal structure of the complex of erythropoietin and its receptor, we designed a peptide, termed Epotris, corresponding to the C α-helix region (amino-acid residues 92-111) of human erythropoietin. The peptide specifically bound to the erythropoietin receptor and promoted neurite outgrowth and survival of primary neurons with the same efficiency as erythropoietin, but with 10(3)-fold lower potency. Knockdown of the erythropoietin receptor or interference with its downstream signalling inhibited the Epotris-induced neuritogenic and pro-survival effect. Similarly to erythropoietin, Epotris penetrated the blood-brain barrier. Moreover, treatment with the peptide attenuated seizures, decreased mortality and reduced neurodegeneration in an in vivo model of kainic acid-induced neurotoxicity. In contrast to erythropoietin, Epotris did not stimulate erythropoiesis upon chronic administration. Thus, Epotris is a novel neuroprotective non-haematopoietic erythropoietin mimetic that may offer new opportunities for the treatment of neurological disorders.


Subject(s)
Erythropoietin/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Erythropoietin/agonists , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Erythropoiesis/drug effects , Erythropoiesis/physiology , Erythropoietin/chemistry , Erythropoietin/metabolism , Erythropoietin/pharmacokinetics , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurites/drug effects , Neurites/physiology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/mortality , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Rats , Rats, Wistar , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Seizures/drug therapy , Seizures/mortality
5.
J Neurosci Res ; 88(10): 2165-76, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20209630

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a key role in neural development, regeneration, and synaptic plasticity. The crystal structure of a fragment of NCAM comprising the three N-terminal immunoglobulin (Ig)-like modules indicates that the first and second Ig modules bind to each other, thereby presumably mediating dimerization of NCAM molecules expressed on the same cell surface (cis-interactions), whereas the third Ig module, through interactions with the first or second Ig module, mediates interactions between NCAM molecules expressed on the surface of opposing cells (trans-interactions). We have designed a new potent peptide ligand of NCAM, termed plannexin, based on a discontinuous sequence in the second NCAM Ig module that represents a homophilic binding site for an opposing third Ig module. The peptide was found by surface plasmon resonance analysis to bind the third NCAM Ig module. It promoted survival of cultured cerebellar granule neurons (CGNs) and also induced neurite extension in cultures of dopaminergic neurons and CGNs; the latter effect was shown to be dependent on NCAM expression, indicating that plannexin mimics the neuritogenic effect of homophilic NCAM binding.


Subject(s)
Neurites/metabolism , Neurons/cytology , Neurons/metabolism , Oligopeptides/metabolism , Animals , Cell Enlargement , Cell Line, Tumor , Cell Survival , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Dopamine/metabolism , Ligands , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Neural Cell Adhesion Molecules/metabolism , Oligopeptides/chemistry , Peptides/metabolism , Protein Binding , Rats , Rats, Wistar
6.
Dev Neurobiol ; 69(13): 837-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19634127

ABSTRACT

Fibroblast growth factor receptor (FGFR) signaling is pivotal in the regulation of neurogenesis, neuronal differentiation and survival, and synaptic plasticity both during development and in adulthood. In order to develop low molecular weight agonists of FGFR, seven peptides, termed hexafins, corresponding to the beta6-beta7 loop region of the FGF 1, 2, 3, 8, 9, 10, and 17, were synthesized. This region shares a homologous amino acid sequence with the FG-loop region of the second fibronectin Type III module of the neural cell adhesion molecule (NCAM) that binds to the FGFR. Hexafins were shown by surface plasmon resonance to bind to FGFR1-IIIc-Ig2-3 and FGFR2-IIIb-Ig2-3. The heparin analog sucrose octasulfate inhibited hexafin binding to FGFR1-IIIc-Ig2-3 indicating overlapping binding sites. Hexafin-binding to FGFR1-IIIc resulted in receptor phosphorylation, but inhibited FGF1-induced FGFR1 phosphorylation, indicating that hexafins act as partial agonists. Hexafin2, 3, 8, 10, and 17 (but not 1 or 9) induced neurite outgrowth from cerebellar granule neurons (CGNs), an effect that was abolished by two inhibitors of FGFR, SU5402 and inositol hexaphosphate (IP6) and a diacylglycerol lipase inhibitor, RHC-80267. The neuritogenic effects of selected hexafins could also be inhibited by FGF1 which by itself did not induce neurite outgrowth. Moreover, hexafin1, 3, 9, 10, and 17 (but not 2 or 8) promoted survival of CGNs induced to undergo apoptosis. Thus, selected hexafins induced neuronal differentiation and survival, making them promising pharmacological tools for the study of functional FGFR regulation in development of the nervous system.


Subject(s)
Cell Survival/drug effects , Fibroblast Growth Factors/metabolism , Neurons/cytology , Receptors, Fibroblast Growth Factor/metabolism , Analysis of Variance , Animals , Binding Sites , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Neural Cell Adhesion Molecules/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phytic Acid/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/agonists , Video Recording
7.
J Neurochem ; 106(5): 2030-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18624916

ABSTRACT

The neural cell adhesion molecule (NCAM) directly interacts with the fibroblast growth factor receptor (FGFR). Both fibronectin type III (FN3) modules of NCAM are involved in this interaction. One of the NCAM-FGFR contact sites has been localized recently to the upper N-terminal part of the second NCAM FN3 module encompassing the F and G beta-strands and the interconnecting loop region. Here, we investigated whether any of the six putative strand-loop-strand regions in the first NCAM FN3 module are involved in FGFR interactions. Peptide sequences encompassing these regions, termed encamins, were synthesized and tested for their ability to bind and activate FGFR. Encamins localized to the N-terminal part of the first FN3 module did not interact with FGFR, whereas encamins localized to the C-terminal part, termed EncaminA, C and E, bound to and activated FGFR. The encamins induced FGFR-dependent neurite outgrowth, and EncaminC and E promoted neuronal survival and enhanced pre-synaptic function. In conclusion, the interaction between NCAM and FGFR probably involves multiple contact sites at an interface formed by the two NCAM FN3 modules and FGFR, and encamins could constitute important pharmacological tools for the study of specific functional aspects of NCAM, including neuroprotection and modulation of plasticity.


Subject(s)
Fibroblast Growth Factors/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , Peptides/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/physiology , Humans , Mice , Neural Cell Adhesion Molecules/chemistry , Neurites/drug effects , Neurites/ultrastructure , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Peptides/chemistry , Protein Structure, Tertiary/physiology , Rats
8.
J Neurochem ; 104(3): 667-82, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199118

ABSTRACT

A series of peptides, termed dekafins, were derived from the beta10-beta11 loop regions of fibroblast growth factors (FGFs) 1, 2, 3, 5, 6, 8, 9, 10, and 17. The dekafins share a homologous amino acid sequence similar to a sequence in the first fibronectin type III module of the neural cell adhesion molecule. All dekafins were shown by surface plasmon resonance analysis to bind fibroblast growth factor receptor (FGFR)1-IIIc-Ig2-3 and FGFR2-IIIb-Ig2-3, respectively, with K(d) values of approximately 10(-7) to 10(-8) mol/L. Binding of dekafin1 to FGFR1-IIIc-Ig2-3 was inhibited by a heparin analog, sucrose octasulfate, indicating that heparin sulfate moiety can modulate dekafin binding to FGFRs. Treatment of transcription and mRNA export (TREX) cells permanently expressing Strep-tag-labeled FGFR1-IIIc with dekafins resulted in receptor phosphorylation. FGF1-induced FGFR1-IIIc phosphorylation was inhibited by dekafin1 and 10 in high concentrations, indicating that dekafins are FGFR partial agonists. The dekafins induced neuronal differentiation as reflected by neurite outgrowth from cerebellar granule neurons, an effect that was abolished by SU5402, a specific inhibitor of the FGFR tyrosine kinase, and by inositolhexaphosphate, an extracellularly acting FGFR antagonist. Some, but not all, dekafins were capable of promoting survival of cerebellar granule neurons induced to undergo apoptosis. Thus, the dekafins are functional FGFR agonists with apparent therapeutic potential.


Subject(s)
Fibroblast Growth Factors/chemistry , Peptide Fragments/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/drug effects , Amino Acid Motifs/physiology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Binding Sites/drug effects , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , Fibroblast Growth Factors/antagonists & inhibitors , Humans , Molecular Sequence Data , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Protein Binding/drug effects , Pyrroles/pharmacology , Rats , Rats, Wistar , Transfection/methods
9.
J Neurosci Res ; 77(6): 777-86, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15334597

ABSTRACT

Mts1 (S100A4) is a calcium-binding protein of the EF-hand type, belonging to the S100 family of proteins. The mts1/S100A4 gene was originally isolated from tumor cell lines, and the protein is believed to play an important role in tumor progression. More recently, oligomeric, but not dimeric, forms of Mts1 have been shown to have a neuritogenic effect when added extracellularly to hippocampal neurons. Here we show increased neurite outgrowth in two other cell types, dopaminergic and cerebellar neurons, in response to treatment with Mts1 oligomers. Moreover, we demonstrate that Mts1 acts as a neuroprotectant in primary cerebellar, dopaminergic, and hippocampal neurons induced to undergo cell death. Interestingly, the survival of the cerebellar and hippocampal neurons increased as a result of treatment with Mts1 not only in oligomeric form but also--although to a lesser extent--in dimeric form. The inhibition of death in cerebellar neurons by Mts1 was accompanied by an inhibition of DNA fragmentation, but Mts1 did not affect the activity of caspases-3 and -6. In hippocampal neurons, cell death induced by the amyloid-beta peptide (Abeta(25-35)) was characterized by an increase in caspase-3 and -6 activity, but no DNA fragmentation was observed. As in cerebellar neurons, the induced increase in caspase activity in hippocampal neurons was not affected by Mts1.


Subject(s)
Hippocampus/drug effects , Neuroprotective Agents/pharmacology , S100 Proteins/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/metabolism , Humans , Rats , Rats, Wistar , S100 Calcium-Binding Protein A4
10.
J Neurosci Res ; 75(1): 55-65, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689448

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a pivotal role in neural development, regeneration, and plasticity. NCAM mediates adhesion and subsequent signal transduction through NCAM-NCAM binding. Recently, a peptide ligand termed P2 corresponding to a 12-amino-acid sequence in the FG loop of the second Ig domain of NCAM was shown to mimic NCAM homophilic binding as reflected by induction of neurite outgrowth in hippocampal neurons. We demonstrate here that in concentrations between 0.1 and 10 microM, P2 also induced neuritogenesis in primary dopaminergic and cerebellar neurons. Furthermore, it enhanced the survival rate of cerebellar neurons although not of mesencephalic dopaminergic neurons. Moreover, our data indicate that the protective effect of P2 in cerebellar neurons was due to an inhibition of the apoptotic process, in that caspase-3 activity and the level of DNA fragmentation were lowered by P2. Finally, treatment of neurons with P2 resulted in phosphorylation of the ser/thr kinase Akt. Thus, a small peptide mimicking homophilic NCAM interaction is capable of inducing differentiation as reflected by neurite outgrowth in several neuronal cell types and inhibiting apoptosis in cerebellar granule neurons.


Subject(s)
Myelin Proteins/pharmacology , Neural Cell Adhesion Molecules/metabolism , Neurites/drug effects , Neurons/drug effects , Protein Serine-Threonine Kinases , Tyrosine 3-Monooxygenase/metabolism , Animals , Animals, Newborn , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , GAP-43 Protein/metabolism , Glial Cell Line-Derived Neurotrophic Factor , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/pharmacology , Male , Mesencephalon/cytology , Mesencephalon/embryology , Mesencephalon/growth & development , Nerve Growth Factors/pharmacology , Neurons/metabolism , Oxidopamine/pharmacology , Pregnancy , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Sympatholytics/pharmacology
11.
Brain Res ; 992(1): 128-36, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14604781

ABSTRACT

Metallothionein I and II (MT-I+II) are antioxidant and tissue protective factors. We have previously shown that MT-I+II prevent oxidative stress and apoptotic cell death and are of therapeutic value in brain inflammation. However, MT-I+II are expressed in glia and it remains to be elucidated if MT-I+II can affect neurons directly. It is likely that MT isoforms could be beneficial also during neurodegenerative disorders. In this study, we have examined if MT-II affects survival and neurite extension of dopaminergic and hippocampal neurons. We show for the first time that MT-II treatment can significantly stimulate neurite extension from both dopaminergic and hippocampal neurons. Moreover, MT-II treatment significantly increases survival of dopaminergic neurons exposed to 6-hydroxydopamine (6-OHDA) and protects significantly hippocampal neurons from amyloid beta-peptide-induced neurotoxicity. Accordingly, treatment with MT-II may be of therapeutic value in neurodegenerative disorders.


Subject(s)
Hippocampus/physiology , Metallothionein/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Adrenergic Agents/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dopamine/metabolism , Embryo, Mammalian , Hippocampus/drug effects , Neurons/cytology , Neurons/physiology , Oxidopamine/pharmacology , Rats , Rats, Wistar
12.
J Neurochem ; 84(3): 546-56, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558974

ABSTRACT

The neural cell adhesion molecule, NCAM, is known to stimulate neurite outgrowth from primary neurones and PC12 cells presumably through signalling pathways involving the fibroblast growth factor receptor (FGFR), protein kinase A (PKA), protein kinase C (PKC), the Ras-mitogen activated protein kinase (MAPK) pathway and an increase in intracellular Ca2+ levels. Stimulation of neurones with the synthetic NCAM-ligand, C3, induces neurite outgrowth through signalling pathways similar to the pathways activated through physiological, homophilic NCAM-stimulation. We present here data indicating that phosphatidylinositol 3-kinase (PI3K) is required for NCAM-mediated neurite outgrowth from PC12-E2 cells and from cerebellar and dopaminergic neurones in primary culture, and that the thr/ser kinase Akt/protein kinase B (PKB) is phosphorylated downstream of PI3K after stimulation with C3. Moreover, we present data indicating a survival-promoting effect of NCAM-stimulation by C3 on cerebellar and dopaminergic neurones induced to undergo apoptosis. This protective effect of C3 included an inhibition of both DNA-fragmentation and caspase-3 activation. The survival-promoting effect of NCAM-stimulation was also shown to be dependent on PI3K.


Subject(s)
Cell Differentiation/physiology , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Adhesion Molecules, Neuronal/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Humans , In Situ Nick-End Labeling , Ligands , Mice , Molecular Sequence Data , Neural Cell Adhesion Molecules/pharmacology , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Peptides/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...