Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Brain ; 14(1): 66, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33832520

RESUMEN

GAP-43 is a vertebrate neuron-specific protein and that is strongly related to axon growth and regeneration; thus, this protein has been utilized as a classical molecular marker of these events and growth cones. Although GAP-43 was biochemically characterized more than a quarter century ago, how this protein is related to these events is still not clear. Recently, we identified many phosphorylation sites in the growth cone membrane proteins of rodent brains. Two phosphorylation sites of GAP-43, S96 and T172, were found within the top 10 hit sites among all proteins. S96 has already been characterized (Kawasaki et al., 2018), and here, phosphorylation of T172 was characterized. In vitro (cultured neurons) and in vivo, an antibody specific to phosphorylated T172 (pT172 antibody) specifically recognized cultured growth cones and growing axons in developing mouse neurons, respectively. Immunoblotting showed that pT172 antigens were more rapidly downregulated throughout development than those of pS96 antibody. From the primary structure, this phosphorylation site was predicted to be conserved in a wide range of animals including primates. In the developing marmoset brainstem and in differentiated neurons derived from human induced pluripotent stem cells, immunoreactivity with pT172 antibody revealed patterns similar to those in mice. pT172 antibody also labeled regenerating axons following sciatic nerve injury. Taken together, the T172 residue is widely conserved in a wide range of mammals including primates, and pT172 is a new candidate molecular marker for growing axons.


Asunto(s)
Axones/metabolismo , Biomarcadores/metabolismo , Proteína GAP-43/metabolismo , Mamíferos/metabolismo , Fosfotreonina/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Encéfalo/embriología , Callithrix , Células Cultivadas , Hurones , Proteína GAP-43/química , Conos de Crecimiento/metabolismo , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos C57BL , Regeneración Nerviosa , Fosforilación , Primates , Nervio Ciático/lesiones
2.
Biosci Rep ; 40(1)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31854448

RESUMEN

Protein S-acylation is a reversible post-translational modification involving the addition of fatty acids to cysteines and is catalyzed by transmembrane protein acyltransferases (PATs) mainly expressed at the Golgi complex. In case of soluble proteins, S-acylation confers stable membrane attachment. Myristoylation or farnesylation of many soluble proteins constitutes the initial transient membrane adsorption step prior to S-acylation. However, some S-acylated soluble proteins, such as the neuronal growth-associated protein Growth-associated protein-43 (GAP-43), lack the hydrophobic modifications required for this initial membrane interaction. The signals for GAP-43 S-acylation are confined to the first 13 amino acids, including the S-acylatable cysteines 3 and 4 embedded in a hydrophobic region, followed by a cluster of basic amino acids. We found that mutation of critical basic amino acids drastically reduced membrane interaction and hence S-acylation of GAP-43. Interestingly, acute depletion of phosphatidylinositol 4-phosphate (PtdIns4P) at the Golgi complex reduced GAP-43 membrane binding, highlighting a new, pivotal role for this anionic lipid and supporting the idea that basic amino acid residues are involved in the electrostatic interactions between GAP-43 and membranes of the Golgi complex where they are S-acylated.


Asunto(s)
Homólogo 4 de la Proteína Discs Large/metabolismo , Proteína GAP-43/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Procesamiento Proteico-Postraduccional , Red trans-Golgi/metabolismo , Acilación , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células CHO , Secuencia Conservada , Cricetulus , Homólogo 4 de la Proteína Discs Large/química , Homólogo 4 de la Proteína Discs Large/genética , Proteína GAP-43/química , Proteína GAP-43/genética , Interacciones Hidrofóbicas e Hidrofílicas , Electricidad Estática , Factores de Tiempo , Red trans-Golgi/genética
3.
J Am Chem Soc ; 140(50): 17374-17378, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30516377

RESUMEN

Post-translational S-palmitoylation plays a central role in protein localization, trafficking, stability, aggregation, and cell signaling. Dysregulation of palmitoylation pathways in cells can alter protein function and is the cause of several diseases. Considering the biological and clinical importance of S-palmitoylation, tools for direct, in vivo modulation of this lipid modification would be extremely valuable. Here, we describe a method for the cleavage of native S-palmitoyl groups from proteins in living cells. Using a cell permeable, cysteine-functionalized amphiphile, we demonstrate the direct depalmitoylation of cellular proteins. We show that amphiphile-mediated depalmitoylation (AMD) can effectively cleave S-palmitoyl groups from the native GTPase HRas and successfully depalmitoylate mislocalized proteins in an infantile neuronal ceroid lipofuscinosis (INCL) disease model. AMD enables direct and facile depalmitoylation of proteins in live cells and has potential therapeutic applications for diseases such as INCL, where native protein thioesterase activity is deficient.


Asunto(s)
Lipoilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Cisteína/análogos & derivados , Cisteína/farmacología , Proteína GAP-43/química , Proteína GAP-43/metabolismo , Humanos , Lipopéptidos/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/química
4.
Brain Struct Funct ; 223(6): 2767-2783, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29633039

RESUMEN

The primary visual cortex (area V1) is an extensively studied part of the cerebral cortex with well-characterized connectivity, cellular and molecular architecture and functions (for recent reviews see Amunts and Zilles, Neuron 88:1086-1107, 2015; Casagrande and Xu, Parallel visual pathways: a comparative perspective. The visual neurosciences, MIT Press, Cambridge, pp 494-506, 2004). In humans, V1 is defined by heavily myelinated fibers arriving from the radiatio optica that form the Gennari stripe in cortical layer IV, which is further subdivided into laminae IVa, IVb, IVcα and IVcß. Due to this unique laminar pattern, V1 represents an excellent region to test whether multimodal mass spectrometric imaging could reveal novel biomolecular markers for a functionally relevant parcellation of the human cerebral cortex. Here we analyzed histological sections of three post-mortem brains with matrix-assisted laser desorption/ionization mass spectrometry imaging and laser ablation inductively coupled plasma mass spectrometry imaging to investigate the distribution of lipids, proteins and metals in human V1. We identified 71 peptides of 13 different proteins by in situ tandem mass spectrometry, of which 5 proteins show a differential laminar distribution pattern revealing the border between V1 and V2. High-accuracy mass measurements identified 123 lipid species, including glycerolipids, glycerophospholipids and sphingolipids, of which at least 20 showed differential distribution within V1 and V2. Specific lipids labeled not only myelinated layer IVb, but also IVa and especially IVc in a layer-specific manner, but also and clearly separated V1 from V2. Elemental imaging further showed a specific accumulation of copper in layer IV. In conclusion, multimodal mass spectrometry imaging identified novel biomolecular and elemental markers with specific laminar and inter-areal differences. We conclude that mass spectrometry imaging provides a promising new approach toward multimodal, molecule-based cortical parcellation.


Asunto(s)
Mapeo Encefálico , Procesamiento de Imagen Asistido por Computador , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Corteza Visual/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Femenino , Proteína GAP-43/química , Proteína GAP-43/metabolismo , Hemoglobinas/metabolismo , Humanos , Metabolismo de los Lípidos , Masculino , Metales/metabolismo , Persona de Mediana Edad , Proteína Básica de Mielina/química , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Péptidos/metabolismo , Cambios Post Mortem , Corteza Visual/anatomía & histología , Vías Visuales/diagnóstico por imagen , Vías Visuales/metabolismo
5.
FEBS J ; 283(8): 1550-69, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26918762

RESUMEN

Brain acid-soluble protein-1 (BASP1) and growth-associated protein-43 (GAP-43) are presynaptic membrane proteins participating in axon guidance, neuroregeneration and synaptic plasticity. They are presumed to sequester phosphatidylinositol-4,5-bisphosphate (PIP2 ) in lipid rafts. Previously we have shown that the proteins form heterogeneously sized oligomers in the presence of anionic phospholipids or SDS at submicellar concentration. BASP1 and GAP-43 are intrinsically disordered proteins (IDPs). In light of this, we investigated the structure of their oligomers. Using partial cross-linking of the oligomers with glutaraldehyde, the aggregation numbers of BASP1 and GAP-43 were estimated as 10-14 and 6-7 monomer subunits, respectively. The cross-linking pattern indicated that the subunits are circularly arranged. The circular dichroism (CD) spectra of the monomers were characteristic of coil-like IDPs showing unordered structure with a high population of polyproline-II conformation. The oligomerization was accompanied by a minor CD spectral change attributable to formation of a small amount of α-helix. The number of residues in the α-helical conformation was estimated as 13 in BASP1 and 18 in GAP-43. However, the overall structure of the oligomers remained disordered, indicating a high degree of 'fuzziness'. This was confirmed by measuring the hydrodynamic dimensions of the oligomers using polyacrylamide gradient gel electrophoresis and size-exclusion chromatography, and by assaying their sensitivity to proteolytic digestion. There is evidence that the observed α-helical folding occurs within the basic effector domains, which are presumably tethered together via anionic molecules of SDS or PIP2 . We conclude that BASP1 and GAP-43 oligomers preserve a mostly disordered structure, which may be of great importance for their function in PIP2 signaling pathway.


Asunto(s)
Encéfalo/metabolismo , Proteína GAP-43/química , Proteínas Intrínsecamente Desordenadas/química , Proteínas del Tejido Nervioso/química , Secuencia de Aminoácidos , Animales , Bovinos , Dicroismo Circular , Reactivos de Enlaces Cruzados/farmacología , Electroforesis en Gel de Poliacrilamida , Proteína GAP-43/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Conformación Proteica , Multimerización de Proteína
6.
Biomol NMR Assign ; 10(1): 171-4, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26748655

RESUMEN

GAP-43 is a 25 kDa neuronal intrinsically disordered protein, highly abundant in the neuronal growth cone during development and regeneration. The exact molecular function(s) of GAP-43 remains unclear but it appears to be involved in growth cone guidance and actin cytoskeleton organization. Therefore, GAP-43 seems to play an important role in neurotransmitter vesicle fusion and recycling, long-term potentiation, spatial memory formation and learning. Here we report the nearly complete assignment of recombinant human GAP-43.


Asunto(s)
Proteína GAP-43/química , Membrana Celular/metabolismo , Proteína GAP-43/metabolismo , Humanos , Ligandos , Resonancia Magnética Nuclear Biomolecular
7.
Mol Cell Biol ; 35(10): 1712-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25755278

RESUMEN

Growth-associated protein 43 (GAP43) is known to regulate axon growth, but whether it also plays a role in synaptogenesis remains unclear. Here, we found that GAP43 regulates the aggregation of gephyrin, a pivotal protein for clustering postsynaptic GABA(A) receptors (GABA(A)Rs), in developing cortical neurons. Pharmacological blockade of either protein kinase C (PKC) or neuronal activity increased both GAP43-gephyrin association and gephyrin misfolding-induced aggregation, suggesting the importance of PKC-dependent regulation of GABAergic synapses. Furthermore, we found that PKC phosphorylation-resistant GAP43(S41A), but not PKC phosphorylation-mimicking GAP43(S41D), interacted with cytosolic gephyrin to trigger gephyrin misfolding and its sequestration into aggresomes. In contrast, GAP43(S41D), but not GAP43(S41A), inhibited the physiological aggregation/clustering of gephyrin, reduced surface GABA(A)Rs under physiological conditions, and attenuated gephyrin misfolding under transient oxygen-glucose deprivation (tOGD) that mimics pathological neonatal hypoxia. Calcineurin-mediated GAP43 dephosphorylation that accompanied tOGD also led to GAP43-gephyrin association and gephyrin misfolding. Thus, PKC-dependent phosphorylation of GAP43 plays a critical role in regulating postsynaptic gephyrin aggregation in developing GABAergic synapses.


Asunto(s)
Proteínas Portadoras/metabolismo , Neuronas GABAérgicas/metabolismo , Proteína GAP-43/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa C/metabolismo , Animales , Proteínas Portadoras/química , Células Cultivadas , Femenino , Flavonoides/farmacología , Neuronas GABAérgicas/citología , Proteína GAP-43/química , Células HEK293 , Humanos , Indoles/farmacología , Proteínas de la Membrana/química , Fosforilación , Embarazo , Pliegue de Proteína/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 9(2): e88554, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24533107

RESUMEN

In a previous study, we generated two monoclonal antibodies (mAbs) in mice, aNogoA-N and aNogo-66 mAb, which were raised against recombinant N-terminal fragments of rat NogoA and Nogo-66, respectively. When compared with the commercial rabbit anti-rat NogoA polyclonal antibody (pAb), which can specifically recognise NogoA, the two mAbs were also specific for the NogoA antigen in immunofluorescence histochemical (IHC) staining and Western blot (WB) analysis. Serial truncations of NogoA covering the N-terminal region of NogoA (aa 570-691) and Nogo-66 (aa 1026-1091) were expressed in E. coli. The epitopes recognised by aNogoA-N and aNogo-66 are located in the aa 634-668 and aa 1026-1055 regions of NogoA, respectively. Both mAbs remarkably enhanced the axon growth and branching of cultured hippocampal neurons in vitro. These results suggest that the antibodies that bind to aa 634-668 and aa 1026-1055 of NogoA may have stimulatory effects on axon growth and branching. Additionally, the two mAbs that we generated are specific for NogoA and significantly block NogoA function. In conclusion, two sites in NogoA located within aa 634-668 and aa 1026-1055 are recognised by our two antibodies and are novel and potentially promising targets for repair after central nervous system (CNS) injury.


Asunto(s)
Anticuerpos Monoclonales/química , Axones/fisiología , Proteínas de la Mielina/química , Neuronas/citología , Animales , Sistema Nervioso Central/lesiones , Mapeo Epitopo , Epítopos/química , Proteína GAP-43/química , Hipocampo/citología , Inmunohistoquímica , Masculino , Microscopía Fluorescente , Vaina de Mielina/química , Proteínas Nogo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química
9.
Stroke ; 44(7): 1951-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23696550

RESUMEN

BACKGROUND AND PURPOSE: We sought to demonstrate the contribution of axonal remodeling of the corticospinal tract (CST) in the spinal cord to functional outcome after stroke. METHODS: Bilateral pyramidotomy (BPT) or sham-BPT was performed in mice with transgenic yellow fluorescent protein labeling in the CST subjected to middle cerebral artery occlusion (MCAo). Foot-fault and single pellet reaching tests were performed 3 days after MCAo and weekly thereafter. Mice were euthanized at day 14 or 28 after stroke. Immunofluorescent staining for growth-associated protein-43 and Synaptophysin was performed on cervical sections. RESULTS: Functional improvements were evident during the initial 14 days in both MCAo-sham-BPT and MCAo-BPT mice (P<0.01, versus day 3). Progressive recovery was present during the subsequent 14 days in MCAo-sham-BPT mice (P<0.001, versus day 14) but not in MCAo-BPT mice. In the stroke-affected cervical gray matter of MCAo-sham-BPT mice, growth-associated protein-43-Cy3 staining on CST axons were significantly increased at day 14 after stroke compared with normal mice (P<0.001), and CST axonal density and Synaptophysin-Cy3 staining of CST-yellow fluorescent protein axonal terminals were significantly increased at day 28 compared with day 14 after MCAo (P<0.001). CONCLUSIONS: Our data demonstrate that voluntary motor recovery is associated with CST axonal outgrowth and synaptic formation in the denervated side of the spinal gray matter during the later phase after stroke, suggesting that the CST axonal plasticity in the spinal cord contributes to neurological recovery.


Asunto(s)
Axones/fisiología , Infarto de la Arteria Cerebral Media/fisiopatología , Tractos Piramidales/fisiopatología , Recuperación de la Función/fisiología , Animales , Modelos Animales de Enfermedad , Proteína GAP-43/química , Infarto de la Arteria Cerebral Media/etiología , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Plasticidad Neuronal/fisiología , Tractos Piramidales/lesiones , Tractos Piramidales/patología , Distribución Aleatoria , Sinaptofisina/química , Factores de Tiempo
10.
Sci Rep ; 3: 1392, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23462742

RESUMEN

Neuromodulin (Nm) and neurogranin (Ng) are neuron-specific substrates of protein kinase C (PKC). Their interactions with Calmodulin (CaM) are crucial for learning and memory formation in neurons. Here, we report the structure of IQ peptides (24aa) of Nm/Ng complexed with CaM and their functional studies with full-length proteins. Nm/Ng and their respective IQ peptides are intrinsically unstructured; however, upon binding with CaM, IQ motifs adopt a helical conformation. Ser41 (Ser36) of Nm (Ng) is located in a negatively charged pocket in the apo CaM and, when phosphorylated, it will repel Nm/Ng from CaM. These observations explain the mechanism by which PKC-induced Ser phosphorylation blocks the association of Nm/Ng with CaM and interrupts several learning- and memory-associated functions. Moreover, the present study identified Arg as a key CaM interacting residue from Nm/Ng. This residue is crucial for CaM-mediated function, as evidenced by the inability of the Ng mutant (Arg-to-Ala) to potentiate synaptic transmission in CA1 hippocampal neurons.


Asunto(s)
Calmodulina/metabolismo , Proteína GAP-43/química , Neurogranina/química , Neuronas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteína GAP-43/metabolismo , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Neurogranina/metabolismo , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Secundaria de Proteína , Desplegamiento Proteico , Ratas , Alineación de Secuencia , Transmisión Sináptica
11.
Nat Neurosci ; 15(2): 243-9, 2011 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-22197832

RESUMEN

Synaptotagmin I (syt1) is required for normal rates of synaptic vesicle endo- and exocytosis. However, whether the kinetic defects observed during endocytosis in Syt1 knockout neurons are secondary to defective exocytosis or whether syt1 directly regulates the rate of vesicle retrieval remains unknown. To address this question, we sought to dissociate these two activities. We uncoupled the function of syt1 in exo- and endocytosis in mouse neurons either by re-targeting the protein or via mutagenesis of its tandem C2 domains. The effect of these manipulations on exo- and endocytosis were analyzed using electrophysiology, in conjunction with optical imaging of the vesicle cycle. Our results indicate that syt1 is directly involved in endocytosis. Notably, either of the C2 domains of syt1, C2A or C2B, was able to function as a Ca(2+) sensor for endocytosis. Thus, syt1 functions as a dual Ca(2+) sensor for both endo- and exocytosis, potentially coupling these two components of the vesicle cycle.


Asunto(s)
Endocitosis/fisiología , Exocitosis/fisiología , Sinapsis/fisiología , Vesículas Sinápticas/fisiología , Sinaptotagmina I/metabolismo , Potenciales de Acción/genética , Animales , Animales Recién Nacidos , Biofisica , Calcio/metabolismo , Células Cultivadas , Células Cromafines , Ácido Egtácico/análogos & derivados , Ácido Egtácico/metabolismo , Estimulación Eléctrica , Endocitosis/genética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Exocitosis/genética , Proteína GAP-43/química , Proteína GAP-43/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Ratones , Modelos Biológicos , Mutagénesis/fisiología , Neuronas/citología , Neuronas/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/genética , Sinapsis/genética , Vesículas Sinápticas/genética , Sinaptotagmina I/deficiencia , Sinaptotagmina I/genética , Transfección
12.
Biochim Biophys Acta ; 1804(12): 2162-76, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20869470

RESUMEN

It has recently been proposed that prolyl oligopeptidase (POP), the cytosolic serine peptidase with neurological implications, binds GAP43 (Growth-Associated Protein 43) and is implicated in neuronal growth cone formation, axon guidance and synaptic plasticity. We investigated the interaction between GAP43 and POP with various biophysical and biochemical methods in vitro and studied the co-localisation of the two proteins in differentiated HeLa cells. GAP43 and POP showed partial co-localisation in the cell body as well as in the potential growth cone structures. We could not detect significant binding between the recombinantly expressed POP and GAP43 using gel filtration, CD, ITC and BIACORE studies, pull-down experiments, glutaraldehyde cross-linking and limited proteolysis. However, glutaraldehyde cross-linking suggested a weak and transient interaction between the proteins. Both POP and GAP43 interacted with artificial lipids in our in vitro model system, but the presence of lipids did not evoke binding between them. In native polyacrylamide gel electrophoresis, GAP43 interacted with one of the three forms of a polyhistidine-tagged prolyl oligopeptidase. The interaction of the two proteins was also evident in ELISA and we have observed co-precipitation of the two proteins during co-incubation at higher concentrations. Our results indicate that there is no strong and direct interaction between POP and GAP43 at physiological conditions.


Asunto(s)
Proteína GAP-43/metabolismo , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Bovinos , Diferenciación Celular , Dicroismo Circular , Reactivos de Enlaces Cruzados/química , Electroforesis en Gel de Poliacrilamida , Proteína GAP-43/química , Proteína GAP-43/genética , Glutaral/química , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Células HeLa , Humanos , Membrana Dobles de Lípidos/metabolismo , Microscopía Electrónica , Datos de Secuencia Molecular , Prolil Oligopeptidasas , Unión Proteica , Proteínas Recombinantes/química , Serina Endopeptidasas/química , Serina Endopeptidasas/genética
13.
Dev Neurobiol ; 70(13): 897-911, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20669323

RESUMEN

Visual activity acts via NMDA Receptors to refine developing retinotectal maps by shaping retinal arbors. Retinal axons add and delete transient branches, and the dynamic rates increase when MK801 blocks NMDARs, as if this prevents release of a stabilizing signal. Ca(++) entry through NMDARs activates phospholipase A2 (cPLA2) to release arachidonic acid (AA), which taps into a presynaptic growth control mechanism. NCAM, L1, N-cadherin, and FGF all stimulate axon growth via AA activation of protein kinase C to phosphorylate GAP43 and polymerize/stabilize F-actin. Our previous results show that blocking cPLA2 mimics NMDAR blockers, whereas exogenous AA reverses the increased dynamics, and PKC inhibitors also arrest growth. To test whether this activity-driven F-actin control mechanism shapes retinotectal arbors in zebrafish, we used the alpha-1-tubulin promoter to express GAP43-GFP fusion proteins in retinal ganglion cells, and imaged arbors in time-lapse to test for effects of GAP43 levels and its phosphorylation. Overexpressing wildtype GAP43 gave faster growth and larger arbors (#branches, spatial extent, total length of branches) at three days and especially four days. Surprisingly, the N-terminal 20 amino acid segment alone caused the same increase in branching, but no increase in growth. Earlier studies implicate this region in activating G(o) resulting in collapse of growth cones, which is now known to precede branch initiation. In contrast, GAP43 with ser41 mutated to ala (S41A) to prevent phosphorylation did not increase either branching or growth but resulted in immature, elongated arbors even at four to five days. In support of this atrophic effect, only half of brain/spinal neurons expressing S41A successfully initiated axonal outgrowth (vs. nearly 100% for wtGAP43). These results suggest that the region around the ser41 phosphorylation site, which binds CaM and PIP2, promotes growth only when phosphorylated, and also activates the branching control region in the first 10-20 amino acids. Whereas phosphorylation introduces a bulky negative charge group, mutation of serine to arginine introduces a bulky positive charge. But this also produced the same growth and branching as phosphorylation, suggesting that the effect of phosphorylation is through hydrophilic bulk rather than negative charge, in agreement with other IQ motifs. The results implicate the cPLA2-AA-PKC-GAP43 pathway as part of an F-actin based mechanism that both stabilizes new synapses and initiates new branches near effective synapses.


Asunto(s)
Proteína GAP-43/metabolismo , Conos de Crecimiento/fisiología , Retina/crecimiento & desarrollo , Transducción de Señal/fisiología , Colículos Superiores/crecimiento & desarrollo , Vías Visuales/crecimiento & desarrollo , Animales , Diferenciación Celular/fisiología , Proteína GAP-43/química , Proteína GAP-43/genética , Neurogénesis/fisiología , Fosforilación/fisiología , Retina/citología , Retina/fisiología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Colículos Superiores/citología , Colículos Superiores/fisiología , Vías Visuales/citología , Vías Visuales/fisiología , Pez Cebra
14.
J Struct Biol ; 170(3): 470-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20109554

RESUMEN

Brain abundant proteins GAP-43 and BASP1 participate in the regulation of actin cytoskeleton dynamics in neuronal axon terminals. The proposed mechanism suggests that the proteins sequester phosphatidylinositol-4,5-diphosphate (PIP(2)) in the inner leaflet of the plasma membrane. We found that model anionic phospholipid membranes in the form of liposomes induce rapid oligomerization of GAP-43 and BASP1 proteins. Multiply charged phosphoinositides produced the most potent effect. Anionic detergent sodium dodecyl sulfate (SDS) at submicellar concentration stimulated formation of similar oligomers in solution. BASP1, but not GAP-43, also formed oligomers at sufficiently high concentration in the absence of lipids and SDS. Electron microscopy study demonstrated that the oligomers have disk-shaped or annular structure of 10-30nm in diameter. BASP1 also formed higher aggregates of linear rod-like structure, with average length of about 100nm. In outward appearance, the oligomers and linear aggregates are reminiscent of oligomers and protofibrils of amyloid proteins. Both the synthetic N-terminal peptide GAP-43(1-40) and the brain-derived fragment GAP-43-3 preserved the ability to oligomerize under the action of acidic phospholipids and SDS. On the contrary, BASP1 fragment truncated by the short N-terminal myristoylated peptide was unable to form oligomers. GAP-43 and BASP1 oligomerization can be regulated by calmodulin, which disrupts the oligomers and displaces the proteins from the membrane. We suggest that in vivo, the role of membrane-bound GAP-43 and BASP1 oligomers consists in accumulation of PIP(2) in functional clusters, which become accessible for other PIP(2)-binding proteins after dissociation of the oligomers.


Asunto(s)
Proteína GAP-43/química , Proteínas del Tejido Nervioso/química , Animales , Calmodulina/metabolismo , Bovinos , Reactivos de Enlaces Cruzados , Electroforesis en Gel de Poliacrilamida , Proteína GAP-43/metabolismo , Proteína GAP-43/ultraestructura , Técnicas In Vitro , Lípidos de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/ultraestructura , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/ultraestructura , Fosfatidilinositol 4,5-Difosfato/metabolismo , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Cuaternaria de Proteína
15.
Biochemistry ; 48(49): 11766-72, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19877718

RESUMEN

The affinities of Ca(2+)-saturated and Ca(2+)-free calmodulin for a fluorescent reporter construct containing the PEP19 IQ domain differ by a factor of approximately 100, with K(d) values of 11.0 +/- 1.2 and 1128.4 +/- 176.5 muM, respectively, while the affinities of a reporter containing the neuromodulin IQ domain are essentially identical, with K(d) values of 2.9 +/- 0.3 and 2.4 +/- 0.3 muM, respectively. When Ca(2+) is bound only to the C-terminal pair of Ca(2+)-binding sites in calmodulin, the K(d) value for the PEP19 reporter complex is decreased approximately 5-fold, while the value for the neuromodulin reporter complex is increased by the same factor. When Ca(2+) is bound only to the N-terminal pair of Ca(2+)-binding sites, the K(d) value for the PEP19 reporter complex is unaffected, but the value for the complex with the neuromodulin reporter is increased approximately 12-fold. These functional differences are largely ascribed to three differences in the CaM-binding sequences of the two reporters. Replacement of a central Gly in the neuromodulin IQ domain with a Lys at this position in PEP19 almost entirely accounts for the distinctive patterns of Ca(2+)-dependent stability changes exhibited by the two complexes. Replacement of a Lys immediately before the "IQ" amino acid pair in the neuromodulin sequence with the Ala in PEP19 accounts for the remaining Ca(2+)-dependent differences. Replacement of an Ala in the N-terminal half of the neuromodulin sequence with the Gln in PEP19 accounts for approximately half of the Ca(2+)-independent difference in the stabilities of the two reporter complexes, with the Ca(2+)-independent effect of the Lys replacement accounting for most of the remainder. Since the central Gly in the neuromodulin sequence is conserved in half of all known IQ domains, these results suggest that the presence or absence of this residue defines two major functional classes.


Asunto(s)
Motivos EF Hand/fisiología , Proteína GAP-43/química , Proteína GAP-43/clasificación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/clasificación , Proteínas Activadoras de ras GTPasa/química , Proteínas Activadoras de ras GTPasa/clasificación , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Calmodulina/química , Calmodulina/genética , Calmodulina/metabolismo , Calmodulina/fisiología , Motivos EF Hand/genética , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Genes Reporteros/fisiología , Glicina/genética , Humanos , Ligandos , Lisina/genética , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/genética , Estabilidad Proteica , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
16.
Dev Neurobiol ; 69(2-3): 124-40, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19023859

RESUMEN

Songbirds are appreciated for the insights they provide into regulated neural plasticity. Here, we describe the comparative analysis and brain expression of two gene sequences encoding probable regulators of synaptic plasticity in songbirds: neuromodulin (GAP-43) and neurogranin (RC3). Both are members of the calpacitin family and share a distinctive conserved core domain that mediates interactions between calcium, calmodulin, and protein kinase C signaling pathways. Comparative sequence analysis is consistent with known phylogenetic relationships, with songbirds most closely related to chicken and progressively more distant from mammals and fish. The C-terminus of neurogranin is different in birds and mammals, and antibodies to the protein reveal high expression in adult zebra finches in cerebellar Purkinje cells, which has not been observed in other species. RNAs for both proteins are generally abundant in the telencephalon yet markedly reduced in certain nuclei of the song control system in adult canaries and zebra finches: neuromodulin RNA is very low in RA and HVC (relative to the surrounding pallial areas), whereas neurogranin RNA is conspicuously low in Area X (relative to surrounding striatum). In both cases, this selective downregulation develops in the zebra finch during the juvenile song learning period, 25-45 days after hatching. These results suggest molecular parallels to the robust stability of the adult avian song control circuit.


Asunto(s)
Proteína GAP-43/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Centro Vocal Superior/crecimiento & desarrollo , Centro Vocal Superior/metabolismo , Neurogranina/metabolismo , Vocalización Animal/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Clonación Molecular , Pinzones , Proteína GAP-43/química , Proteína GAP-43/genética , Espacio Intracelular/metabolismo , Neurogranina/química , Neurogranina/genética , ARN Mensajero/metabolismo , Análisis de Secuencia
17.
Gene Expr Patterns ; 8(6): 382-388, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18599366

RESUMEN

Unlike mammals, fish have the capacity for functional adult CNS regeneration, which is due, in part, to their ability to express axon growth-related genes in response to nerve injury. One such axon growth-associated gene is gap43, which is expressed during periods of developmental and regenerative axon growth, but is not expressed in CNS neurons that do not regenerate in adult mammals. We previously demonstrated that cis-regulatory elements of gap43 that are sufficient for developmental expression are not sufficient for regenerative expression in the zebrafish. Here we have identified a 3.6kb genomic sequence from Fugu rubripes that can promote reporter gene expression in the nervous system during both development and regeneration in zebrafish. This compact sequence is advantageous for functional dissection of regions important for axon growth-associated gene expression during development and/or regeneration. In addition, this sequence will also be useful for targeting gene expression to neurons during periods of growth and plasticity.


Asunto(s)
Axones/fisiología , Proteínas de Peces/genética , Proteína GAP-43/genética , Regeneración Nerviosa/genética , Sistema Nervioso/embriología , Neuronas/metabolismo , Takifugu/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Elementos de Facilitación Genéticos , Evolución Molecular , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Proteína GAP-43/química , Proteína GAP-43/metabolismo , Expresión Génica , Genoma , Datos de Secuencia Molecular , Sistema Nervioso/metabolismo , Regiones Promotoras Genéticas , Retina/embriología , Retina/fisiología , Homología de Secuencia de Aminoácido , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
18.
Hippocampus ; 18(8): 814-23, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18493953

RESUMEN

HuD is a neuronal RNA-binding protein associated with the stabilization of mRNAs for GAP-43 and other neuronal proteins that are important for nervous system development and learning and memory mechanisms. To better understand the function of this protein, we generated transgenic mice expressing human HuD (HuD-Tg) in adult forebrain neurons. We have previously shown that expression of HuD in adult dentate granule cells results in an abnormal accumulation of GAP-43 mRNA via posttranscriptional mechanisms. Here we show that this mRNA accumulation leads to the ectopic expression of GAP-43 protein in mossy fibers. Electrophysiological analyses of the mossy fiber to CA3 synapse of HuD-Tg mice revealed increases in paired-pulse facilitation (PPF) at short interpulse intervals and no change in long-term potentiation (LTP). Presynaptic calcium transients at the same synapses exhibited faster time constants of decay, suggesting a decrease in the endogenous Ca(2+) buffer capacity of mossy fiber terminals of HuD-Tg mice. Under resting conditions, GAP-43 binds very tightly to calmodulin sequestering it and then releasing it upon PKC-dependent phosphorylation. Therefore, subsequent studies examined the extent of GAP-43 phosphorylation and its association to calmodulin. We found that despite the increased GAP-43 expression in HuD-Tg mice, the levels of PKC-phosphorylated GAP-43 were decreased in these animals. Furthermore, in agreement with the increased proportion of nonphosphorylated GAP-43, HuD-Tg mice showed increased binding of calmodulin to this protein. These results suggest that a significant amount of calmodulin may be trapped in an inactive state, unable to bind free calcium, and activate downstream signaling pathways. In conclusion, we propose that an unregulated expression of HuD disrupts mossy fiber physiology in adult mice in part by altering the expression and phosphorylation of GAP-43 and the amount of free calmodulin available at the synaptic terminal.


Asunto(s)
Proteínas ELAV/genética , Proteínas ELAV/fisiología , Proteína GAP-43/genética , Proteína GAP-43/fisiología , Fibras Musgosas del Hipocampo/fisiología , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Proteína 4 Similar a ELAV , Electrofisiología , Proteína GAP-43/química , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Terminales Presinápticos/metabolismo , Unión Proteica , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Biophys J ; 94(1): 125-33, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17827240

RESUMEN

The lipid phosphatidylinositol 4,5-bisphosphate (PIP(2)) is critical for a number of physiological functions, and its presence in membrane microdomains (rafts) appears to be important for several of these spatially localized events. However, lipids like PIP(2) that contain polyunsaturated hydrocarbon chains are usually excluded from rafts, which are enriched in phospholipids (such as sphingomyelin) containing saturated or monounsaturated chains. Here we tested a mechanism by which multivalent PIP(2) molecules could be transferred into rafts through electrostatic interactions with polybasic cytoplasmic proteins, such as GAP-43, which bind to rafts via their acylated N-termini. We analyzed the interactions between lipid membranes containing raft microdomains and a peptide (GAP-43P) containing the linked N-terminus and the basic effector domain of GAP-43. In the absence or presence of nonacylated GAP-43P, PIP(2) was found primarily in detergent-soluble membranes thought to correspond to nonraft microdomains. However, when GAP-43P was acylated by palmitoyl coenzyme A, both the peptide and PIP(2) were greatly enriched in detergent-resistant membranes that correspond to rafts; acylation of GAP-43P changed the free energy of transfer of PIP(2) from detergent-soluble membranes to detergent-resistant membranes by -1.3 kcal/mol. Confocal microscopy of intact giant unilamellar vesicles verified that in the absence of GAP-43P PIP(2) was in nonraft microdomains, whereas acylated GAP-43P laterally sequestered PIP(2) into rafts. These data indicate that sequestration of PIP(2) to raft microdomains could involve interactions with acylated basic proteins such as GAP-43.


Asunto(s)
Proteína GAP-43/química , Microdominios de Membrana/química , Fosfatidilinositol 4,5-Difosfato/química , Liposomas Unilamelares/química , Unión Proteica
20.
Biochemistry ; 45(22): 6987-95, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16734434

RESUMEN

The relationship between the free Ca2+ concentration and the apparent dissociation constant for the complex between calmodulin (CaM) and the neuromodulin IQ domain consists of two phases. In the first phase, Ca2+ bound to the C-ter EF hand pair in CaM increases the Kd for the complex from the Ca2+-free value of 2.3 +/- 0.1 microM to a value of 14.4 +/- 1.3 microM. In the second phase, Ca2+ bound to the N-ter EF hand pair reduces the Kd for the complex to a value of 2.5 +/- 0.1 microM, reversing the effect of the first phase. Due to energy coupling effects associated with these phases, the mean dissociation constant for binding of Ca2+ to the C-ter EF hand pair is increased approximately 3-fold, from 1.8 +/- 0.1 to 5.1 +/- 0.7 microM, and the mean dissociation constant for binding of Ca2+ to the N-ter EF hand pair is decreased by the same factor, from 11.2 +/- 1.0 to 3.5 +/- 0.6 microM. These characteristics produce a bell-shaped relationship between the apparent dissociation constant for the complex and the free Ca2+ concentration, with a distance of 5-6 microM between the midpoints of the rising and falling phases. Release of CaM from the neuromodulin IQ domain therefore appears to be promoted over a relatively narrow range of free Ca2+ concentrations. Our results demonstrate that CaM-IQ domain complexes can function as biphasic Ca2+ switches through opposing effects of Ca2+ bound sequentially to the two EF hand pairs in CaM.


Asunto(s)
Calcio/química , Proteínas de Unión a Calmodulina/química , Calmodulina/química , Proteína GAP-43/química , Calmodulina/genética , Mutación , Conformación Proteica , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...