Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Semin Cell Dev Biol ; 27: 44-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24239676

RESUMEN

The transmission of information in the brain depends on the highly polarized architecture of neurons. A number of cellular transport processes support this organization, including active targeting of proteins and passive corralling between compartments. The axon initial segment (AIS), which separates the somatodendritic and axonal compartments, has a key role in neuronal physiology, as both the initiation site of action potentials and the gatekeeper of the axonal arborization. Over the years, the AIS main components and their interactions have been progressively unraveled, as well as their role in the AIS assembly and maintenance. Two mechanisms have been shown to contribute to the regulation of protein transport at the AIS: a surface diffusion barrier and an intracellular traffic filter. However, a molecular understanding of these processes is still lacking. In the view of recent results on the AIS cytoskeleton structure, we will discuss how a better knowledge of the AIS architecture can help understanding its role in the regulation of protein transport and the maintenance of axonal identity.


Asunto(s)
Axones/metabolismo , Animales , Transporte Axonal , Axones/ultraestructura , Polaridad Celular , Proteínas del Citoesqueleto/fisiología , Humanos , Proteínas de la Membrana/metabolismo , Microtúbulos/fisiología , Transporte de Proteínas
2.
J Neurochem ; 134(3): 527-37, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25950943

RESUMEN

The axon initial segment (AIS) plays a central role in electrogenesis and in the maintenance of neuronal polarity. Its molecular organization is dependent on the scaffolding protein ankyrin (Ank) G and is regulated by kinases. For example, the phosphorylation of voltage-gated sodium channels by the protein kinase CK2 regulates their interaction with AnkG and, consequently, their accumulation at the AIS. We previously showed that IQ motif containing J-Schwannomin-Interacting Protein 1 (IQCJ-SCHIP-1), an isoform of the SCHIP-1, accumulated at the AIS in vivo. Here, we analyzed the molecular mechanisms involved in IQCJ-SCHIP-1-specific axonal location. We showed that IQCJ-SCHIP-1 accumulation in the AIS of cultured hippocampal neurons depended on AnkG expression. Pull-down assays and surface plasmon resonance analysis demonstrated that AnkG binds to CK2-phosphorylated IQCJ-SCHIP-1 but not to the non-phosphorylated protein. Surface plasmon resonance approaches using IQCJ-SCHIP-1, SCHIP-1a, another SCHIP-1 isoform, and their C-terminus tail mutants revealed that a segment including multiple CK2-phosphorylatable sites was directly involved in the interaction with AnkG. Pharmacological inhibition of CK2 diminished both IQCJ-SCHIP-1 and AnkG accumulation in the AIS. Silencing SCHIP-1 expression reduced AnkG cluster at the AIS. Finally, over-expression of IQCJ-SCHIP-1 decreased AnkG concentration at the AIS, whereas a mutant deleted of the CK2-regulated AnkG interaction site did not. Our study reveals that CK2-regulated IQJC-SCHIP-1 association with AnkG contributes to AIS maintenance. The axon initial segment (AIS) organization depends on ankyrin (Ank) G and kinases. Here we showed that AnkG binds to CK2-phosphorylated IQCJ-SCHIP-1, in a segment including 12 CK2-phosphorylatable sites. In cultured neurons, either pharmacological inhibition of CK2 or IQCJ-SCHIP-1 silencing reduced AnkG clustering. Overexpressed IQCJ-SCHIP-1 decreased AnkG concentration at the AIS whereas a mutant deleted of the CK2-regulated AnkG interaction site did not. Thus, CK2-regulated IQJC-SCHIP-1 association with AnkG contributes to AIS maintenance.


Asunto(s)
Ancirinas/metabolismo , Axones/metabolismo , Proteínas Portadoras/metabolismo , Quinasa de la Caseína II/metabolismo , Animales , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Hipocampo/metabolismo , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Ratas , Ratas Wistar , Resonancia por Plasmón de Superficie , Transfección
3.
J Neurochem ; 131(1): 33-41, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24903831

RESUMEN

The tetrodotoxin-resistant (TTX-R) voltage-gated sodium channel Nav 1.8 is predominantly expressed in peripheral afferent neurons, but in case of neuronal injury an ectopic and detrimental expression of Nav 1.8 occurs in neurons of the CNS. In CNS neurons, Nav 1.2 and Nav 1.6 channels accumulate at the axon initial segment, the site of the generation of the action potential, through a direct interaction with the scaffolding protein ankyrin G (ankG). This interaction is regulated by protein kinase CK2 phosphorylation. In this study, we quantitatively analyzed the interaction between Nav 1.8 and ankG. GST pull-down assay and surface plasmon resonance technology revealed that Nav 1.8 strongly and constitutively interacts with ankG, in comparison to what observed for Nav 1.2. An ion channel bearing the ankyrin-binding motif of Nav 1.8 displaced the endogenous Nav 1 accumulation at the axon initial segment of hippocampal neurons. Finally, Nav 1.8 and ankG co-localized in skin nerves fibers. Altogether, these results indicate that Nav 1.8 carries all the information required for its localization at ankG micro-domains. The constitutive binding of Nav 1.8 with ankG could contribute to the pathological aspects of illnesses where Nav 1.8 is ectopically expressed in CNS neurons.


Asunto(s)
Ancirinas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Datos de Secuencia Molecular , Embarazo , Unión Proteica/fisiología , Ratas , Ratas Wistar
4.
Proc Natl Acad Sci U S A ; 108(21): 8826-31, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21551097

RESUMEN

The axon initial segment (AIS) plays a key role in maintaining the molecular and functional polarity of the neuron. The relationship between the AIS architecture and the microtubules (MTs) supporting axonal transport is unknown. Here we provide evidence that the MT plus-end-binding (EB) proteins EB1 and EB3 have a role in the AIS in addition to their MT plus-end tracking protein behavior in other neuronal compartments. In mature neurons, EB3 is concentrated and stabilized in the AIS. We identified a direct interaction between EB3/EB1 and the AIS scaffold protein ankyrin G (ankG). In addition, EB3 and EB1 participate in AIS maintenance, and AIS disassembly through ankG knockdown leads to cell-wide up-regulation of EB3 and EB1 comets. Thus, EB3 and EB1 coordinate a molecular and functional interplay between ankG and the AIS MTs that supports the central role of ankG in the maintenance of neuronal polarity.


Asunto(s)
Ancirinas/metabolismo , Axones/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Animales , Polaridad Celular , Neuronas/ultraestructura , Unión Proteica , Ratas , Ratas Wistar
5.
Semin Cell Dev Biol ; 22(2): 171-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20934527

RESUMEN

In mammalian neurons, the generation and propagation of the action potential result from the presence of dense clusters of voltage-gated sodium channels (Nav) at the axonal initial segment (AIS) and nodes of Ranvier. In these two structures, the assembly of specific supra-molecular complexes composed of numerous partners, such as cytoskeletal scaffold proteins and signaling proteins ensures the high concentration of Nav channels. Understanding how neurons regulate the expression and discrete localization of Nav channels is critical to understanding the diversity of normal neuronal function as well as neuronal dysfunction caused by defects in these processes. Here, we review the mechanisms establishing the clustering of Nav channels at the AIS and in the node and discuss how the alterations of Nav channel clustering can lead to certain pathophysiologies.


Asunto(s)
Neuronas/metabolismo , Canales de Sodio/metabolismo , Animales , Humanos , Activación del Canal Iónico , Unión Proteica , Transporte de Proteínas
6.
Eur J Neurosci ; 36(4): 2400-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22642323

RESUMEN

Postnatal formation of the neuromuscular synapse requires complex interactions among nerve terminal, muscle fibres and terminal Schwann cells. In motor endplate disease (med) mice, neuromuscular transmission is severely impaired without alteration of axonal conduction and a lethal paralytic phenotype occurs during the postnatal period. The med phenotype appears at a crucial stage of the neuromuscular junction development, corresponding to the increase in terminal Schwann cell number, the elimination of the multiple innervations and the pre- and postsynaptic maturation. Here we investigated the early cellular and molecular consequences of the med mutation on neuromuscular junction development. We observed that cellular defects preceded overt clinical phenotype. The first detectable cellular effect of the mutation at the onset of the clinical phenotype was a drastic reduction in the number of terminal Schwann cells, in part due to an increase in glial apoptosis, and a delayed maturation of motor endplates. We also showed that, in terminally ill animals, mono-innervation was not achieved, synaptic vesicles had accumulated in the presynaptic compartment and, finally, the size of motor endplates was reduced. All together, our findings suggested that the clinical weakness in these mutant mice was likely to be related to postnatal structural abnormalities of the neuromuscular junction maturation.


Asunto(s)
Enfermedades de la Unión Neuromuscular/patología , Unión Neuromuscular/crecimiento & desarrollo , Animales , Apoptosis , Ratones , Fibras Musculares Esqueléticas/patología , Mutación , Canal de Sodio Activado por Voltaje NAV1.6/genética , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Terminales Presinápticos/patología , Células de Schwann/patología , Vesículas Sinápticas/patología
7.
J Neurosci ; 28(24): 6111-7, 2008 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-18550753

RESUMEN

Axon initial segments (AISs) and nodes of Ranvier (NRs) are essential regions for saltatory conduction of the action potential along the axon. These two domains are enriched in similar multimolecular complexes, which include voltage-gated sodium channels (Na(v)), NF186 (neurofascin 186), NrCAM (neuron glia-related cell adhesion molecule), and cytoskeleton linkers ankyrin G (AnkG) and betaIV-spectrin. Identification of novel members of these complexes is critical to better understand their formation, function, and maintenance. Here we report that IQCJ-SCHIP-1, a recently identified isoform of schwannomin-interacting protein-1 (SCHIP-1), is a novel component of both AISs and NRs in the central and peripheral nervous systems. We show that IQCJ-SCHIP-1 binds calmodulin in the absence of Ca(2+) and is highly enriched at AISs and NRs. IQCJ-SCHIP-1 accumulation at AISs and NRs is a late event, suggesting that IQCJ-SCHIP-1 is likely to play a role in mature AISs and NRs rather than during their formation. IQCJ-SCHIP-1 was not detected at AISs in the absence of AnkG and interacted in vitro with this protein. IQCJ-SCHIP-1 was also absent from central NRs and AISs of quivering mice, which have a mutation of betaIV-spectrin. We suggest that IQCJ-SCHIP-1 might participate, along with AnkG and betaIV-spectrin, in the stabilization or function of the multimolecular complexes of AISs and NRs, possibly by participating in Ca(2+)-mediated responses.


Asunto(s)
Axones/metabolismo , Proteínas Portadoras/metabolismo , Neuronas/citología , Nódulos de Ranvier/metabolismo , Animales , Ancirinas/deficiencia , Línea Celular Transformada , Chlorocebus aethiops , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/biosíntesis , Hipocampo/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteínas Asociadas a Microtúbulos/metabolismo , Espectrina/genética , Transfección/métodos
8.
J Cell Biol ; 166(4): 571-8, 2004 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-15302857

RESUMEN

The axonal initial segment is a unique subdomain of the neuron that maintains cellular polarization and contributes to electrogenesis. To obtain new insights into the mechanisms that determine protein segregation in this subdomain, we analyzed the trafficking of a reporter protein containing the cytoplasmic II-III linker sequence involved in sodium channel targeting and clustering. Here, we show that this reporter protein is preferentially inserted in the somatodendritic domain and is trapped at the axonal initial segment by tethering to the cytoskeleton, before its insertion in the axonal tips. The nontethered population in dendrites, soma, and the distal part of axons is subsequently eliminated by endocytosis. We provide evidence for the involvement of two independent determinants in the II-III linker of sodium channels. These findings indicate that endocytotic elimination and domain-selective tethering constitute a potential mechanism of protein segregation at the axonal initial segment of hippocampal neurons.


Asunto(s)
Axones/metabolismo , Endocitosis , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Axones/química , Brefeldino A/farmacología , Antígenos CD4/biosíntesis , Células COS , Citoplasma/metabolismo , ADN/metabolismo , Detergentes/farmacología , Ácido Glutámico/química , Hipocampo/citología , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/patología , Cinética , Microscopía Confocal , Microscopía Fluorescente , Datos de Secuencia Molecular , Neuronas/metabolismo , Neuronas/patología , Mutación Puntual , Estructura Terciaria de Proteína , Ratas , Canales de Sodio/química , Canales de Sodio/genética , Canales de Sodio/metabolismo , Factores de Tiempo , Transfección
9.
J Neurochem ; 106(2): 969-77, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18466332

RESUMEN

Members of the striatin family are scaffolding proteins involved in numerous signaling pathways principally in neurons. Zinedin is the only member of this protein family for which the brain distribution has not been determined so far. Here, we have validated a specific antibody against zinedin and used this tool to study the localization of zinedin at cellular and sub-cellular levels in the rat brain. Zinedin is primarily expressed in neurons of the hippocampus, cerebral cortex, olfactory bulb and caudate putamen nucleus. Like other members of the striatin family, zinedin displays a polarized distribution in the somato-dendritic compartment of neurons and is enriched in dendritic spines. The rostral expression of zinedin as well as its compartmented distribution in dendritic spines may have important implications not only for zinedin function but also in the physiology of dendritic spines of a particular subset of neurons.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Autoantígenos/metabolismo , Encéfalo/citología , Proteínas de Unión a Calmodulina/genética , Línea Celular Transformada , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Inmunoelectrónica/métodos , Proteínas del Tejido Nervioso/genética , Neuronas/ultraestructura , Ratas , Transfección/métodos
10.
Front Cell Neurosci ; 11: 6, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28184187

RESUMEN

The axon initial segment (AIS) is a highly specialized neuronal compartment that plays a key role in neuronal development and excitability. It concentrates multiple membrane proteins such as ion channels and cell adhesion molecules (CAMs) that are recruited to the AIS by the scaffold protein ankyrin G (ankG). The crucial function of ankG in the anchoring of AIS membrane components is well established, but a reciprocal role of membrane partners in ankG targeting and stabilization remained elusive. In rat cultured hippocampal neurons and cortical organotypic slices, we found that shRNA-mediated knockdown of ankG membrane partners (voltage-gated sodium channels (Nav) or neurofascin-186) led to a decrease of ankG concentration and perturbed the AIS formation and maintenance. These effects were rescued by expressing a recombinant AIS-targeted Nav or by a minimal construct containing the ankyrin-binding domain of Nav1.2 and a membrane anchor (mABD). Moreover, overexpressing mABD in mature neurons led to ankG mislocalization. Altogether, these results demonstrate that a tight and precocious association of ankG with its membrane partners is a key step for the establishment and maintenance of the AIS.

11.
Cell Rep ; 13(12): 2781-93, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26711344

RESUMEN

The axon initial segment (AIS), located within the first 30 µm of the axon, has two essential roles in generating action potentials and maintaining axonal identity. AIS assembly depends on a ßIV-spectrin/ankyrin G scaffold, but its macromolecular arrangement is not well understood. Here, we quantitatively determined the AIS nanoscale architecture by using stochastic optical reconstruction microscopy (STORM). First, we directly demonstrate that the 190-nm periodicity of the AIS submembrane lattice results from longitudinal, head-to-head ßIV-spectrin molecules connecting actin rings. Using multicolor 3D-STORM, we resolve the nanoscale organization of ankyrin G: its amino terminus associates with the submembrane lattice, whereas the C terminus radially extends (∼ 32 nm on average) toward the cytosol. This AIS nano-architecture is highly resistant to cytoskeletal perturbations, indicating its role in structural stabilization. Our findings provide a comprehensive view of AIS molecular architecture and will help reveal the crucial physiological functions of this compartment.


Asunto(s)
Axones/fisiología , Axones/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Animales , Ancirinas/fisiología , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Ratas , Ratas Wistar
12.
J Cell Biol ; 210(3): 401-17, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26216902

RESUMEN

Although actin at neuronal growth cones is well-studied, much less is known about actin organization and dynamics along axon shafts and presynaptic boutons. Using probes that selectively label filamentous-actin (F-actin), we found focal "actin hotspots" along axons-spaced ∼3-4 µm apart-where actin undergoes continuous assembly/disassembly. These foci are a nidus for vigorous actin polymerization, generating long filaments spurting bidirectionally along axons-a phenomenon we call "actin trails." Super-resolution microscopy reveals intra-axonal deep actin filaments in addition to the subplasmalemmal "actin rings" described recently. F-actin hotspots colocalize with stationary axonal endosomes, and blocking vesicle transport diminishes the actin trails, suggesting mechanistic links between vesicles and F-actin kinetics. Actin trails are formin-but not Arp2/3-dependent and help enrich actin at presynaptic boutons. Finally, formin inhibition dramatically disrupts synaptic recycling. Collectively, available data suggest a two-tier F-actin organization in axons, with stable "actin rings" providing mechanical support to the plasma membrane and dynamic "actin trails" generating a flexible cytoskeletal network with putative physiological roles.


Asunto(s)
Actinas/metabolismo , Axones/metabolismo , Proteínas Fetales/metabolismo , Conos de Crecimiento/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Vesículas Sinápticas/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/biosíntesis , Animales , Membrana Celular/fisiología , Proteínas Fetales/antagonistas & inhibidores , Forminas , Proteínas Fluorescentes Verdes , Ratones , Proteínas de Microfilamentos/antagonistas & inhibidores , Microtúbulos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Terminales Presinápticos/metabolismo
13.
J Cell Biol ; 192(5): 813-24, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21357749

RESUMEN

Kv1 channels are concentrated at specific sites in the axonal membrane, where they regulate neuronal excitability. Establishing these distributions requires regulated dissociation of Kv1 channels from the neuronal trafficking machinery and their subsequent insertion into the axonal membrane. We find that the auxiliary Kvß2 subunit of Kv1 channels purified from brain is phosphorylated on serine residues 9 and 31, and that cyclin-dependent kinase (Cdk)-mediated phosphorylation at these sites negatively regulates the interaction of Kvß2 with the microtubule plus end-tracking protein EB1. Endogenous Cdks, EB1, and Kvß2 phosphorylated at serine 31 are colocalized in the axons of cultured hippocampal neurons, with enrichment at the axon initial segment (AIS). Acute inhibition of Cdk activity leads to intracellular accumulation of EB1, Kvß2, and Kv1 channel subunits within the AIS. These studies reveal a new regulatory mechanism for the targeting of Kv1 complexes to the axonal membrane through the reversible Cdk phosphorylation-dependent binding of Kvß2 to EB1.


Asunto(s)
Axones/metabolismo , Quinasas Ciclina-Dependientes/fisiología , Neuronas/metabolismo , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Potasio de la Superfamilia Shaker/metabolismo , Secuencia de Aminoácidos , Animales , Axones/química , Cromatografía Liquida , Hipocampo/química , Hipocampo/metabolismo , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Neuronas/química , Fosforilación , Canales de Potasio con Entrada de Voltaje/análisis , Canales de Potasio con Entrada de Voltaje/química , Subunidades de Proteína/análisis , Subunidades de Proteína/química , Subunidades de Proteína/fisiología , Transporte de Proteínas/fisiología , Ratas , Nervio Ciático/química , Nervio Ciático/metabolismo , Serina/química , Espectrometría de Masas en Tándem
14.
Neurosci Lett ; 486(2): 92-100, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20817077

RESUMEN

In neurons, voltage-gated sodium (Nav) channels underlie the generation and propagation of the action potential. The proper targeting and concentration of Nav channels at the axon initial segment (AIS) and at the nodes of Ranvier are therefore vital for neuronal function. In AIS and nodes, Nav channels are part of specific supra-molecular complexes that include accessory proteins, adhesion proteins and cytoskeletal adaptors. Multiple approaches, from biochemical characterization of protein-protein interactions to functional studies using mutant mice, have addressed the mechanisms of Nav channel targeting to AIS and nodes. This review summarizes our current knowledge of both the intrinsic determinants and the role of partner proteins in Nav targeting. A few fundamental trafficking mechanisms, such as selective endocytosis and diffusion/retention, have been characterized. However, a lot of exciting questions are still open, such as the mechanism of differentiated Nav subtype localization and targeting, and the possible interplay between electrogenesis properties and Nav concentration at the AIS and the nodes.


Asunto(s)
Neuronas/metabolismo , Canales de Sodio/fisiología , Animales , Axones/metabolismo , Difusión , Endocitosis , Humanos , Activación del Canal Iónico , Ratones , Complejos Multiproteicos/metabolismo , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Nódulos de Ranvier/metabolismo , Transducción de Señal , Canales de Sodio/genética
15.
J Cell Biol ; 191(2): 383-95, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20956383

RESUMEN

In mammalian neurons, the precise accumulation of sodium channels at the axonal initial segment (AIS) ensures action potential initiation. This accumulation precedes the immobilization of membrane proteins and lipids by a diffusion barrier at the AIS. Using single-particle tracking, we measured the mobility of a chimeric ion channel bearing the ankyrin-binding motif of the Nav1.2 sodium channel. We found that ankyrin G (ankG) limits membrane diffusion of ion channels when coexpressed in neuroblastoma cells. Site-directed mutants with decreased affinity for ankG exhibit increased diffusion speeds. In immature hippocampal neurons, we demonstrated that ion channel immobilization by ankG is regulated by protein kinase CK2 and occurs as soon as ankG accumulates at the AIS of elongating axons. Once the diffusion barrier is formed, ankG is still required to stabilize ion channels. In conclusion, our findings indicate that specific binding to ankG constitutes the initial step for Nav channel immobilization at the AIS membrane and precedes the establishment of the diffusion barrier.


Asunto(s)
Ancirinas/fisiología , Axones/metabolismo , Membrana Celular/metabolismo , Canales de Sodio/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Quinasa de la Caseína II/metabolismo , Quinasa de la Caseína II/fisiología , Línea Celular , Ratones , Datos de Secuencia Molecular , Fosforilación , Transporte de Proteínas , Alineación de Secuencia , Canales de Sodio/química
16.
PLoS One ; 4(5): e5509, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19434240

RESUMEN

The neuropeptide somatostatin has been suggested to play an important role during neuronal development in addition to its established modulatory impact on neuroendocrine, motor and cognitive functions in adults. Although six somatostatin G protein-coupled receptors have been discovered, little is known about their distribution and function in the developing mammalian brain. In this study, we have first characterized the developmental expression of the somatostatin receptor sst2A, the subtype found most prominently in the adult rat and human nervous system. In the rat, the sst2A receptor expression appears as early as E12 and is restricted to post-mitotic neuronal populations leaving the ventricular zone. From E12 on, migrating neuronal populations immunopositive for the receptor were observed in numerous developing regions including the cerebral cortex, hippocampus and ganglionic eminences. Intense but transient immunoreactive signals were detected in the deep part of the external granular layer of the cerebellum, the rostral migratory stream and in tyrosine hydroxylase- and serotonin- positive neurons and axons. Activation of the sst2A receptor in vitro in rat cerebellar microexplants and primary hippocampal neurons revealed stimulatory effects on neuronal migration and axonal growth, respectively. In the human cortex, receptor immunoreactivity was located in the preplate at early development stages (8 gestational weeks) and was enriched to the outer part of the germinal zone at later stages. In the cerebellum, the deep part of the external granular layer was strongly immunoreactive at 19 gestational weeks, similar to the finding in rodents. In addition, migrating granule cells in the internal granular layer were also receptor-positive. Together, theses results strongly suggest that the somatostatin sst2A receptor participates in the development and maturation of specific neuronal populations during rat and human brain ontogenesis.


Asunto(s)
Axones/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Movimiento Celular , Neuronas/citología , Neuronas/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/citología , Dendritas/metabolismo , Técnica del Anticuerpo Fluorescente , Edad Gestacional , Humanos , Especificidad de Órganos , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo
17.
J Cell Biol ; 183(6): 1101-14, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19064667

RESUMEN

In neurons, generation and propagation of action potentials requires the precise accumulation of sodium channels at the axonal initial segment (AIS) and in the nodes of Ranvier through ankyrin G scaffolding. We found that the ankyrin-binding motif of Na(v)1.2 that determines channel concentration at the AIS depends on a glutamate residue (E1111), but also on several serine residues (S1112, S1124, and S1126). We showed that phosphorylation of these residues by protein kinase CK2 (CK2) regulates Na(v) channel interaction with ankyrins. Furthermore, we observed that CK2 is highly enriched at the AIS and the nodes of Ranvier in vivo. An ion channel chimera containing the Na(v)1.2 ankyrin-binding motif perturbed endogenous sodium channel accumulation at the AIS, whereas phosphorylation-deficient chimeras did not. Finally, inhibition of CK2 activity reduced sodium channel accumulation at the AIS of neurons. In conclusion, CK2 contributes to sodium channel organization by regulating their interaction with ankyrin G.


Asunto(s)
Ancirinas/metabolismo , Axones/metabolismo , Quinasa de la Caseína II/metabolismo , Membrana Celular/metabolismo , Canales de Sodio/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Axones/efectos de los fármacos , Axones/enzimología , Quinasa de la Caseína II/antagonistas & inhibidores , Membrana Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Ácido Glutámico/metabolismo , Hipocampo/citología , Activación del Canal Iónico/efectos de los fármacos , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Mutación Puntual/genética , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Nódulos de Ranvier/efectos de los fármacos , Nódulos de Ranvier/enzimología , Ratas , Serina/metabolismo , Canales de Sodio/química
18.
Traffic ; 7(1): 74-84, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16445688

RESUMEN

Striatin, SG2NA and zinedin, the three mammalian members of the striatin family are multimodular WD-repeat, calmodulin and calveolin-binding proteins. These scaffolding proteins, involved in both signaling and trafficking, are highly expressed in neurons. Using ultrastructural immunolabeling, we showed that, in Purkinje cells and hippocampal neurons, SG2NA is confined to the somatodendritic compartment with the highest density in dendritic spines. In cultured hippocampal neurons, SG2NA is also highly concentrated in dendritic spines. By expressing truncated forms of HA-tagged SG2NAbeta, we demonstrated that the coiled-coil domain plays an essential role in the targeting of SG2NA within spines. Furthermore, co-immunoprecipitation experiments indicate that this coiled-coil domain is also crucial for the homo- and hetero-oligomerization of these proteins. Thus, oligomerization of the striatin family proteins is probably an obligatory step for their routing to the dendritic spines, and hetero-oligomerization explains why all these proteins are often co-expressed in the neurons of the rat brain and spinal cord.


Asunto(s)
Autoantígenos/biosíntesis , Proteínas de Unión a Calmodulina/biosíntesis , Espinas Dendríticas/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Espinas Dendríticas/química , Técnicas de Transferencia de Gen , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/fisiología , Ratas , Transducción de Señal
19.
Biol Cell ; 95(7): 437-45, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14597261

RESUMEN

One of the major physiological roles of the neuronal voltage-gated sodium channel is to generate action potentials at the axon hillock/initial segment and to ensure propagation along myelinated or unmyelinated fibers to nerve terminal. These processes require a precise distribution of sodium channels accumulated at high density in discrete subdomains of the nerve membrane. In neurons, information relevant to ion channel trafficking and compartmentalization into sub-domains of the plasma membrane is far from being elucidated. Besides, whereas information on dendritic targeting is beginning to emerge, less is known about the mechanisms leading to the polarized distribution of proteins in axon. To obtain a better understanding of how neurons selectively target sodium channels to discrete subdomains of the nerve, we addressed the question as to whether any of the large intracellular regions of Nav1.2 contain axonal sorting and/or clustering signals. We first obtained evidence showing that addition of the cytoplasmic carboxy-terminal region of Nav1.2 restricted the distribution of a dendritic-axonal reporter protein to axons of hippocampal neurons. The analysis of mutants revealed that a di-leucine-based motif mediates chimera compartmentalization in axons and its elimination in soma and dendrites by endocytosis. The analysis of the others generated chimeras showed that the determinant conferring sodium channel clustering at the axonal initial segment is contained within the cytoplasmic loop connecting domains II-III of Nav1.2. Expression of a soluble Nav1.2 II-III linker protein led to the disorganization of endogenous sodium channels. The motif was sufficient to redirect a somatodendritic potassium channel to the axonal initial segment, a process involving association with ankyrin G. Thus, it is conceivable that concerted action of the two determinants is required for sodium channel compartmentalization in axons.


Asunto(s)
Axones/química , Proteínas del Tejido Nervioso/análisis , Canales de Sodio/análisis , Secuencias de Aminoácidos , Animales , Compartimento Celular , Endocitosis , Péptidos y Proteínas de Señalización Intercelular/análisis , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/química , Ratas , Canales de Sodio/química
20.
Science ; 300(5628): 2091-4, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12829783

RESUMEN

The sorting of sodium channels to axons and the formation of clusters are of primary importance for neuronal electrogenesis. Here, we showed that the cytoplasmic loop connecting domains II and III of the Nav1 subunit contains a determinant conferring compartmentalization in the axonal initial segment of rat hippocampal neurons. Expression of a soluble Nav1.2II-III linker protein led to the disorganization of endogenous sodium channels. The motif was sufficient to redirect a somatodendritic potassium channel to the axonal initial segment, a process involving association with ankyrin G. Thus, this motif may play a fundamental role in controlling electrical excitability during development and plasticity.


Asunto(s)
Axones/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Sodio/química , Canales de Sodio/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Ancirinas/metabolismo , Membrana Celular/metabolismo , Canales de Potasio de Tipo Rectificador Tardío , Hipocampo/citología , Humanos , Activación del Canal Iónico , Datos de Secuencia Molecular , Mutación , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Canales de Sodio/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA