Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(7): 378, 2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35739402

RESUMEN

SUMOylation is a post-translational modification essential to cell homeostasis. A tightly controlled equilibrium between SUMOylation and deSUMOylation processes is also critical to the neuronal function including neurotransmitter release and synaptic transmission and plasticity. Disruption of the SUMOylation homeostasis in neurons is associated with several neurological disorders. The balance between the SUMOylation and deSUMOylation of substrate proteins is maintained by a group of deSUMOylation enzymes called SENPs. We previously showed that the activation of type 5 metabotropic glutamate receptors (mGlu5R) first triggers a rapid increase in synaptic SUMOylation and then upon the sustained activation of these receptors, the deSUMOylase activity of SENP1 allows the increased synaptic SUMOylation to get back to basal levels. Here, we combined the use of pharmacological tools with subcellular fractionation and live-cell imaging of individual hippocampal dendritic spines to demonstrate that the synaptic accumulation of the deSUMOylation enzyme SENP1 is bidirectionally controlled by the activation of type 1 mGlu1 and mGlu5 receptors. Indeed, the pharmacological blockade of mGlu1R activation during type 1 mGluR stimulation leads to a faster and greater accumulation of SENP1 at synapses indicating that mGlu1R acts as a brake to the mGlu5R-dependent deSUMOylation process at the post-synapse. Altogether, our findings reveal that type 1 mGluRs work in opposition to dynamically tune the homeostasis of SUMOylation at the mammalian synapse.


Asunto(s)
Receptores de Glutamato Metabotrópico , Sumoilación , Animales , Hipocampo/metabolismo , Mamíferos/metabolismo , Neuronas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/metabolismo
2.
Nat Commun ; 12(1): 1557, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692361

RESUMEN

Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and the best-described monogenic cause of autism. CGG-repeat expansion in the FMR1 gene leads to FMR1 silencing, loss-of-expression of the Fragile X Mental Retardation Protein (FMRP), and is a common cause of FXS. Missense mutations in the FMR1 gene were also identified in FXS patients, including the recurrent FMRP-R138Q mutation. To investigate the mechanisms underlying FXS caused by this mutation, we generated a knock-in mouse model (Fmr1R138Q) expressing the FMRP-R138Q protein. We demonstrate that, in the hippocampus of the Fmr1R138Q mice, neurons show an increased spine density associated with synaptic ultrastructural defects and increased AMPA receptor-surface expression. Combining biochemical assays, high-resolution imaging, electrophysiological recordings, and behavioural testing, we also show that the R138Q mutation results in impaired hippocampal long-term potentiation and socio-cognitive deficits in mice. These findings reveal the functional impact of the FMRP-R138Q mutation in a mouse model of FXS.


Asunto(s)
Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Mutación Missense/fisiología , Receptores de Glutamato/metabolismo , Animales , Biotinilación , Encéfalo/metabolismo , Encéfalo/fisiopatología , Células Cultivadas , Disfunción Cognitiva/metabolismo , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Immunoblotting , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Mutación Missense/genética , Técnicas de Placa-Clamp , Receptores de Glutamato/genética
3.
eNeuro ; 7(4)2020.
Artículo en Inglés | MEDLINE | ID: mdl-32719102

RESUMEN

Posttranslational modifications (PTMs) represent a dynamic regulatory system that precisely modulates the functional organization of synapses. PTMs consist in target modifications by small chemical moieties or conjugation of lipids, sugars or polypeptides. Among them, ubiquitin and a large family of ubiquitin-like proteins (UBLs) share several features such as the structure of the small protein modifiers, the enzymatic cascades mediating the conjugation process, and the targeted aminoacidic residue. In the brain, ubiquitination and two UBLs, namely sumoylation and the recently discovered neddylation orchestrate fundamental processes including synapse formation, maturation and plasticity, and their alteration is thought to contribute to the development of neurological disorders. Remarkably, emerging evidence suggests that these pathways tightly interplay to modulate the function of several proteins that possess pivotal roles for brain homeostasis as well as failure of this crosstalk seems to be implicated in the development of brain pathologies. In this review, we outline the role of ubiquitination, sumoylation, neddylation, and their functional interplay in synapse physiology and discuss their implication in the molecular pathogenesis of intellectual disability (ID), a neurodevelopmental disorder that is frequently comorbid with a wide spectrum of brain pathologies. Finally, we propose a few outlooks that might contribute to better understand the complexity of these regulatory systems in regard to neuronal circuit pathophysiology.


Asunto(s)
Discapacidad Intelectual , Ubiquitina , Humanos , Sumoilación , Sinapsis/metabolismo , Ubiquitinas/metabolismo
4.
Mol Psychiatry ; 25(8): 1688-1703, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31822816

RESUMEN

The Fragile X Mental Retardation Protein (FMRP) is an RNA-binding protein essential to the regulation of local translation at synapses. In the mammalian brain, synapses are constantly formed and eliminated throughout development to achieve functional neuronal networks. At the molecular level, thousands of proteins cooperate to accomplish efficient neuronal communication. Therefore, synaptic protein levels and their functional interactions need to be tightly regulated. FMRP generally acts as a translational repressor of its mRNA targets. FMRP is the target of several post-translational modifications (PTMs) that dynamically regulate its function. Here we provide an overview of the PTMs controlling the FMRP function and discuss how their spatiotemporal interplay contributes to the physiological regulation of FMRP. Importantly, FMRP loss-of-function leads to Fragile X syndrome (FXS), a rare genetic developmental condition causing a range of neurological alterations including intellectual disability (ID), learning and memory impairments, autistic-like features and seizures. Here, we also explore the possibility that recently reported missense mutations in the FMR1 gene disrupt the PTM homoeostasis of FMRP, thus participating in the aetiology of FXS. This suggests that the pharmacological targeting of PTMs may be a promising strategy to develop innovative therapies for patients carrying such missense mutations.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/química , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Procesamiento Proteico-Postraduccional , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Humanos , Sinapsis/metabolismo , Sinapsis/patología
5.
Cell Mol Life Sci ; 76(15): 3019-3031, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30904951

RESUMEN

Sumoylation is a reversible post-translational modification essential to the modulation of neuronal function, including neurotransmitter release and synaptic plasticity. A tightly regulated equilibrium between the sumoylation and desumoylation processes is critical to the brain function and its disruption has been associated with several neurological disorders. This sumoylation/desumoylation balance is governed by the activity of the sole SUMO-conjugating enzyme Ubc9 and a group of desumoylases called SENPs, respectively. We previously demonstrated that the activation of type 5 metabotropic glutamate receptors (mGlu5R) triggers the transient trapping of Ubc9 in dendritic spines, leading to a rapid increase in the overall synaptic sumoylation. However, the mechanisms balancing this increased synaptic sumoylation are still not known. Here, we examined the diffusion properties of the SENP1 enzyme using a combination of advanced biochemical approaches and restricted photobleaching/photoconversion of individual hippocampal spines. We demonstrated that the activation of mGlu5R leads to a time-dependent decrease in the exit rate of SENP1 from dendritic spines. The resulting post-synaptic accumulation of SENP1 restores synaptic sumoylation to initial levels. Altogether, our findings reveal the mGlu5R system as a central activity-dependent mechanism to maintaining the homeostasis of sumoylation at the mammalian synapse.


Asunto(s)
Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Cisteína Endopeptidasas/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Microscopía Fluorescente , Neuronas/citología , Neuronas/metabolismo , Ratas Wistar , Proteína SUMO-1/metabolismo , Sumoilación , Enzimas Ubiquitina-Conjugadoras/metabolismo
6.
Front Mol Neurosci ; 11: 342, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319351

RESUMEN

Fragile X syndrome (FXS), the most common form of inherited intellectual disability (ID) and a leading cause of autism, results from the loss of expression of the Fmr1 gene which encodes the RNA-binding protein Fragile X Mental Retardation Protein (FMRP). Among the thousands mRNA targets of FMRP, numerous encode regulators of ion homeostasis. It has also been described that FMRP directly interacts with Ca2+ channels modulating their activity. Collectively these findings suggest that FMRP plays critical roles in Ca2+ homeostasis during nervous system development. We carried out a functional analysis of Ca2+ regulation using a calcium imaging approach in Fmr1-KO cultured neurons and we show that these cells display impaired steady state Ca2+ concentration and an altered entry of Ca2+ after KCl-triggered depolarization. Consistent with these data, we show that the protein product of the Cacna1a gene, the pore-forming subunit of the Cav2.1 channel, is less expressed at the plasma membrane of Fmr1-KO neurons compared to wild-type (WT). Thus, our findings point out the critical role that Cav2.1 plays in the altered Ca2+ flux in Fmr1-KO neurons, impacting Ca2+ homeostasis of these cells. Remarkably, we highlight a new phenotype of cultured Fmr1-KO neurons that can be considered a novel cellular biomarker and is amenable to small molecule screening and identification of new drugs to treat FXS.

7.
Nat Commun ; 9(1): 757, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29472612

RESUMEN

Fragile X syndrome (FXS) is the most frequent inherited cause of intellectual disability and the best-studied monogenic cause of autism. FXS results from the functional absence of the fragile X mental retardation protein (FMRP) leading to abnormal pruning and consequently to synaptic communication defects. Here we show that FMRP is a substrate of the small ubiquitin-like modifier (SUMO) pathway in the brain and identify its active SUMO sites. We unravel the functional consequences of FMRP sumoylation in neurons by combining molecular replacement strategy, biochemical reconstitution assays with advanced live-cell imaging. We first demonstrate that FMRP sumoylation is promoted by activation of metabotropic glutamate receptors. We then show that this increase in sumoylation controls the homomerization of FMRP within dendritic mRNA granules which, in turn, regulates spine elimination and maturation. Altogether, our findings reveal the sumoylation of FMRP as a critical activity-dependent regulatory mechanism of FMRP-mediated neuronal function.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Sumoilación , Secuencia de Aminoácidos , Animales , Células Cultivadas , Espinas Dendríticas/genética , Espinas Dendríticas/patología , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/química , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Modelos Neurológicos , Fenotipo , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vesículas Secretoras/metabolismo , Homología de Secuencia de Aminoácido
8.
Hum Mol Genet ; 27(6): 1027-1038, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29360992

RESUMEN

The PCDH19 gene (Xp22.1) encodes the cell-adhesion protein protocadherin-19 (PCDH19) and is responsible for a neurodevelopmental pathology characterized by female-limited epilepsy, cognitive impairment and autistic features, the pathogenic mechanisms of which remain to be elucidated. Here, we identified a new interaction between PCDH19 and GABAA receptor (GABAAR) alpha subunits in the rat brain. PCDH19 shRNA-mediated downregulation reduces GABAAR surface expression and affects the frequency and kinetics of miniature inhibitory postsynaptic currents (mIPSCs) in cultured hippocampal neurons. In vivo, PCDH19 downregulation impairs migration, orientation and dendritic arborization of CA1 hippocampal neurons and increases rat seizure susceptibility. In sum, these data indicate a role for PCDH19 in GABAergic transmission as well as migration and morphological maturation of neurons.


Asunto(s)
Cadherinas/metabolismo , Moduladores del GABA/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Animales , Células COS , Chlorocebus aethiops , Epilepsia/genética , Femenino , Células HEK293 , Hipocampo/citología , Humanos , Potenciales Postsinápticos Inhibidores , Plasticidad Neuronal , Protocadherinas , Ratas , Ratas Sprague-Dawley , Convulsiones/metabolismo
9.
J Biol Chem ; 293(3): 1040-1053, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29180451

RESUMEN

L-type voltage-gated CaV1.2 calcium channels (CaV1.2) are key regulators of neuronal excitability, synaptic plasticity, and excitation-transcription coupling. Surface-exposed CaV1.2 distributes in clusters along the dendrites of hippocampal neurons. A permanent exchange between stably clustered and laterally diffusive extra-clustered channels maintains steady-state levels of CaV1.2 at dendritic signaling domains. A dynamic equilibrium between anchored and diffusive receptors is a common feature among ion channels and is crucial to modulate signaling transduction. Despite the importance of this fine regulatory system, the molecular mechanisms underlying the surface dynamics of CaV1.2 are completely unexplored. Here, we examined the dynamic states of CaV1.2 depending on phosphorylation on Ser-1700 and Ser-1928 at the channel C terminus. Phosphorylation at these sites is strongly involved in CaV1.2-mediated nuclear factor of activated T cells (NFAT) signaling, long-term potentiation, and responsiveness to adrenergic stimulation. We engineered CaV1.2 constructs mimicking phosphorylation at Ser-1700 and Ser-1928 and analyzed their behavior at the membrane by immunolabeling protocols, fluorescence recovery after photobleaching, and single particle tracking. We found that the phosphomimetic S1928E variant increases the mobility of CaV1.2 without altering the steady-state maintenance of cluster in young neurons and favors channel stabilization later in differentiation. Instead, mimicking phosphorylation at Ser-1700 promoted the diffusive state of CaV1.2 irrespective of the differentiation stage. Together, these results reveal that phosphorylation could contribute to the establishment of channel anchoring mechanisms depending on the neuronal differentiation state. Finally, our findings suggest a novel mechanism by which phosphorylation at the C terminus regulates calcium signaling by tuning the content of CaV1.2 at signaling complexes.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Hipocampo/citología , Neuronas/citología , Neuronas/metabolismo , Animales , Electrofisiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Simulación de Dinámica Molecular , Fosforilación , Ratas , Ratas Sprague-Dawley
10.
Cereb Cortex ; 27(11): 5369-5384, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28968657

RESUMEN

Intellectual disability affects 2-3% of the world's population and typically begins during childhood, causing impairments in social skills and cognitive abilities. Mutations in the TM4SF2 gene, which encodes the TSPAN7 protein, cause a severe form of intellectual disability, and currently, no therapy is able to ameliorate this cognitive impairment. We previously reported that, in cultured neurons, shRNA-mediated down-regulation of TSPAN7 affects AMPAR trafficking by enhancing PICK1-GluA2 interaction, thereby increasing the intracellular retention of AMPAR. Here, we found that loss of TSPAN7 function in mice causes alterations in hippocampal excitatory synapse structure and functionality as well as cognitive impairment. These changes occurred along with alterations in AMPAR expression levels. We also found that interfering with PICK1-GluA2 binding restored synaptic function in Tm4sf2-/y mice. Moreover, potentiation of AMPAR activity via the administration of the ampakine CX516 reverted the neurological phenotype observed in Tm4sf2-/y mice, suggesting that pharmacological modulation of AMPAR may represent a new approach for treating patients affected by TM4SF2 mutations and intellectual disability.


Asunto(s)
Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Psicotrópicos/farmacología , Receptores AMPA/metabolismo , Regulación Alostérica , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/ultraestructura , Discapacidad Intelectual/patología , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos
11.
Front Mol Neurosci ; 9: 1, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26834556

RESUMEN

Myosin IXa (Myo9a) is a motor protein that is highly expressed in the brain. However, the role of Myo9a in neurons remains unknown. Here, we investigated Myo9a function in hippocampal synapses. In rat hippocampal neurons, Myo9a localizes to the postsynaptic density (PSD) and binds the alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) GluA2 subunit. Myo9a(+/-) mice displayed a thicker PSD and increased levels of PSD95 and surface AMPAR expression. Furthermore, synaptic transmission, long-term potentiation (LTP) and cognitive functions were impaired in Myo9a(+/-) mice. Together, these results support a key role for Myo9a in controlling the molecular structure and function of hippocampal synapses.

12.
Nat Commun ; 5: 5182, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25316086

RESUMEN

Although loss-of-function mutations in the PARK2 gene, the gene that encodes the protein parkin, cause autosomal recessive juvenile parkinsonism, the responsible molecular mechanisms remain unclear. Evidence suggests that a loss of parkin dysregulates excitatory synapses. Here we show that parkin interacts with the kainate receptor (KAR) GluK2 subunit and regulates KAR function. Loss of parkin function in primary cultured neurons causes GluK2 protein to accumulate in the plasma membrane, potentiates KAR currents and increases KAR-dependent excitotoxicity. Expression in the mouse brain of a parkin mutant causing autosomal recessive juvenile parkinsonism results in GluK2 protein accumulation and excitotoxicity. These findings show that parkin regulates KAR function in vitro and in vivo, and suggest that KAR upregulation may have a pathogenetic role in parkin-related autosomal recessive juvenile parkinsonism.


Asunto(s)
Trastornos Parkinsonianos/metabolismo , Receptores de Ácido Kaínico/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Membrana Celular/genética , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Trastornos Parkinsonianos/genética , Unión Proteica , Ratas , Receptores de Ácido Kaínico/genética , Ubiquitina-Proteína Ligasas/genética , Receptor de Ácido Kaínico GluK2
13.
J Neurosci ; 34(27): 9088-95, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24990929

RESUMEN

Heterogeneous nuclear ribonucleoprotein K (hnRNP K) is an RNA-binding protein implicated in RNA metabolism. Here, we investigated the role of hnRNP K in synapse function. We demonstrated that hnRNP K regulates dendritic spine density and long-term potentiation (LTP) in cultured hippocampal neurons from embryonic rats. LTP requires the extracellular signal-regulated kinase (ERK)1/2-mediated phosphorylation and cytoplasmic accumulation of hnRNP K. Moreover, hnRNP K knockdown prevents ERK cascade activation and GluA1-S845 phosphorylation and surface delivery, which are essential steps for LTP. These findings establish hnRNP K as a new critical regulator of synaptic transmission and plasticity in hippocampal neurons.


Asunto(s)
Potenciación a Largo Plazo/fisiología , Proteínas del Tejido Nervioso/fisiología , Ribonucleoproteínas/fisiología , Transmisión Sináptica/fisiología , Animales , Señalización del Calcio , Células Cultivadas , Dendritas/ultraestructura , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipocampo/citología , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Neuronas/ultraestructura , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Receptores AMPA/metabolismo , Ribonucleoproteínas/antagonistas & inhibidores , Ribonucleoproteínas/genética , Transfección
14.
Cell Mol Life Sci ; 70(23): 4411-30, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23475111

RESUMEN

Glutamate ionotropic alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPARs) mediate most fast excitatory synaptic transmission in the central nervous system. The content and composition of AMPARs in postsynaptic membranes (which determine synaptic strength) are dependent on the regulated trafficking of AMPAR subunits in and out of the membranes. AMPAR trafficking is a key mechanism that drives nascent synapse development, and is the main determinant of both Hebbian and homeostatic plasticity in mature synapses. Hebbian plasticity seems to be the biological substrate of at least some forms of learning and memory; while homeostatic plasticity (also known as synaptic scaling) keeps neuronal circuits stable by maintaining changes within a physiological range. In this review, we examine recent findings that provide further understanding of the role of AMPAR trafficking in synapse maturation, Hebbian plasticity, and homeostatic plasticity.


Asunto(s)
Plasticidad Neuronal/fisiología , Receptores AMPA/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Homeostasis/fisiología , Humanos , Modelos Neurológicos , Transporte de Proteínas/fisiología , Receptores AMPA/metabolismo , Sinapsis/metabolismo
15.
Neuron ; 73(6): 1143-58, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22445342

RESUMEN

Mutations in TSPAN7--a member of the tetraspanin protein superfamily--are implicated in some forms of X-linked intellectual disability. Here we show that TSPAN7 overexpression promotes the formation of filopodia and dendritic spines in cultured hippocampal neurons from embryonic rats, whereas TSPAN7 silencing reduces head size and stability of spines and AMPA receptor currents. Via its C terminus, TSPAN7 interacts with the PDZ domain of protein interacting with C kinase 1 (PICK1), to regulate PICK1 and GluR2/3 association and AMPA receptor trafficking. These findings indicate that, in hippocampal neurons, TSPAN7 regulates AMPA receptor trafficking by limiting PICK1 accessibility to AMPA receptors and suggest an additional mechanism for the functional maturation of glutamatergic synapses, whose impairment is implicated in intellectual disability.


Asunto(s)
Espinas Dendríticas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Receptores AMPA/metabolismo , Sinapsis/fisiología , Tetraspaninas/metabolismo , Análisis de Varianza , Animales , Biofisica , Proteínas Portadoras/metabolismo , Células Cultivadas , Chlorocebus aethiops , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/genética , Homólogo 4 de la Proteína Discs Large , Estimulación Eléctrica , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Hidrazonas/farmacología , Inmunoprecipitación , Técnicas In Vitro , Integrina beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Técnicas de Placa-Clamp , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Seudópodos/efectos de los fármacos , Seudópodos/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Sinapsis/genética , Tetraspaninas/genética , Factores de Tiempo , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...