Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Pharmacol ; 95(1): 20-32, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30404891

RESUMEN

Calcineurin (CaN) phosphatase signaling is regulated by targeting CaN to substrates, inhibitors, and scaffold proteins containing docking motifs with the consensus sequence of PxIxIT. Here, we identify the docking of CaN to the γ isoform of MKK7, a component of the c-Jun N-terminal kinase (JNK) pathway. Because of alternative splicing of a single exon within the N-terminal domain, MKK7γ encodes a unique PxIxIT motif (PIIVIT) that is not present in MKK7α or ß We found that MKK7γ bound directly to CaN through this PIIVIT motif in vitro, immunoprecipitated with CaN from cell extracts, and exhibited fluorescence resonance energy transfer (FRET) with CaN in the cytoplasm but not in the nucleus of living cells. In contrast, MKK7α and ß exhibited no direct binding or FRET with CaN and were localized more in the nucleus than the cytoplasm. Furthermore, the inhibition of CaN phosphatase activity increased the basal phosphorylation of MKK7γ but not MKK7ß Deletion of the MKK7γ PIIVIT motif eliminated FRET with CaN and promoted MKK7γ redistribution to the nucleus; however, the inhibition of CaN activity did not alter MKK7γ localization, indicating that MKK7γ cytoplasmic retention by CaN is phosphatase activity independent. Finally, the inhibition of CaN phosphatase activity in vascular smooth muscle cells, which express MKK7γ mRNA, enhances JNK activation. Overall, we conclude that the MKK7γ-specific PxIxIT motif promotes high-affinity CaN binding that could promote novel cross talk between CaN and JNK signaling by limiting MKK7γ phosphorylation and restricting its localization to the cytoplasm.


Asunto(s)
MAP Quinasa Quinasa 7/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica/fisiología , Isoformas de Proteínas/metabolismo , Empalme Alternativo/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Células COS , Línea Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosforilación/fisiología , Transducción de Señal/fisiología
2.
J Neurosci ; 36(45): 11532-11543, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27911757

RESUMEN

Beta amyloid (Aß) triggers the elimination of excitatory synaptic connections in the CNS, an early manifestation of Alzheimer's disease. Oligomeric assemblies of Aß peptide associate with excitatory synapses resulting in synapse elimination through a process that requires NMDA-type glutamate receptor activation. Whether Aß affects synaptic NMDA receptor (NMDAR) function directly and acts locally at synapses to which it has bound and whether synaptic activity influences Aß synaptic binding and synaptotoxicity have remained fundamental questions. Here, we used subcellular Ca2+ imaging in rat hippocampal neurons to visualize NMDAR function at individual synapses before and after Aß application. Aß triggered a robust impairment of NMDAR Ca2+ entry at most, but not all, synapses. NMDAR function was more severely impaired at highly active synapses and synapses with bound Aß, but activity was not required for Aß synapse binding. Blocking NMDARs during Aß exposure prevented Aß-mediated impairment. Finally, Aß impaired NMDAR Ca2+ entry at doses much lower than those required for NMDAR internalization, revealing a novel, potent mode of NMDAR regulation by Aß. SIGNIFICANCE STATEMENT: Amyloid ß (Aß) is strongly implicated in Alzheimer's disease. Aß triggers the elimination of excitatory synapses through a mechanism that requires NMDA receptors (NMDARs). However, little is known about how or whether Aß influences synaptic NMDAR function. We used an imaging-based assay to investigate the relationship among Aß binding, activity, and NMDAR function at individual synapses. Aß triggered a robust impairment of NMDAR Ca2+ entry at most, but not all, synapses. NMDAR function was more severely impaired at highly active synapses and synapses with bound Aß. Blocking NMDARs during Aß exposure prevented Aß-mediated impairment. Together, our experiments reveal a novel use-dependent, potent, and local mode of Aß-mediated NMDAR impairment.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Señalización del Calcio/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Imagen de Colorante Sensible al Voltaje/métodos , Animales , Células Cultivadas , Femenino , Masculino , Ratas , Ratas Sprague-Dawley
3.
Mol Pharmacol ; 89(5): 552-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26957206

RESUMEN

Alzheimer's disease (AD) is characterized by neurofibrillary tangles, amyloid plaques, and neurodegeneration. However, this pathology is preceded by increased soluble amyloid beta (Aß) 1-42 oligomers that interfere with the glutamatergic synaptic plasticity required for learning and memory, includingN-methyl-d-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP). In particular, soluble Aß(1-42) acutely inhibits LTP and chronically causes synapse loss. Many mechanisms have been proposed for Aß-induced synaptic dysfunction, but we recently found that Aß(1-42) inhibits the microtubule motor protein Eg5/kinesin-5. Here we compared the impacts of Aß(1-42) and monastrol, a small-molecule Eg5 inhibitor, on LTP in hippocampal slices and synapse loss in neuronal cultures. Acute (20-minute) treatment with monastrol, like Aß, completely inhibited LTP at doses >100 nM. In addition, 1 nM Aß(1-42) or 50 nM monastrol inhibited LTP #x223c;50%, and when applied together caused complete LTP inhibition. At concentrations that impaired LTP, neither Aß(1-42) nor monastrol inhibited NMDAR synaptic responses until #x223c;60 minutes, when only #x223c;25% inhibition was seen for monastrol, indicating that NMDAR inhibition was not responsible for LTP inhibition by either agent when applied for only 20 minutes. Finally, 48 hours of treatment with either 0.5-1.0µM Aß(1-42) or 1-5µM monastrol reduced the dendritic spine/synapse density in hippocampal cultures up to a maximum of #x223c;40%, and when applied together at maximal concentrations, no additional spine loss resulted. Thus, monastrol can mimic and in some cases occlude the impact of Aßon LTP and synapse loss, suggesting that Aßinduces acute and chronic synaptic dysfunction in part through inhibiting Eg5.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Amiloide/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Hipocampo/efectos de los fármacos , Cinesinas/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Amiloide/metabolismo , Animales , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Técnicas In Vitro , Cinesinas/metabolismo , Cinética , Potenciación a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Pirimidinas/toxicidad , Tionas/toxicidad
4.
J Neurosci ; 32(43): 15036-52, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-23100425

RESUMEN

AMPA receptors (AMPARs) are tetrameric ion channels assembled from GluA1-GluA4 subunits that mediate the majority of fast excitatory synaptic transmission in the brain. In the hippocampus, most synaptic AMPARs are composed of GluA1/2 or GluA2/3 with the GluA2 subunit preventing Ca(2+) influx. However, a small number of Ca(2+)-permeable GluA1 homomeric receptors reside in extrasynaptic locations where they can be rapidly recruited to synapses during synaptic plasticity. Phosphorylation of GluA1 S845 by the cAMP-dependent protein kinase (PKA) primes extrasynaptic receptors for synaptic insertion in response to NMDA receptor Ca(2+) signaling during long-term potentiation (LTP), while phosphatases dephosphorylate S845 and remove synaptic and extrasynaptic GluA1 during long-term depression (LTD). PKA and the Ca(2+)-activated phosphatase calcineurin (CaN) are targeted to GluA1 through binding to A-kinase anchoring protein 150 (AKAP150) in a complex with PSD-95, but we do not understand how the opposing activities of these enzymes are balanced to control plasticity. Here, we generated AKAP150ΔPIX knock-in mice to selectively disrupt CaN anchoring in vivo. We found that AKAP150ΔPIX mice lack LTD but express enhanced LTP at CA1 synapses. Accordingly, basal GluA1 S845 phosphorylation is elevated in AKAP150ΔPIX hippocampus, and LTD-induced dephosphorylation and removal of GluA1, AKAP150, and PSD-95 from synapses are impaired. In addition, basal synaptic activity of GluA2-lacking AMPARs is increased in AKAP150ΔPIX mice and pharmacologic antagonism of these receptors restores normal LTD and inhibits the enhanced LTP. Thus, AKAP150-anchored CaN opposes PKA phosphorylation of GluA1 to restrict synaptic incorporation of Ca(2+)-permeable AMPARs both basally and during LTP and LTD.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Calcineurina/metabolismo , Calcio/metabolismo , Plasticidad Neuronal/fisiología , Receptores AMPA/metabolismo , Sinapsis/fisiología , Potenciales de Acción/genética , Análisis de Varianza , Animales , Biofisica , Calcineurina/genética , Células Cultivadas , Espinas Dendríticas/ultraestructura , Homólogo 4 de la Proteína Discs Large , Estimulación Eléctrica , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Inmunoprecipitación , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , N-Metilaspartato/farmacología , Plasticidad Neuronal/genética , Neuronas/ultraestructura , Técnicas de Placa-Clamp , Fosforilación , Quinoxalinas/farmacología , Serina/metabolismo , Tinción con Nitrato de Plata , Bloqueadores de los Canales de Sodio/farmacología , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura , Sinapsis/ultraestructura , Tetrodotoxina/farmacología
5.
J Neurosci ; 32(21): 7119-36, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22623657

RESUMEN

NMDA receptor-dependent long-term potentiation (LTP) and depression (LTD) are forms of synaptic plasticity underlying learning and memory that are expressed through increases and decreases, respectively, in dendritic spine size and AMPA receptor (AMPAR) phosphorylation and postsynaptic localization. The A-kinase anchoring protein 79/150 (AKAP79/150) signaling scaffold regulates AMPAR phosphorylation, channel activity, and endosomal trafficking associated with LTP and LTD. AKAP79/150 is targeted to dendritic spine plasma membranes by an N-terminal polybasic domain that binds phosphoinositide lipids, F-actin, and cadherin cell adhesion molecules. However, we do not understand how regulation of AKAP targeting controls AMPAR endosomal trafficking. Here, we report that palmitoylation of the AKAP N-terminal polybasic domain targets it to postsynaptic lipid rafts and dendritic recycling endosomes. AKAP palmitoylation was regulated by seizure activity in vivo and LTP/LTD plasticity-inducing stimuli in cultured rat hippocampal neurons. With chemical LTP induction, we observed AKAP79 dendritic spine recruitment that required palmityolation and Rab11-regulated endosome recycling coincident with spine enlargement and AMPAR surface delivery. Importantly, a palmitoylation-deficient AKAP79 mutant impaired regulation of spine size, endosome recycling, AMPAR trafficking, and synaptic potentiation. These findings emphasize the emerging importance of palmitoylation in controlling synaptic function and reveal novel roles for the AKAP79/150 signaling complex in dendritic endosomes.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/fisiología , Dendritas/metabolismo , Endosomas/metabolismo , Plasticidad Neuronal/fisiología , Transporte de Proteínas/fisiología , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Espinas Dendríticas/ultraestructura , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/metabolismo , Hipocampo/fisiología , Ácido Kaínico/farmacología , Lipoilación/fisiología , Potenciación a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores AMPA/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/fisiopatología
6.
J Neurosci ; 29(24): 7929-43, 2009 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-19535604

RESUMEN

A-kinase anchoring protein (AKAP) 79/150 is a scaffold protein found in dendritic spines that recruits the cAMP-dependent protein kinase (PKA) and protein phosphatase 2B-calcineurin (CaN) to membrane-associated guanylate kinase (MAGUK)-linked AMPA receptors (AMPARs) to control receptor phosphorylation and synaptic plasticity. However, AKAP79/150 may also coordinate regulation of AMPAR activity with spine structure directly through MAGUK binding and membrane-cytoskeletal interactions of its N-terminal targeting domain. In cultured hippocampal neurons, we observed that rat AKAP150 expression was low early in development but then increased coincident with spine formation and maturation. Overexpression of human AKAP79 in immature or mature neurons increased the number of dendritic filopodia and spines and enlarged spine area. However, RNA interference knockdown of AKAP150 decreased dendritic spine area only in mature neurons. Importantly, AKAP79 overexpression in immature neurons increased AMPAR postsynaptic localization and activity. Neither the AKAP79 PKA nor CaN anchoring domain was required for increasing dendritic protrusion numbers, spine area, or AMPAR synaptic localization; however, an internal region identified as the MAGUK binding domain was found to be essential as shown by expression of a MAGUK binding mutant that formed mainly filopodia and decreased AMPAR synaptic localization and activity. Expression of the AKAP79 N-terminal targeting domain alone also increased filopodia numbers but not spine area. Overall, these results demonstrate a novel structural role for AKAP79/150 in which the N-terminal targeting domain induces dendritic filopodia and binding to MAGUKs promotes spine enlargement and AMPAR recruitment.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Guanilato-Quinasas/metabolismo , Neuronas/citología , Terminales Presinápticos/fisiología , Sinapsis/fisiología , Proteínas de Anclaje a la Quinasa A/genética , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Calcineurina/genética , Calcineurina/metabolismo , Células Cultivadas , Espinas Dendríticas/metabolismo , Homólogo 4 de la Proteína Discs Large , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Regulación del Desarrollo de la Expresión Génica/genética , Guanilato-Quinasas/genética , Hipocampo/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Luminiscentes/genética , Proteínas de la Membrana/genética , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/genética , Mutación/fisiología , Neuronas/fisiología , Terminales Presinápticos/efectos de los fármacos , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Interferencia de ARN/fisiología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores AMPA/metabolismo , Sinapsis/genética , Factores de Tiempo , Transfección/métodos
7.
J Pharmacol Exp Ther ; 332(3): 720-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20008487

RESUMEN

Alterations in N-methyl-d-aspartate receptor (NMDAR) protein levels or subcellular localization in brain after chronic ethanol exposure may contribute to withdrawal-associated seizures and neurotoxicity. We have investigated synaptic localization of NMDARs in cultured hippocampal pyramidal neurons after prolonged (7 days) exposure to, and acute withdrawal from, 80 mM ethanol using fluorescence immunocytochemistry techniques. After chronic ethanol exposure, there was a significant increase in the clustering of NR1 and NR2B subunits and their colocalization with the synaptic proteins synaptophysin and postsynaptic density protein 95, respectively. There was also increased expression of NR1 variants containing the C2' cassette after chronic ethanol exposure. The ethanol-induced synaptic clustering and colocalization were rapidly reversed within 4 h after ethanol withdrawal. Surface labeling of NR2B subunits suggested that this rapid reversal involved lateral receptor movement to extrasynaptic sites rather than internalization of receptors. Receptor removal from the synapse during ethanol withdrawal was associated with changes in the phosphorylation state of NR2B Ser1480, controlled by the protein kinase CK2. The redistribution of NMDAR to synapses produced by long-term ethanol exposure, as well as the rapid removal during withdrawal, may not only affect neuronal withdrawal hyperexcitability but also may sensitize the system to subsequent synaptic plasticity.


Asunto(s)
Etanol/farmacología , Células Piramidales/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Sinapsis/efectos de los fármacos , Animales , Animales Recién Nacidos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Fosforilación , Subunidades de Proteína/metabolismo , Transporte de Proteínas , Células Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo
8.
Neuron ; 101(5): 863-875.e6, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30704911

RESUMEN

Regulated secretion is critical for diverse biological processes ranging from immune and endocrine signaling to synaptic transmission. Botulinum and tetanus neurotoxins, which specifically proteolyze vesicle fusion proteins involved in regulated secretion, have been widely used as experimental tools to block these processes. Genetic expression of these toxins in the nervous system has been a powerful approach for disrupting neurotransmitter release within defined circuitry, but their current utility in the brain and elsewhere remains limited by lack of spatial and temporal control. Here we engineered botulinum neurotoxin B so that it can be activated with blue light. We demonstrate the utility of this approach for inducibly disrupting excitatory neurotransmission, providing a first-in-class optogenetic tool for persistent, light-triggered synaptic inhibition. In addition to blocking neurotransmitter release, this approach will have broad utility for conditionally disrupting regulated secretion of diverse bioactive molecules, including neuropeptides, neuromodulators, hormones, and immune molecules. VIDEO ABSTRACT.


Asunto(s)
Toxinas Botulínicas/farmacología , Optogenética/métodos , Transmisión Sináptica/efectos de los fármacos , Animales , Proteínas de Arabidopsis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Toxinas Botulínicas/genética , Toxinas Botulínicas/efectos de la radiación , Caenorhabditis elegans , Células Cultivadas , Criptocromos/genética , Femenino , Células HEK293 , Humanos , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/efectos de la radiación , Proteínas SNARE/metabolismo , Sinapsis/metabolismo , Sinapsis/fisiología , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
9.
J Neurosci ; 26(9): 2391-402, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510716

RESUMEN

NMDA receptor-dependent long-term potentiation and long-term depression (LTD) involve changes in AMPA receptor activity and postsynaptic localization that are in part controlled by glutamate receptor 1 (GluR1) subunit phosphorylation. The scaffolding molecule A-kinase anchoring protein (AKAP)79/150 targets both the cAMP-dependent protein kinase (PKA) and protein phosphatase 2B/calcineurin (PP2B/CaN) to AMPA receptors to regulate GluR1 phosphorylation. Here, we report that brief NMDA receptor activation leads to persistent redistribution of AKAP79/150 and PKA-RII, but not PP2B/CaN, from postsynaptic membranes to the cytoplasm in hippocampal slices. Similar to LTD, AKAP79/150 redistribution requires PP2B/CaN activation and is accompanied by GluR1 dephosphorylation and internalization. Using fluorescence resonance energy transfer microscopy in hippocampal neurons, we demonstrate that PKA anchoring to AKAP79/150 is required for NMDA receptor regulation of PKA-RII localization and that movement of AKAP-PKA complexes underlies PKA redistribution. These findings suggest that LTD involves removal of AKAP79/150 and PKA from synapses in addition to activation of PP2B/CaN. Movement of AKAP79/150-PKA complexes from the synapse could further favor the actions of phosphatases in maintaining dephosphorylation of postsynaptic substrates, such as GluR1, that are important for LTD induction and expression. In addition, our observations demonstrate that AKAPs serve not solely as stationary anchors in cells but also as dynamic signaling components.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Neuronas/citología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Proteínas de Anclaje a la Quinasa A , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Proteínas Bacterianas/metabolismo , Western Blotting/métodos , Calcineurina/metabolismo , Células Cultivadas , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Dendritas/metabolismo , Homólogo 4 de la Proteína Discs Large , Interacciones Farmacológicas , Activación Enzimática , Agonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/efectos de la radiación , Proteínas Luminiscentes/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Mutagénesis , N-Metilaspartato/farmacología , Neuronas/fisiología , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Sinapsis/efectos de los fármacos , Factores de Tiempo , Transfección/métodos , Valina/análogos & derivados , Valina/farmacología
10.
Biotechnol Biofuels ; 10: 29, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28184244

RESUMEN

BACKGROUND: Solid-binding peptides (SBPs) bind strongly to a diverse range of solid materials without the need for any chemical reactions. They have been used mainly for the functionalisation of nanomaterials but little is known about their use for the immobilisation of thermostable enzymes and their feasibility in industrial-scale biocatalysis. RESULTS: A silica-binding SBP sequence was fused genetically to three thermostable hemicellulases. The resulting enzymes were active after fusion and exhibited identical pH and temperature optima but differing thermostabilities when compared to their corresponding unmodified enzymes. The silica-binding peptide mediated the efficient immobilisation of each enzyme onto zeolite, demonstrating the construction of single enzyme biocatalytic modules. Cross-linked enzyme aggregates (CLEAs) of enzyme preparations either with or without zeolite immobilisation displayed greater activity retention during enzyme recycling than those of free enzymes (without silica-binding peptide) or zeolite-bound enzymes without any crosslinking. CLEA preparations comprising all three enzymes simultaneously immobilised onto zeolite enabled the formation of multiple enzyme biocatalytic modules which were shown to degrade several hemicellulosic substrates. CONCLUSIONS: The current work introduced the construction of functional biocatalytic modules for the hydrolysis of simple and complex polysaccharides. This technology exploited a silica-binding SBP to mediate effectively the rapid and simple immobilisation of thermostable enzymes onto readily-available and inexpensive silica-based matrices. A conceptual application of biocatalytic modules consisting of single or multiple enzymes was validated by hydrolysing various hemicellulosic polysaccharides.

11.
Neuron ; 93(3): 646-660.e5, 2017 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-28132827

RESUMEN

The molecular composition of the postsynaptic membrane is sculpted by synaptic activity. During synaptic plasticity at excitatory synapses, numerous structural, signaling, and receptor molecules concentrate at the postsynaptic density (PSD) to regulate synaptic strength. We developed an approach that uses light to tune the abundance of specific molecules in the PSD. We used this approach to investigate the relationship between the number of AMPA-type glutamate receptors in the PSD and synaptic strength. Surprisingly, adding more AMPA receptors to excitatory contacts had little effect on synaptic strength. Instead, we observed increased excitatory input through the apparent addition of new functional sites. Our data support a model where adding AMPA receptors is sufficient to activate synapses that had few receptors to begin with, but that additional remodeling events are required to strengthen established synapses. More broadly, this approach introduces the precise spatiotemporal control of optogenetics to the molecular control of synaptic function.


Asunto(s)
Plasticidad Neuronal/genética , Neuronas/metabolismo , Optogenética/métodos , Densidad Postsináptica/metabolismo , Receptores AMPA/genética , Sinapsis/metabolismo , Membranas Sinápticas/metabolismo , Animales , Proteínas de Arabidopsis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Criptocromos/genética , Hipocampo/citología , Potenciación a Largo Plazo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Sinapsis/fisiología
12.
Cell Rep ; 21(8): 2134-2146, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166605

RESUMEN

The repertoire and abundance of proteins displayed on the surface of neuronal dendrites are tuned by regulated fusion of recycling endosomes (REs) with the dendritic plasma membrane. While this process is critical for neuronal function and plasticity, how synaptic activity drives RE fusion remains unexplored. We demonstrate a multistep fusion mechanism that requires Ca2+ from distinct sources. NMDA receptor Ca2+ initiates RE fusion with the plasma membrane, while L-type voltage-gated Ca2+ channels (L-VGCCs) regulate whether fused REs collapse into the membrane or reform without transferring their cargo to the cell surface. Accordingly, NMDA receptor activation triggered AMPA-type glutamate receptor trafficking to the dendritic surface in an L-VGCC-dependent manner. Conversely, potentiating L-VGCCs enhanced AMPA receptor surface expression only when NMDA receptors were also active. Thus L-VGCCs play a role in tuning activity-triggered surface expression of key synaptic proteins by gating the mode of RE fusion.


Asunto(s)
Dendritas/metabolismo , Potenciación a Largo Plazo/fisiología , Animales , Células Cultivadas , Endosomas/metabolismo , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
13.
Eur J Cell Biol ; 85(7): 627-33, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16504338

RESUMEN

Central to organization of signaling pathways are scaffolding, anchoring and adaptor proteins that mediate localized assembly of multi-protein complexes containing receptors, second messenger-generating enzymes, kinases, phosphatases, and substrates. At the postsynaptic density (PSD) of excitatory synapses, AMPA (AMPAR) and NMDA (NMDAR) glutamate receptors are linked to signaling proteins, the actin cytoskeleton, and synaptic adhesion molecules on dendritic spines through a network of scaffolding proteins that may play important roles regulating synaptic structure and receptor functions in synaptic plasticity underlying learning and memory. AMPARs are rapidly recruited to dendritic spines through NMDAR activation during induction of long-term potentiation (LTP) through pathways that also increase the size and F-actin content of spines. Phosphorylation of AMPAR-GluR1 subunits by the cAMP-dependent protein kinase (PKA) helps stabilize AMPARs recruited during LTP. In contrast, induction of long-term depression (LTD) leads to rapid calcineurin-protein phosphatase 2B (CaN) mediated dephosphorylation of PKA-phosphorylated GluR1 receptors, endocytic removal of AMPAR from synapses, and a reduction in spine size. However, mechanisms for coordinately regulating AMPAR localization, phosphorylation, and synaptic structure by PKA and CaN are not well understood. A kinase-anchoring protein (AKAP) 79/150 is a PKA- and CaN-anchoring protein that is linked to NMDARs and AMPARs through PSD-95 and SAP97 membrane-associated guanylate kinase (MAGUK) scaffolds. Importantly, disruption of PKA-anchoring in neurons and functional analysis of GluR1-MAGUK-AKAP79 complexes in heterologous cells suggests that AKAP79/150-anchored PKA and CaN may regulate AMPARs in LTD. In the work presented at the "First International Meeting on Anchored cAMP Signaling Pathways" (Berlin-Buch, Germany, October 15-16, 2005), we demonstrate that AKAP79/150 is targeted to dendritic spines by an N-terminal basic region that binds phosphatidylinositol-4,5-bisphosphate (PIP(2)), F-actin, and actin-linked cadherin adhesion molecules. Thus, anchoring of PKA and CaN as well as physical linkage of the AKAP to both cadherin-cytoskeletal and MAGUK-receptor complexes could play roles in coordinating changes in synaptic structure and receptor signaling functions underlying plasticity. Importantly, we provide evidence showing that NMDAR-CaN signaling pathways implicated in AMPAR regulation during LTD lead to a disruption of AKAP79/150 interactions with actin, MAGUKs, and cadherins and lead to a loss of the AKAP and anchored PKA from postsynapses. Our studies thus far indicate that this AKAP79/150 translocation depends on activation of CaN, F-actin reorganization, and possibly Ca(2+)-CaM binding to the N-terminal basic regions. Importantly, this tranlocation of the AKAP79/150-PKA complex from spines may shift the balance of PKA kinase and CaN/PP1 phosphatase activity at the postsynapse in favor of the phosphatases. This loss of PKA could then promote actions of CaN and PP1 during induction of LTD including maintaining AMPAR dephosphorylation, promoting AMPAR endocytosis, and preventing AMPAR recycling. Overall, these findings challenge the accepted notion that AKAPs are static anchors that position signaling proteins near fixed target substrates and instead suggest that AKAPs can function in more dynamic manners to regulate local signaling events.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neuronas/metabolismo , Transducción de Señal , Proteínas de Anclaje a la Quinasa A , Actinas/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Humanos , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal , Neurotoxinas/efectos adversos , Proteínas Asociadas a Matriz Nuclear/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Distribución Tisular
14.
J Cell Biol ; 201(4): 631-40, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23671313

RESUMEN

Optical control of protein interactions has emerged as a powerful experimental paradigm for manipulating and studying various cellular processes. Tools are now available for controlling a number of cellular functions, but some fundamental processes, such as protein secretion, have been difficult to engineer using current optical tools. Here we use UVR8, a plant photoreceptor protein that forms photolabile homodimers, to engineer the first light-triggered protein secretion system. UVR8 fusion proteins were conditionally sequestered in the endoplasmic reticulum, and a brief pulse of light triggered robust forward trafficking through the secretory pathway to the plasma membrane. UVR8 was not responsive to excitation light used to image cyan, green, or red fluorescent protein variants, allowing multicolor visualization of cellular markers and secreted protein cargo as it traverses the cellular secretory pathway. We implemented this novel tool in neurons to demonstrate restricted, local trafficking of secretory cargo near dendritic branch points.


Asunto(s)
Luz , Óptica y Fotónica/métodos , Proteínas/metabolismo , Animales , Animales Recién Nacidos , Proteínas de Arabidopsis/metabolismo , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Proteínas Cromosómicas no Histona/metabolismo , Dendritas/metabolismo , Genes de Plantas , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hipocampo/citología , Humanos , Microscopía Fluorescente , Neuronas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Transporte de Proteínas/efectos de la radiación , Ratas , Ratas Sprague-Dawley , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA