Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nat Rev Neurosci ; 22(4): 237-255, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33712727

RESUMEN

The brain is arguably the most complex organ. The branched and extended morphology of nerve cells, their subcellular complexity, the multiplicity of brain cell types as well as their intricate connectivity and the scattering properties of brain tissue present formidable challenges to the understanding of brain function. Neuroscientists have often been at the forefront of technological and methodological developments to overcome these hurdles to visualize, quantify and modify cell and network properties. Over the last few decades, the development of advanced imaging methods has revolutionized our approach to explore the brain. Super-resolution microscopy and tissue imaging approaches have recently exploded. These instrumentation-based innovations have occurred in parallel with the development of new molecular approaches to label protein targets, to evolve new biosensors and to target them to appropriate cell types or subcellular compartments. We review the latest developments for labelling and functionalizing proteins with small localization and functionalized reporters. We present how these molecular tools are combined with the development of a wide variety of imaging methods that break either the diffraction barrier or the tissue penetration depth limits. We put these developments in perspective to emphasize how they will enable step changes in our understanding of the brain.


Asunto(s)
Encéfalo/citología , Microscopía/métodos , Neuroglía/citología , Neuronas/citología , Coloración y Etiquetado/métodos , Animales , Humanos
2.
Biol Cell ; 113(12): 492-506, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34498765

RESUMEN

Leucine Rich Repeat Transmembrane proteins (LRRTMs) are neuronal cell adhesion molecules involved in synapse development and plasticity. LRRTM2 is the most synaptogenic isoform of the family, and its expression is strongly restricted to excitatory synapses in mature neurons. However, the mechanisms by which LRRTM2 is trafficked and stabilized at synapses remain unknown. Here, we examine the role of LRRTM2 intracellular domain on its membrane expression and stabilization at excitatory synapses, using a knock-down strategy combined to single molecule tracking and super-resolution dSTORM microscopy. We show that LRRTM2 operates an important shift in mobility after synaptogenesis in hippocampal neurons. Knock-down of LRRTM2 during synapse formation reduced excitatory synapse density in mature neurons. Deletion of LRRTM2 C-terminal domain abolished the compartmentalization of LRRTM2 in dendrites and disrupted its synaptic enrichment. Furtheremore, we show that LRRTM2 diffusion is increased in the absence of its intracellular domain, and that the protein is more dispersed at synapses. Surprisingly, LRRTM2 confinement at synapses was strongly dependent on a YxxC motif in the C-terminal domain, but was independent of the PDZ-like binding motif ECEV. Finally, the nanoscale organization of LRRTM2 at excitatory synapses depended on its C-terminal domain, with involvement of both the PDZ-binding and YxxC motifs. Altogether, these results demonstrate that LRRTM2 trafficking and enrichment at excitatory synapses are dependent on its intracellular domain.


Asunto(s)
Proteínas del Tejido Nervioso , Moléculas de Adhesión de Célula Nerviosa , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Sinapsis
3.
Elife ; 132024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38167295

RESUMEN

Despite the constant advances in fluorescence imaging techniques, monitoring endogenous proteins still constitutes a major challenge in particular when considering dynamics studies or super-resolution imaging. We have recently evolved specific protein-based binders for PSD-95, the main postsynaptic scaffold proteins at excitatory synapses. Since the synthetic recombinant binders recognize epitopes not directly involved in the target protein activity, we consider them here as tools to develop endogenous PSD-95 imaging probes. After confirming their lack of impact on PSD-95 function, we validated their use as intrabody fluorescent probes. We further engineered the probes and demonstrated their usefulness in different super-resolution imaging modalities (STED, PALM, and DNA-PAINT) in both live and fixed neurons. Finally, we exploited the binders to enrich at the synapse genetically encoded calcium reporters. Overall, we demonstrate that these evolved binders constitute a robust and efficient platform to selectively target and monitor endogenous PSD-95 using various fluorescence imaging techniques.


Asunto(s)
Colorantes Fluorescentes , Neuronas , Homólogo 4 de la Proteína Discs Large/genética , Homólogo 4 de la Proteína Discs Large/metabolismo , Neuronas/metabolismo , Colorantes Fluorescentes/metabolismo , Sinapsis/metabolismo
4.
J Am Chem Soc ; 135(12): 4580-3, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23480637

RESUMEN

We report a general method for light-assisted control of interactions of PDZ domain binding motifs with their cognate domains by the incorporation of a photolabile caging group onto the essential C-terminal carboxylate binding determinant of the motif. The strategy was implemented and validated for both simple monovalent and biomimetic divalent ligands, which have recently been established as powerful tools for acute perturbation of native PDZ domain-dependent interactions in live cells.


Asunto(s)
Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Dominios PDZ , Péptidos/química , Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Fluorescencia , Colorantes Fluorescentes/síntesis química , Ligandos , Luz , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/síntesis química , Fotólisis , Unión Proteica , Mapeo de Interacción de Proteínas , Ratas
5.
Nat Chem Biol ; 7(2): 81-91, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186349

RESUMEN

The interactions of the AMPA receptor (AMPAR) auxiliary subunit Stargazin with PDZ domain-containing scaffold proteins such as PSD-95 are critical for the synaptic stabilization of AMPARs. To investigate these interactions, we have developed biomimetic competing ligands that are assembled from two Stargazin-derived PSD-95/DLG/ZO-1 (PDZ) domain-binding motifs using 'click' chemistry. Characterization of the ligands in vitro and in a cellular FRET-based model revealed an enhanced affinity for the multiple PDZ domains of PSD-95 compared to monovalent peptides. In cultured neurons, the divalent ligands competed with transmembrane AMPAR regulatory protein (TARP) for the intracellular membrane-associated guanylate kinase resulting in increased lateral diffusion and endocytosis of surface AMPARs, while showing strong inhibition of synaptic AMPAR currents. This provides evidence for a model in which the TARP-containing AMPARs are stabilized at the synapse by engaging in multivalent interactions. In light of the prevalence of PDZ domain clusters, these new biomimetic chemical tools could find broad application for acutely perturbing multivalent complexes.


Asunto(s)
Biomimética , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Ligandos , Modelos Moleculares
6.
Drug Discov Today Technol ; 10(4): e531-40, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24451645

RESUMEN

Modulating protein-protein interactions constitutes a promising strategy both for the investigation of biological mechanisms and for developing new therapeutic approaches. Among the many types of inter-actions, PDZ domain-mediated interactions (PDMIs) have emerged over the last decade as attractive targets in the drug discovery field. Indeed, these small domains are involved in the regulation of many signaling pathways and possess structural properties which are favorable for the design of competing ligands. Herein, we describe the recent approaches developed to inhibit this class of protein-protein interactions.


Asunto(s)
Descubrimiento de Drogas , Dominios PDZ , Animales , Humanos , Ligandos , Péptidos/farmacología , Mapeo de Interacción de Proteínas
7.
Proc Natl Acad Sci U S A ; 107(45): 19561-6, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974938

RESUMEN

The relative content of NR2 subunits in the NMDA receptor confers specific signaling properties and plasticity to synapses. However, the mechanisms that dynamically govern the retention of synaptic NMDARs, in particular 2A-NMDARs, remain poorly understood. Here, we investigate the dynamic interaction between NR2 C termini and proteins containing PSD-95/Discs-large/ZO-1 homology (PDZ) scaffold proteins at the single molecule level by using high-resolution imaging. We report that a biomimetic divalent competing ligand, mimicking the last 15 amino acids of NR2A C terminus, specifically and efficiently disrupts the interaction between 2A-NMDARs, but not 2B-NMDARs, and PDZ proteins on the time scale of minutes. Furthermore, displacing 2A-NMDARs out of synapses lead to a compensatory increase in synaptic NR2B-NMDARs, providing functional evidence that the anchoring mechanism of 2A- or 2B-NMDARs is different. These data reveal an unexpected role of the NR2 subunit divalent arrangement in providing specific anchoring within synapses, highlighting the need to study such dynamic interactions in native conditions.


Asunto(s)
Dominios PDZ , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/química , Animales , Cinética , Plasticidad Neuronal , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Unión Proteica , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica
8.
Front Synaptic Neurosci ; 14: 835427, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35546899

RESUMEN

Neuroligins (NLGNs) form a family of cell adhesion molecules implicated in synapse development, but the mechanisms that retain these proteins at synapses are still incompletely understood. Recent studies indicate that surface-associated NLGN1 is diffusionally trapped at synapses, where it interacts with quasi-static scaffolding elements of the post-synaptic density. Whereas single molecule tracking reveals rapid diffusion and transient immobilization of NLGN1 at synapses within seconds, fluorescence recovery after photobleaching experiments indicate instead a long-term turnover of NLGN1 at synapse, in the hour time range. To gain insight into the mechanisms supporting NLGN1 anchorage at post-synapses and try to reconcile those experimental paradigms, we quantitatively analyzed here live-cell and super-resolution imaging experiments performed on NLGN1 using a newly released simulator of membrane protein dynamics for fluorescence microscopy, FluoSim. Based on a small set of parameters including diffusion coefficients, binding constants, and photophysical rates, the framework describes fairly well the dynamic behavior of extra-synaptic and synaptic NLGN1 over both short and long time ranges, and provides an estimate of NLGN1 copy numbers in post-synaptic densities at steady-state (around 50 dimers). One striking result is that the residence time of NLGN1 at synapses is much longer than what can be expected from extracellular interactions with pre-synaptic neurexins only, suggesting that NLGN1 is stabilized at synapses through multivalent interactions with intracellular post-synaptic scaffolding proteins.

9.
Elife ; 112022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35532105

RESUMEN

MDGA molecules can bind neuroligins and interfere with trans-synaptic interactions to neurexins, thereby impairing synapse development. However, the subcellular localization and dynamics of MDGAs, or their specific action mode in neurons remain unclear. Here, surface immunostaining of endogenous MDGAs and single molecule tracking of recombinant MDGAs in dissociated hippocampal neurons reveal that MDGAs are homogeneously distributed and exhibit fast membrane diffusion, with a small reduction in mobility across neuronal maturation. Knocking-down/out MDGAs using shRNAs and CRISPR/Cas9 strategies increases the density of excitatory synapses, the membrane confinement of neuroligin-1, and the phosphotyrosine level of neuroligins associated with excitatory post-synaptic differentiation. Finally, MDGA silencing reduces the mobility of AMPA receptors, increases the frequency of miniature EPSCs (but not IPSCs), and selectively enhances evoked AMPA-receptor-mediated EPSCs in CA1 pyramidal neurons. Overall, our results support a mechanism by which interactions between MDGAs and neuroligin-1 delays the assembly of functional excitatory synapses containing AMPA receptors.


Asunto(s)
Proteínas del Tejido Nervioso , Receptores AMPA , Moléculas de Adhesión Celular Neuronal/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores AMPA/genética , Receptores AMPA/metabolismo , Sinapsis/fisiología
10.
Sci Adv ; 8(30): eabm5298, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35895810

RESUMEN

Regulation of synaptic neurotransmitter receptor content is a fundamental mechanism for tuning synaptic efficacy during experience-dependent plasticity and behavioral adaptation. However, experimental approaches to track and modify receptor movements in integrated experimental systems are limited. Exploiting AMPA-type glutamate receptors (AMPARs) as a model, we generated a knock-in mouse expressing the biotin acceptor peptide (AP) tag on the GluA2 extracellular N-terminal. Cell-specific introduction of biotin ligase allows the use of monovalent or tetravalent avidin variants to respectively monitor or manipulate the surface mobility of endogenous AMPAR containing biotinylated AP-GluA2 in neuronal subsets. AMPAR immobilization precluded the expression of long-term potentiation and formation of contextual fear memory, allowing target-specific control of the expression of synaptic plasticity and animal behavior. The AP tag knock-in model offers unprecedented access to resolve and control the spatiotemporal dynamics of endogenous receptors, and opens new avenues to study the molecular mechanisms of synaptic plasticity and learning.

11.
Nat Cell Biol ; 23(11): 1148-1162, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34737443

RESUMEN

Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell migration.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Movimiento Celular , Fibroblastos/metabolismo , Mecanotransducción Celular , Seudópodos/metabolismo , Citoesqueleto de Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/genética , Animales , Línea Celular Transformada , Ratones , Microscopía Fluorescente , Pinzas Ópticas , Imagen Individual de Molécula , Estrés Mecánico , Factores de Tiempo
12.
Neuropharmacology ; 169: 107555, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30831159

RESUMEN

Adhesion proteins play crucial roles at synapses, not only by providing a physical trans-synaptic linkage between axonal and dendritic membranes, but also by connecting to functional elements including the pre-synaptic neurotransmitter release machinery and post-synaptic receptors. To mediate these functions, adhesion proteins must be organized on the neuronal surface in a precise and controlled manner. Recent studies have started to describe the mobility, nanoscale organization, and turnover rate of key synaptic adhesion molecules including cadherins, neurexins, neuroligins, SynCAMs, and LRRTMs, and show that some of these proteins are highly mobile in the plasma membrane while others are confined at sub-synaptic compartments, providing evidence for different regulatory pathways. In this review article, we provide a biophysical view of the diffusional trapping of adhesion molecules at synapses, involving both extracellular and intracellular protein interactions. We review the methodology underlying these measurements, including biomimetic systems with purified adhesion proteins, means to perturb protein expression or function, single molecule imaging in cultured neurons, and analytical models to interpret the data. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.


Asunto(s)
Fenómenos Biofísicos/fisiología , Moléculas de Adhesión Celular Neuronal/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/análisis , Moléculas de Adhesión Celular Neuronal/genética , Humanos , Neuronas/química , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Sinapsis/química , Sinapsis/genética
13.
J Am Chem Soc ; 131(19): 6680-2, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19388649

RESUMEN

A systematic and general approach for identifying efficient probes for class I PDZ domains based on environment-sensitive chromophores is presented. A series of peptides derived from the C-terminal sequence of Stargazin was first used with PDZ domains of PSD-95 and Shank3 to identify the optimal position and linker length for the 4-DMAP chromophore. The results were applied to well-characterized ligand sequences for each set of domains to generate high affinity probes that retain their native sequence specificity and yield remarkable fluorescence increases upon binding. These probes constitute efficient tools to study the dynamics and regulatory mechanisms of PDZ domain-mediated interactions.


Asunto(s)
Colorantes Fluorescentes/aislamiento & purificación , Dominios PDZ , Biblioteca de Péptidos , Péptidos/aislamiento & purificación , 4-Aminopiridina/análogos & derivados , Secuencia de Aminoácidos , Colorantes Fluorescentes/química , Ligandos , Datos de Secuencia Molecular , Péptidos/química
14.
Cell Rep ; 29(5): 1130-1146.e8, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665629

RESUMEN

Tetraspanins are a class of evolutionarily conserved transmembrane proteins with 33 members identified in mammals that have the ability to organize specific membrane domains, named tetraspanin-enriched microdomains (TEMs). Despite the relative abundance of different tetraspanins in the CNS, few studies have explored their role at synapses. Here, we investigate the function of TSPAN5, a member of the tetraspanin superfamily for which mRNA transcripts are found at high levels in the mouse brain. We demonstrate that TSPAN5 is localized in dendritic spines of pyramidal excitatory neurons and that TSPAN5 knockdown induces a dramatic decrease in spine number because of defects in the spine maturation process. Moreover, we show that TSPAN5 interacts with the postsynaptic adhesion molecule neuroligin-1, promoting its correct surface clustering. We propose that membrane compartmentalization by tetraspanins represents an additional mechanism for regulating excitatory synapses.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Espinas Dendríticas/metabolismo , Microdominios de Membrana/metabolismo , Tetraspaninas/química , Tetraspaninas/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Hipocampo/metabolismo , Humanos , Ratones Endogámicos C57BL , Unión Proteica , Células Piramidales/metabolismo , Ratas Wistar , Sinapsis/metabolismo
15.
Nat Commun ; 10(1): 4462, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575863

RESUMEN

During clathrin mediated endocytosis (CME), the concerted action of dynamin and its interacting partners drives membrane scission. Essential interactions occur between the proline/arginine-rich domain of dynamin (dynPRD) and the Src-homology domain 3 (SH3) of various proteins including amphiphysins. Here we show that multiple SH3 domains must bind simultaneously to dynPRD through three adjacent motifs for dynamin's efficient recruitment and function. First, we show that mutant dynamins modified in a single motif, including the central amphiphysin SH3 (amphSH3) binding motif, partially rescue CME in dynamin triple knock-out cells. However, mutating two motifs largely prevents that ability. Furthermore, we designed divalent dynPRD-derived peptides. These ligands bind multimers of amphSH3 with >100-fold higher affinity than monovalent ones in vitro. Accordingly, dialyzing living cells with these divalent peptides through a patch-clamp pipette blocks CME much more effectively than with monovalent ones. We conclude that dynamin drives vesicle scission via multivalent interactions in cells.


Asunto(s)
Dinaminas/química , Dinaminas/metabolismo , Endocitosis/fisiología , Dominios y Motivos de Interacción de Proteínas , Animales , Sitios de Unión , Clatrina/farmacología , Dinaminas/genética , Endocitosis/efectos de los fármacos , Técnicas de Inactivación de Genes , Cinética , Ligandos , Ratones , Células 3T3 NIH , Unión Proteica , Dominios Proteicos , Proteómica , Dominios Homologos src
16.
Nat Commun ; 10(1): 4521, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586061

RESUMEN

Designing highly specific modulators of protein-protein interactions (PPIs) is especially challenging in the context of multiple paralogs and conserved interaction surfaces. In this case, direct generation of selective and competitive inhibitors is hindered by high similarity within the evolutionary-related protein interfaces. We report here a strategy that uses a semi-rational approach to separate the modulator design into two functional parts. We first achieve specificity toward a region outside of the interface by using phage display selection coupled with molecular and cellular validation. Highly selective competition is then generated by appending the more degenerate interaction peptide to contact the target interface. We apply this approach to specifically bind a single PDZ domain within the postsynaptic protein PSD-95 over highly similar PDZ domains in PSD-93, SAP-97 and SAP-102. Our work provides a paralog-selective and domain specific inhibitor of PSD-95, and describes a method to efficiently target other conserved PPI modules.


Asunto(s)
Anticuerpos/química , Dominios PDZ , Péptidos/química , Ingeniería de Proteínas , Mapas de Interacción de Proteínas/efectos de los fármacos , Animales , Anticuerpos/farmacología , Células COS , Chlorocebus aethiops , Homólogo 4 de la Proteína Discs Large/antagonistas & inhibidores , Homólogo 4 de la Proteína Discs Large/metabolismo , Diseño de Fármacos , Mapeo Epitopo , Modelos Moleculares , Biblioteca de Péptidos , Péptidos/farmacología , Unión Proteica , Proteínas Recombinantes/metabolismo
17.
Nat Commun ; 9(1): 3979, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30266896

RESUMEN

To better understand the molecular mechanisms by which early neuronal connections mature into synapses, we examined the impact of neuroligin-1 (Nlg1) phosphorylation on synapse differentiation, focusing on a unique intracellular tyrosine (Y782), which differentially regulates Nlg1 binding to PSD-95 and gephyrin. By expressing Nlg1 point mutants (Y782A/F) in hippocampal neurons, we show using imaging and electrophysiology that Y782 modulates the recruitment of functional AMPA receptors (AMPARs). Nlg1-Y782F impaired both dendritic spine formation and AMPAR diffusional trapping, but not NMDA receptor recruitment, revealing the assembly of silent synapses. Furthermore, replacing endogenous Nlg1 with either Nlg1-Y782A or -Y782F in CA1 hippocampal neurons impaired long-term potentiation (LTP), demonstrating a critical role of AMPAR synaptic retention. Screening of tyrosine kinases combined with pharmacological inhibitors point to Trk family members as major regulators of endogenous Nlg1 phosphorylation and synaptogenic function. Thus, Nlg1 tyrosine phosphorylation signaling is a critical event in excitatory synapse differentiation and LTP.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Potenciación a Largo Plazo/fisiología , Receptores AMPA/metabolismo , Sinapsis/fisiología , Tirosina/metabolismo , Animales , Células COS , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Chlorocebus aethiops , Hipocampo/citología , Potenciación a Largo Plazo/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neuronas/metabolismo , Neuronas/fisiología , Ratas Sprague-Dawley , Sinapsis/metabolismo , Tirosina/genética
18.
Nat Commun ; 9(1): 4272, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30323233

RESUMEN

Impaired hippocampal synaptic plasticity contributes to cognitive impairment in Huntington's disease (HD). However, the molecular basis of such synaptic plasticity defects is not fully understood. Combining live-cell nanoparticle tracking and super-resolution imaging, we show that AMPAR surface diffusion, a key player in synaptic plasticity, is disturbed in various rodent models of HD. We demonstrate that defects in the brain-derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling pathway contribute to the deregulated AMPAR trafficking by reducing the interaction between transmembrane AMPA receptor regulatory proteins (TARPs) and the PDZ-domain scaffold protein PSD95. The disturbed AMPAR surface diffusion is rescued by the antidepressant drug tianeptine via the BDNF signaling pathway. Tianeptine also restores the impaired LTP and hippocampus-dependent memory in different HD mouse models. These findings unravel a mechanism underlying hippocampal synaptic and memory dysfunction in HD, and highlight AMPAR surface diffusion as a promising therapeutic target.


Asunto(s)
Hipocampo/fisiopatología , Enfermedad de Huntington/fisiopatología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Receptores AMPA/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Difusión , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Potenciación a Largo Plazo/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Receptor trkB/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Tiazepinas/farmacología
19.
Elife ; 72018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044218

RESUMEN

The nanoscale organization of neurotransmitter receptors regarding pre-synaptic release sites is a fundamental determinant of the synaptic transmission amplitude and reliability. How modifications in the pre- and post-synaptic machinery alignments affects synaptic currents, has only been addressed with computer modelling. Using single molecule super-resolution microscopy, we found a strong spatial correlation between AMPA receptor (AMPAR) nanodomains and the post-synaptic adhesion protein neuroligin-1 (NLG1). Expression of a truncated form of NLG1 disrupted this correlation without affecting the intrinsic AMPAR organization, shifting the pre-synaptic release machinery away from AMPAR nanodomains. Electrophysiology in dissociated and organotypic hippocampal rodent cultures shows these treatments significantly decrease AMPAR-mediated miniature and EPSC amplitudes. Computer modelling predicts that ~100 nm lateral shift between AMPAR nanoclusters and glutamate release sites induces a significant reduction in AMPAR-mediated currents. Thus, our results suggest the synapses necessity to release glutamate precisely in front of AMPAR nanodomains, to maintain a high synaptic responses efficiency.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Sinapsis/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Potenciales Postsinápticos Excitadores , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neuronas/citología , Ratas , Transmisión Sináptica
20.
Cell Rep ; 23(11): 3137-3145, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29898386

RESUMEN

Alzheimer's disease (AD) is emerging as a synaptopathology driven by metaplasticity. Indeed, reminiscent of metaplasticity, oligomeric forms of the amyloid-ß peptide (oAß) prevent induction of long-term potentiation (LTP) via the prior activation of GluN2B-containing NMDA receptors (NMDARs). However, the downstream Ca2+-dependent signaling molecules that mediate aberrant metaplasticity are unknown. In this study, we show that oAß promotes the activation of Ca2+/calmodulin-dependent kinase II (CaMKII) via GluN2B-containing NMDARs. Importantly, we find that CaMKII inhibition rescues both the LTP impairment and the dendritic spine loss mediated by oAß. Mechanistically resembling metaplasticity, oAß prevents subsequent rounds of plasticity from inducing CaMKII T286 autophosphorylation, as well as the associated anchoring and accumulation of synaptic AMPA receptors (AMPARs). Finally, prolonged oAß treatment-induced CaMKII misactivation leads to dendritic spine loss via the destabilization of surface AMPARs. Thus, our study demonstrates that oAß engages synaptic metaplasticity via aberrant CaMKII activation.


Asunto(s)
Péptidos beta-Amiloides/química , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Fragmentos de Péptidos/química , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Células Cultivadas , Espinas Dendríticas/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/química , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA