Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2831: 209-217, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39134852

RESUMEN

Plasticity of synaptic transmission underlies learning and memory. It is accompanied by changes in the density and size of synapses, collectively called structural plasticity. Therefore, understanding the mechanism of structural plasticity is critical for understanding the mechanism of synaptic plasticity. In this chapter, we describe the procedures and equipment required to image structural plasticity of a single dendritic spine, which hosts excitatory synapses in the central nervous system, and underlying molecular interactions/biochemical reactions using two-photon fluorescence lifetime microscopy (2P-FLIM) in combination with Förster resonance energy transfer (FRET)-based biosensors.


Asunto(s)
Espinas Dendríticas , Transferencia Resonante de Energía de Fluorescencia , Microscopía de Fluorescencia por Excitación Multifotónica , Plasticidad Neuronal , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Espinas Dendríticas/fisiología , Plasticidad Neuronal/fisiología , Animales , Transferencia Resonante de Energía de Fluorescencia/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Sinapsis/metabolismo , Sinapsis/fisiología , Ratones , Técnicas Biosensibles/métodos
2.
PLoS Biol ; 22(3): e3002006, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38452102

RESUMEN

Proteome analyses of the postsynaptic density (PSD), a proteinaceous specialization beneath the postsynaptic membrane of excitatory synapses, have identified several thousands of proteins. While proteins with predictable functions have been well studied, functionally uncharacterized proteins are mostly overlooked. In this study, we conducted a comprehensive meta-analysis of 35 PSD proteome datasets, encompassing a total of 5,869 proteins. Employing a ranking methodology, we identified 97 proteins that remain inadequately characterized. From this selection, we focused our detailed analysis on the highest-ranked protein, FAM81A. FAM81A interacts with PSD proteins, including PSD-95, SynGAP, and NMDA receptors, and promotes liquid-liquid phase separation of those proteins in cultured cells or in vitro. Down-regulation of FAM81A in cultured neurons causes a decrease in the size of PSD-95 puncta and the frequency of neuronal firing. Our findings suggest that FAM81A plays a crucial role in facilitating the interaction and assembly of proteins within the PSD, and its presence is important for maintaining normal synaptic function. Additionally, our methodology underscores the necessity for further characterization of numerous synaptic proteins that still lack comprehensive understanding.


Asunto(s)
Separación de Fases , Proteoma , Proteoma/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Sinapsis/metabolismo , Membranas Sinápticas
3.
Sci Adv ; 9(46): eadh1110, 2023 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-37967196

RESUMEN

Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.


Asunto(s)
Actinas , Hipocampo , Ratones , Animales , Actinas/metabolismo , Hipocampo/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Fosforilación/fisiología , Plasticidad Neuronal/fisiología
4.
Adv Neurobiol ; 33: 333-356, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37615873

RESUMEN

The synapse is a highly specialized asymmetric structure that transmits and stores information in the brain. The size of pre- and postsynaptic structures and function is well coordinated at the individual synapse level. For example, large postsynaptic dendritic spines have a larger postsynaptic density with higher α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) number on their surface, while juxtaposing presynaptic terminals have a larger active zone and higher release probability. This indicates that pre- and postsynaptic domains bidirectionally communicate to coordinate assembly of specific molecules on both sides of the synaptic cleft. Cell adhesion molecules (CAMs) that localize at synapses form transsynaptic protein interactions across the synaptic cleft and play important roles in synapse formation and regulation. The extracellular domain of CAMs is essential for specific synapse formation and function. In contrast, the intracellular domain is necessary for binding with synaptic molecules and signal transduction. Therefore, CAMs play an essential role on synapse function and structure. In fact, ample evidence indicates that transsynaptic CAMs instruct and modulate functions at presynaptic sites. This chapter focuses on transsynaptic protein interactions that regulate presynaptic functions emphasizing the role of neuronal CAMs and the intracellular mechanism of their regulation.


Asunto(s)
Encéfalo , Moléculas de Adhesión Celular , Humanos , Transducción de Señal , Sinapsis
5.
Neurosci Res ; 189: 29-36, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36584924

RESUMEN

After initial formation during learning, memories are further processed in the brain during subsequent days for long-term consolidation, with sleep playing a key role in this process. Studies have shown that neuronal activity patterns during the awake period are repeated in the hippocampus during sleep, which may coordinate brain-wide reactivation leading to memory consolidation. Consistently, perturbation of this activity blocks the formation of long-term memory. This 'replay' of activity during sleep likely triggers plastic changes in synaptic transmission, a cellular substrate of memory, in multiple brain regions, which likely plays a critical role in long-term memory. Two forms of synaptic plasticity, potentiation and depression of synaptic transmission, are induced in parallel during sleep and is termed "offline synaptic plasticity", as opposed to the "online synaptic plasticity" that occurs immediately following a memory event.


Asunto(s)
Consolidación de la Memoria , Consolidación de la Memoria/fisiología , Sueño/fisiología , Plasticidad Neuronal/fisiología , Encéfalo/fisiología , Neuronas/fisiología , Hipocampo/fisiología
6.
Nat Rev Neurosci ; 23(11): 666-682, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36056211

RESUMEN

Calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is the most abundant protein in excitatory synapses and is central to synaptic plasticity, learning and memory. It is activated by intracellular increases in calcium ion levels and triggers molecular processes necessary for synaptic plasticity. CaMKII phosphorylates numerous synaptic proteins, thereby regulating their structure and functions. This leads to molecular events crucial for synaptic plasticity, such as receptor trafficking, localization and activity; actin cytoskeletal dynamics; translation; and even transcription through synapse-nucleus shuttling. Several new tools affording increasingly greater spatiotemporal resolution have revealed the link between CaMKII activity and downstream signalling processes in dendritic spines during synaptic and behavioural plasticity. These technologies have provided insights into the function of CaMKII in learning and memory.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Calmodulina , Humanos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/análisis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/análisis , Calmodulina/metabolismo , Calcio/metabolismo , Actinas/análisis , Actinas/metabolismo , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo , Hipocampo
7.
Cell Rep ; 40(2): 111064, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35830796

RESUMEN

Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a signaling protein required for long-term memory. When activated by Ca2+/CaM, it sustains activity even after the Ca2+ dissipates. In addition to the well-known autophosphorylation-mediated mechanism, interaction with specific binding partners also persistently activates CaMKII. A long-standing model invokes two distinct S and T sites. If an interactor binds at the T-site, then it will preclude autoinhibition and allow substrates to be phosphorylated at the S site. Here, we specifically test this model with X-ray crystallography, molecular dynamics simulations, and biochemistry. Our data are inconsistent with this model. Co-crystal structures of four different activators or substrates show that they all bind to a single continuous site across the kinase domain. We propose a mechanistic model where persistent CaMKII activity is facilitated by high-affinity binding partners that kinetically compete with autoinhibition by the regulatory segment to allow substrate phosphorylation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Procesamiento Proteico-Postraduccional , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dominio Catalítico , Fosforilación
8.
Neurosci Res ; 175: 3-15, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34375719

RESUMEN

Long-term potentiation (LTP) of synaptic transmission is considered to be a cellular counterpart of learning and memory. Activation of postsynaptic NMDA type glutamate receptor (NMDA-R) induces trafficking of AMPA type glutamate receptors (AMPA-R) and other proteins to the synapse in sequential fashion. At the same time, the dendritic spine expands for long-term and modulation of actin underlies this (structural LTP or sLTP). How these changes persist despite constant diffusion and turnover of the component proteins have been the central focus of the current LTP research. Signaling triggered by Ca2+-influx via NMDA-R triggers kinase including Ca2+/calmodulin-dependent protein kinase II (CaMKII). CaMKII can sustain longer-term biochemical signaling by forming a reciprocally-activating kinase-effector complex with its substrate proteins including Tiam1, thereby regulating persistence of the downstream signaling. Furthermore, activated CaMKII can condense at the synapse through the mechanism of liquid-liquid phase separation (LLPS). This increases the binding capacity at the synapse, thereby contributing to the maintenance of enlarged protein complexes. It may also serve as the synapse tag, which captures newly synthesized proteins.


Asunto(s)
Aprendizaje , Potenciación a Largo Plazo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/metabolismo , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Sinapsis/metabolismo
9.
Science ; 374(6569): 857-863, 2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34762472

RESUMEN

Memories are initially encoded in the hippocampus but subsequently consolidated to the cortex. Although synaptic plasticity is key to these processes, its precise spatiotemporal profile remains poorly understood. Using optogenetics to selectively erase long-term potentiation (LTP) within a defined temporal window, we found that distinct phases of synaptic plasticity play differential roles. The first wave acts locally in the hippocampus to confer context specificity. The second wave, during sleep on the same day, organizes these neurons into synchronously firing assemblies. Finally, LTP in the anterior cingulate cortex during sleep on the second day is required for further stabilization of the memory. This demonstrates the precise localization, timing, and characteristic contributions of the plasticity events that underlie the early phase of memory consolidation.


Asunto(s)
Región CA1 Hipocampal/fisiología , Consolidación de la Memoria , Plasticidad Neuronal , Animales , Inactivación por Luz Asistida por Cromóforo , Potenciales Postsinápticos Excitadores , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética , Células Piramidales/fisiología , Ratas , Sueño , Sinapsis/fisiología
10.
Cell Rep ; 35(10): 109229, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107264

RESUMEN

Hippocampal sclerosis, the major neuropathological hallmark of temporal lobe epilepsy, is characterized by different patterns of neuronal loss. The mechanisms of cell-type-specific vulnerability and their progression and histopathological classification remain controversial. Using single-cell electrophysiology in vivo and immediate-early gene expression, we reveal that superficial CA1 pyramidal neurons are overactive in epileptic rodents. Bulk tissue and single-nucleus expression profiling disclose sublayer-specific transcriptomic signatures and robust microglial pro-inflammatory responses. Transcripts regulating neuronal processes such as voltage channels, synaptic signaling, and cell adhesion are deregulated differently by epilepsy across sublayers, whereas neurodegenerative signatures primarily involve superficial cells. Pseudotime analysis of gene expression in single nuclei and in situ validation reveal separated trajectories from health to epilepsy across cell types and identify a subset of superficial cells undergoing a later stage in neurodegeneration. Our findings indicate that sublayer- and cell-type-specific changes associated with selective CA1 neuronal damage contribute to progression of hippocampal sclerosis.


Asunto(s)
Epilepsia/patología , Hipocampo/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/patología , Esclerosis/genética , Animales , Humanos , Ratones
11.
Cell Rep ; 35(7): 109130, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34010643

RESUMEN

Dendritic spines constitute the major compartments of excitatory post-synapses. They undergo activity-dependent enlargement, which is thought to increase the synaptic efficacy underlying learning and memory. The activity-dependent spine enlargement requires activation of signaling pathways leading to promotion of actin polymerization within the spines. However, the molecular machinery that suffices for that structural plasticity remains unclear. Here, we demonstrate that shootin1a links polymerizing actin filaments in spines with the cell-adhesion molecules N-cadherin and L1-CAM, thereby mechanically coupling the filaments to the extracellular environment. Synaptic activation enhances shootin1a-mediated actin-adhesion coupling in spines. Promotion of actin polymerization is insufficient for the plasticity; the enhanced actin-adhesion coupling is required for polymerizing actin filaments to push against the membrane for spine enlargement. By integrating cell signaling, cell adhesion, and force generation into the current model of actin-based machinery, we propose molecular machinery that is sufficient to trigger the activity-dependent spine structural plasticity.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Actinas/metabolismo , Animales , Espinas Dendríticas/metabolismo , Humanos , Ratones , Plasticidad Neuronal
12.
J Neurosci ; 41(24): 5315-5329, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33980545

RESUMEN

Impairment of episodic memory, a class of memory for spatiotemporal context of an event, is an early symptom of Alzheimer's disease. Both spatial and temporal information are encoded and represented in the hippocampal neurons, but how these representations are impaired under amyloid ß (Aß) pathology remains elusive. We performed chronic imaging of the hippocampus in awake male amyloid precursor protein (App) knock-in mice behaving in a virtual reality environment to simultaneously monitor spatiotemporal representations and the progression of Aß depositions. We found that temporal representation is preserved, whereas spatial representation is significantly impaired in the App knock-in mice. This is because of the overall reduction of active place cells, but not time cells, and compensatory hyperactivation of remaining place cells near Aß aggregates. These results indicate the differential impact of Aß aggregates on two major modalities of episodic memory, suggesting different mechanisms for forming and maintaining these two representations in the hippocampus.


Asunto(s)
Enfermedad de Alzheimer/patología , Región CA1 Hipocampal/patología , Trastornos de la Memoria/patología , Neuronas/patología , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Modelos Animales de Enfermedad , Masculino , Memoria Episódica , Ratones
13.
Nat Neurosci ; 24(6): 777-785, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33927400

RESUMEN

Transient information input to the brain leads to persistent changes in synaptic circuits, contributing to the formation of memory engrams. Pre- and postsynaptic structures undergo coordinated functional and structural changes during this process, but how such changes are achieved by their component molecules remains largely unknown. We found that activated CaMKII, a central player of synaptic plasticity, undergoes liquid-liquid phase separation with the NMDA-type glutamate receptor subunit GluN2B. Due to CaMKII autophosphorylation, the condensate stably persists even after Ca2+ is removed. The selective binding of activated CaMKII with GluN2B cosegregates AMPA receptors and the synaptic adhesion molecule neuroligin into a phase-in-phase assembly. In this way, Ca2+-induced liquid-liquid phase separation of CaMKII has the potential to act as an activity-dependent mechanism to crosslink postsynaptic proteins, which may serve as a platform for synaptic reorganization associated with synaptic plasticity.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/análisis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Extracción Líquido-Líquido/métodos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/metabolismo , Secuencia de Aminoácidos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Activación Enzimática/fisiología , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratas , Ratas Sprague-Dawley , Receptores AMPA/análisis , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/análisis , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
14.
Curr Opin Neurobiol ; 69: 84-92, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33752045

RESUMEN

Advances in microscopy techniques have revealed the details of synaptic nanodomains as defined by the segregation of specific molecules on or beneath both presynaptic and postsynaptic membranes. However, it is yet to be clarified how such segregation is accomplished without demarcating membrane and how nanodomains respond to the neuronal activity. It was recently discovered that proteins at the synapse undergo liquid-liquid phase separation (LLPS), which not only contributes to the accumulation of synaptic proteins but also to further segregating the proteins into subdomains by forming phase-in-phase structures. More specifically, CaMKII, a postsynaptic multifunctional kinase that serves as a signaling molecule, acts as a synaptic cross-linker which segregates certain molecules through LLPS in a manner triggered by Ca2+. Nanodomain formation contributes to the establishment of trans-synaptic nanocolumns, which may be involved in the optimization of spatial arrangement of the transmitter release site and receptor, thereby serving as a new mechanism of synaptic plasticity.


Asunto(s)
Plasticidad Neuronal , Sinapsis , Neuronas , Transducción de Señal , Membranas Sinápticas , Transmisión Sináptica
15.
Hippocampus ; 31(3): 235-243, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33452849

RESUMEN

In the hippocampus, spatial and nonspatial information are jointly represented as a neural map in which locations associated with salient features are over-represented by increased densities of relevant place cells. Although we recently demonstrated that experience-dependent establishment of these disproportionate maps is governed by selective stabilization of salient place cells following their conversion from non-place cells, the underlying mechanism for pre-established map reorganization remained to be understood. To this end, we investigated the changes in CA1 functional cellular maps imaged using two-photon calcium imaging in mice performing a reward-rearrangement task in virtual reality. Mice were pre-trained on a virtual linear track with a visual landmark and a reward in two distinct locations. Then, they were re-trained on the same track with the exception that the location of reward was shifted to match the landmark location. We found that, in contrast to de novo map formation, robust map reorganization occurred through parallel coordination of new place field formation, lateral shifting of existing place fields, and selective stabilization of place fields encoding salient locations. Our findings demonstrate that intricate interplay between multiple forms of cellular dynamics enables rapid updating of information stored in hippocampal maps.


Asunto(s)
Región CA1 Hipocampal , Células de Lugar , Animales , Hipocampo , Ratones , Recompensa
16.
J Neurosci ; 41(5): 834-844, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33472825

RESUMEN

Molecules within cells are segregated into functional domains to form various organelles. While some of those organelles are delimited by lipid membranes demarcating their constituents, others lack a membrane enclosure. Recently, liquid-liquid phase separation (LLPS) revolutionized our view of how segregation of macromolecules can produce membraneless organelles. While the concept of LLPS has been well studied in the areas of soft matter physics and polymer chemistry, its significance has only recently been recognized in the field of biology. It occurs typically between macromolecules that have multivalent interactions. Interestingly, these features are present in many molecules that exert key functions within neurons. In this review, we cover recent topics of LLPS in different contexts of neuronal physiology and pathology.


Asunto(s)
Extracción Líquido-Líquido/métodos , Enfermedades del Sistema Nervioso/fisiopatología , Neuronas/fisiología , Orgánulos/fisiología , Animales , Humanos
17.
FEBS J ; 288(9): 2930-2955, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33175445

RESUMEN

Activity-regulated cytoskeleton-associated protein (Arc) is a protein interaction hub with diverse roles in intracellular neuronal signaling, and important functions in neuronal synaptic plasticity, memory, and postnatal cortical development. Arc has homology to retroviral Gag protein and is capable of self-assembly into virus-like capsids implicated in the intercellular transfer of RNA. However, the molecular basis of Arc self-association and capsid formation is largely unknown. Here, we identified a 28-amino-acid stretch in the mammalian Arc N-terminal (NT) domain that is necessary and sufficient for self-association. Within this region, we identified a 7-residue oligomerization motif, critical for the formation of virus-like capsids. Purified wild-type Arc formed capsids as shown by transmission and cryo-electron microscopy, whereas mutant Arc with disruption of the oligomerization motif formed homogenous dimers. An atomic-resolution crystal structure of the oligomerization region peptide demonstrated an antiparallel coiled-coil interface, strongly supporting NT-NT domain interactions in Arc oligomerization. The NT coil-coil interaction was also validated in live neurons using fluorescence lifetime FRET imaging, and mutation of the oligomerization motif disrupted Arc-facilitated endocytosis. Furthermore, using single-molecule photobleaching, we show that Arc mRNA greatly enhances higher-order oligomerization in a manner dependent on the oligomerization motif. In conclusion, a helical coil in the Arc NT domain supports self-association above the dimer stage, mRNA-induced oligomerization, and formation of virus-like capsids. DATABASE: The coordinates and structure factors for crystallographic analysis of the oligomerization region were deposited at the Protein Data Bank with the entry code 6YTU.


Asunto(s)
Secuencias de Aminoácidos/genética , Proteínas del Citoesqueleto/ultraestructura , Proteínas de Drosophila/genética , Proteínas del Tejido Nervioso/ultraestructura , Neuronas/metabolismo , Conformación Proteica , Animales , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Cristalografía por Rayos X , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/ultraestructura , Humanos , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Dominios Proteicos/genética , ARN/genética , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Virión/genética
18.
Cell Rep ; 32(1): 107864, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640229

RESUMEN

In the hippocampus, locations associated with salient features are represented by a disproportionately large number of neurons, but the cellular and molecular mechanisms underlying this over-representation remain elusive. Using longitudinal calcium imaging in mice learning to navigate in virtual reality, we find that the over-representation of reward and landmark locations are mediated by persistent and separable subsets of neurons, with distinct time courses of emergence and differing underlying molecular mechanisms. Strikingly, we find that in mice lacking Shank2, an autism spectrum disorder (ASD)-linked gene encoding an excitatory postsynaptic scaffold protein, the learning-induced over-representation of landmarks was absent whereas the over-representation of rewards was substantially increased, as was goal-directed behavior. These findings demonstrate that multiple hippocampal coding processes for unique types of salient features are distinguished by a Shank2-dependent mechanism and suggest that abnormally distorted hippocampal salience mapping may underlie cognitive and behavioral abnormalities in a subset of ASDs.


Asunto(s)
Puntos Anatómicos de Referencia , Hipocampo/anatomía & histología , Animales , Conducta Animal , Cognición , Femenino , Objetivos , Hipocampo/citología , Masculino , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Recompensa , Análisis y Desempeño de Tareas , Factores de Tiempo
19.
J Neurosci ; 40(25): 4936-4944, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32414785

RESUMEN

Hippocampus receives dense serotonergic input specifically from raphe nuclei. However, what information is carried by this input and its impact on behavior has not been fully elucidated. Here we used in vivo two-photon imaging of activity of hippocampal median raphe projection fibers in behaving male and female mice and identified two distinct populations: one linked to reward delivery and the other to locomotion. Local optogenetic manipulation of these fibers confirmed a functional role for these projections in the modulation of reward-induced behavior. The diverse function of serotonergic inputs suggests a key role in integrating locomotion and reward information into the hippocampal CA1.SIGNIFICANCE STATEMENT Information constantly flows in the hippocampus, but only some of it is captured as a memory. One potential process that discriminates which information should be remembered is concomitance with reward. In this work, we report a neuromodulatory pathway, which delivers reward signal as well as locomotion signal to the hippocampal CA1. We found that the serotonergic system delivers heterogeneous input that may be integrated by the hippocampus to support its mnemonic functions. It is dynamically involved in regulating behavior through interaction with the hippocampus. Our results suggest that the serotonergic system interacts with the hippocampus in a dynamic and behaviorally specific manner to regulate reward-related information processing.


Asunto(s)
Conducta Animal/fisiología , Hipocampo/fisiología , Locomoción/fisiología , Vías Nerviosas/fisiología , Recompensa , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Serotoninérgicas/fisiología
20.
Structure ; 28(3): 290-300.e4, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-31879129

RESUMEN

Shank1/2/3, major scaffold proteins in excitatory synapses, are frequently mutated in patients with psychiatric disorders. Although the Shank N-terminal domain and ankyrin repeats domain tandem (NTD-ANK) is known to bind to Ras and Rap1, the molecular mechanism underlying and functional significance of the bindings in synapses are unknown. Here, we demonstrate that Shank3 NTD-ANK specifically binds to the guanosine triphosphate (GTP)-bound form of HRas and Rap1. In addition to the canonical site mediated by the Ras-association domain and common to both GTPases, Shank3 contains an unconventional Rap1 binding site formed by NTD and ANK together. Binding of Shank3 to the GTP-loaded Rap1 slows down its GTP hydrolysis by SynGAP. We further show that the interactions between Shank3 and HRas/Rap1 at excitatory synapses are promoted by synaptic activation. Thus, Shank3 may be able to modulate signaling of the Ras family proteins via directly binding to and stabilizing the GTP-bound form of the enzymes.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Estabilidad de Enzimas , Proteínas Activadoras de GTPasa/química , Humanos , Hidrólisis , Unión Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Activadoras de ras GTPasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...