Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 219
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Rev Neurosci ; 25(2): 131-139, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38172626

RESUMEN

Synapses are a key component of neural circuits, facilitating rapid and specific signalling between neurons. Synaptic engineering - the synthetic insertion of new synaptic connections into in vivo neural circuits - is an emerging approach for neural circuit interrogation. This approach is especially powerful for establishing causality in neural circuit structure-function relationships, for emulating synaptic plasticity and for exploring novel patterns of circuit connectivity. Contrary to other approaches for neural circuit manipulation, synaptic engineering targets specific connections between neurons and functions autonomously with no user-controlled external activation. Synaptic engineering has been successfully implemented in several systems and in different forms, including electrical synapses constructed from ectopically expressed connexin gap junction proteins, synthetic optical synapses composed of presynaptic photon-emitting luciferase coupled with postsynaptic light-gated channels, and artificial neuropeptide signalling pathways. This Perspective describes these different methods and how they have been applied, and examines how the field may advance.


Asunto(s)
Sinapsis Eléctricas , Sinapsis , Humanos , Sinapsis/fisiología , Sinapsis Eléctricas/fisiología , Neuronas/fisiología , Sistema Nervioso , Transducción de Señal , Plasticidad Neuronal/fisiología
2.
eNeuro ; 11(1)2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38164593

RESUMEN

The thalamic reticular nucleus (TRN) inhibits sensory thalamocortical relay neurons and is a key regulator of sensory attention as well as sleep and wake states. Recent developments have identified two distinct genetic subtypes of TRN neurons, calbindin-expressing (CB) and somatostatin-expressing (SOM) neurons. These subtypes differ in localization within the TRN, electrophysiological properties, and importantly, targeting of thalamocortical relay channels. CB neurons send inhibition to and receive excitation from first-order thalamic relay nuclei, while SOM neurons send inhibition to and receive excitation from higher-order thalamic areas. These differences create distinct channels of information flow. It is unknown whether TRN neurons form electrical synapses between SOM and CB neurons and consequently bridge first-order and higher-order thalamic channels. Here, we use GFP reporter mice to label and record from CB-expressing and SOM-expressing TRN neurons. We confirm that GFP expression properly differentiates TRN subtypes based on electrophysiological differences, and we identified electrical synapses between pairs of neurons with and without common GFP expression for both CB and SOM types. That is, electrical synapses link both within and across subtypes of neurons in the TRN, forming either homocellular or heterocellular synapses. Therefore, we conclude that electrical synapses within the TRN provide a substrate for functionally linking thalamocortical first-order and higher-order channels within the TRN.


Asunto(s)
Sinapsis Eléctricas , Núcleos Talámicos , Ratones , Animales , Sinapsis Eléctricas/fisiología , Potenciales de Acción/fisiología , Núcleos Talámicos/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Tálamo
3.
PLoS Genet ; 19(11): e1011045, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38011265

RESUMEN

Electrical synapses are neuronal gap junction (GJ) channels associated with a macromolecular complex called the electrical synapse density (ESD), which regulates development and dynamically modifies electrical transmission. However, the proteomic makeup and molecular mechanisms utilized by the ESD that direct electrical synapse formation are not well understood. Using the Mauthner cell of zebrafish as a model, we previously found that the intracellular scaffolding protein ZO1b is a member of the ESD, localizing postsynaptically, where it is required for GJ channel localization, electrical communication, neural network function, and behavior. Here, we show that the complexity of the ESD is further diversified by the genomic structure of the ZO1b gene locus. The ZO1b gene is alternatively initiated at three transcriptional start sites resulting in isoforms with unique N-termini that we call ZO1b-Alpha, -Beta, and -Gamma. We demonstrate that ZO1b-Beta and ZO1b-Gamma are broadly expressed throughout the nervous system and localize to electrical synapses. By contrast, ZO1b-Alpha is expressed mainly non-neuronally and is not found at synapses. We generate mutants in all individual isoforms, as well as double mutant combinations in cis on individual chromosomes, and find that ZO1b-Beta is necessary and sufficient for robust GJ channel localization. ZO1b-Gamma, despite its localization to the synapse, plays an auxiliary role in channel localization. This study expands the notion of molecular complexity at the ESD, revealing that an individual genomic locus can contribute distinct isoforms to the macromolecular complex at electrical synapses. Further, independent scaffold isoforms have differential contributions to developmental assembly of the interneuronal GJ channels. We propose that ESD molecular complexity arises both from the diversity of unique genes and from distinct isoforms encoded by single genes. Overall, ESD proteomic diversity is expected to have critical impacts on the development, structure, function, and plasticity of electrical transmission.


Asunto(s)
Sinapsis Eléctricas , Pez Cebra , Animales , Sinapsis Eléctricas/fisiología , Pez Cebra/genética , Proteómica , Sinapsis/genética , Uniones Comunicantes/fisiología , Canales Iónicos , Isoformas de Proteínas/genética
4.
J Biol Chem ; 299(11): 105282, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37742923

RESUMEN

The intracellular domains of connexins are essential for the assembly of gap junctions. For connexin 36 (Cx36), the major neuronal connexin, it has been shown that a dysfunctional PDZ-binding motif interferes with electrical synapse formation. However, it is still unknown how this motif coordinates the transport of Cx36. In the present study, we characterize a phenotype of Cx36 mutants that lack a functional PDZ-binding motif using HEK293T cells as an expression system. We provide evidence that an intact PDZ-binding motif is critical for proper endoplasmic reticulum (ER) export of Cx36. Removing the PDZ-binding motif of Cx36 results in ER retention and the formation of multimembrane vesicles containing gap junction-like connexin aggregates. Using a combination of site-directed mutagenesis and electron micrographs, we reveal that these vesicles consist of Cx36 channels that docked prematurely in the ER. Our data suggest a model in which ER-retained Cx36 channels reshape the ER membrane into concentric whorls that are released into the cytoplasm.


Asunto(s)
Conexinas , Retículo Endoplásmico , Uniones Comunicantes , Humanos , Conexinas/genética , Conexinas/metabolismo , Retículo Endoplásmico/metabolismo , Uniones Comunicantes/metabolismo , Células HEK293 , Dominios Proteicos , Secuencias de Aminoácidos , Sinapsis Eléctricas/fisiología , Mutación , Transporte de Proteínas/genética , Vesículas Sinápticas/patología , Vesículas Sinápticas/ultraestructura , Microscopía Electrónica de Rastreo , Proteína delta-6 de Union Comunicante
5.
Curr Biol ; 33(10): 2063-2074.e4, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37172585

RESUMEN

The subcellular positioning of synapses and their specialized molecular compositions form the fundamental basis of neural circuits. Like chemical synapses, electrical synapses are constructed from an assortment of adhesion, scaffolding, and regulatory molecules, yet little is known about how these molecules localize to specific neuronal compartments. Here, we investigate the relationship between the autism- and epilepsy-associated gene Neurobeachin, the neuronal gap junction channel-forming Connexins, and the electrical synapse scaffold ZO1. Using the zebrafish Mauthner circuit, we find Neurobeachin localizes to the electrical synapse independently of ZO1 and Connexins. By contrast, we show Neurobeachin is required postsynaptically for the robust localization of ZO1 and Connexins. We demonstrate that Neurobeachin binds ZO1 but not Connexins. Finally, we find Neurobeachin is required to restrict electrical postsynaptic proteins to dendrites, but not electrical presynaptic proteins to axons. Together, the results reveal an expanded understanding of electrical synapse molecular complexity and the hierarchical interactions required to build neuronal gap junctions. Further, these findings provide novel insight into the mechanisms by which neurons compartmentalize the localization of electrical synapse proteins and provide a cell biological mechanism for the subcellular specificity of electrical synapse formation and function.


Asunto(s)
Sinapsis Eléctricas , Pez Cebra , Animales , Conexinas/metabolismo , Sinapsis Eléctricas/fisiología , Uniones Comunicantes/metabolismo , Neuronas/fisiología , Sinapsis/fisiología , Pez Cebra/metabolismo
6.
Nature ; 618(7963): 118-125, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225999

RESUMEN

Insect asynchronous flight is one of the most prevalent forms of animal locomotion used by more than 600,000 species. Despite profound insights into the motor patterns1, biomechanics2,3 and aerodynamics underlying asynchronous flight4,5, the architecture and function of the central-pattern-generating (CPG) neural network remain unclear. Here, on the basis of an experiment-theory approach including electrophysiology, optophysiology, Drosophila genetics and mathematical modelling, we identify a miniaturized circuit solution with unexpected properties. The CPG network consists of motoneurons interconnected by electrical synapses that, in contrast to doctrine, produce network activity splayed out in time instead of synchronized across neurons. Experimental and mathematical evidence support a generic mechanism for network desynchronization that relies on weak electrical synapses and specific excitability dynamics of the coupled neurons. In small networks, electrical synapses can synchronize or desynchronize network activity, depending on the neuron-intrinsic dynamics and ion channel composition. In the asynchronous flight CPG, this mechanism translates unpatterned premotor input into stereotyped neuronal firing with fixed sequences of cell activation that ensure stable wingbeat power and, as we show, is conserved across multiple species. Our findings prove a wider functional versatility of electrical synapses in the dynamic control of neural circuits and highlight the relevance of detecting electrical synapses in connectomics.


Asunto(s)
Drosophila melanogaster , Sinapsis Eléctricas , Vuelo Animal , Uniones Comunicantes , Vías Nerviosas , Animales , Sinapsis Eléctricas/fisiología , Fenómenos Electrofisiológicos , Vuelo Animal/fisiología , Uniones Comunicantes/metabolismo , Neuronas Motoras/fisiología , Drosophila melanogaster/fisiología
7.
Curr Biol ; 32(9): 2022-2036.e4, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35385694

RESUMEN

Electrical synapses are present in almost all organisms that have a nervous system. However, their brain-wide expression patterns and the full range of contributions to neural function are unknown in most species. Here, we first provide a light-microscopic, immunohistochemistry-based anatomical map of all innexin gap junction proteins-the building blocks of electrical synapses-in the central nervous system of Drosophila melanogaster. Of those innexin types that are expressed in the nervous system, some localize to glial cells, whereas others are predominantly expressed in neurons, with shakB being the most widely expressed neuronal innexin. We then focus on the function of shakB in VS/HS cells-a class of visual projection neurons-thereby uncovering an unexpected role for electrical synapses. Removing shakB from these neurons leads to spontaneous, cell-autonomous voltage and calcium oscillations, demonstrating that electrical synapses are required for these cells' intrinsic stability. Furthermore, we investigate the role of shakB-type electrical synapses in early visual processing. We find that the loss of shakB from the visual circuits upstream of VS/HS cells differentially impairs ON and OFF visual motion processing pathways but is not required for the computation of direction selectivity per se. Taken together, our study demonstrates that electrical synapses are widespread across the Drosophila nervous system and that they play essential roles in neuronal function and visual information processing.


Asunto(s)
Proteínas de Drosophila , Sinapsis Eléctricas , Animales , Conexinas/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Sinapsis Eléctricas/fisiología , Uniones Comunicantes/metabolismo , Sinapsis/metabolismo
8.
eNeuro ; 9(2)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35135867

RESUMEN

Electrical synapses couple inhibitory neurons across the brain, underlying a variety of functions that are modifiable by activity. Despite recent advances, many functions and contributions of electrical synapses within neural circuitry remain underappreciated. Among these are the sources and impacts of electrical synapse asymmetry. Using multi-compartmental models of neurons coupled through dendritic electrical synapses, we investigated intrinsic factors that contribute to effective synaptic asymmetry and that result in modulation of spike timing and synchrony between coupled cells. We show that electrical synapse location along a dendrite, input resistance, internal dendritic resistance, or directional conduction of the electrical synapse itself each alter asymmetry as measured by coupling between cell somas. Conversely, we note that asymmetrical gap junction (GJ) conductance can be masked by each of these properties. Furthermore, we show that asymmetry modulates spike timing and latency of coupled cells by up to tens of milliseconds, depending on direction of conduction or dendritic location of the electrical synapse. Coordination of rhythmic activity between two cells also depends on asymmetry. These simulations illustrate that causes of asymmetry are diverse, may not be apparent in somatic measurements of electrical coupling, influence dendritic processing, and produce a variety of outcomes on spiking and synchrony of coupled cells. Our findings highlight aspects of electrical synapses that should always be included in experimental demonstrations of coupling, and when assembling simulated networks containing electrical synapses.


Asunto(s)
Sinapsis Eléctricas , Uniones Comunicantes , Sinapsis Eléctricas/fisiología , Uniones Comunicantes/fisiología , Neuronas/fisiología , Sinapsis/fisiología
9.
PLoS Comput Biol ; 17(12): e1009644, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34871315

RESUMEN

Peristalsis, the coordinated contraction-relaxation of the muscles of the stomach is important for normal gastric motility and is impaired in motility disorders. Coordinated electrical depolarizations that originate and propagate within a network of interconnected layers of interstitial cells of Cajal (ICC) and smooth muscle (SM) cells of the stomach wall as a slow-wave, underly peristalsis. Normally, the gastric slow-wave oscillates with a single period and uniform rostrocaudal lag, exhibiting network entrainment. Understanding of the integrative role of neurotransmission and intercellular coupling in the propagation of an entrained gastric slow-wave, important for understanding motility disorders, however, remains incomplete. Using a computational framework constituted of a novel gastric motility network (GMN) model we address the hypothesis that engaging biological oscillators (i.e., ICCs) by constitutive gap junction coupling mechanisms and enteric neural innervation activated signals can confer a robust entrained gastric slow-wave. We demonstrate that while a decreasing enteric neural innervation gradient that modulates the intracellular IP3 concentration in the ICCs can guide the aboral slow-wave propagation essential for peristalsis, engaging ICCs by recruiting the exchange of second messengers (inositol trisphosphate (IP3) and Ca2+) ensures a robust entrained longitudinal slow-wave, even in the presence of biological variability in electrical coupling strengths. Our GMN with the distinct intercellular coupling in conjunction with the intracellular feedback pathways and a rostrocaudal enteric neural innervation gradient allows gastric slow waves to oscillate with a moderate range of frequencies and to propagate with a broad range of velocities, thus preventing decoupling observed in motility disorders. Overall, the findings provide a mechanistic explanation for the emergence of decoupled slow waves associated with motility impairments of the stomach, offer directions for future experiments and theoretical work, and can potentially aid in the design of new interventional pharmacological and neuromodulation device treatments for addressing gastric motility disorders.


Asunto(s)
Relojes Biológicos/fisiología , Tracto Gastrointestinal , Músculo Liso , Peristaltismo/fisiología , Sistemas de Mensajero Secundario/fisiología , Animales , Calcio/metabolismo , Biología Computacional , Sinapsis Eléctricas/fisiología , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Células Intersticiales de Cajal/fisiología , Potenciales de la Membrana/fisiología , Modelos Biológicos , Contracción Muscular/fisiología , Músculo Liso/inervación , Músculo Liso/fisiología
10.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34830020

RESUMEN

Two distinct types of neuronal activity result in long-term depression (LTD) of electrical synapses, with overlapping biochemical intracellular signaling pathways that link activity to synaptic strength, in electrically coupled neurons of the thalamic reticular nucleus (TRN). Because components of both signaling pathways can also be modulated by GABAB receptor activity, here we examined the impact of GABAB receptor activation on the two established inductors of LTD in electrical synapses. Recording from patched pairs of coupled rat neurons in vitro, we show that GABAB receptor inactivation itself induces a modest depression of electrical synapses and occludes LTD induction by either paired bursting or metabotropic glutamate receptor (mGluR) activation. GABAB activation also occludes LTD from either paired bursting or mGluR activation. Together, these results indicate that afferent sources of GABA, such as those from the forebrain or substantia nigra to the reticular nucleus, gate the induction of LTD from either neuronal activity or afferent glutamatergic receptor activation. These results add to a growing body of evidence that the regulation of thalamocortical transmission and sensory attention by TRN is modulated and controlled by other brain regions. Significance: We show that electrical synapse plasticity is gated by GABAB receptors in the thalamic reticular nucleus. This effect is a novel way for afferent GABAergic input from the basal ganglia to modulate thalamocortical relay and is a possible mediator of intra-TRN inhibitory effects.


Asunto(s)
Sinapsis Eléctricas/fisiología , Depresión Sináptica a Largo Plazo/genética , Plasticidad Neuronal/genética , Receptores de GABA-B/genética , Animales , Humanos , Depresión Sináptica a Largo Plazo/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Ratas , Tálamo/metabolismo , Tálamo/fisiopatología , Núcleos Talámicos Ventrales/metabolismo , Núcleos Talámicos Ventrales/fisiopatología
11.
Int Immunopharmacol ; 101(Pt B): 108335, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34781121

RESUMEN

The accumulation of amyloid beta (Aß) in the brain is thought to be associated with cognitive deficits in Alzheimer's disease (AD). However, current methods to combat Aß neurotoxicity are still lacking. G protein-coupled receptor 17 (GPR17) has become a target for treating inflammation in brain diseases, but it is unclear whether it has a role in AD. Here, we investigated the effects of cangrelor, a GPR17 antagonist, on neurotoxicity and memory impairment induced by intracerebroventricular (i.c.v.) injection of Aß1-42 in mice. The behavior results showed that cangrelor (2.0 or 4.0 µg/mouse, i.c.v.) treatment reversed the deficits in memory and learning ability induced by Aß1-42 in mice. Importantly, we demonstrated for the first time that GPR17 expression in the hippocampus and frontal cortex is increased in response to Aß1-42 exposures. We also found that cangrelor treatment reduced the activity of ß-secretase 1 (BACE1) and the levels of soluble Aß1-42 in the hippocampus and frontal cortex. Meanwhile, cangrelor treatment suppressed oxidative stress induced by Aß1-42, as proved by reduced production of malondialdehyde (MDA), and increased glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), and promoted the expression of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Furthermore, cangrelor also suppressed Aß1-42-induced neuroinflammation, characterized by suppressed activation of microglia, decreased the levels of pro-inflammatory cytokines, and nuclear translocation of NF-κB p65, as well as ameliorated synaptic deficits by promoting the upregulation of synaptic proteins, and increasing the number of Golgi-Cox stained dendritic spines. These results suggest that cangrelor may reverse Aß1-42-induced cognition deficits via inhibiting oxidative stress, neuroinflammation, and synaptic dysfunction mediated by Nrf2/HO-1 and NF-κB signaling.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Sinapsis Eléctricas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Inhibidores de Agregación Plaquetaria/uso terapéutico , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Monofosfato/uso terapéutico , Animales , Hemo-Oxigenasa 1/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal
12.
Dev Cell ; 56(23): 3235-3249.e4, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34741804

RESUMEN

Electrical synapses are established between specific neurons and within distinct subcellular compartments, but the mechanisms that direct gap junction assembly in the nervous system are largely unknown. Here, we show that a developmental program tunes cAMP signaling to direct the neuron-specific assembly and placement of electrical synapses in the C. elegans motor circuit. We use live-cell imaging to visualize electrical synapses in vivo and an optogenetic assay to confirm that they are functional. In ventral A class (VA) motor neurons, the UNC-4 transcription factor blocks expression of cAMP antagonists that promote gap junction miswiring. In unc-4 mutants, VA electrical synapses are established with an alternative synaptic partner and are repositioned from the VA axon to soma. cAMP counters these effects by driving gap junction trafficking into the VA axon for electrical synapse assembly. Thus, our experiments establish that cAMP regulates gap junction trafficking for the biogenesis of functional electrical synapses.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , AMP Cíclico/farmacología , Sinapsis Eléctricas/fisiología , Proteínas de Homeodominio/metabolismo , Neuronas Motoras/fisiología , Fracciones Subcelulares/fisiología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Axones/efectos de los fármacos , Axones/fisiología , Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Conexinas/genética , Conexinas/metabolismo , Sinapsis Eléctricas/efectos de los fármacos , Uniones Comunicantes , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Neuronas Motoras/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos
13.
Cell Rep ; 37(3): 109853, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686323

RESUMEN

Currently, many genetic methods are available for mapping chemical connectivity, but analogous methods for electrical synapses are lacking. Here, we present pupylation-based interaction labeling (PUPIL), a genetically encoded system for noninvasively mapping and stamping transient electrical synapses in the mouse brain. Upon fusion of connexin 26 (CX26) with the ligase PafA, pupylation yields tag puncta following conjugation of its substrate, a biotin- or fluorescent-protein-tagged PupE, to the neighboring proteins of electrical synapses containing CX26-PafA. Tag puncta are validated to correlate well with functional electrical synapses in immature neurons. Furthermore, puncta are retained in mature neurons when electrical synapses mostly disappear-suggesting successful stamping. We use PUPIL to uncover spatial subcellular localizations of electrical synapses and approach their physiological functions during development. Thus, PUPIL is a powerful tool for probing electrical connectivity patterns in complex nervous systems and has great potential for transient receptors and ion channels as well.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Sinapsis Eléctricas/fisiología , Uniones Comunicantes/fisiología , Neuronas/fisiología , Optogenética , Factores de Edad , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Animales Recién Nacidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Conexina 26/genética , Conexina 26/metabolismo , Conexinas/genética , Conexinas/metabolismo , Conductividad Eléctrica , Sinapsis Eléctricas/metabolismo , Sinapsis Eléctricas/ultraestructura , Femenino , Uniones Comunicantes/metabolismo , Uniones Comunicantes/ultraestructura , Edad Gestacional , Células HEK293 , Células HeLa , Humanos , Ratones Endogámicos ICR , Ratones Noqueados , Microscopía Confocal , Neuronas/metabolismo , Neuronas/ultraestructura , Embarazo , Potenciales Sinápticos , Proteína delta-6 de Union Comunicante
14.
PLoS Comput Biol ; 17(9): e1009378, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34529674

RESUMEN

Activity-dependent regulation of intrinsic excitability has been shown to greatly contribute to the overall plasticity of neuronal circuits. Such neuroadaptations are commonly investigated in patch clamp experiments using current step stimulation and the resulting input-output functions are analyzed to quantify alterations in intrinsic excitability. However, it is rarely addressed, how such changes translate to the function of neurons when they operate under natural synaptic inputs. Still, it is reasonable to expect that a strong correlation and near proportional relationship exist between static firing responses and those evoked by synaptic drive. We challenge this view by performing a high-yield electrophysiological analysis of cultured mouse hippocampal neurons using both standard protocols and simulated synaptic inputs via dynamic clamp. We find that under these conditions the neurons exhibit vastly different firing responses with surprisingly weak correlation between static and dynamic firing intensities. These contrasting responses are regulated by two intrinsic K-currents mediated by Kv1 and Kir channels, respectively. Pharmacological manipulation of the K-currents produces differential regulation of the firing output of neurons. Static firing responses are greatly increased in stuttering type neurons under blocking their Kv1 channels, while the synaptic responses of the same neurons are less affected. Pharmacological blocking of Kir-channels in delayed firing type neurons, on the other hand, exhibit the opposite effects. Our subsequent computational model simulations confirm the findings in the electrophysiological experiments and also show that adaptive changes in the kinetic properties of such currents can even produce paradoxical regulation of the firing output.


Asunto(s)
Modelos Neurológicos , Neuronas/fisiología , Potenciales de Acción/fisiología , Animales , Células Cultivadas , Biología Computacional , Simulación por Computador , Sinapsis Eléctricas/fisiología , Fenómenos Electrofisiológicos , Hipocampo/citología , Hipocampo/fisiología , Cinética , Ratones , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología
15.
Cells ; 10(9)2021 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-34572046

RESUMEN

The retinas of many species show regional specialisations that are evident in the differences in the processing of visual input from different parts of the visual field. Regional specialisation is thought to reflect an adaptation to the natural visual environment, optical constraints, and lifestyle of the species. Yet, little is known about regional differences in synaptic circuitry. Here, we were interested in the topographical distribution of connexin-36 (Cx36), the major constituent of electrical synapses in the retina. We compared the retinas of mice, rats, and cats to include species with different patterns of regional specialisations in the analysis. First, we used the density of Prox1-immunoreactive amacrine cells as a marker of any regional specialisation, with higher cell density signifying more central regions. Double-labelling experiments showed that Prox1 is expressed in AII amacrine cells in all three species. Interestingly, large Cx36 plaques were attached to about 8-10% of Prox1-positive amacrine cell somata, suggesting the strong electrical coupling of pairs or small clusters of cell bodies. When analysing the regional changes in the volumetric density of Cx36-immunoreactive plaques, we found a tight correlation with the density of Prox1-expressing amacrine cells in the ON, but not in the OFF sublamina in all three species. The results suggest that the relative contribution of electrical synapses to the ON- and OFF-pathways of the retina changes with retinal location, which may contribute to functional ON/OFF asymmetries across the visual field.


Asunto(s)
Células Amacrinas/fisiología , Conexinas/metabolismo , Dendritas/fisiología , Sinapsis Eléctricas/fisiología , Uniones Comunicantes/fisiología , Proteínas de Homeodominio/metabolismo , Retina/fisiología , Proteínas Supresoras de Tumor/metabolismo , Células Amacrinas/citología , Animales , Conexinas/genética , Femenino , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Retina/citología , Proteínas Supresoras de Tumor/genética , Proteína delta-6 de Union Comunicante
16.
Cells ; 10(8)2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34440662

RESUMEN

Neuronal culture was used to investigate neuronal function in physiological and pathological conditions. Despite its inevitability, primary neuronal culture remained a gold standard method that requires laborious preparation, intensive training, and animal resources. To circumvent the shortfalls of primary neuronal preparations and efficiently give rise to functional neurons, we combine a neural stem cell culture method with a direct cell type-conversion approach. The lucidity of this method enables the efficient preparation of functional neurons from mouse neural progenitor cells on demand. We demonstrate that induced neurons (NPC-iNs) by this method make synaptic connections, elicit neuronal activity-dependent cellular responses, and develop functional neuronal networks. This method will provide a concise platform for functional neuronal assessments. This indeed offers a perspective for using these characterized neuronal networks for investigating plasticity mechanisms, drug screening assays, and probing the molecular and biophysical basis of neurodevelopmental and neurodegenerative diseases.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis , Animales , Técnicas de Cultivo de Célula , Línea Celular , Sinapsis Eléctricas/fisiología , Potenciales Evocados , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Red Nerviosa/fisiología , Neurogénesis/genética , Fenotipo , Transmisión Sináptica
17.
PLoS Comput Biol ; 17(6): e1009163, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34181653

RESUMEN

Synchronous oscillations in neural populations are considered being controlled by inhibitory neurons. In the granular layer of the cerebellum, two major types of cells are excitatory granular cells (GCs) and inhibitory Golgi cells (GoCs). GC spatiotemporal dynamics, as the output of the granular layer, is highly regulated by GoCs. However, there are various types of inhibition implemented by GoCs. With inputs from mossy fibers, GCs and GoCs are reciprocally connected to exhibit different network motifs of synaptic connections. From the view of GCs, feedforward inhibition is expressed as the direct input from GoCs excited by mossy fibers, whereas feedback inhibition is from GoCs via GCs themselves. In addition, there are abundant gap junctions between GoCs showing another form of inhibition. It remains unclear how these diverse copies of inhibition regulate neural population oscillation changes. Leveraging a computational model of the granular layer network, we addressed this question to examine the emergence and modulation of network oscillation using different types of inhibition. We show that at the network level, feedback inhibition is crucial to generate neural oscillation. When short-term plasticity was equipped on GoC-GC synapses, oscillations were largely diminished. Robust oscillations can only appear with additional gap junctions. Moreover, there was a substantial level of cross-frequency coupling in oscillation dynamics. Such a coupling was adjusted and strengthened by GoCs through feedback inhibition. Taken together, our results suggest that the cooperation of distinct types of GoC inhibition plays an essential role in regulating synchronous oscillations of the GC population. With GCs as the sole output of the granular network, their oscillation dynamics could potentially enhance the computational capability of downstream neurons.


Asunto(s)
Corteza Cerebelosa/citología , Corteza Cerebelosa/fisiología , Modelos Neurológicos , Animales , Biología Computacional , Sinapsis Eléctricas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Retroalimentación Fisiológica , Humanos , Potenciales Postsinápticos Inhibidores/fisiología , Fibras Nerviosas/fisiología , Red Nerviosa/citología , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Análisis de la Célula Individual/estadística & datos numéricos , Sinapsis/fisiología
18.
Int J Mol Sci ; 22(9)2021 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-33922931

RESUMEN

Anatomical and electrophysiological evidence that gap junctions and electrical coupling occur between neurons was initially confined to invertebrates and nonmammals and was thought to be a primitive form of synaptic transmission. More recent studies revealed that electrical communication is common in the mammalian central nervous system (CNS), often coexisting with chemical synaptic transmission. The subsequent progress indicated that electrical synapses formed by the gap junction protein connexin-36 (Cx36) and its paralogs in nonmammals constitute vital elements in mammalian and fish synaptic circuitry. They govern the collective activity of ensembles of coupled neurons, and Cx36 gap junctions endow them with enormous adaptive plasticity, like that seen at chemical synapses. Moreover, they orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie the fundamental integrative processes, such as memory and learning. Here, we review the available mechanistic evidence and models that argue for the essential roles of calcium, calmodulin, and the Ca2+/calmodulin-dependent protein kinase II in integrating calcium signals to modulate the strength of electrical synapses through interactions with the gap junction protein Cx36.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/fisiología , Conexinas/metabolismo , Sinapsis Eléctricas/fisiología , Animales , Calcio/metabolismo , Conexinas/genética , Sinapsis Eléctricas/metabolismo , Uniones Comunicantes/metabolismo , Humanos , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Mapas de Interacción de Proteínas , Transmisión Sináptica , Proteína delta-6 de Union Comunicante
19.
Elife ; 102021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33908867

RESUMEN

Electrical synaptic transmission relies on neuronal gap junctions containing channels constructed by Connexins. While at chemical synapses neurotransmitter-gated ion channels are critically supported by scaffolding proteins, it is unknown if channels at electrical synapses require similar scaffold support. Here, we investigated the functional relationship between neuronal Connexins and Zonula Occludens 1 (ZO1), an intracellular scaffolding protein localized to electrical synapses. Using model electrical synapses in zebrafish Mauthner cells, we demonstrated that ZO1 is required for robust synaptic Connexin localization, but Connexins are dispensable for ZO1 localization. Disrupting this hierarchical ZO1/Connexin relationship abolishes electrical transmission and disrupts Mauthner cell-initiated escape responses. We found that ZO1 is asymmetrically localized exclusively postsynaptically at neuronal contacts where it functions to assemble intercellular channels. Thus, forming functional neuronal gap junctions requires a postsynaptic scaffolding protein. The critical function of a scaffolding molecule reveals an unanticipated complexity of molecular and functional organization at electrical synapses.


Neurons 'talk' with each another at junctions called synapses, which can either be chemical or electrical. Communication across a chemical synapse involves a 'sending' neuron releasing chemicals that diffuse between the cells and subsequently bind to specialized receptors on the receiving neuron. These complex junctions involve a large number of well-studied molecular actors. Electrical synapses, on the other hand, are believed to be simpler. There, neurons are physically connected via channels formed of 'connexin' proteins, which allow electrically charged ions to flow between the cells. However, it is likely that other proteins help to create these structures. In particular, recent evidence shows that without a structurally supporting 'scaffolding' protein called ZO1, electrical synapses cannot form in the brain of a tiny freshwater fish known as zebrafish. As their name implies, scaffolding proteins help cells organize their internal structure, for example by anchoring other molecules to the cell membrane. By studying electrical synapses in zebrafish, Lasseigne, Echeverry, Ijaz, Michel et al. now show that these structures are more complex than previously assumed. In particular, the experiments reveal that ZO1 proteins are only present on one side of electrical synapses; despite their deceptively symmetrical anatomical organization, these junctions can be asymmetric, like their chemical cousins. The results also show that ZO1 must be present for connexins to gather at electrical synapses, whereas the converse is not true. This suggests that when a new electrical synapse forms, ZO1 moves into position first: it then recruits or stabilizes connexins to form the channels connecting the two cells. In many animals with a spine, electrical synapses account for about 20% of all neural junctions. Understanding how these structures form and work could help to find new treatments for disorders linked to impaired electrical synapses, such as epilepsy.


Asunto(s)
Conexinas/metabolismo , Sinapsis Eléctricas/fisiología , Transmisión Sináptica/genética , Proteínas de Pez Cebra/genética , Pez Cebra/fisiología , Proteína de la Zonula Occludens-1/genética , Animales , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
20.
Commun Biol ; 4(1): 241, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33623091

RESUMEN

Short-term plasticity preserves a brief history of synaptic activity that is communicated to the postsynaptic neuron. This is primarily regulated by a calcium signal initiated by voltage dependent calcium channels in the presynaptic terminal. Imaging studies of CA3-CA1 synapses reveal the presence of another source of calcium, the endoplasmic reticulum (ER) in all presynaptic terminals. However, the precise role of the ER in modifying STP remains unexplored. We performed in-silico experiments in synaptic geometries based on reconstructions of the rat CA3-CA1 synapses to investigate the contribution of ER. Our model predicts that presynaptic ER is critical in generating the observed short-term plasticity profile of CA3-CA1 synapses and allows synapses with low release probability to operate more reliably. Blocking the ER lowers facilitation in a manner similar to what has been previously characterized in animal models of Alzheimer's disease and underscores the important role played by presynaptic stores in normal function.


Asunto(s)
Sinapsis Eléctricas/fisiología , Retículo Endoplásmico/fisiología , Hipocampo/fisiología , Modelos Neurológicos , Plasticidad Neuronal , Terminales Presinápticos/fisiología , Animales , Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Canales de Calcio/metabolismo , Señalización del Calcio , Simulación por Computador , Sinapsis Eléctricas/metabolismo , Retículo Endoplásmico/metabolismo , Hipocampo/metabolismo , Humanos , Método de Montecarlo , Terminales Presinápticos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA