Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 754: 109959, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490311

RESUMEN

Electrical synapses are essential components of neural circuits. Neuronal signal transduction across electrical synapses is primarily mediated by gap junction channels composed of Connexin36 (Cx36), the lack of which causes impaired electrical coupling between certain neurons including cortical interneurons and thalamic reticular nucleus (TRN) neurons. However, the structural basis underlying Cx36 function and assembly remains elusive. Recently, Lee et al. reported cryo-EM structures of Cx36, thus provided first insights of its gating mechanism. Here, we report a consistent cryo-EM structure of Cx36 determined in parallel, and describe unique interactions underpinning its assembly mechanism in complementary to the competing work. In particular, we found non-canonical electrostatic interactions between protomers from opposing hemichannels and a steric complementary site between adjacent protomers within a hemichannel, which together provide a structural explanation for the assembly specificity in homomeric and heteromeric gap junction channels.


Asunto(s)
Sinapsis Eléctricas , Proteína delta-6 de Union Comunicante , Conexinas/química , Conexinas/metabolismo , Microscopía por Crioelectrón , Sinapsis Eléctricas/metabolismo , Uniones Comunicantes/metabolismo , Canales Iónicos , Neuronas/metabolismo , Subunidades de Proteína , Humanos
2.
Neuroscience ; 523: 31-46, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37225049

RESUMEN

Spinal sympathetic preganglionic neurons (SPNs) are among the many neuronal populations in the mammalian central nervous system (CNS) where there is evidence for electrical coupling between cell pairs linked by gap junctions composed of connexin36 (Cx36). Understanding the organization of this coupling in relation to autonomic functions of spinal sympathetic systems requires knowledge of how these junctions are deployed among SPNs. Here, we document the distribution of immunofluorescence detection of Cx36 among SPNs identified by immunolabelling of their various markers, including choline acetyltransferase, nitric oxide and peripherin in adult and developing mouse and rat. In adult animals, labelling of Cx36 was exclusively punctate and dense concentrations of Cx36-puncta were distributed along the entire length of the spinal thoracic intermediolateral cell column (IML). These puncta were also seen in association with SPN dendritic processes in the lateral funiculus, the intercalated and central autonomic areas and those within and extending medially from the IML. All labelling for Cx36 was absent in spinal cords of Cx36 knockout mice. High densities of Cx36-puncta were already evident among clusters of SPNs in the IML of mouse and rat at postnatal days 10-12. In Cx36BAC::eGFP mice, eGFP reporter was absent in SPNs, thus representing false negative detection, but was localized to some glutamatergic and GABAergic synaptic terminals. Some eGFP+ terminals were found contacting SPN dendrites. These results indicate widespread Cx36 expression in SPNs, further supporting evidence of electrical coupling between these cells, and suggest that SPNs are innervated by neurons that themselves may be electrically coupled.


Asunto(s)
Sinapsis Eléctricas , Uniones Comunicantes , Ratones , Ratas , Animales , Sinapsis Eléctricas/metabolismo , Ratas Sprague-Dawley , Uniones Comunicantes/metabolismo , Conexinas/metabolismo , Neuronas/metabolismo , Médula Espinal/metabolismo , Ratones Noqueados , Mamíferos/metabolismo , Proteína delta-6 de Union Comunicante
3.
Neuroscience ; 521: 58-76, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37100373

RESUMEN

Sexually dimorphic motoneurons (MNs) located in lower lumbar spinal cord are involved in mating and reproductive behaviours and are known to be coupled by electrical synapses. The cremaster motor nucleus in upper lumbar spinal cord has also been suggested to support physiological processes associated with sexual behaviours in addition to its thermoregulatory and protective role in maintaining testes integrity. Using immunofluorescence approaches, we investigated whether cremaster MNs also exhibit features reflecting their potential for electrical synaptic communication and examined some of their other synaptic characteristics. Both mice and rats displayed punctate immunolabelling of Cx36 associated with cremaster MNs, indicative of gap junction formation. Transgenic mice with enhanced green fluorescent protein (eGFP) reporter for connexin36 expression showed that subpopulations of cremaster MNs in both male and female mice express eGFP, with greater proportions of those in male mice. The eGFP+ MNs within the cremaster nucleus vs. eGFP- MNs inside and outside this nucleus displayed a 5-fold greater density of serotonergic innervation and exhibited a paucity of innervation by C-terminals arising from cholinergic V0c interneurons. All MNs within the cremaster motor nucleus displayed prominent patches of immunolabelling for SK3 (K+) channels around their periphery, suggestive of their identity as slow MNs, many though not all of which were in apposition to C-terminals. The results provide evidence for electrical coupling of a large proportion of cremaster MNs and suggest the existence of two populations of these MNs with possibly differential innervation of their peripheral target muscles serving different functions.


Asunto(s)
Sinapsis Eléctricas , Médula Espinal , Ratones , Ratas , Masculino , Femenino , Animales , Sinapsis Eléctricas/metabolismo , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Neuronas Motoras/metabolismo , Uniones Comunicantes/metabolismo , Ratones Transgénicos
4.
Neuroscientist ; 29(5): 554-568, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36125001

RESUMEN

Gap junctions between neurons of the brain are thought to be present in only certain cell types, and they mostly connect dendrites, somata, and axons. Synapses with gap junctions serve bidirectional metabolic and electrical coupling between connected neuronal compartments. Although plasticity of electrical synapses has been described, recent evidence of the presence of silent, but activatable, gap junctions suggests that electrical nodes in a neuronal circuit can be added or suppressed by changes in the synaptic microenvironment. This opens the possibility of reconfiguration of neuronal ensembles in response to activity. Moreover, the coexistence of gap junctions in a glutamatergic synapse may add electric and metabolic coupling to a neuronal aggregate and may serve to constitute primed ensembles within a higher-order neural network. The interaction of chemical with electrical synapses should be further explored to find, especially, emerging properties of neuronal ensembles. It will be worth to reexamine in a new light the "functional" implications of the "anatomic" concepts: "continuity" and "contiguity," which were championed by Golgi and Ramón y Cajal, respectively. In any case, exploring the versatility of the gap junctions will likely enrich the heuristic aspects of the neural and network postulates.


Asunto(s)
Sinapsis Eléctricas , Uniones Comunicantes , Humanos , Uniones Comunicantes/metabolismo , Sinapsis Eléctricas/metabolismo , Sinapsis/metabolismo , Neuronas/fisiología , Encéfalo/metabolismo , Axones/metabolismo
5.
Immunity ; 55(1): 159-173.e9, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34982959

RESUMEN

To accommodate the changing needs of the developing brain, microglia must undergo substantial morphological, phenotypic, and functional reprogramming. Here, we examined whether cellular metabolism regulates microglial function during neurodevelopment. Microglial mitochondria bioenergetics correlated with and were functionally coupled to phagocytic activity in the developing brain. Transcriptional profiling of microglia with diverse metabolic profiles revealed an activation signature wherein the interleukin (IL)-33 signaling axis is associated with phagocytic activity. Genetic perturbation of IL-33 or its receptor ST2 led to microglial dystrophy, impaired synaptic function, and behavioral abnormalities. Conditional deletion of Il33 from astrocytes or Il1rl1, encoding ST2, in microglia increased susceptibility to seizures. Mechanistically, IL-33 promoted mitochondrial activity and phagocytosis in an AKT-dependent manner. Mitochondrial metabolism and AKT activity were temporally regulated in vivo. Thus, a microglia-astrocyte circuit mediated by the IL-33-ST2-AKT signaling axis supports microglial metabolic adaptation and phagocytic function during early development, with implications for neurodevelopmental and neuropsychiatric disorders.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Convulsiones/inmunología , Animales , Conducta Animal , Susceptibilidad a Enfermedades , Sinapsis Eléctricas/metabolismo , Metabolismo Energético , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/genética , Ratones , Ratones Noqueados , Microglía/patología , Neurogénesis/genética , Proteína Oncogénica v-akt/metabolismo , Fagocitosis , Transducción de Señal
6.
Sci Rep ; 11(1): 22568, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799629

RESUMEN

WDR45 plays an essential role in the early stage of autophagy. De novo heterozygous mutations in WDR45 have been known to cause ß-propeller protein-associated neurodegeneration (BPAN), a subtype of neurodegeneration with brain iron accumulation (NBIA). Although BPAN patients display global developmental delay with intellectual disability, the neurodevelopmental pathophysiology of BPAN remains largely unknown. In the present study, we analyzed the physiological role of Wdr45 and pathophysiological significance of the gene abnormality during mouse brain development. Morphological and biochemical analyses revealed that Wdr45 is expressed in a developmental stage-dependent manner in mouse brain. Wdr45 was also found to be located in excitatory synapses by biochemical fractionation. Since WDR45 mutations are thought to cause protein degradation, we conducted acute knockdown experiments by in utero electroporation in mice to recapitulate the pathophysiological conditions of BPAN. Knockdown of Wdr45 caused abnormal dendritic development and synaptogenesis during corticogenesis, both of which were significantly rescued by co-expression with RNAi-resistant version of Wdr45. In addition, terminal arbors of callosal axons were less developed in Wdr45-deficient cortical neurons of adult mouse when compared to control cells. These results strongly suggest a pathophysiological significance of WDR45 gene abnormalities in neurodevelopmental aspects of BPAN.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Degeneración Nerviosa , Distrofias Neuroaxonales/metabolismo , Neurogénesis , Animales , Axones/metabolismo , Axones/patología , Encéfalo/embriología , Células COS , Proteínas Portadoras/genética , Chlorocebus aethiops , Dendritas/metabolismo , Dendritas/patología , Sinapsis Eléctricas/metabolismo , Sinapsis Eléctricas/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Edad Gestacional , Trastornos del Metabolismo del Hierro/embriología , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Ratones Endogámicos ICR , Distrofias Neuroaxonales/embriología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Transducción de Señal
7.
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
8.
ACS Appl Mater Interfaces ; 13(33): 39641-39651, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34374517

RESUMEN

Biomaterial-based memristors (bio-memristors) are often adopted to emulate biological synapse functions and applied to construct neural computing networks in brain-inspired chip systems. However, the randomness of conductive filament formation in bio-memristors inhibits their switching performance by causing the dispersion of the device-switching parameters. In this case, a facile porous silk fibroin (p-SF) memristor was obtained through a protein surface reconstruction strategy, in which the size of the hole can be adjusted by the density of hybrid nanoseeds. The porous SF memristors exhibit greatly enhanced electrical characteristics, including uniform I-V cycles, centralized distribution of the switching voltages, and both high and low resistances, compared to devices without pores. The results of three-dimensional (3D) simulations based on classical density functional theory (cDFT) suggest that the reconstructed pores in the SF layers guide the formation and fracture of Ag filaments under an electric field and enhance the overall conductivity by separating Ag+ ion and electron diffusion pathways. Ag+ ions are predicted to preferentially diffuse through pores, whereas electrons diffuse through the SF network. Interestingly, the device conductance can be bidirectionally modulated gradually by positive and negative voltages, can faithfully simulate short-term and long-term plasticity, and can even realize the triplet-spike-timing-dependent plasticity (triplet-STDP) rule, which can be used for pattern recognition in biological systems. The simulation results reveal that a memristor network of this type has an accuracy of ∼95.78% in memory learning and the capability of pattern learning. This work provides a facile technology route to improve the performance of bionic-material memristors.


Asunto(s)
Sinapsis Eléctricas/química , Sinapsis Eléctricas/metabolismo , Fibroínas/química , Encéfalo , Cationes/química , Simulación por Computador , Teoría Funcional de la Densidad , Conductividad Eléctrica , Modelos Biológicos , Redes Neurales de la Computación , Plasticidad Neuronal/fisiología , Porosidad , Plata/química , Propiedades de Superficie
9.
Cells ; 10(8)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34440631

RESUMEN

The extracellular matrix (ECM) plays a key role in synaptogenesis and the regulation of synaptic functions in the central nervous system. Recent studies revealed that in addition to dopaminergic and serotoninergic neuromodulatory systems, microglia also contribute to the regulation of ECM remodeling. In the present work, we investigated the physiological role of microglia in the remodeling of perineuronal nets (PNNs), predominantly associated with parvalbumin-immunopositive (PV+) interneurons, and the perisynaptic ECM around pyramidal neurons in the hippocampus. Adult mice were treated with PLX3397 (pexidartinib), as the inhibitor of colony-stimulating factor 1 receptor (CSF1-R), to deplete microglia. Then, confocal analysis of the ECM and synapses was performed. Although the elimination of microglia did not alter the overall number or intensity of PNNs in the CA1 region of the hippocampus, it decreased the size of PNN holes and elevated the expression of the surrounding ECM. In the neuropil area in the CA1 str. radiatum, the depletion of microglia increased the expression of perisynaptic ECM proteoglycan brevican, which was accompanied by the elevated expression of presynaptic marker vGluT1 and the increased density of dendritic spines. Thus, microglia regulate the homeostasis of pre- and postsynaptic excitatory terminals and the surrounding perisynaptic ECM as well as the fine structure of PNNs enveloping perisomatic-predominantly GABAergic-synapses.


Asunto(s)
Región CA1 Hipocampal/patología , Sinapsis Eléctricas/patología , Potenciales Postsinápticos Excitadores , Matriz Extracelular/patología , Microglía/patología , Aminopiridinas/toxicidad , Animales , Brevicano/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Sinapsis Eléctricas/metabolismo , Matriz Extracelular/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Red Nerviosa/metabolismo , Red Nerviosa/patología , Pirroles/toxicidad , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteína Fluorescente Roja
10.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972428

RESUMEN

Electrical synapses are specialized structures that mediate the flow of electrical currents between neurons and have well known roles in synchronizing the activities of neuronal populations, both by mediating the current transfer from more active to less active neurons and by shunting currents from active neurons to their less active neighbors. However, how these positive and negative functions of electrical synapses are coordinated to shape rhythmic synaptic outputs and behavior is not well understood. Here, using a combination of genetics, behavioral analysis, and live calcium imaging in Caenorhabditis elegans, we show that electrical synapses formed by the gap junction protein INX-1/innexin couple the presynaptic terminals of a pair of motor neurons (AVL and DVB) to synchronize their activation in response to a pacemaker signal. Live calcium imaging reveals that inx-1/innexin mutations lead to asynchronous activation of AVL and DVB, due, in part, to loss of AVL-mediated activation of DVB by the pacemaker. In addition, loss of inx-1 leads to the ectopic activation of DVB at inappropriate times during the cycle through the activation of the L-type voltage-gated calcium channel EGL-19. We propose that electrical synapses between AVL and DVB presynaptic terminals function to ensure the precise and robust execution of a specific step in a rhythmic behavior by both synchronizing the activities of presynaptic terminals in response to pacemaker signaling and by inhibiting their activation in between cycles when pacemaker signaling is low.


Asunto(s)
Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Sinapsis Eléctricas/metabolismo , Neuronas Motoras/metabolismo , Terminales Presinápticos/metabolismo , Transmisión Sináptica/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Conexinas/genética , Conexinas/metabolismo , Sinapsis Eléctricas/ultraestructura , Uniones Comunicantes/metabolismo , Uniones Comunicantes/ultraestructura , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Imagen Molecular , Neuronas Motoras/citología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Periodicidad , Terminales Presinápticos/ultraestructura , Proteína Fluorescente Roja
11.
PLoS One ; 16(4): e0244902, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33857131

RESUMEN

Intercellular gap junction channels and single-membrane channels have been reported to regulate electrical synapse and the brain function. Innexin is known as a gap junction-related protein in invertebrates and is involved in the formation of intercellular gap junction channels and single-cell membrane channels. Multiple isoforms of innexin protein in each species enable the precise regulation of channel function. In molluscan species, sequence information of innexins is still limited and the sequences of multiple innexin isoforms have not been classified. This study examined the innexin transcripts expressed in the central nervous system of the terrestrial slug Limax valentianus and identified 16 transcripts of 12 innexin isoforms, including the splicing variants. We performed phylogenetic analysis and classified the isoforms with other molluscan innexin sequences. Next, the phosphorylation, N-glycosylation, and S-nitrosylation sites were predicted to characterize the innexin isoforms. Further, we identified 16 circular RNA sequences of nine innexin isoforms in the central nervous system of Limax. The identification and classification of molluscan innexin isoforms provided novel insights for understanding the regulatory mechanism of innexin in this phylum.


Asunto(s)
Conexinas/clasificación , Conexinas/genética , Gastrópodos/genética , Animales , Transporte Biológico/fisiología , Sistema Nervioso Central/fisiología , Sinapsis Eléctricas/metabolismo , Uniones Comunicantes/genética , Uniones Comunicantes/metabolismo , Gastrópodos/metabolismo , Expresión Génica/genética , Canales Iónicos/metabolismo , Filogenia , Isoformas de Proteínas/genética , Transcriptoma/genética
12.
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
13.
Elife ; 102021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33871358

RESUMEN

Most research on neurodegenerative diseases has focused on neurons, yet glia help form and maintain the synapses whose loss is so prominent in these conditions. To investigate the contributions of glia to Huntington's disease (HD), we profiled the gene expression alterations of Drosophila expressing human mutant Huntingtin (mHTT) in either glia or neurons and compared these changes to what is observed in HD human and HD mice striata. A large portion of conserved genes are concordantly dysregulated across the three species; we tested these genes in a high-throughput behavioral assay and found that downregulation of genes involved in synapse assembly mitigated pathogenesis and behavioral deficits. To our surprise, reducing dNRXN3 function in glia was sufficient to improve the phenotype of flies expressing mHTT in neurons, suggesting that mHTT's toxic effects in glia ramify throughout the brain. This supports a model in which dampening synaptic function is protective because it attenuates the excitotoxicity that characterizes HD.


When a neuron dies, through injury or disease, the body loses all communication that passes through it. The brain compensates by rerouting the flow of information through other neurons in the network. Eventually, if the loss of neurons becomes too great, compensation becomes impossible. This process happens in Alzheimer's, Parkinson's, and Huntington's disease. In the case of Huntington's disease, the cause is mutation to a single gene known as huntingtin. The mutation is present in every cell in the body but causes particular damage to parts of the brain involved in mood, thinking and movement. Neurons and other cells respond to mutations in the huntingtin gene by turning the activities of other genes up or down, but it is not clear whether all of these changes contribute to the damage seen in Huntington's disease. In fact, it is possible that some of the changes are a result of the brain trying to protect itself. So far, most research on this subject has focused on neurons because the huntingtin gene plays a role in maintaining healthy neuronal connections. But, given that all cells carry the mutated gene, it is likely that other cells are also involved. The glia are a diverse group of cells that support the brain, providing care and sustenance to neurons. These cells have a known role in maintaining the connections between neurons and may also have play a role in either causing or correcting the damage seen in Huntington's disease. The aim of Onur et al. was to find out which genes are affected by having a mutant huntingtin gene in neurons or glia, and whether severity of Huntington's disease improved or worsened when the activity of these genes changed. First, Onur et al. identified genes affected by mutant huntingtin by comparing healthy human brains to the brains of people with Huntington's disease. Repeating the same comparison in mice and fruit flies identified genes affected in the same way across all three species, revealing that, in Huntington's disease, the brain dials down glial cell genes involved in maintaining neuronal connections. To find out how these changes in gene activity affect disease severity and progression, Onur et al. manipulated the activity of each of the genes they had identified in fruit flies that carried mutant versions of huntingtin either in neurons, in glial cells or in both cell types. They then filmed the flies to see the effects of the manipulation on movement behaviors, which are affected by Huntington's disease. This revealed that purposely lowering the activity of the glial genes involved in maintaining connections between neurons improved the symptoms of the disease, but only in flies who had mutant huntingtin in their glial cells. This indicates that the drop in activity of these genes observed in Huntington's disease is the brain trying to protect itself. This work suggests that it is important to include glial cells in studies of neurological disorders. It also highlights the fact that changes in gene expression as a result of a disease are not always bad. Many alterations are compensatory, and try to either make up for or protect cells affected by the disease. Therefore, it may be important to consider whether drugs designed to treat a condition by changing levels of gene activity might undo some of the body's natural protection. Working out which changes drive disease and which changes are protective will be essential for designing effective treatments.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Sinapsis Eléctricas/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Neuroglía/metabolismo , Transmisión Sináptica , Animales , Conducta Animal , Encéfalo/patología , Encéfalo/fisiopatología , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Sinapsis Eléctricas/patología , Femenino , Redes Reguladoras de Genes , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Locomoción , Masculino , Ratones Transgénicos , Mutación , Neuroglía/patología , Transcriptoma , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
14.
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
15.
Cell Rep ; 34(8): 108773, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33626349

RESUMEN

Mutations in genes essential for synaptic function, such as the presynaptic adhesion molecule Neurexin1α (Nrxn1α), are strongly implicated in neuropsychiatric pathophysiology. As the input nucleus of the basal ganglia, the striatum integrates diverse excitatory projections governing cognitive and motor control, and its impairment may represent a recurrent pathway to disease. Here, we test the functional relevance of Nrxn1α in striatal circuits by employing optogenetic-mediated afferent recruitment of dorsal prefrontal cortical (dPFC) and parafascicular thalamic connections onto dorsomedial striatal (DMS) spiny projection neurons (SPNs). For dPFC-DMS circuits, we find decreased synaptic strength specifically onto indirect pathway SPNs in both Nrxn1α+/- and Nrxn1α-/- mice, driven by reductions in neurotransmitter release. In contrast, thalamic excitatory inputs to DMS exhibit relatively normal excitatory synaptic strength despite changes in synaptic N-methyl-D-aspartate receptor (NMDAR) content. These findings suggest that dysregulation of Nrxn1α modulates striatal function in an input- and target-specific manner.


Asunto(s)
Vías Aferentes/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cuerpo Estriado/metabolismo , Sinapsis Eléctricas/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Transmisión Sináptica , Vías Aferentes/citología , Animales , Proteínas de Unión al Calcio/genética , Cuerpo Estriado/citología , Sinapsis Eléctricas/genética , Potenciales Postsinápticos Excitadores , Ácido Glutámico/metabolismo , Heterocigoto , Homocigoto , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Moléculas de Adhesión de Célula Nerviosa/genética , Optogenética , Receptores de N-Metil-D-Aspartato/metabolismo
16.
Cell Syst ; 12(3): 263-271.e4, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33472027

RESUMEN

Neuronal loss can considerably diminish neural circuit function, impairing normal behavior by disrupting information flow in the circuit. Here, we use genetically engineered electrical synapses to reroute the flow of information in a C. elegans damaged chemosensory circuit in order to restore organism behavior. We impaired chemotaxis by removing one pair of interneurons from the circuit then artificially coupled two other adjacent neuron pairs by ectopically expressing the gap junction protein, connexin, in them. This restored chemotaxis in the animals. We expected to observe linear and direct information flow between the connexin-coupled neurons in the recovered circuit but also revealed the formation of new potent left-right lateral electrical connections within the connexin-expressing neuron pairs. Our analysis suggests that these additional electrical synapses help restore circuit function by amplifying weakened neuronal signals in the damaged circuit in addition to emulating the wild-type circuit. A record of this paper's transparent peer review process is included in the Supplemental Information.


Asunto(s)
Sinapsis Eléctricas/metabolismo , Ingeniería Genética/métodos , Animales , Caenorhabditis elegans
17.
Methods Mol Biol ; 2258: 93-103, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33340356

RESUMEN

Embryogenesis, as well as regeneration, is increasingly recognized to be orchestrated by an interplay of transcriptional and bioelectric networks. Spatiotemporal patterns of resting potentials direct the size, shape, and locations of numerous organ primordia during patterning. These bioelectrical properties are established by the function of ion channels and pumps that set voltage potentials of individual cells, and gap junctions (electrical synapses) that enable physiological states to propagate across tissue networks. Functional experiments to probe the roles of bioelectrical states can be carried out by targeting endogenous ion channels during development. Here, we describe protocols, optimized for the highly tractable Xenopus laevis embryo, for molecular genetic targeting of ion channels and connexins based on CRISPR, and monitoring of resting potential states using voltage-sensing fluorescent dye. Similar strategies can be adapted to other model species.


Asunto(s)
Sistemas CRISPR-Cas , Conexinas/metabolismo , Sinapsis Eléctricas/metabolismo , Edición Génica , Canales Iónicos/metabolismo , Xenopus laevis/metabolismo , Animales , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Conexinas/genética , Sinapsis Eléctricas/genética , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Canales Iónicos/genética , Potenciales de la Membrana , Microscopía Fluorescente , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Factores de Tiempo , Xenopus laevis/embriología , Xenopus laevis/genética
18.
Sci Rep ; 10(1): 18378, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110101

RESUMEN

Connexin36 (Cx36) is the most abundant connexin in central nervous system neurons. It forms gap junction channels that act as electrical synapses. Similar to chemical synapses, Cx36-containing gap junctions undergo activity-dependent plasticity and complex regulation. Cx36 gap junctions represent multimolecular complexes and contain cytoskeletal, regulatory and scaffolding proteins, which regulate channel conductance, assembly and turnover. The amino acid sequence of mammalian Cx36 harbors a phosphorylation site for the Ca2+/calmodulin-dependent kinase II at serine 315. This regulatory site is homologous to the serine 298 in perch Cx35 and in close vicinity to a PDZ binding domain at the very C-terminal end of the protein. We hypothesized that this phosphorylation site may serve as a molecular switch, influencing the affinity of the PDZ binding domain for its binding partners. Protein microarray and pulldown experiments revealed that this is indeed the case: phosphorylation of serine 298 decreased the binding affinity for MUPP1, a known scaffolding partner of connexin36, and increased the binding affinity for two different 14-3-3 proteins. Although we did not find the same effect in cell culture experiments, our data suggest that phosphorylation of serine 315/298 may serve to recruit different proteins to connexin36/35-containing gap junctions in an activity-dependent manner.


Asunto(s)
Proteínas 14-3-3/metabolismo , Conexinas/metabolismo , Dominios PDZ , Animales , Conexinas/química , Sinapsis Eléctricas/metabolismo , Uniones Comunicantes/metabolismo , Células HeLa , Humanos , Fosforilación , Unión Proteica , Proteína delta-6 de Union Comunicante
19.
Mol Pharmacol ; 98(4): 454-461, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32606204

RESUMEN

Endogenous opioid peptides in the amygdala regulate many of our behaviors and emotional responses. In particular, the endogenous opioid enkephalin plays a significant role in regulating amygdala activity, but its action is strongly limited by peptidases, which degrade enkephalin into inactive fragments. Inhibiting peptidases may be an attractive method to enhance endogenous opioid signaling; however, we do not know which specific peptidase(s) to target. Using inhibition of glutamate release onto the intercalated cells of the amygdala as an assay for enkephalin activity, we applied specific peptidase inhibitors to determine which peptidase(s) regulate enkephalin signaling in this region. Thiorphan (10 µM), captopril (1 µM), or bestatin (10 µM) were used to inhibit the activity of neprilysin, angiotensin-converting enzyme, or aminopeptidase N, respectively. In rat brain slices containing the intercalated cells, we found that inhibition of glutamate release by a submaximal concentration of enkephalin was doubled by application of all three peptidase inhibitors combined. Then, we tested inhibitors individually and found that inhibition of neprilysin alone could enhance enkephalin responses to the same extent as inhibitors of all three peptidases combined. This indicates neprilysin is the predominant peptidase responsible for degrading enkephalins in the intercalated cells of the amygdala. This differs from the striatum, locus coeruleus, and spinal cord, where multiple peptidases metabolize enkephalin. These data highlight the importance of knowing which specific peptidase(s) control opioid actions in the relevant neural circuit and how they change in disease states to allow rational choices of drugs targeting the specific peptidase of interest. SIGNIFICANCE STATEMENT: Endogenous opioids modulate many of our emotional and behavioral responses. In the amygdala, they modulate our pain, fear, and addictive behaviors. Their actions are terminated when they are catabolized into inactive fragments by at least three different peptidases. In this study, we found that neprilysin selectively controls endogenous opioid concentrations at synapses in the intercalated cells of the amygdala. This peptidase may be a target for regulation of endogenous opioid modulation of amygdala-mediated emotional and behavioral responses.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Encefalinas/metabolismo , Neprilisina/metabolismo , Inhibidores de Proteasas/farmacología , Animales , Captopril/farmacología , Sinapsis Eléctricas/efectos de los fármacos , Sinapsis Eléctricas/metabolismo , Ácido Glutámico/metabolismo , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Neprilisina/antagonistas & inhibidores , Proteolisis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tiorfan/farmacología
20.
J Neurosci ; 40(24): 4661-4672, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32393538

RESUMEN

It is widely assumed that electrical synapses in the mammalian brain, especially between interneurons, underlie neuronal synchrony. In the hippocampus, principal cells also establish electrical synapses with each other and have also been implicated in network oscillations, whereby the origin of fast electrical activity has been attributed to ectopic spikelets and dendro-dendritic or axo-axonal gap junctions. However, if electrical synapses were in axo-dendritic connections, where chemical synapses occur, the synaptic events would be mixed, having an electrical component preceding the chemical one. This type of communication is less well studied, mainly because it is not easily detected. Moreover, a possible scenario could be that an electrical synapse coexisted with a chemical one, but in a nonconductive state; hence, it would be considered inexistent. Could chemical synapses have a quiescent electrical component? If so, can silent electrical synapses be activated to be detected? We addressed this possibility, and we here report that, indeed, the connexin-36-containing glutamatergic mossy fiber synapses of the rat hippocampus express previously unrecognized electrical synapses, which are normally silent. We reveal that these synapses are pH sensitive, actuate in vitro and in vivo, and that the electrical signaling is bidirectional. With the simultaneous recording of hundreds of cells, we could reveal the existence of an electrical circuit in the hippocampus of adult rats of either sex consisting of principal cells where the nodes are interregional glutamatergic synapses containing silent but ready-to-use gap junctions.SIGNIFICANCE STATEMENT In this work, we present a series of experiments, both in vitro and in vivo, that reveal previously unrecognized silent pH-sensitive electrical synapses coexisting in one of the best studied glutamatergic synapses of the brain, the mossy fiber synapse of the hippocampus. This type of connectivity underlies an "electrical circuit" between two substructures of the adult rat hippocampus consisting of principal cells where the nodes are glutamatergic synapses containing silent but ready-to-use gap junctions. Its identification will allow us to explore the participation of such a circuit in physiological and pathophysiological functions and will provide valuable conceptual tools to understanding computational and regulatory mechanisms that may underlie network activity.


Asunto(s)
Sinapsis Eléctricas/fisiología , Uniones Comunicantes/fisiología , Ácido Glutámico/metabolismo , Hipocampo/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Transmisión Sináptica/fisiología , Animales , Células Cultivadas , Sinapsis Eléctricas/metabolismo , Uniones Comunicantes/metabolismo , Hipocampo/metabolismo , Masculino , Red Nerviosa/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA