Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Neural Circuits ; 17: 1125071, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37035505

RESUMEN

Acetylcholine and GABA are often co-released, including from VIP-expressing neurons of the cortex, cortically-projecting neurons of the globus pallidus externus and basal forebrain, and hippocampal-projecting neurons of the medial septum. The co-release of the functionally antagonistic neurotransmitters GABA and acetylcholine (ACh) greatly expands the possible functional effects of cholinergic neurons and provides an additional exogenous source of inhibition to the cortex. Transgene expression suggests that nearly all forebrain cholinergic neurons in mice at some point in development express Slc32a1, which encodes the vesicular GABA transporter (VGAT). To determine the degree of co-expression of GABA and Ach handling proteins, we measured expression in adult mice of Slc32a1, Gad1 and Gad2 (which encode GAD67 and GAD65, respectively, the GABA synthetic enzymes) in cholinergic neurons using fluorescent in situ hybridization. We found that only a subset of cholinergic neurons express the necessary machinery for GABA release at a single time in adult mice. This suggests that GABA co-release from cholinergic neurons is dynamic and potentially developmentally regulated. By measuring expression of Slc32a1, Gad1, Gad2, and Chat in the basal forebrain and medial septum in mice from post-natal day 0 to 28, we noted abundant yet variable expressions of GABAergic markers across early development, which are subsequently downregulated in adulthood. This is in contrast with the forebrain-projecting pedunculopontine nucleus, which showed no evidence of co-expression of GABAergic genes. These results suggest that expression of GABA signaling machinery in the cortically-projecting cholinergic system peaks during early development before settling at a non-zero level that is maintained through adulthood.


Asunto(s)
Acetilcolina , Ácido gamma-Aminobutírico , Ratones , Animales , Acetilcolina/metabolismo , Hibridación Fluorescente in Situ , Neuronas Colinérgicas/fisiología , Corteza Cerebral/metabolismo , Expresión Génica , Colina O-Acetiltransferasa/metabolismo
2.
Elife ; 92020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33043885

RESUMEN

Neurons communicate by the activity-dependent release of small-molecule neurotransmitters packaged into synaptic vesicles (SVs). Although many molecules have been identified as neurotransmitters, technical limitations have precluded a full metabolomic analysis of SV content. Here, we present a workflow to rapidly isolate SVs and to interrogate their metabolic contents at high-resolution using mass spectrometry. We validated the enrichment of glutamate in SVs of primary cortical neurons using targeted polar metabolomics. Unbiased and extensive global profiling of SVs isolated from these neurons revealed that the only detectable polar metabolites they contain are the established neurotransmitters glutamate and GABA. In addition, we adapted the approach to enable quick capture of SVs directly from brain tissue and determined the neurotransmitter profiles of diverse brain regions in a cell-type-specific manner. The speed, robustness, and precision of this method to interrogate SV contents will facilitate novel insights into the chemical basis of neurotransmission.


Asunto(s)
Encéfalo/metabolismo , Espectrometría de Masas/métodos , Metaboloma , Metabolómica/métodos , Vesículas Sinápticas/metabolismo , Animales , Femenino , Ácido Glutámico/metabolismo , Masculino , Ratones
4.
Nat Chem Biol ; 14(4): 352-360, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29483642

RESUMEN

We developed a new way to engineer complex proteins toward multidimensional specifications using a simple, yet scalable, directed evolution strategy. By robotically picking mammalian cells that were identified, under a microscope, as expressing proteins that simultaneously exhibit several specific properties, we can screen hundreds of thousands of proteins in a library in just a few hours, evaluating each along multiple performance axes. To demonstrate the power of this approach, we created a genetically encoded fluorescent voltage indicator, simultaneously optimizing its brightness and membrane localization using our microscopy-guided cell-picking strategy. We produced the high-performance opsin-based fluorescent voltage reporter Archon1 and demonstrated its utility by imaging spiking and millivolt-scale subthreshold and synaptic activity in acute mouse brain slices and in larval zebrafish in vivo. We also measured postsynaptic responses downstream of optogenetically controlled neurons in C. elegans.


Asunto(s)
Evolución Molecular Dirigida/métodos , Proteínas Luminiscentes/química , Ingeniería de Proteínas/métodos , Robótica , Pez Cebra/embriología , Animales , Encéfalo/diagnóstico por imagen , Caenorhabditis elegans , Separación Celular , Femenino , Citometría de Flujo , Fluorescencia , Biblioteca de Genes , Genes Reporteros , Células HEK293 , Hipocampo/citología , Humanos , Masculino , Ratones , Microscopía Fluorescente , Neuronas/citología , Optogenética
5.
Neuron ; 96(5): 1070-1083.e5, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29154125

RESUMEN

Protein kinase A (PKA) integrates inputs from G-protein-coupled neuromodulator receptors to modulate synaptic and cellular function. Gαs signaling stimulates PKA activity, whereas Gαi inhibits PKA activity. Gαq, on the other hand, signals through phospholipase C, and it remains unclear whether Gαq-coupled receptors signal to PKA in their native context. Here, using two independent optical reporters of PKA activity in acute mouse hippocampus slices, we show that endogenous Gαq-coupled muscarinic acetylcholine receptors activate PKA. Mechanistically, this effect is mediated by parallel signaling via either calcium or protein kinase C. Furthermore, multiple Gαq-coupled receptors modulate phosphorylation by PKA, a classical Gαs/Gαi effector. Thus, these results highlight PKA as a biochemical integrator of three major types of GPCRs and necessitate reconsideration of classic models used to predict neuronal signaling in response to the large family of Gαq-coupled receptors.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Receptores de Neurotransmisores/metabolismo , Animales , Señalización del Calcio/genética , Señalización del Calcio/fisiología , Activación Enzimática/genética , Femenino , Células HEK293 , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Embarazo , Proteína Quinasa C/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Transducción de Señal/genética
6.
Front Pharmacol ; 7: 46, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26941646

RESUMEN

[This corrects the article on p. 56 in vol. 5, PMID: 24765076.].

7.
PLoS One ; 9(8): e105584, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25140704

RESUMEN

The prokaryotic adaptive immune system CRISPR/Cas9 has recently been adapted for genome editing in eukaryotic cells. This technique allows for sequence-specific induction of double-strand breaks in genomic DNA of individual cells, effectively resulting in knock-out of targeted genes. It thus promises to be an ideal candidate for application in neuroscience where constitutive genetic modifications are frequently either lethal or ineffective due to adaptive changes of the brain. Here we use CRISPR/Cas9 to knock-out Grin1, the gene encoding the obligatory NMDA receptor subunit protein GluN1, in a sparse population of mouse pyramidal neurons. Within this genetically mosaic tissue, manipulated cells lack synaptic current mediated by NMDA-type glutamate receptors consistent with complete knock-out of the targeted gene. Our results show the first proof-of-principle demonstration of CRISPR/Cas9-mediated knock-down in neurons in vivo, where it can be a useful tool to study the function of specific proteins in neuronal circuits.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen/métodos , Proteínas del Tejido Nervioso/genética , Células Piramidales/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animales , Ratones , Proteínas del Tejido Nervioso/metabolismo , Células Piramidales/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciales Sinápticos
8.
Nat Methods ; 11(8): 825-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24952910

RESUMEN

All-optical electrophysiology-spatially resolved simultaneous optical perturbation and measurement of membrane voltage-would open new vistas in neuroscience research. We evolved two archaerhodopsin-based voltage indicators, QuasAr1 and QuasAr2, which show improved brightness and voltage sensitivity, have microsecond response times and produce no photocurrent. We engineered a channelrhodopsin actuator, CheRiff, which shows high light sensitivity and rapid kinetics and is spectrally orthogonal to the QuasArs. A coexpression vector, Optopatch, enabled cross-talk-free genetically targeted all-optical electrophysiology. In cultured rat neurons, we combined Optopatch with patterned optical excitation to probe back-propagating action potentials (APs) in dendritic spines, synaptic transmission, subcellular microsecond-timescale details of AP propagation, and simultaneous firing of many neurons in a network. Optopatch measurements revealed homeostatic tuning of intrinsic excitability in human stem cell-derived neurons. In rat brain slices, Optopatch induced and reported APs and subthreshold events with high signal-to-noise ratios. The Optopatch platform enables high-throughput, spatially resolved electrophysiology without the use of conventional electrodes.


Asunto(s)
Mamíferos/fisiología , Neuronas/fisiología , Rodopsina/fisiología , Animales , Evolución Molecular Dirigida , Proteínas Recombinantes/metabolismo , Transmisión Sináptica
9.
Front Pharmacol ; 5: 56, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24765076

RESUMEN

Neuromodulators have profound effects on behavior, but the dynamics of their intracellular effectors has remained unclear. Most neuromodulators exert their function via G-protein-coupled receptors (GPCRs). One major challenge for understanding neuromodulator action is the lack of dynamic readouts of the biochemical signals produced by GPCR activation. The adenylate cyclase/cyclic AMP/protein kinase A (PKA) module is a central component of such biochemical signaling. This module is regulated by several behaviorally important neuromodulator receptors. Furthermore, PKA activity is necessary for the induction of many forms of synaptic plasticity as well as for the formation of long-term memory. In order to monitor PKA activity in brain tissue, we have developed a 2-photon fluorescence lifetime imaging microscopy (2pFLIM) compatible PKA sensor termed FLIM-AKAR, which is based on the ratiometric FRET sensor AKAR3. FLIM-AKAR shows a large dynamic range and little pH sensitivity. In addition, it is a rapidly diffusible cytoplasmic protein that specifically reports net PKA activity in situ. FLIM-AKAR expresses robustly in various brain regions with multiple transfection methods, can be targeted to genetically identified cell types, and responds to activation of both endogenous GPCRs and spatial-temporally specific delivery of glutamate. Initial experiments reveal differential regulation of PKA activity across subcellular compartments in response to neuromodulator inputs. Therefore, the reporter FLIM-AKAR, coupled with 2pFLIM, enables the study of PKA activity in response to neuromodulator inputs in genetically identified neurons in the brain, and sheds light on the intracellular dynamics of endogenous GPCR activation.

10.
J Neurosci ; 34(3): 869-79, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431445

RESUMEN

The NMDA-type glutamate receptor (NMDAR) is essential for synaptogenesis, synaptic plasticity, and higher cognitive function. Emerging evidence indicates that NMDAR Ca(2+) permeability is under the control of cAMP/protein kinase A (PKA) signaling. Whereas the functional impact of PKA on NMDAR-dependent Ca(2+) signaling is well established, the molecular target remains unknown. Here we identify serine residue 1166 (Ser1166) in the carboxy-terminal tail of the NMDAR subunit GluN2B to be a direct molecular and functional target of PKA phosphorylation critical to NMDAR-dependent Ca(2+) permeation and Ca(2+) signaling in spines. Activation of ß-adrenergic and D1/D5-dopamine receptors induces Ser1166 phosphorylation. Loss of this single phosphorylation site abolishes PKA-dependent potentiation of NMDAR Ca(2+) permeation, synaptic currents, and Ca(2+) rises in dendritic spines. We further show that adverse experience in the form of forced swim, but not exposure to fox urine, elicits striking phosphorylation of Ser1166 in vivo, indicating differential impact of different forms of stress. Our data identify a novel molecular and functional target of PKA essential to NMDAR-mediated Ca(2+) signaling at synapses and regulated by the emotional response to stress.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Espinas Dendríticas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Espinas Dendríticas/genética , Zorros , Células HEK293 , Hipocampo/metabolismo , Humanos , Inhibición Neural/fisiología , Fosforilación/fisiología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/fisiología , Serina/genética , Estrés Psicológico/genética , Estrés Psicológico/metabolismo
11.
Front Mol Neurosci ; 6: 28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24065881

RESUMEN

Neurons use a variety of mechanisms to homeostatically regulate neural network activity in order to maintain firing in a bounded range. One such process involves the bi-directional modulation of excitatory synaptic drive in response to chronic changes in network activity. Down-scaling of excitatory synapses in response to high activity requires Arc-dependent endocytosis of glutamate receptors. However, the temporal dynamics and signaling pathways regulating Arc during homeostatic plasticity are not well understood. Here we determine the relative contribution of transcriptional and translational control in the regulation of Arc, the signaling pathways responsible for the activity-dependent production of Arc, and the time course of these signaling events as they relate to the homeostatic adjustment of network activity in hippocampal neurons. We find that an ERK1/2-dependent transcriptional pathway active within 1-2 h of up-regulated network activity induces Arc leading to a restoration of network spiking rates within 12 h. Under basal and low activity conditions, specialized mechanisms are in place to rapidly degrade Arc mRNA and protein such that they have half-lives of less than 1 h. In addition, we find that while mTOR signaling is regulated by network activity on a similar time scale, mTOR-dependent translational control is not a major regulator of Arc production or degradation suggesting that the signaling pathways underlying homeostatic plasticity are distinct from those mediating synapse-specific forms of synaptic depression.

12.
Neuron ; 78(3): 510-22, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23664616

RESUMEN

Neural circuits are regulated by activity-dependent feedback systems that tightly control network excitability and which are thought to be crucial for proper brain development. Defects in the ability to establish and maintain network homeostasis may be central to the pathogenesis of neurodevelopmental disorders. Here, we examine the function of the tuberous sclerosis complex (TSC)-mTOR signaling pathway, a common target of mutations associated with epilepsy and autism spectrum disorder, in regulating activity-dependent processes in the mouse hippocampus. We find that the TSC-mTOR pathway is a central component of a positive feedback loop that promotes network activity by repressing inhibitory synapses onto excitatory neurons. In Tsc1 KO neurons, weakened inhibition caused by deregulated mTOR alters the balance of excitatory and inhibitory synaptic transmission, leading to hippocampal hyperexcitability. These findings identify the TSC-mTOR pathway as a regulator of neural network activity and have implications for the neurological dysfunction in disorders exhibiting deregulated mTOR signaling.


Asunto(s)
Hipocampo/fisiopatología , Sinapsis/fisiología , Esclerosis Tuberosa/fisiopatología , Proteínas Supresoras de Tumor/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
13.
Glia ; 61(6): 985-1002, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536313

RESUMEN

Transforming growth factor ß1 (TGF-ß1) is a pleiotropic cytokine expressed throughout the CNS. Previous studies demonstrated that TGF-ß1 contributes to maintain neuronal survival, but mechanistically this effect is not well understood. We generated a CNS-specific TGF-ß1-deficient mouse model to investigate the functional consequences of TGF-ß1-deficiency in the adult mouse brain. We found that depletion of TGF-ß1 in the CNS resulted in a loss of the astrocyte glutamate transporter (GluT) proteins GLT-1 (EAAT2) and GLAST (EAAT1) and decreased glutamate uptake in the mouse hippocampus. Treatment with TGF-ß1 induced the expression of GLAST and GLT-1 in cultured astrocytes and enhanced astroglial glutamate uptake. Similar to GLT-1-deficient mice, CNS-TGF-ß1-deficient mice had reduced brain weight and neuronal loss in the CA1 hippocampal region. CNS-TGF-ß1-deficient mice showed GluN2B-dependent aberrant synaptic plasticity in the CA1 area of the hippocampus similar to the glutamate transport inhibitor DL-TBOA and these mice were highly sensitive to excitotoxic injury. In addition, hippocampal neurons from TGF-ß1-deficient mice had elevated GluN2B-mediated calcium signals in response to extrasynaptic glutamate receptor stimulation, whereas cells treated with TGF-ß1 exhibited reduced GluN2B-mediated calcium signals. In summary, our study demonstrates a previously unrecognized function of TGF-ß1 in the CNS to control extracellular glutamate homeostasis and GluN2B-mediated calcium responses in the mouse hippocampus.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Apoptosis/fisiología , Espinas Dendríticas/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Transmisión Sináptica/fisiología , Factor de Crecimiento Transformador beta1/genética
14.
Nat Neurosci ; 15(12): 1667-74, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23143522

RESUMEN

Members of the neuroligin family of cell-adhesion proteins are found at excitatory and inhibitory synapses and are mutated in some familial forms of autism spectrum disorders. Although they display synaptogenic properties in heterologous systems, the function of neuroligins in vivo in the regulation of synapse formation and synapse number has been difficult to establish. We found that neuroligin-1 (NL1), which is located at excitatory postsynaptic densities, regulates activity-dependent synaptogenesis and mature synapse number on cortical layer 2/3 pyramidal neurons in vivo. However, synapse number was not sensitive to absolute NL1 levels but instead depended on transcellular differences in the relative amounts of NL1. These effects were independent of the cell-autonomous regulation of NMDA-type glutamate receptors by absolute levels of NL1. Our data indicate that transcellular competitive processes govern synapse formation and number in developing cortex and that NL1 has a central function in these processes.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Neurogénesis/fisiología , Sinapsis/fisiología , Animales , Comunicación Celular/fisiología , Recuento de Células , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Embarazo , Ratas , Ratas Sprague-Dawley
15.
J Neurosci ; 31(24): 8862-9, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677170

RESUMEN

The autism spectrum disorder tuberous sclerosis complex (TSC) is caused by mutations in the Tsc1 or Tsc2 genes, whose protein products form a heterodimeric complex that negatively regulates mammalian target of rapamycin-dependent protein translation. Although several forms of synaptic plasticity, including metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD), depend on protein translation at the time of induction, it is unknown whether these forms of plasticity require signaling through the Tsc1/2 complex. To examine this possibility, we postnatally deleted Tsc1 in vivo in a subset of hippocampal CA1 neurons using viral delivery of Cre recombinase in mice. We found that hippocampal mGluR-LTD was abolished by loss of Tsc1, whereas a protein synthesis-independent form of NMDA receptor-dependent LTD was preserved. Additionally, AMPA and NMDA receptor-mediated EPSCs and miniature spontaneous EPSC frequency were enhanced in Tsc1 KO neurons. These changes in synaptic function occurred in the absence of alterations in spine density, morphology, or presynaptic release probability. Our findings indicate that signaling through Tsc1/2 is required for the expression of specific forms of hippocampal synaptic plasticity as well as the maintenance of normal excitatory synaptic strength. Furthermore, these data suggest that perturbations of synaptic signaling may contribute to the pathogenesis of TSC.


Asunto(s)
Región CA1 Hipocampal/citología , Depresión Sináptica a Largo Plazo/genética , Neuronas/fisiología , Receptores de Glutamato/metabolismo , Simportadores/deficiencia , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Espinas Dendríticas/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Proteínas Fluorescentes Verdes/genética , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos , N-Metilaspartato/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Receptores de Glutamato/genética , Serina/genética , Sinapsis/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...