Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Cell ; 186(19): 4117-4133.e22, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37591239

RESUMEN

Aging is the key risk factor for cognitive decline, yet the molecular changes underlying brain aging remain poorly understood. Here, we conducted spatiotemporal RNA sequencing of the mouse brain, profiling 1,076 samples from 15 regions across 7 ages and 2 rejuvenation interventions. Our analysis identified a brain-wide gene signature of aging in glial cells, which exhibited spatially defined changes in magnitude. By integrating spatial and single-nucleus transcriptomics, we found that glial aging was particularly accelerated in white matter compared with cortical regions, whereas specialized neuronal populations showed region-specific expression changes. Rejuvenation interventions, including young plasma injection and dietary restriction, exhibited distinct effects on gene expression in specific brain regions. Furthermore, we discovered differential gene expression patterns associated with three human neurodegenerative diseases, highlighting the importance of regional aging as a potential modulator of disease. Our findings identify molecular foci of brain aging, providing a foundation to target age-related cognitive decline.


Asunto(s)
Envejecimiento , Disfunción Cognitiva , Sustancia Blanca , Animales , Humanos , Ratones , Disfunción Cognitiva/genética , Perfilación de la Expresión Génica , Núcleo Solitario , Sustancia Blanca/patología , Análisis de Expresión Génica de una Sola Célula , Encéfalo/patología
2.
Nature ; 610(7931): 319-326, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36224417

RESUMEN

Self-organizing neural organoids represent a promising in vitro platform with which to model human development and disease1-5. However, organoids lack the connectivity that exists in vivo, which limits maturation and makes integration with other circuits that control behaviour impossible. Here we show that human stem cell-derived cortical organoids transplanted into the somatosensory cortex of newborn athymic rats develop mature cell types that integrate into sensory and motivation-related circuits. MRI reveals post-transplantation organoid growth across multiple stem cell lines and animals, whereas single-nucleus profiling shows progression of corticogenesis and the emergence of activity-dependent transcriptional programs. Indeed, transplanted cortical neurons display more complex morphological, synaptic and intrinsic membrane properties than their in vitro counterparts, which enables the discovery of defects in neurons derived from individuals with Timothy syndrome. Anatomical and functional tracings show that transplanted organoids receive thalamocortical and corticocortical inputs, and in vivo recordings of neural activity demonstrate that these inputs can produce sensory responses in human cells. Finally, cortical organoids extend axons throughout the rat brain and their optogenetic activation can drive reward-seeking behaviour. Thus, transplanted human cortical neurons mature and engage host circuits that control behaviour. We anticipate that this approach will be useful for detecting circuit-level phenotypes in patient-derived cells that cannot otherwise be uncovered.


Asunto(s)
Vías Nerviosas , Organoides , Animales , Animales Recién Nacidos , Trastorno Autístico , Humanos , Síndrome de QT Prolongado , Motivación , Neuronas/fisiología , Optogenética , Organoides/citología , Organoides/inervación , Organoides/trasplante , Ratas , Recompensa , Corteza Somatosensorial/citología , Corteza Somatosensorial/fisiología , Células Madre/citología , Sindactilia
3.
Nature ; 594(7862): 277-282, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34040258

RESUMEN

Neurons have recently emerged as essential cellular constituents of the tumour microenvironment, and their activity has been shown to increase the growth of a diverse number of solid tumours1. Although the role of neurons in tumour progression has previously been demonstrated2, the importance of neuronal activity to tumour initiation is less clear-particularly in the setting of cancer predisposition syndromes. Fifteen per cent of individuals with the neurofibromatosis 1 (NF1) cancer predisposition syndrome (in which tumours arise in close association with nerves) develop low-grade neoplasms of the optic pathway (known as optic pathway gliomas (OPGs)) during early childhood3,4, raising  the possibility that postnatal light-induced activity of the optic nerve drives tumour initiation. Here we use an authenticated mouse model of OPG driven by mutations in the neurofibromatosis 1 tumour suppressor gene (Nf1)5 to demonstrate that stimulation of optic nerve activity increases optic glioma growth, and that decreasing visual experience via light deprivation prevents tumour formation and maintenance. We show that the initiation of Nf1-driven OPGs (Nf1-OPGs) depends on visual experience during a developmental period in which Nf1-mutant mice are susceptible to tumorigenesis. Germline Nf1 mutation in retinal neurons results in aberrantly increased shedding of neuroligin 3 (NLGN3) within the optic nerve in response to retinal neuronal activity. Moreover, genetic Nlgn3 loss or pharmacological inhibition of NLGN3 shedding blocks the formation and progression of Nf1-OPGs. Collectively, our studies establish an obligate role for neuronal activity in the development of some types of brain tumours, elucidate a therapeutic strategy to reduce OPG incidence or mitigate tumour progression, and underscore the role of Nf1mutation-mediated dysregulation of neuronal signalling pathways in mouse models of the NF1 cancer predisposition syndrome.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes de Neurofibromatosis 1 , Mutación , Neurofibromina 1/genética , Neuronas/metabolismo , Glioma del Nervio Óptico/genética , Glioma del Nervio Óptico/patología , Animales , Astrocitoma/genética , Astrocitoma/patología , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Transformación Celular Neoplásica/efectos de la radiación , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de la radiación , Nervio Óptico/citología , Nervio Óptico/efectos de la radiación , Estimulación Luminosa , Retina/citología , Retina/efectos de la radiación
4.
Proc Natl Acad Sci U S A ; 119(43): e2210122119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36256819

RESUMEN

Hyperexcitability of brain circuits is a common feature of autism spectrum disorders (ASDs). Genetic deletion of a chromatin-binding protein, retinoic acid induced 1 (RAI1), causes Smith-Magenis syndrome (SMS). SMS is a syndromic ASD associated with intellectual disability, autistic features, maladaptive behaviors, overt seizures, and abnormal electroencephalogram (EEG) patterns. The molecular and neural mechanisms underlying abnormal brain activity in SMS remain unclear. Here we show that panneural Rai1 deletions in mice result in increased seizure susceptibility and prolonged hippocampal seizure duration in vivo and increased dentate gyrus population spikes ex vivo. Brain-wide mapping of neuronal activity pinpointed selective cell types within the limbic system, including the hippocampal dentate gyrus granule cells (dGCs) that are hyperactivated by chemoconvulsant administration or sensory experience in Rai1-deficient brains. Deletion of Rai1 from glutamatergic neurons, but not from gamma-aminobutyric acidergic (GABAergic) neurons, was responsible for increased seizure susceptibility. Deleting Rai1 from the Emx1Cre-lineage glutamatergic neurons resulted in abnormal dGC properties, including increased excitatory synaptic transmission and increased intrinsic excitability. Our work uncovers the mechanism of neuronal hyperexcitability in SMS by identifying Rai1 as a negative regulator of dGC intrinsic and synaptic excitability.


Asunto(s)
Síndrome de Smith-Magenis , Ratones , Animales , Síndrome de Smith-Magenis/genética , Transactivadores/genética , Transactivadores/metabolismo , Fenotipo , Modelos Animales de Enfermedad , Cromatina , Hipocampo/metabolismo , Convulsiones/genética , Tretinoina
5.
Nature ; 545(7652): 54-59, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28445465

RESUMEN

The development of the nervous system involves a coordinated succession of events including the migration of GABAergic (γ-aminobutyric-acid-releasing) neurons from ventral to dorsal forebrain and their integration into cortical circuits. However, these interregional interactions have not yet been modelled with human cells. Here we generate three-dimensional spheroids from human pluripotent stem cells that resemble either the dorsal or ventral forebrain and contain cortical glutamatergic or GABAergic neurons. These subdomain-specific forebrain spheroids can be assembled in vitro to recapitulate the saltatory migration of interneurons observed in the fetal forebrain. Using this system, we find that in Timothy syndrome-a neurodevelopmental disorder that is caused by mutations in the CaV1.2 calcium channel-interneurons display abnormal migratory saltations. We also show that after migration, interneurons functionally integrate with glutamatergic neurons to form a microphysiological system. We anticipate that this approach will be useful for studying neural development and disease, and for deriving spheroids that resemble other brain regions to assemble circuits in vitro.


Asunto(s)
Neuronas/citología , Prosencéfalo/citología , Prosencéfalo/crecimiento & desarrollo , Esferoides Celulares/citología , Trastorno Autístico/genética , Trastorno Autístico/patología , Línea Celular , Movimiento Celular , Células Cultivadas , Femenino , Neuronas GABAérgicas/citología , Ácido Glutámico/metabolismo , Humanos , Interneuronas/citología , Interneuronas/patología , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/patología , Masculino , Modelos Biológicos , Neurogénesis , Neuronas/patología , Células Madre Pluripotentes/citología , Prosencéfalo/anatomía & histología , Sinapsis/fisiología , Sindactilia/genética , Sindactilia/patología
6.
Proc Natl Acad Sci U S A ; 117(51): 32711-32721, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33277431

RESUMEN

CLC-2 is a voltage-gated chloride channel that is widely expressed in mammalian tissues. In the central nervous system, CLC-2 appears in neurons and glia. Studies to define how this channel contributes to normal and pathophysiological function in the central nervous system raise questions that remain unresolved, in part due to the absence of precise pharmacological tools for modulating CLC-2 activity. Herein, we describe the development and optimization of AK-42, a specific small-molecule inhibitor of CLC-2 with nanomolar potency (IC50 = 17 ± 1 nM). AK-42 displays unprecedented selectivity (>1,000-fold) over CLC-1, the closest CLC-2 homolog, and exhibits no off-target engagement against a panel of 61 common channels, receptors, and transporters expressed in brain tissue. Computational docking, validated by mutagenesis and kinetic studies, indicates that AK-42 binds to an extracellular vestibule above the channel pore. In electrophysiological recordings of mouse CA1 hippocampal pyramidal neurons, AK-42 acutely and reversibly inhibits CLC-2 currents; no effect on current is observed on brain slices taken from CLC-2 knockout mice. These results establish AK-42 as a powerful tool for investigating CLC-2 neurophysiology.


Asunto(s)
Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Sitios de Unión , Células CHO , Canales de Cloruro CLC-2 , Línea Celular , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cricetulus , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Hipocampo/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación del Acoplamiento Molecular , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad
7.
Nat Methods ; 16(1): 75-78, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573846

RESUMEN

The differentiation of pluripotent stem cells in three-dimensional cultures can recapitulate key aspects of brain development, but protocols are prone to variable results. Here we differentiated multiple human pluripotent stem cell lines for over 100 d using our previously developed approach to generate brain-region-specific organoids called cortical spheroids and, using several assays, found that spheroid generation was highly reliable and consistent. We anticipate the use of this approach for large-scale differentiation experiments and disease modeling.


Asunto(s)
Organoides/crecimiento & desarrollo , Ingeniería de Tejidos , Línea Celular , Humanos , Células Madre Pluripotentes/citología , Prosencéfalo/fisiología , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos
8.
Nat Methods ; 12(7): 671-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26005811

RESUMEN

The human cerebral cortex develops through an elaborate succession of cellular events that, when disrupted, can lead to neuropsychiatric disease. The ability to reprogram somatic cells into pluripotent cells that can be differentiated in vitro provides a unique opportunity to study normal and abnormal corticogenesis. Here, we present a simple and reproducible 3D culture approach for generating a laminated cerebral cortex-like structure, named human cortical spheroids (hCSs), from pluripotent stem cells. hCSs contain neurons from both deep and superficial cortical layers and map transcriptionally to in vivo fetal development. These neurons are electrophysiologically mature, display spontaneous activity, are surrounded by nonreactive astrocytes and form functional synapses. Experiments in acute hCS slices demonstrate that cortical neurons participate in network activity and produce complex synaptic events. These 3D cultures should allow a detailed interrogation of human cortical development, function and disease, and may prove a versatile platform for generating other neuronal and glial subtypes in vitro.


Asunto(s)
Astrocitos/fisiología , Corteza Cerebral/fisiología , Células Madre Pluripotentes/citología , Astrocitos/citología , Células Cultivadas , Corteza Cerebral/citología , Humanos , Esferoides Celulares , Sinapsis/fisiología
9.
Proc Natl Acad Sci U S A ; 112(12): 3805-10, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25775522

RESUMEN

Initiating and regulating vertebrate reproduction requires pulsatile release of gonadotropin-releasing hormone (GnRH1) from the hypothalamus. Coordinated GnRH1 release, not simply elevated absolute levels, effects the release of pituitary gonadotropins that drive steroid production in the gonads. However, the mechanisms underlying synchronization of GnRH1 neurons are unknown. Control of synchronicity by gap junctions between GnRH1 neurons has been proposed but not previously found. We recorded simultaneously from pairs of transgenically labeled GnRH1 neurons in adult male Astatotilapia burtoni cichlid fish. We report that GnRH1 neurons are strongly and uniformly interconnected by electrical synapses that can drive spiking in connected cells and can be reversibly blocked by meclofenamic acid. Our results suggest that electrical synapses could promote coordinated spike firing in a cellular assemblage of GnRH1 neurons to produce the pulsatile output necessary for activation of the pituitary and reproduction.


Asunto(s)
Cíclidos/fisiología , Sinapsis Eléctricas , Hormona Liberadora de Gonadotropina/metabolismo , Animales , Conexinas/metabolismo , Femenino , Uniones Comunicantes , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/metabolismo , Hibridación in Situ , Masculino , Ácido Meclofenámico/química , Modelos Neurológicos , Neuronas/fisiología , Hipófisis/metabolismo , Transmisión Sináptica , Transgenes
10.
Brain ; 139(Pt 2): 468-80, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26685158

RESUMEN

Ischaemic stroke is the leading cause of severe long-term disability yet lacks drug therapies that promote the repair phase of recovery. This repair phase of stroke occurs days to months after stroke onset and involves brain remapping and plasticity within the peri-infarct zone. Elucidating mechanisms that promote this plasticity is critical for the development of new therapeutics with a broad treatment window. Inhibiting tonic (extrasynaptic) GABA signalling during the repair phase was reported to enhance functional recovery in mice suggesting that GABA plays an important function in modulating brain repair. While tonic GABA appears to suppress brain repair after stroke, less is known about the role of phasic (synaptic) GABA during the repair phase. We observed an increase in postsynaptic phasic GABA signalling in mice within the peri-infarct cortex specific to layer 5; we found increased numbers of α1 receptor subunit-containing GABAergic synapses detected using array tomography, and an associated increased efficacy of spontaneous and miniature inhibitory postsynaptic currents in pyramidal neurons. Furthermore, we demonstrate that enhancing phasic GABA signalling using zolpidem, a Food and Drug Administration (FDA)-approved GABA-positive allosteric modulator, during the repair phase improved behavioural recovery. These data identify potentiation of phasic GABA signalling as a novel therapeutic strategy, indicate zolpidem's potential to improve recovery, and underscore the necessity to distinguish the role of tonic and phasic GABA signalling in stroke recovery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Agonistas de Receptores de GABA-A/administración & dosificación , Inhibición Neural/fisiología , Piridinas/administración & dosificación , Receptores de GABA-A/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Sistemas de Liberación de Medicamentos/tendencias , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/efectos de los fármacos , Neocórtex/fisiología , Inhibición Neural/efectos de los fármacos , Técnicas de Cultivo de Órganos , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Zolpidem
11.
Nature ; 477(7363): 171-8, 2011 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-21796121

RESUMEN

Severe behavioural deficits in psychiatric diseases such as autism and schizophrenia have been hypothesized to arise from elevations in the cellular balance of excitation and inhibition (E/I balance) within neural microcircuitry. This hypothesis could unify diverse streams of pathophysiological and genetic evidence, but has not been susceptible to direct testing. Here we design and use several novel optogenetic tools to causally investigate the cellular E/I balance hypothesis in freely moving mammals, and explore the associated circuit physiology. Elevation, but not reduction, of cellular E/I balance within the mouse medial prefrontal cortex was found to elicit a profound impairment in cellular information processing, associated with specific behavioural impairments and increased high-frequency power in the 30-80 Hz range, which have both been observed in clinical conditions in humans. Consistent with the E/I balance hypothesis, compensatory elevation of inhibitory cell excitability partially rescued social deficits caused by E/I balance elevation. These results provide support for the elevated cellular E/I balance hypothesis of severe neuropsychiatric disease-related symptoms.


Asunto(s)
Modelos Neurológicos , Inhibición Neural/fisiología , Neuronas/metabolismo , Corteza Prefrontal/fisiología , Corteza Prefrontal/fisiopatología , Conducta Social , Animales , Trastorno Autístico/fisiopatología , Modelos Animales de Enfermedad , Células HEK293 , Hipocampo/citología , Humanos , Aprendizaje , Trastornos Mentales/fisiopatología , Ratones , Actividad Motora , Opsinas/metabolismo , Esquizofrenia/fisiopatología
12.
J Neurosci ; 35(2): 761-75, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589769

RESUMEN

The modulation of gamma power (25-90 Hz) is associated with attention and has been observed across species and brain areas. However, mechanisms that control these modulations are poorly understood. The midbrain spatial attention network in birds generates high-amplitude gamma oscillations in the local field potential that are thought to represent the highest priority location for attention. Here we explore, in midbrain slices from chickens, mechanisms that regulate the power of these oscillations, using high-resolution techniques including intracellular recordings from neurons targeted by calcium imaging. The results identify a specific subtype of neuron, expressing non-α7 nicotinic acetylcholine receptors, that directly drives inhibition in the gamma-generating circuit and switches the network into a primed state capable of producing high-amplitude oscillations. The special properties of this mechanism enable rapid, persistent changes in gamma power. The brain may employ this mechanism wherever rapid modulations of gamma power are critical to information processing.


Asunto(s)
Atención , Neuronas Colinérgicas/fisiología , Ritmo Gamma , Mesencéfalo/fisiología , Animales , Células Cultivadas , Pollos , Neuronas Colinérgicas/metabolismo , Femenino , Masculino , Mesencéfalo/citología , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo
13.
J Neurophysiol ; 116(3): 995-1011, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27281752

RESUMEN

The thalamic reticular nucleus (nRt), composed of GABAergic cells providing inhibition of relay neurons in the dorsal thalamus, receives excitation from the neocortex and thalamus. The two excitatory pathways promoting feedback or feedforward inhibition of thalamocortical neurons contribute to sensory processing and rhythm generation. While synaptic inhibition within the nRt has been carefully characterized, little is known regarding the biophysics of synaptic excitation. To characterize the functional properties of thalamocortical and corticothalamic connections to the nRt, we recorded minimal electrically evoked excitatory postsynaptic currents from nRt cells in vitro. A hierarchical clustering algorithm distinguished two types of events. Type 1 events had larger amplitudes and faster kinetics, largely mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, whereas type 2 responses had more prominent N-methyl-d-aspartate (NMDA) receptor contribution. Type 1 responses showed subnormal axonal propagation and paired pulse depression, consistent with thalamocortical inputs. Furthermore, responses kinetically similar to type 1 events were evoked by glutamate-mediated activation of thalamic neurons. Type 2 responses, in contrast, likely arise from corticothalamic inputs, with larger NMDA conductance and weak Mg(2+)-dependent block, suggesting that NMDA receptors are critical for the cortical excitation of reticular neurons. The long-lasting action of NMDA receptors would promote reticular cell burst firing and produce powerful inhibitory output to relay neurons proposed to be important in triggering epilepsy. This work provides the first complete voltage-clamp analysis of the kinetics and voltage dependence of AMPA and NMDA responses of thalamocortical and corticothalamic synapses in the nRt and will be critical in optimizing biologically realistic neural network models of thalamocortical circuits relevant to sensory processing and thalamocortical oscillations.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Neuronas/fisiología , Sinapsis/clasificación , Sinapsis/fisiología , Núcleos Talámicos/citología , Análisis de Varianza , Animales , Animales Recién Nacidos , Biofisica , Análisis por Conglomerados , Estimulación Eléctrica , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/farmacología , Técnicas In Vitro , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
J Neurophysiol ; 115(3): 1183-95, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631150

RESUMEN

The GABAergic neurons of the thalamic reticular nucleus (nRt) provide the primary source of inhibition within the thalamus. Using physiology, pharmacology, and immunohistochemistry in mice, we characterized postsynaptic developmental changes in these inhibitory projection neurons. First, at postnatal days 3-5 (P3-5), inhibitory postsynaptic currents (IPSCs) decayed very slowly, followed by a biphasic developmental progression, becoming faster at P6-8 and then slower again at P9-11 before stabilizing in a mature form around P12. Second, the pharmacological profile of GABA(A) receptor (GABA(A)R)-mediated IPSCs differed between neonatal and mature nRt neurons, and this was accompanied by reciprocal changes in α3 (late) and α5 (early) subunit expression in nRt. Zolpidem, selective for α1- and α3-containing GABA(A)Rs, augmented only mature IPSCs, whereas clonazepam enhanced IPSCs at all stages. This effect was blocked by the α5-specific inverse agonist L-655,708, but only in immature neurons. In α3(H126R) mice, in which α3-subunits were mutated to become benzodiazepine insensitive, IPSCs were enhanced compared with those in wild-type animals in early development. Third, tonic GABA(A)R activation in nRt is age dependent and more prominent in immature neurons, which correlates with early expression of α5-containing GABA(A)Rs. Thus neonatal nRt neurons show relatively high expression of α5-subunits, which contributes to both slow synaptic and tonic extrasynaptic inhibition. The postnatal switch in GABA(A)R subunits from α5 to α3 could facilitate spontaneous network activity in nRt that occurs at this developmental time point and which is proposed to play a role in early circuit development.


Asunto(s)
Núcleos Talámicos Intralaminares/metabolismo , Receptores de GABA-A/metabolismo , Animales , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Potenciales Postsinápticos Inhibidores , Núcleos Talámicos Intralaminares/citología , Núcleos Talámicos Intralaminares/crecimiento & desarrollo , Núcleos Talámicos Intralaminares/fisiología , Ratones , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de GABA-A/genética
15.
Cereb Cortex ; 25(10): 3406-19, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25037921

RESUMEN

The chromatin-remodeling protein Satb2 plays a role in the generation of distinct subtypes of neocortical pyramidal neurons. Previous studies have shown that Satb2 is required for normal development of callosal projection neurons (CPNs), which fail to extend axons callosally in the absence of Satb2 and instead project subcortically. Here we conditionally delete Satb2 from the developing neocortex and find that neurons in the upper layers adopt some electrophysiological properties characteristic of deep layer neurons, but projections from the superficial layers do not contribute to the aberrant subcortical projections seen in Satb2 mutants. Instead, axons from deep layer CPNs descend subcortically in the absence of Satb2. These data demonstrate distinct developmental roles of Satb2 in regulating the fates of upper and deep layer neurons. Unexpectedly, Satb2 mutant brains also display changes in gene expression by subcerebral projection neurons (SCPNs), accompanied by a failure of corticospinal tract (CST) formation. Altering the timing of Satb2 ablation reveals that SCPNs require an early expression of Satb2 for differentiation and extension of the CST, suggesting that early transient expression of Satb2 in these cells plays an essential role in development. Collectively these data show that Satb2 is required by both CPNs and SCPNs for proper differentiation and axon pathfinding.


Asunto(s)
Axones/fisiología , Diferenciación Celular , Corteza Cerebral/embriología , Cuerpo Calloso/embriología , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Neuronas/fisiología , Factores de Transcripción/fisiología , Animales , Axones/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Cuerpo Calloso/metabolismo , Femenino , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones Transgénicos , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Corteza Somatosensorial/embriología , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Proc Natl Acad Sci U S A ; 110(50): 20278-83, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24262146

RESUMEN

Emerging evidence indicates that diazepam-binding inhibitor (DBI) mediates an endogenous benzodiazepine-mimicking (endozepine) effect on synaptic inhibition in the thalamic reticular nucleus (nRT). Here we demonstrate that DBI peptide colocalizes with both astrocytic and neuronal markers in mouse nRT, and investigate the role of astrocytic function in endozepine modulation in this nucleus by testing the effects of the gliotoxin fluorocitrate (FC) on synaptic inhibition and endozepine signaling in the nRT using patch-clamp recordings. FC treatment reduced the effective inhibitory charge of GABAA receptor (GABAAR)-mediated spontaneous inhibitory postsynaptic currents in WT mice, indicating that astrocytes enhance GABAAR responses in the nRT. This effect was abolished by both a point mutation that inhibits classical benzodiazepine binding to GABAARs containing the α3 subunit (predominant in the nRT) and a chromosomal deletion that removes the Dbi gene. Thus, astrocytes are required for positive allosteric modulation via the α3 subunit benzodiazepine-binding site by DBI peptide family endozepines. Outside-out sniffer patches pulled from neurons in the adjacent ventrobasal nucleus, which does not contain endozepines, show a potentiated response to laser photostimulation of caged GABA when placed in the nRT. FC treatment blocked the nRT-dependent potentiation of this response, as did the benzodiazepine site antagonist flumazenil. When sniffer patches were placed in the ventrobasal nucleus, however, subsequent treatment with FC led to potentiation of the uncaged GABA response, suggesting nucleus-specific roles for thalamic astrocytes in regulating inhibition. Taken together, these results suggest that astrocytes are required for endozepine actions in the nRT, and as such can be positive modulators of synaptic inhibition.


Asunto(s)
Astrocitos/fisiología , Inhibidor de la Unión a Diazepam/metabolismo , Neuronas GABAérgicas/fisiología , Núcleos Talámicos Intralaminares/fisiología , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Regulación Alostérica/fisiología , Animales , Citratos/farmacología , Neuronas GABAérgicas/metabolismo , Gliotoxina/análogos & derivados , Gliotoxina/farmacología , Núcleos Talámicos Intralaminares/citología , Ratones , Técnicas de Placa-Clamp , Receptores de GABA-A/metabolismo , Transmisión Sináptica/efectos de los fármacos
17.
J Neurosci ; 34(2): 675-87, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24403165

RESUMEN

Recurrent connections in the corticothalamic circuit underlie oscillatory behavior in this network and range from normal sleep rhythms to the abnormal spike-wave discharges seen in absence epilepsy. The propensity of thalamic neurons to fire postinhibitory rebound bursts mediated by low-threshold calcium spikes renders the circuit vulnerable to both increased excitation and increased inhibition, such as excessive excitatory cortical drive to thalamic reticular (RT) neurons or heightened inhibition of thalamocortical relay (TC) neurons by RT. In this context, a protective role may be played by group III metabotropic receptors (mGluRs), which are uniquely located in the presynaptic active zone and typically act as autoreceptors or heteroceptors to depress synaptic release. Here, we report that these receptors regulate short-term plasticity at two loci in the corticothalamic circuit in rats: glutamatergic cortical synapses onto RT neurons and GABAergic synapses onto TC neurons in somatosensory ventrobasal thalamus. The net effect of group III mGluR activation at these synapses is to suppress thalamic oscillations as assayed in vitro. These findings suggest a functional role of these receptors to modulate corticothalamic transmission and protect against prolonged activity in the network.


Asunto(s)
Corteza Cerebral/fisiología , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Tálamo/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Inmunohistoquímica , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
18.
J Neurosci ; 34(35): 11769-80, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25164672

RESUMEN

Hippocampal oscillations are critical for information processing, and are strongly influenced by inputs from the medial septum. Hippocamposeptal neurons provide direct inhibitory feedback from the hippocampus onto septal cells, and are therefore likely to also play an important role in the circuit; these neurons fire at either low or high frequency, reflecting hippocampal network activity during theta oscillations or ripple events, respectively. Here, we optogenetically target the long-range GABAergic projection from the hippocampus to the medial septum in rats, and thereby simulate hippocampal input onto downstream septal cells in an acute slice preparation. In response to optogenetic activation of hippocamposeptal fibers at theta and ripple frequencies, we elicit postsynaptic GABAergic responses in a subset (24%) of septal cells, most predominantly in fast-spiking cells. In addition, in another subset of septal cells (19%) corresponding primarily to cholinergic cells, we observe a slow hyperpolarization of the resting membrane potential and a decrease in input resistance, particularly in response to prolonged high-frequency (ripple range) stimulation. This slow response is partially sensitive to GIRK channel and D2 dopamine receptor block. Our results suggest that two independent populations of septal cells distinctly encode hippocampal feedback, enabling the septum to monitor ongoing patterns of activity in the hippocampus.


Asunto(s)
Hipocampo/fisiología , Vías Nerviosas/fisiología , Núcleos Septales/fisiología , Transducción de Señal/fisiología , Animales , Inmunohistoquímica , Potenciales de la Membrana/fisiología , Ratones , Optogenética , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Ratas , Ratas Long-Evans
19.
Neurobiol Dis ; 78: 115-25, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25836421

RESUMEN

Post-injury epilepsy (PIE) is a common complication following brain insults, including ischemic, and traumatic brain injuries. At present, there are no means to identify the patients at risk to develop PIE or to prevent its development. Seizures can occur months or years after the insult, do not respond to anti-seizure medications in over third of the patients, and are often associated with significant neuropsychiatric morbidities. We have previously established the critical role of blood-brain barrier dysfunction in PIE, demonstrating that exposure of brain tissue to extravasated serum albumin induces activation of inflammatory transforming growth factor beta (TGF-ß) signaling in astrocytes and eventually seizures. However, the link between the acute astrocytic inflammatory responses and reorganization of neural networks that underlie recurrent spontaneous seizures remains unknown. Here we demonstrate in vitro and in vivo that activation of the astrocytic ALK5/TGF-ß-pathway induces excitatory, but not inhibitory, synaptogenesis that precedes the appearance of seizures. Moreover, we show that treatment with SJN2511, a specific ALK5/TGF-ß inhibitor, prevents synaptogenesis and epilepsy. Our findings point to astrocyte-mediated synaptogenesis as a key epileptogenic process and highlight the manipulation of the TGF-ß-pathway as a potential strategy for the prevention of PIE.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Epilepsia/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Albúmina Sérica/administración & dosificación , Sinapsis/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Astrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Convulsiones/inducido químicamente , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 109(39): E2635-44, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22955882

RESUMEN

Current models of sleep/wake regulation posit that Hypocretin (Hcrt)-expressing neurons in the lateral hypothalamus promote and stabilize wakefulness by projecting to subcortical arousal centers. However, the critical downstream effectors of Hcrt neurons are unknown. Here we use optogenetic, pharmacological, and computational tools to investigate the functional connectivity between Hcrt neurons and downstream noradrenergic neurons in the locus coeruleus (LC) during nonrapid eye movement (NREM) sleep. We found that photoinhibiting LC neurons during Hcrt stimulation blocked Hcrt-mediated sleep-to-wake transitions. In contrast, when LC neurons were optically stimulated to increase membrane excitability, concomitant photostimulation of Hcrt neurons significantly increased the probability of sleep-to-wake transitions compared with Hcrt stimulation alone. We also built a conductance-based computational model of Hcrt-LC circuitry that recapitulates our behavioral results using LC neurons as the main effectors of Hcrt signaling. These results establish the Hcrt-LC connection as a critical integrator-effector circuit that regulates NREM sleep/wake behavior during the inactive period. This coupling of distinct neuronal systems can be generalized to other hypothalamic integrator nuclei with downstream effector/output populations in the brain.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Locus Coeruleus/metabolismo , Neuropéptidos/metabolismo , Transducción de Señal/fisiología , Sueño REM/fisiología , Vigilia/fisiología , Neuronas Adrenérgicas/citología , Animales , Locus Coeruleus/citología , Ratones , Ratones Noqueados , Orexinas , Estimulación Luminosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA