Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Neurosci ; 45: 447-469, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35440143

RESUMEN

Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types.


Asunto(s)
Dependovirus , Vectores Genéticos , Encéfalo , Cápside/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen
2.
Nature ; 544(7650): 362-366, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28405024

RESUMEN

There are no disease-modifying treatments for adult human neurodegenerative diseases. Here we test RNA-targeted therapies in two mouse models of spinocerebellar ataxia type 2 (SCA2), an autosomal dominant polyglutamine disease. Both models recreate the progressive adult-onset dysfunction and degeneration of a neuronal network that are seen in patients, including decreased firing frequency of cerebellar Purkinje cells and a decline in motor function. We developed a potential therapy directed at the ATXN2 gene by screening 152 antisense oligonucleotides (ASOs). The most promising oligonucleotide, ASO7, downregulated ATXN2 mRNA and protein, which resulted in delayed onset of the SCA2 phenotype. After delivery by intracerebroventricular injection to ATXN2-Q127 mice, ASO7 localized to Purkinje cells, reduced cerebellar ATXN2 expression below 75% for more than 10 weeks without microglial activation, and reduced the levels of cerebellar ATXN2. Treatment of symptomatic mice with ASO7 improved motor function compared to saline-treated mice. ASO7 had a similar effect in the BAC-Q72 SCA2 mouse model, and in both mouse models it normalized protein levels of several SCA2-related proteins expressed in Purkinje cells, including Rgs8, Pcp2, Pcp4, Homer3, Cep76 and Fam107b. Notably, the firing frequency of Purkinje cells returned to normal even when treatment was initiated more than 12 weeks after the onset of the motor phenotype in BAC-Q72 mice. These findings support ASOs as a promising approach for treating some human neurodegenerative diseases.


Asunto(s)
Oligonucleótidos Antisentido/uso terapéutico , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/terapia , Potenciales de Acción , Animales , Ataxina-2/deficiencia , Ataxina-2/genética , Ataxina-2/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Movimiento , Fenotipo , Células de Purkinje/metabolismo , Células de Purkinje/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología
3.
Proc Natl Acad Sci U S A ; 115(52): E12407-E12416, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30530649

RESUMEN

The genetically heterogeneous spinocerebellar ataxias (SCAs) are caused by Purkinje neuron dysfunction and degeneration, but their underlying pathological mechanisms remain elusive. The Src family of nonreceptor tyrosine kinases (SFK) are essential for nervous system homeostasis and are increasingly implicated in degenerative disease. Here we reveal that the SFK suppressor Missing-in-metastasis (MTSS1) is an ataxia locus that links multiple SCAs. MTSS1 loss results in increased SFK activity, reduced Purkinje neuron arborization, and low basal firing rates, followed by cell death. Surprisingly, mouse models for SCA1, SCA2, and SCA5 show elevated SFK activity, with SCA1 and SCA2 displaying dramatically reduced MTSS1 protein levels through reduced gene expression and protein translation, respectively. Treatment of each SCA model with a clinically approved Src inhibitor corrects Purkinje neuron basal firing and delays ataxia progression in MTSS1 mutants. Our results identify a common SCA therapeutic target and demonstrate a key role for MTSS1/SFK in Purkinje neuron survival and ataxia progression.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología , Animales , Ataxia/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Neoplasias/genética , Proteínas/metabolismo , Células de Purkinje/fisiología , Ataxias Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/fisiopatología , Familia-src Quinasas/metabolismo
4.
Genes Dev ; 26(5): 445-60, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22357600

RESUMEN

The Rbfox proteins (Rbfox1, Rbfox2, and Rbfox3) regulate the alternative splicing of many important neuronal transcripts and have been implicated in a variety of neurological disorders. However, their roles in brain development and function are not well understood, in part due to redundancy in their activities. Here we show that, unlike Rbfox1 deletion, the CNS-specific deletion of Rbfox2 disrupts cerebellar development. Genome-wide analysis of Rbfox2(-/-) brain RNA identifies numerous splicing changes altering proteins important both for brain development and mature neuronal function. To separate developmental defects from alterations in the physiology of mature cells, Rbfox1 and Rbfox2 were deleted from mature Purkinje cells, resulting in highly irregular firing. Notably, the Scn8a mRNA encoding the Na(v)1.6 sodium channel, a key mediator of Purkinje cell pacemaking, is improperly spliced in RbFox2 and Rbfox1 mutant brains, leading to highly reduced protein expression. Thus, Rbfox2 protein controls a post-transcriptional program required for proper brain development. Rbfox2 is subsequently required with Rbfox1 to maintain mature neuronal physiology, specifically Purkinje cell pacemaking, through their shared control of sodium channel transcript splicing.


Asunto(s)
Cerebelo/embriología , Neuronas Motoras/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Cerebelo/citología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ratones , Canal de Sodio Activado por Voltaje NAV1.6 , Proteínas del Tejido Nervioso/metabolismo , Células de Purkinje/metabolismo , Empalme del ARN/genética , Factores de Empalme de ARN , Canales de Sodio/metabolismo
5.
Hum Mol Genet ; 26(16): 3069-3080, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28525545

RESUMEN

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominant neurodegenerative disease caused by CAG repeat expansion in the ATXN2 gene. The repeat resides in an encoded region of the gene resulting in polyglutamine (polyQ) expansion which has been assumed to result in gain of function, predominantly, for the ATXN2 protein. We evaluated temporal cerebellar expression profiles by RNA sequencing of ATXN2Q127 mice versus wild-type (WT) littermates. ATXN2Q127 mice are characterized by a progressive motor phenotype onset, and have progressive cerebellar molecular and neurophysiological (Purkinje cell firing frequency) phenotypes. Our analysis revealed previously uncharacterized early and progressive abnormal patterning of cerebellar gene expression. Weighted Gene Coexpression Network Analysis revealed four gene modules that were significantly correlated with disease status, composed primarily of genes associated with GTPase signaling, calcium signaling and cell death. Of these genes, few overlapped with differentially expressed cerebellar genes that we identified in Atxn2-/- knockout mice versus WT littermates, suggesting that loss-of-function is not a significant component of disease pathology. We conclude that SCA2 is a disease characterized by gain of function for ATXN2.


Asunto(s)
Redes Reguladoras de Genes , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-2/genética , Ataxina-2/metabolismo , Ataxinas/genética , Secuencia de Bases , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Expresión Génica , Perfilación de la Expresión Génica , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Células de Purkinje/metabolismo , Análisis de Secuencia de ARN , Repeticiones de Trinucleótidos
6.
Neuroimage ; 173: 370-383, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29496611

RESUMEN

Complex animal behavior is produced by dynamic interactions between discrete regions of the brain. As such, defining functional connections between brain regions is critical in gaining a full understanding of how the brain generates behavior. Evidence suggests that discrete regions of the cerebellar cortex functionally project to the forebrain, mediating long-range communication potentially important in motor and non-motor behaviors. However, the connectivity map remains largely incomplete owing to the challenge of driving both reliable and selective output from the cerebellar cortex, as well as the need for methods to detect region specific activation across the entire forebrain. Here we utilize a paired optogenetic and fMRI (ofMRI) approach to elucidate the downstream forebrain regions modulated by activating a region of the cerebellum that induces stereotypical, ipsilateral forelimb movements. We demonstrate with ofMRI, that activating this forelimb motor region of the cerebellar cortex results in functional activation of a variety of forebrain and midbrain areas of the brain, including the hippocampus and primary motor, retrosplenial and anterior cingulate cortices. We further validate these findings using optogenetic stimulation paired with multi-electrode array recordings and post-hoc staining for molecular markers of activated neurons (i.e. c-Fos). Together, these findings demonstrate that a single discrete region of the cerebellar cortex is capable of influencing motor output and the activity of a number of downstream forebrain as well as midbrain regions thought to be involved in different aspects of behavior.


Asunto(s)
Corteza Cerebelosa/anatomía & histología , Imagen por Resonancia Magnética/métodos , Vías Nerviosas/anatomía & histología , Optogenética/métodos , Prosencéfalo/anatomía & histología , Animales , Mapeo Encefálico/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Movimiento/fisiología
7.
Cerebellum ; 16(1): 230-252, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27193702

RESUMEN

For many decades, the predominant view in the cerebellar field has been that the olivocerebellar system's primary function is to induce plasticity in the cerebellar cortex, specifically, at the parallel fiber-Purkinje cell synapse. However, it has also long been proposed that the olivocerebellar system participates directly in motor control by helping to shape ongoing motor commands being issued by the cerebellum. Evidence consistent with both hypotheses exists; however, they are often investigated as mutually exclusive alternatives. In contrast, here, we take the perspective that the olivocerebellar system can contribute to both the motor learning and motor control functions of the cerebellum and might also play a role in development. We then consider the potential problems and benefits of it having multiple functions. Moreover, we discuss how its distinctive characteristics (e.g., low firing rates, synchronization, and variable complex spike waveforms) make it more or less suitable for one or the other of these functions, and why having multiple functions makes sense from an evolutionary perspective. We did not attempt to reach a consensus on the specific role(s) the olivocerebellar system plays in different types of movements, as that will ultimately be determined experimentally; however, collectively, the various contributions highlight the flexibility of the olivocerebellar system, and thereby suggest that it has the potential to act in both the motor learning and motor control functions of the cerebellum.


Asunto(s)
Cerebelo/fisiología , Aprendizaje/fisiología , Actividad Motora/fisiología , Núcleo Olivar/fisiología , Animales , Consenso , Humanos , Vías Nerviosas/fisiología
8.
J Neurosci ; 35(49): 16142-58, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26658866

RESUMEN

The role of GABAA receptor (GABAAR)-mediated tonic inhibition in interneurons remains unclear and may vary among subgroups. Somatostatin (SOM) interneurons in the hilus of the dentate gyrus show negligible expression of nonsynaptic GABAAR subunits and very low tonic inhibition. To determine the effects of ectopic expression of tonic GABAAR subtypes in these neurons, Cre-dependent viral vectors were used to express GFP-tagged GABAAR subunits (α6 and δ) selectively in hilar SOM neurons in SOM-Cre mice. In single-transfected animals, immunohistochemistry demonstrated strong expression of either the α6 or δ subunit; in cotransfected animals, both subunits were consistently expressed in the same neurons. Electrophysiology revealed a robust increase of tonic current, with progressively larger increases following transfection of δ, α6, and α6/δ subunits, respectively, indicating formation of functional receptors in all conditions and likely coassembly of the subunits in the same receptor following cotransfection. An in vitro model of repetitive bursting was used to determine the effects of increased tonic inhibition in hilar SOM interneurons on circuit activity in the dentate gyrus. Upon cotransfection, the frequency of GABAAR-mediated bursting in granule cells was reduced, consistent with a reduction in synchronous firing among hilar SOM interneurons. Moreover, in vivo studies of Fos expression demonstrated reduced activation of α6/δ-cotransfected neurons following acute seizure induction by pentylenetetrazole. The findings demonstrate that increasing tonic inhibition in hilar SOM interneurons can alter dentate gyrus circuit activity during strong stimulation and suggest that tonic inhibition of interneurons could play a role in regulating excessive synchrony within the network. SIGNIFICANCE STATEMENT: In contrast to many hippocampal interneurons, somatostatin (SOM) neurons in the hilus of the dentate gyrus have very low levels of nonsynaptic GABAARs and exhibit very little tonic inhibition. In an effort to increase tonic inhibition selectively in these interneurons, we used Cre-dependent viral vectors in SOM-Cre mice to achieve interneuron-specific expression of the nonsynaptic GABAAR subunits (α6 and δ) in vivo. We show, for the first time, that such recombinant GFP-tagged GABAAR subunits are expressed robustly, assemble to form functional receptors, substantially increase tonic inhibition in SOM interneurons, and alter circuit activity within the dentate gyrus.


Asunto(s)
Giro Dentado/citología , Red Nerviosa/metabolismo , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Somatostatina/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Giro Dentado/efectos de los fármacos , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Vectores Genéticos/metabolismo , Humanos , Isoxazoles/farmacología , Masculino , Ratones , Ratones Transgénicos , Inhibición Neural/efectos de los fármacos , Inhibición Neural/genética , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Pentilenotetrazol/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Pirimidinas/farmacología , Receptores de GABA-A/genética , Somatostatina/genética
9.
J Physiol ; 594(16): 4653-60, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27198167

RESUMEN

Degenerative ataxias are a common form of neurodegenerative disease that affect about 20 individuals per 100,000. The autosomal dominant spinocerebellar ataxias (SCAs) are caused by a variety of protein coding mutations (single nucleotide changes, deletions and expansions) in single genes. Affected genes encode plasma membrane and intracellular ion channels, membrane receptors, protein kinases, protein phosphatases and proteins of unknown function. Although SCA-linked genes are quite diverse they share two key features: first, they are highly, although not exclusively, expressed in cerebellar Purkinje neurons (PNs), and second, when mutated they lead ultimately to the degeneration of PNs. In this review we summarize ataxia-related changes in PN neurophysiology that have been observed in various mouse knockout lines and in transgenic models of human SCA. We also highlight emerging evidence that altered metabotropic glutamate receptor signalling and disrupted calcium homeostasis in PNs form a common, early pathophysiological mechanism in SCAs. Together these findings indicate that aberrant calcium signalling and profound changes in PN neurophysiology precede PN cell loss and are likely to lead to cerebellar circuit dysfunction that explains behavioural signs of ataxia characteristic of the disease.


Asunto(s)
Ataxias Espinocerebelosas/fisiopatología , Potenciales de Acción , Animales , Calcio/fisiología , Humanos , Células de Purkinje/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal
10.
Biophys J ; 108(3): 520-9, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25650920

RESUMEN

In recent years, optical sensors for tracking neural activity have been developed and offer great utility. However, developing microscopy techniques that have several kHz bandwidth necessary to reliably capture optically reported action potentials (APs) at multiple locations in parallel remains a significant challenge. To our knowledge, we describe a novel microscope optimized to measure spatially distributed optical signals with submillisecond and near diffraction-limit resolution. Our design uses a spatial light modulator to generate patterned illumination to simultaneously excite multiple user-defined targets. A galvanometer driven mirror in the emission path streaks the fluorescence emanating from each excitation point during the camera exposure, using unused camera pixels to capture time varying fluorescence at rates that are ∼1000 times faster than the camera's native frame rate. We demonstrate that this approach is capable of recording Ca(2+) transients resulting from APs in neurons labeled with the Ca(2+) sensor Oregon Green Bapta-1 (OGB-1), and can localize the timing of these events with millisecond resolution. Furthermore, optically reported APs can be detected with the voltage sensitive dye DiO-DPA in multiple locations within a neuron with a signal/noise ratio up to ∼40, resolving delays in arrival time along dendrites. Thus, the microscope provides a powerful tool for photometric measurements of dynamics requiring submillisecond sampling at multiple locations.


Asunto(s)
Potenciales de Acción/fisiología , Microscopía Fluorescente/métodos , Neuronas/fisiología , Fenómenos Ópticos , Animales , Calcio/metabolismo , Ratones Endogámicos C57BL , Factores de Tiempo
11.
Hum Mol Genet ; 22(2): 271-83, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23087021

RESUMEN

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominantly inherited disorder, which is caused by a pathological expansion of a polyglutamine (polyQ) tract in the coding region of the ATXN2 gene. Like other ataxias, SCA2 most overtly affects Purkinje cells (PCs) in the cerebellum. Using a transgenic mouse model expressing a full-length ATXN2(Q127)-complementary DNA under control of the Pcp2 promoter (a PC-specific promoter), we examined the time course of behavioral, morphologic, biochemical and physiological changes with particular attention to PC firing in the cerebellar slice. Although motor performance began to deteriorate at 8 weeks of age, reductions in PC number were not seen until after 12 weeks. Decreases in the PC firing frequency first showed at 6 weeks and paralleled deterioration of motor performance with progression of disease. Transcription changes in several PC-specific genes such as Calb1 and Pcp2 mirrored the time course of changes in PC physiology with calbindin-28 K changes showing the first small, but significant decreases at 4 weeks. These results emphasize that in this model of SCA2, physiological and behavioral phenotypes precede morphological changes by several weeks and provide a rationale for future studies examining the effects of restoration of firing frequency on motor function and prevention of future loss of PCs.


Asunto(s)
Expresión Génica , Desempeño Psicomotor , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología , Animales , Ataxinas , Cerebelo/metabolismo , Cerebelo/patología , Cerebelo/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/genética , Mutación , Proteínas del Tejido Nervioso/genética , Fenotipo , Células de Purkinje/patología , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Factores de Tiempo
12.
Proc Natl Acad Sci U S A ; 109(41): 16726-30, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22988089

RESUMEN

Climbing fiber input to the cerebellum is believed to serve as a teaching signal during associative, cerebellum-dependent forms of motor learning. However, it is not understood how this neural pathway coordinates changes in cerebellar circuitry during learning. Here, we use pharmacological manipulations to prolong the postcomplex spike pause, a component of the climbing fiber signal in Purkinje neurons, and show that these manipulations enhance the rate of learning in classical eyelid conditioning. Our findings elucidate an unappreciated aspect of the climbing fiber teaching signal, and are consistent with a model in which convergent postcomplex spike pauses drive learning-related plasticity in the deep cerebellar nucleus. They also suggest a physiological mechanism that could modulate motor learning rates.


Asunto(s)
Parpadeo/fisiología , Condicionamiento Palpebral/fisiología , Fibras Nerviosas/fisiología , Células de Purkinje/fisiología , Análisis de Varianza , Animales , Bencimidazoles/farmacología , Parpadeo/efectos de los fármacos , Agonistas de los Canales de Calcio/farmacología , Fármacos Cardiovasculares/farmacología , Núcleos Cerebelosos/efectos de los fármacos , Núcleos Cerebelosos/fisiología , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/fisiología , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Antagonistas del GABA/farmacología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Ratones , Picrotoxina/farmacología , Células de Purkinje/efectos de los fármacos , Pirimidinas/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
J Neurosci ; 33(36): 14392-405, 2013 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-24005292

RESUMEN

Axonal sprouting of excitatory neurons is frequently observed in temporal lobe epilepsy, but the extent to which inhibitory interneurons undergo similar axonal reorganization remains unclear. The goal of this study was to determine whether somatostatin (SOM)-expressing neurons in stratum (s.) oriens of the hippocampus exhibit axonal sprouting beyond their normal territory and innervate granule cells of the dentate gyrus in a pilocarpine model of epilepsy. To obtain selective labeling of SOM-expressing neurons in s. oriens, a Cre recombinase-dependent construct for channelrhodopsin2 fused to enhanced yellow fluorescent protein (ChR2-eYFP) was virally delivered to this region in SOM-Cre mice. In control mice, labeled axons were restricted primarily to s. lacunosum-moleculare. However, in pilocarpine-treated animals, a rich plexus of ChR2-eYFP-labeled fibers and boutons extended into the dentate molecular layer. Electron microscopy with immunogold labeling demonstrated labeled axon terminals that formed symmetric synapses on dendritic profiles in this region, consistent with innervation of granule cells. Patterned illumination of ChR2-labeled fibers in s. lacunosum-moleculare of CA1 and the dentate molecular layer elicited GABAergic inhibitory responses in dentate granule cells in pilocarpine-treated mice but not in controls. Similar optical stimulation in the dentate hilus evoked no significant responses in granule cells of either group of mice. These findings indicate that under pathological conditions, SOM/GABAergic neurons can undergo substantial axonal reorganization beyond their normal territory and establish aberrant synaptic connections. Such reorganized circuitry could contribute to functional deficits in inhibition in epilepsy, despite the presence of numerous GABAergic terminals in the region.


Asunto(s)
Neuronas GABAérgicas/patología , Interneuronas/patología , Somatostatina/metabolismo , Estado Epiléptico/patología , Animales , Axones/ultraestructura , Dendritas/ultraestructura , Giro Dentado/patología , Neuronas GABAérgicas/fisiología , Hipocampo/patología , Interneuronas/metabolismo , Interneuronas/fisiología , Ratones , Ratones Endogámicos C57BL , Inhibición Neural , Optogenética , Estimulación Luminosa , Pilocarpina/toxicidad , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Somatostatina/genética , Estado Epiléptico/inducido químicamente
14.
J Neurosci ; 32(50): 17988-97, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23238715

RESUMEN

Climbing fiber (CF) input to the cerebellum is thought to instruct associative motor memory formation through its effects on multiple sites within the cerebellar circuit. We used adeno-associated viral delivery of channelrhodopsin-2 (ChR2) to inferior olivary neurons to selectively express ChR2 in CFs, achieving nearly complete transfection of CFs in the caudal cerebellar lobules of rats. As expected, optical stimulation of ChR2-expressing CFs generates complex spike responses in individual Purkinje neurons (PNs); in addition we found that such stimulation recruits a network of inhibitory interneurons in the molecular layer. This CF-driven disynaptic inhibition prolongs the postcomplex spike pause observed when spontaneously firing PNs receive direct CF input; such inhibition also elicits pauses in spontaneously firing PNs not receiving direct CF input. Baseline firing rates of PNs are strongly suppressed by low-frequency (2 Hz) stimulation of CFs, and this suppression is partly relieved by blocking synaptic inhibition. We conclude that CF-driven, disynaptic inhibition has a major influence on PN excitability and contributes to the widely observed negative correlation between complex and simple spike rates. Because they receive input from many CFs, molecular layer interneurons are well positioned to detect the spatiotemporal patterns of CF activity believed to encode error signals. Together, our findings suggest that such inhibition may bind together groups of Purkinje neurons to provide instructive signals to downstream sites in the cerebellar circuit.


Asunto(s)
Vías Nerviosas/fisiología , Neuronas/fisiología , Núcleo Olivar/fisiología , Células de Purkinje/fisiología , Animales , Femenino , Masculino , Vías Nerviosas/citología , Neuronas/citología , Núcleo Olivar/citología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
15.
J Neurophysiol ; 107(11): 2958-70, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22378171

RESUMEN

In the adult mammalian brain, GABA(A) receptors (GABA(A)Rs) are responsible for the predominant forms of synaptic inhibition, but these receptors can excite neurons when the chloride equilibrium potential (E(Cl)) is depolarized. In many mature neurons, GABA(A)Rs are found on presynaptic terminals where they exert depolarizing effects. To understand whether excitatory GABA action affects axonal function, we used transverse cerebellar slices to measure the effects of photolysis of caged GABA on the initiation and propagation of compound parallel fiber (PF) action potentials (APs). Photolysis of caged GABA increased the amplitude and conduction velocity of PF APs; GABA reuptake blockers and a positive modulator of GABA(A)Rs enhanced these effects. In contrast, a modulator selective for δ-subunit-containing GABA(A)Rs did not enhance these effects and responsiveness remained in δ(-/-) mice, arguing that δ-subunit-containing GABA(A)Rs are not required. Synaptically released GABA also increased PF excitability, indicating that the mechanism is engaged by physiological signals. A Hodgkin-Huxley-style compartmental model of the PF axon and granule cell body was constructed, and this model recapitulated the GABA-dependent decrease in AP threshold and the increase in conduction velocity, features that were sensitive to E(Cl) and to the voltage dependence of sodium channel inactivation. The model also predicts that axonal GABA(A)Rs could affect orthodromic spike initiation. We conclude that GABA acting on cerebellar PFs facilitates both spike generation and propagation, allowing axons of granule cells to passively integrate signals from inhibitory interneurons and influence information flow in the input layer to the cerebellar cortex.


Asunto(s)
Potenciales de Acción/fisiología , Axones/fisiología , Fibras Nerviosas Mielínicas/fisiología , Receptores de GABA-A/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Estimulación Eléctrica/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Nerviosas Mielínicas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fotólisis , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
16.
J Neurophysiol ; 106(4): 2057-64, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21795619

RESUMEN

Extrasynaptic GABA(A) receptors (eGABARs) allow ambient GABA to tonically regulate neuronal excitability and are implicated as targets for ethanol and anesthetics. These receptors are thought to be heteropentameric proteins made up of two α subunits-either α4 or α6-two ß2 or ß3 subunits, and one δ subunit. The GABA analog 4,5,6,7-tetrahydroisoxazolo (5,4-c)pyridin-3(-ol) (THIP) has been proposed as a selective ligand for eGABARs. Behavioral and in vitro studies suggest that eGABARs have nanomolar affinity for THIP; however, all published studies on recombinant versions of eGABARs report micromolar affinities. Here, we examine THIP sensitivity of native eGABARs on cerebellar neurons and on reconstituted GABARs in heterologous systems. Concentration-response data for THIP, obtained from cerebellar granule cells and molecular layer interneurons in wild-type and δ subunit knockout slices, confirm that submicromolar THIP sensitivity requires δ subunits. In recombinant experiments, we find that δ subunit coexpression leads to receptors activated by nanomolar THIP concentrations (EC(50) of 30-50 nM for α4ß3δ and α6ß3δ), a sensitivity almost 1,000-fold higher than receptors formed by α4/6 and ß3 subunits. In contrast, γ2 subunit expression significantly reduces THIP sensitivity. Even when δ subunit cDNA or cRNA was supplied in excess, high- and low-sensitivity THIP responses were often apparent, indicative of variable mixtures of low-affinity αß and high-affinity αßδ receptors. We conclude that δ subunit incorporation into GABARs leads to a dramatic increase in THIP sensitivity, a defining feature that accounts for the unique behavioral and neurophysiological properties of THIP.


Asunto(s)
Agonistas del GABA/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Isoxazoles/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/fisiología , Animales , Corteza Cerebelosa/citología , Relación Dosis-Respuesta a Droga , Agonistas del GABA/administración & dosificación , Agonistas del GABA/farmacocinética , Neuronas GABAérgicas/fisiología , Células HEK293 , Humanos , Isoxazoles/administración & dosificación , Isoxazoles/farmacocinética , Ratones , Ratones Noqueados , Oocitos , Técnicas de Placa-Clamp , Isoformas de Proteínas/fisiología , Multimerización de Proteína , Subunidades de Proteína , Receptores de GABA-A/química , Receptores de GABA-A/deficiencia , Receptores de GABA-A/genética , Proteínas Recombinantes de Fusión/fisiología , Solubilidad , Xenopus laevis
17.
Nat Methods ; 5(4): 331-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18311146

RESUMEN

Light-activated ion channels provide a precise and noninvasive optical means for controlling action potential firing, but the genes encoding these channels must first be delivered and expressed in target cells. Here we describe a method for bestowing light sensitivity onto endogenous ion channels that does not rely on exogenous gene expression. The method uses a synthetic photoisomerizable small molecule, or photoswitchable affinity label (PAL), that specifically targets K+ channels. PALs contain a reactive electrophile, enabling covalent attachment of the photoswitch to naturally occurring nucleophiles in K+ channels. Ion flow through PAL-modified channels is turned on or off by photoisomerizing PAL with different wavelengths of light. We showed that PAL treatment confers light sensitivity onto endogenous K+ channels in isolated rat neurons and in intact neural structures from rat and leech, allowing rapid optical regulation of excitability without genetic modification.


Asunto(s)
Potenciales de Acción/efectos de la radiación , Activación del Canal Iónico/efectos de la radiación , Neuronas , Canales de Potasio/metabolismo , Marcadores de Afinidad/química , Animales , Compuestos Azo/química , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Cerebelo/efectos de la radiación , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Sanguijuelas , Neuronas/metabolismo , Neuronas/efectos de la radiación , Estimulación Luminosa , Fotoquímica , Compuestos de Amonio Cuaternario/química , Ratas
18.
J Neurosci ; 29(29): 9197-209, 2009 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-19625510

RESUMEN

A major goal in neuroscience is the development of optical reporters of membrane potential that are easy to use, have limited phototoxicity, and achieve the speed and sensitivity necessary for detection of individual action potentials in single neurons. Here we present a novel, two-component optical approach that attains these goals. By combining DiO, a fluorescent neuronal tracer dye, with dipicrylamine (DPA), a molecule whose membrane partitioning is voltage-sensitive, optical signals related to changes in membrane potential based on FRET (Förster resonance energy transfer) are reported. Using DiO/DPA in HEK-293 cells with diffraction-limited laser spot illumination, depolarization-induced fluorescence changes of 56% per 100 mV (tau approximately 0.1 ms) were obtained, while in neuronal cultures and brain slices, action potentials (APs) generated a Delta F/F per 100 mV of >25%. The high sensitivity provided by DiO/DPA enabled the detection of subthreshold activity and high-frequency APs in single trials from somatic, axonal, or dendritic membrane compartments. Recognizing that DPA can depress excitability, we assayed the amplitude and duration of single APs, burst properties, and spontaneous firing in neurons of primary cultures and brain slices and found that they are undetectably altered by up to 2 microm DPA and only slightly perturbed by 5 microm DPA. These findings substantiate a simple, noninvasive method that relies on a neuronal tracer dye for monitoring electrical signal flow, and offers unique flexibility for the study of signaling within intact neuronal circuits.


Asunto(s)
Técnicas Citológicas/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes , Potenciales de la Membrana , Picratos , Potenciales de Acción , Animales , Línea Celular , Cerebelo/fisiología , Relación Dosis-Respuesta a Droga , Fluorescencia , Hipocampo/fisiología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Neuronas/fisiología , Técnicas de Placa-Clamp , Picratos/administración & dosificación , Células de Purkinje/efectos de los fármacos , Células de Purkinje/fisiología , Factores de Tiempo
19.
Mol Pharmacol ; 78(5): 918-24, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20699325

RESUMEN

GABA(A) receptors (GABA(A)Rs) have long been a focus as targets for alcohol actions. Recent work suggests that tonic GABAergic inhibition mediated by extrasynaptic δ subunit-containing GABA(A)Rs is uniquely sensitive to ethanol and enhanced at concentrations relevant for human alcohol consumption. Ethanol enhancement of recombinant α4ß3δ receptors is blocked by the behavioral alcohol antagonist 8-azido-5,6-dihydro-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylic acid ethyl ester (Ro15-4513), suggesting that EtOH/Ro15-4513-sensitive receptors mediate important behavioral alcohol actions. Here we confirm alcohol/alcohol antagonist sensitivity of α4ß3δ receptors using human clones expressed in a human cell line and test the hypothesis that discrepant findings concerning the high alcohol sensitivity of these receptors are due to difficulties incorporating δ subunits into functional receptors. To track δ subunit incorporation, we used a functional tag, a single amino acid change (H68A) in a benzodiazepine binding residue in which a histidine in the δ subunit is replaced by an alanine residue found at the homologous position in γ subunits. We demonstrate that the δH68A substitution confers diazepam sensitivity to otherwise diazepam-insensitive α4ß3δ receptors. The extent of enhancement of α4ß3δH68A receptors by 1 µM diazepam, 30 mM EtOH, and 1 µM ß-carboline-3-carboxy ethyl ester (but not 1 µM Zn(2+) block) is correlated in individual recordings, suggesting that δ subunit incorporation into recombinant GABA(A)Rs varies from cell to cell and that this variation accounts for the variable pharmacological profile. These data are consistent with the notion that δ subunit-incorporation is often incomplete in recombinant systems yet is necessary for high ethanol sensitivity, one of the features of native δ subunit-containing GABA(A)Rs.


Asunto(s)
Azidas/farmacología , Benzodiazepinas/farmacología , Etanol/antagonistas & inhibidores , Etanol/farmacología , Receptores de GABA-A/biosíntesis , Regulación Alostérica , Animales , Carbolinas/farmacología , Cationes Bivalentes , Línea Celular , Diazepam/farmacología , Humanos , Mutación , Técnicas de Placa-Clamp , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Ratas , Receptores de GABA-A/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección , Zinc/farmacología
20.
Cell Cycle ; 19(2): 153-159, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31876231

RESUMEN

Spinocerebellar ataxias (SCA) are a genetically heterogeneous family of cerebellar neurodegenerative diseases characterized by abnormal firing of Purkinje neurons and degeneration. We recently demonstrated the slowed firing rates seen in several SCAs share a common etiology of hyper-activation of the Src family of non-receptor tyrosine kinases (SFKs). However, the lack of clinically available neuroactive SFK inhibitors lead us to investigate alternative mechanisms to modulate SFK activity. Previous studies demonstrate that SFK activity can be enhanced by the removal of inhibitory phospho-marks by receptor-protein-tyrosine phosphatases (RPTPs). In this Extra View we show that MTSS1 inhibits SFK activity through the binding and inhibition of a subset of the RPTP family members, and lowering RPTP activity in cerebellar slices with peptide inhibitors increases the suppressed Purkinje neuron basal firing rates seen in two different SCA models. Together these results identify RPTPs as novel effectors of Purkinje neuron basal firing, extending the MTSS1/SFK regulatory circuit we previously described and expanding the therapeutic targets for SCA patients.


Asunto(s)
Potenciales de Acción/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Células de Purkinje/enzimología , Potenciales de Acción/efectos de los fármacos , Animales , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Células de Purkinje/efectos de los fármacos , Ataxias Espinocerebelosas/enzimología , Ataxias Espinocerebelosas/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA