Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Biol ; 22(1): e3002483, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38295323

RESUMO

Synaptic transmission mediated by GABAA receptors (GABAARs) in adult, principal striatal spiny projection neurons (SPNs) can suppress ongoing spiking, but its effect on synaptic integration at subthreshold membrane potentials is less well characterized, particularly those near the resting down-state. To fill this gap, a combination of molecular, optogenetic, optical, and electrophysiological approaches were used to study SPNs in mouse ex vivo brain slices, and computational tools were used to model somatodendritic synaptic integration. In perforated patch recordings, activation of GABAARs, either by uncaging of GABA or by optogenetic stimulation of GABAergic synapses, evoked currents with a reversal potential near -60 mV in both juvenile and adult SPNs. Transcriptomic analysis and pharmacological work suggested that this relatively positive GABAAR reversal potential was not attributable to NKCC1 expression, but rather to HCO3- permeability. Regardless, from down-state potentials, optogenetic activation of dendritic GABAergic synapses depolarized SPNs. This GABAAR-mediated depolarization summed with trailing ionotropic glutamate receptor (iGluR) stimulation, promoting dendritic spikes and increasing somatic depolarization. Simulations revealed that a diffuse dendritic GABAergic input to SPNs effectively enhanced the response to dendritic iGluR signaling and promoted dendritic spikes. Taken together, our results demonstrate that GABAARs can work in concert with iGluRs to excite adult SPNs when they are in the resting down-state, suggesting that their inhibitory role is limited to brief periods near spike threshold. This state-dependence calls for a reformulation for the role of intrastriatal GABAergic circuits.


Assuntos
Interneurônios , Receptores de GABA-A , Camundongos , Animais , Corpo Estriado/fisiologia , Neostriado , Transmissão Sináptica/fisiologia , Neurônios GABAérgicos/fisiologia
2.
Nature ; 599(7886): 650-656, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34732887

RESUMO

Loss of functional mitochondrial complex I (MCI) in the dopaminergic neurons of the substantia nigra is a hallmark of Parkinson's disease1. Yet, whether this change contributes to Parkinson's disease pathogenesis is unclear2. Here we used intersectional genetics to disrupt the function of MCI in mouse dopaminergic neurons. Disruption of MCI induced a Warburg-like shift in metabolism that enabled neuronal survival, but triggered a progressive loss of the dopaminergic phenotype that was first evident in nigrostriatal axons. This axonal deficit was accompanied by motor learning and fine motor deficits, but not by clear levodopa-responsive parkinsonism-which emerged only after the later loss of dopamine release in the substantia nigra. Thus, MCI dysfunction alone is sufficient to cause progressive, human-like parkinsonism in which the loss of nigral dopamine release makes a critical contribution to motor dysfunction, contrary to the current Parkinson's disease paradigm3,4.


Assuntos
Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Morte Celular , Dendritos/metabolismo , Dendritos/patologia , Modelos Animais de Doenças , Progressão da Doença , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Feminino , Levodopa/farmacologia , Levodopa/uso terapêutico , Masculino , Camundongos , Destreza Motora/efeitos dos fármacos , NADH Desidrogenase/deficiência , NADH Desidrogenase/genética , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/fisiopatologia , Fenótipo , Substância Negra/citologia , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(26): E5167-E5176, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28611221

RESUMO

Key mitochondrial functions such as ATP production, Ca2+ uptake and release, and substrate accumulation depend on the proton electrochemical gradient (ΔµH+) across the inner membrane. Although several drugs can modulate ΔµH+, their effects are hardly reversible, and lack cellular specificity and spatial resolution. Although channelrhodopsins are widely used to modulate the plasma membrane potential of excitable cells, mitochondria have thus far eluded optogenetic control. Here we describe a toolkit of optometabolic constructs based on selective targeting of channelrhodopsins with distinct functional properties to the inner mitochondrial membrane of intact cells. We show that our strategy enables a light-dependent control of the mitochondrial membrane potential (Δψm) and coupled mitochondrial functions such as ATP synthesis by oxidative phosphorylation, Ca2+ dynamics, and respiratory metabolism. By directly modulating Δψm, the mitochondria-targeted opsins were used to control complex physiological processes such as spontaneous beats in cardiac myocytes and glucose-dependent ATP increase in pancreatic ß-cells. Furthermore, our optometabolic tools allow modulation of mitochondrial functions in single cells and defined cell regions.


Assuntos
Sinalização do Cálcio/fisiologia , Channelrhodopsins/metabolismo , Células Secretoras de Insulina/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Optogenética , Animais , Células HEK293 , Células HeLa , Humanos , Células Secretoras de Insulina/citologia , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley
5.
Hum Mol Genet ; 22(11): 2186-99, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23406870

RESUMO

Synapsin I (SynI) is a synaptic vesicle (SV) phosphoprotein playing multiple roles in synaptic transmission and plasticity by differentially affecting crucial steps of SV trafficking in excitatory and inhibitory synapses. SynI knockout (KO) mice are epileptic, and nonsense and missense mutations in the human SYN1 gene have a causal role in idiopathic epilepsy and autism. To get insights into the mechanisms of epileptogenesis linked to SYN1 mutations, we analyzed the effects of the recently identified Q555X mutation on neurotransmitter release dynamics and short-term plasticity (STP) in excitatory and inhibitory synapses. We used patch-clamp electrophysiology coupled to electron microscopy and multi-electrode arrays to dissect synaptic transmission of primary SynI KO hippocampal neurons in which the human wild-type and mutant SynI were expressed by lentiviral transduction. A parallel decrease in the SV readily releasable pool in inhibitory synapses and in the release probability in excitatory synapses caused a marked reduction in the evoked synchronous release. This effect was accompanied by an increase in asynchronous release that was much more intense in excitatory synapses and associated with an increased total charge transfer. Q555X-hSynI induced larger facilitation and post-tetanic potentiation in excitatory synapses and stronger depression after long trains in inhibitory synapses. These changes were associated with higher network excitability and firing/bursting activity. Our data indicate that imbalances in STP and release dynamics of inhibitory and excitatory synapses trigger network hyperexcitability potentially leading to epilepsy/autism manifestations.


Assuntos
Epilepsia/genética , Epilepsia/metabolismo , Plasticidade Neuronal/genética , Sinapses/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Animais , Feminino , Expressão Gênica , Hipocampo/metabolismo , Humanos , Espaço Intracelular/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Multimerização Proteica , Transporte Proteico , Sinapsinas/química , Potenciais Sinápticos , Vesículas Sinápticas/metabolismo
6.
bioRxiv ; 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38712260

RESUMO

Long-term synaptic plasticity at glutamatergic synapses on striatal spiny projection neurons (SPNs) is central to learning goal-directed behaviors and habits. Although considerable attention has been paid to the mechanisms underlying synaptic strengthening and new learning, little scrutiny has been given to those involved in the attenuation of synaptic strength that attends suppression of a previously learned association. Our studies revealed a novel, non-Hebbian, long-term, postsynaptic depression of glutamatergic SPN synapses induced by interneuronal nitric oxide (NO) signaling (NO-LTD) that was preferentially engaged at quiescent synapses. This form of plasticity was gated by local Ca 2+ influx through CaV1.3 Ca 2+ channels and stimulation of phosphodiesterase 1 (PDE1), which degraded cyclic guanosine monophosphate (cGMP) and blunted NO signaling. Consistent with this model, mice harboring a gain-of-function mutation in the gene coding for the pore-forming subunit of CaV1.3 channels had elevated depolarization-induced dendritic Ca 2+ entry and impaired NO-LTD. Extracellular uncaging of glutamate and intracellular uncaging of cGMP suggested that this Ca 2+ -dependent regulation of PDE1 activity allowed for local regulation of dendritic NO signaling. This inference was supported by simulation of SPN dendritic integration, which revealed that dendritic spikes engaged PDE1 in a branch-specific manner. In a mouse model of Parkinson's disease (PD), NO-LTD was absent not because of a postsynaptic deficit in NO signaling machinery, but rather due to impaired interneuronal NO release. Re-balancing intrastriatal neuromodulatory signaling in the PD model restored NO release and NO-LTD. Taken together, these studies provide novel insights into the mechanisms governing NO-LTD in SPN and its role in psychomotor disorders, like PD.

7.
J Cell Sci ; 124(Pt 18): 3174-86, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21868364

RESUMO

The RE-1-specific silencing transcription factor (REST or NRSF) is a transcription repressor that orchestrates differentiation and also operates in differentiated neurons and neurosecretory cells (neural cells). Its role in proliferation has been investigated so far only in rapidly growing tumors, with conflicting results: suppression in non-neural tumors, stimulation in medulloblastomas. Working with two clones of chromaffin-neuronal PC12 cells, which express different levels of REST, and using genetic complementation and knockdown approaches, we show that REST also promotes proliferation in differentiated neural cells. Mechanistically, this occurs by a signaling pathway involving REST, the GTPase-activating protein tuberin (TSC2) and the transcription co-factor ß-catenin. In PC12 cells, raised expression of REST correlates with reduced TSC2 levels, nuclear accumulation and co-transcriptional activation of ß-catenin, and increased expression of its target oncogenes Myc and Ccnd1, which might account for the proliferation advantage and the distinct morphology. Rest transcription is also increased, unveiling the existence of a self-sustaining, feed-forward REST-TSC2-ß-catenin signaling loop that is also operative in another neural cell model, NT2/D1 cells. Transfection of REST, knockdown of TSC2 or forced expression of active ß-catenin recapitulated the biochemical, functional and morphological properties of the high-expressing REST clone in wild-type PC12 cells. Upregulation of REST promoted proliferation and phenotypic changes, thus hindering neurosecretion. The new REST-TSC2-ß-catenin signaling paradigm might have an important role in various aspects of neural cell physiology and pathology, including the regulation of proliferation and neurosecretion.


Assuntos
Retroalimentação Fisiológica , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neurônios/patologia , Neurossecreção/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Interferente Pequeno/genética , Ratos , Proteínas Repressoras/genética , Transdução de Sinais/genética , Transgenes/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , beta Catenina/genética
8.
Nature ; 447(7148): 1081-6, 2007 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-17558391

RESUMO

Why dopamine-containing neurons of the brain's substantia nigra pars compacta die in Parkinson's disease has been an enduring mystery. Our studies suggest that the unusual reliance of these neurons on L-type Ca(v)1.3 Ca2+ channels to drive their maintained, rhythmic pacemaking renders them vulnerable to stressors thought to contribute to disease progression. The reliance on these channels increases with age, as juvenile dopamine-containing neurons in the substantia nigra pars compacta use pacemaking mechanisms common to neurons not affected in Parkinson's disease. These mechanisms remain latent in adulthood, and blocking Ca(v)1.3 Ca2+ channels in adult neurons induces a reversion to the juvenile form of pacemaking. Such blocking ('rejuvenation') protects these neurons in both in vitro and in vivo models of Parkinson's disease, pointing to a new strategy that could slow or stop the progression of the disease.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Modelos Animais de Doenças , Modelos Neurológicos , Neurônios/citologia , Neurônios/patologia , Doença de Parkinson/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Envelhecimento , Animais , Antiparkinsonianos/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Canais de Cálcio Tipo L/deficiência , Canais de Cálcio Tipo L/genética , Dendritos/metabolismo , Progressão da Doença , Dopamina/metabolismo , Condutividade Elétrica , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Doença de Parkinson/prevenção & controle , Rotenona/farmacologia , Substância Negra/citologia , Substância Negra/metabolismo , Substância Negra/patologia
9.
bioRxiv ; 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36993489

RESUMO

Synaptic transmission mediated by GABA A receptors (GABA A Rs) in adult, principal striatal spiny projection neurons (SPNs) can suppress ongoing spiking, but its effect on synaptic integration at sub-threshold membrane potentials is less well characterized, particularly those near the resting down-state. To fill this gap, a combination of molecular, optogenetic, optical and electrophysiological approaches were used to study SPNs in mouse ex vivo brain slices, and computational tools were used to model somatodendritic synaptic integration. Activation of GABA A Rs, either by uncaging of GABA or by optogenetic stimulation of GABAergic synapses, evoked currents with a reversal potential near -60 mV in perforated patch recordings from both juvenile and adult SPNs. Molecular profiling of SPNs suggested that this relatively positive reversal potential was not attributable to NKCC1 expression, but rather to a dynamic equilibrium between KCC2 and Cl-/HCO3-cotransporters. Regardless, from down-state potentials, optogenetic activation of dendritic GABAergic synapses depolarized SPNs. This GABAAR-mediated depolarization summed with trailing ionotropic glutamate receptor (iGluR) stimulation, promoting dendritic spikes and increasing somatic depolarization. Simulations revealed that a diffuse dendritic GABAergic input to SPNs effectively enhanced the response to coincident glutamatergic input. Taken together, our results demonstrate that GABA A Rs can work in concert with iGluRs to excite adult SPNs when they are in the resting down-state, suggesting that their inhibitory role is limited to brief periods near spike threshold. This state-dependence calls for a reformulation of the role intrastriatal GABAergic circuits.

10.
Nat Commun ; 14(1): 1398, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36914640

RESUMO

Huntington's disease (HD) is a progressive, neurodegenerative disease caused by a CAG triplet expansion in huntingtin. Although corticostriatal dysfunction has long been implicated in HD, the determinants and pathway specificity of this pathophysiology are not fully understood. Here, using a male zQ175+/- knock-in mouse model of HD we carry out optogenetic interrogation of intratelencephalic and pyramidal tract synapses with principal striatal spiny projection neurons (SPNs). These studies reveal that the connectivity of intratelencephalic, but not pyramidal tract, neurons with direct and indirect pathway SPNs increased in early symptomatic zQ175+/- HD mice. This enhancement was attributable to reduced pre-synaptic inhibitory control of intratelencephalic terminals by striatal cholinergic interneurons. Lowering mutant huntingtin selectively in striatal cholinergic interneurons with a virally-delivered zinc finger repressor protein normalized striatal acetylcholine release and intratelencephalic functional connectivity, revealing a node in the network underlying corticostriatal pathophysiology in a HD mouse model.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Camundongos , Masculino , Animais , Doença de Huntington/metabolismo , Doenças Neurodegenerativas/metabolismo , Corpo Estriado/metabolismo , Neostriado/metabolismo , Colinérgicos/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
11.
bioRxiv ; 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38168401

RESUMO

Background: Pathological accumulation of aggregated α-synuclein (aSYN) is a common feature of Parkinson's disease (PD). However, the mechanisms by which intracellular aSYN pathology contributes to dysfunction and degeneration of neurons in the brain are still unclear. A potentially relevant target of aSYN is the mitochondrion. To test this hypothesis, genetic and physiological methods were used to monitor mitochondrial function in substantia nigra pars compacta (SNc) dopaminergic and pedunculopontine nucleus (PPN) cholinergic neurons after stereotaxic injection of aSYN pre-formed fibrils (PFFs) into the mouse brain. Methods: aSYN PPFs were stereotaxically injected into the SNc or PPN of mice. Twelve weeks later, mice were studied using a combination of approaches, including immunocytochemical analysis, cell- type specific transcriptomic profiling, electron microscopy, electrophysiology and two-photon-laser- scanning microscopy of genetically encoded sensors for bioenergetic and redox status. Results: In addition to inducing a significant neuronal loss, SNc injection of PFFs induced the formation of intracellular, phosphorylated aSYN aggregates selectively in dopaminergic neurons. In these neurons, PFF-exposure decreased mitochondrial gene expression, reduced the number of mitochondria, increased oxidant stress, and profoundly disrupted mitochondrial adenosine triphosphate production. Consistent with an aSYN-induced bioenergetic deficit, the autonomous spiking of dopaminergic neurons slowed or stopped. PFFs also up-regulated lysosomal gene expression and increased lysosomal abundance, leading to the formation of Lewy-like inclusions. Similar changes were observed in PPN cholinergic neurons following aSYN PFF exposure. Conclusions: Taken together, our findings suggest that disruption of mitochondrial function, and the subsequent bioenergetic deficit, is a proximal step in the cascade of events induced by aSYN pathology leading to dysfunction and degeneration of neurons at-risk in PD.

12.
Sci Rep ; 12(1): 17851, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36284123

RESUMO

Recombinant adeno-associated viruses (rAAV) are extensively used in both research and clinical applications. Despite significant advances, there is a lack of short promoters able to drive the expression of virus delivered genes in specific classes of neurons. We designed an efficient rAAV vector suitable for the rAAV-mediated gene expression in cortical interneurons, mainly in the parvalbumin expressing cells. The vector includes a short parvalbumin promoter and a specialized poly(A) sequence. The degree of conservation of the parvalbumin gene adjoining non-coding regions was used in both the promoter design and the selection of the poly(A) sequence. The specificity was established by co-localizing the fluorescence of the virus delivered eGFP and the antibody for a neuronal marker. rAAV particles were injected in the visual cortex area V1/V2 of adult rats (2-4 months old). Neurons expressing the virus delivered eGFP were mainly positive for interneuronal markers: 66.5 ± 2.8% for parvalbumin, 14.6 ± 2.4% for somatostatin, 7.1 ± 1.2% for vasoactive intestinal peptide, 2.8 ± 0.6% for cholecystokinin. Meanwhile, only 2.1 ± 0.5% were positive for CaMKII, a marker for principal cells in the cortex. The efficiency of the construct was verified by optogenetic experiments: the expression of the virus delivered ChR2 channels was sufficient to evoke by blue light laser high frequency bursts of action potentials in putative fast spiking neurons. We conclude that our promoter allows highly specific expression of the rAAV delivered cDNAs in cortical interneurons with a strong preference for the parvalbumin positive cells.


Assuntos
Parvalbuminas , Peptídeo Intestinal Vasoativo , Animais , Ratos , Parvalbuminas/genética , Peptídeo Intestinal Vasoativo/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Interneurônios/metabolismo , Dependovirus/genética , Somatostatina/metabolismo , Colecistocinina/metabolismo
13.
Sci Adv ; 8(39): eabp8701, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36179023

RESUMO

How do neurons match generation of adenosine triphosphate by mitochondria to the bioenergetic demands of regenerative activity? Although the subject of speculation, this coupling is still poorly understood, particularly in neurons that are tonically active. To help fill this gap, pacemaking substantia nigra dopaminergic neurons were studied using a combination of optical, electrophysiological, and molecular approaches. In these neurons, spike-activated calcium (Ca2+) entry through Cav1 channels triggered Ca2+ release from the endoplasmic reticulum, which stimulated mitochondrial oxidative phosphorylation through two complementary Ca2+-dependent mechanisms: one mediated by the mitochondrial uniporter and another by the malate-aspartate shuttle. Disrupting either mechanism impaired the ability of dopaminergic neurons to sustain spike activity. While this feedforward control helps dopaminergic neurons meet the bioenergetic demands associated with sustained spiking, it is also responsible for their elevated oxidant stress and possibly to their decline with aging and disease.


Assuntos
Cálcio , Neurônios Dopaminérgicos , Trifosfato de Adenosina/metabolismo , Ácido Aspártico , Cálcio/metabolismo , Neurônios Dopaminérgicos/metabolismo , Malatos/metabolismo , Malatos/farmacologia , Mitocôndrias/metabolismo , Oxidantes , Substância Negra/metabolismo
14.
J Neurosci ; 30(47): 16025-40, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106841

RESUMO

The activity patterns of subthalamic nucleus (STN) neurons are intimately linked to motor function and dysfunction and arise through the complex interaction of intrinsic properties and inhibitory and excitatory synaptic inputs. In many neurons, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play key roles in intrinsic excitability and synaptic integration both under normal conditions and in disease states. However, in STN neurons, which strongly express HCN channels, their roles remain relatively obscure. To address this deficit, complementary molecular and cellular electrophysiological, imaging, and computational approaches were applied to the rat STN. Molecular profiling demonstrated that individual STN neurons express mRNA encoding several HCN subunits, with HCN2 and 3 being the most abundant. Light and electron microscopic analysis showed that HCN2 subunits are strongly expressed and distributed throughout the somatodendritic plasma membrane. Voltage-, current-, and dynamic-clamp analysis, two-photon Ca(2+) imaging, and computational modeling revealed that HCN channels are activated by GABA(A) receptor-mediated inputs and thus limit synaptic hyperpolarization and deinactivation of low-voltage-activated Ca(2+) channels. Although HCN channels also limited the temporal summation of EPSPs, generated through two-photon uncaging of glutamate, this action was largely shunted by GABAergic inhibition that was necessary for HCN channel activation. Together the data demonstrate that HCN channels in STN neurons selectively counteract GABA(A) receptor-mediated inhibition arising from the globus pallidus and thus promote single-spike activity rather than rebound burst firing.


Assuntos
Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Canais Iônicos/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Núcleo Subtalâmico/fisiologia , Transmissão Sináptica/fisiologia , Animais , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Canais de Potássio , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Núcleo Subtalâmico/efeitos dos fármacos , Núcleo Subtalâmico/ultraestrutura , Transmissão Sináptica/efeitos dos fármacos
15.
Nat Neurosci ; 10(11): 1458-66, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17906621

RESUMO

Dopamine-depleting lesions of the striatum that mimic Parkinson's disease induce a profound pruning of spines and glutamatergic synapses in striatopallidal medium spiny neurons, leaving striatonigral medium spiny neurons intact. The mechanisms that underlie this cell type-specific loss of connectivity are poorly understood. The Kir2 K(+) channel is an important determinant of dendritic excitability in these cells. Here we show that opening of these channels is potently reduced by signaling through M1 muscarinic receptors in striatopallidal neurons, but not in striatonigral neurons. This asymmetry could be attributed to differences in the subunit composition of Kir2 channels. Dopamine depletion alters the subunit composition further, rendering Kir2 channels in striatopallidal neurons even more susceptible to modulation. Reduced opening of Kir2 channels enhances dendritic excitability and synaptic integration. This cell type-specific enhancement of dendritic excitability is an essential trigger for synaptic pruning after dopamine depletion, as pruning was prevented by genetic deletion of M1 muscarinic receptors.


Assuntos
Corpo Estriado/citologia , Dendritos/fisiologia , Neurônios/ultraestrutura , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Receptor Muscarínico M1/fisiologia , Animais , Animais Recém-Nascidos , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Potenciais da Membrana/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muscarina/farmacologia , Agonistas Muscarínicos/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Oxidopamina/farmacologia , Técnicas de Patch-Clamp/métodos , Receptor Muscarínico M1/deficiência , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Reserpina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
Neuron ; 50(3): 443-52, 2006 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-16675398

RESUMO

Long-term depression (LTD) of the synapse formed between cortical pyramidal neurons and striatal medium spiny neurons is central to many theories of motor plasticity and associative learning. The induction of LTD at this synapse is thought to depend upon D(2) dopamine receptors localized in the postsynaptic membrane. If this were true, LTD should be inducible in neurons from only one of the two projection systems of the striatum. Using transgenic mice in which neurons that contribute to these two systems are labeled, we show that this is not the case. Rather, in both cell types, the D(2) receptor dependence of LTD induction reflects the need to lower M(1) muscarinic receptor activity-a goal accomplished by D(2) receptors on cholinergic interneurons. In addition to reconciling discordant tracts of the striatal literature, these findings point to cholinergic interneurons as key mediators of dopamine-dependent striatal plasticity and learning.


Assuntos
Acetilcolina/metabolismo , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Interneurônios/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Córtex Cerebral/citologia , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios , Interneurônios/efeitos dos fármacos , Aprendizagem/fisiologia , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Técnicas de Cultura de Órgãos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Receptor Muscarínico M1/metabolismo , Receptores de Dopamina D2/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
17.
Nat Neurosci ; 9(6): 832-42, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16699510

RESUMO

Parkinson disease is a neurodegenerative disorder whose symptoms are caused by the loss of dopaminergic neurons innervating the striatum. As striatal dopamine levels fall, striatal acetylcholine release rises, exacerbating motor symptoms. This adaptation is commonly attributed to the loss of interneuronal regulation by inhibitory D(2) dopamine receptors. Our results point to a completely different, new mechanism. After striatal dopamine depletion, D(2) dopamine receptor modulation of calcium (Ca(2+)) channels controlling vesicular acetylcholine release in interneurons was unchanged, but M(4) muscarinic autoreceptor coupling to these same channels was markedly attenuated. This adaptation was attributable to the upregulation of RGS4-an autoreceptor-associated, GTPase-accelerating protein. This specific signaling adaptation extended to a broader loss of autoreceptor control of interneuron spiking. These observations suggest that RGS4-dependent attenuation of interneuronal autoreceptor signaling is a major factor in the elevation of striatal acetylcholine release in Parkinson disease.


Assuntos
Acetilcolina/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Interneurônios/metabolismo , Proteínas RGS/metabolismo , Receptor Muscarínico M4/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Autorreceptores/efeitos dos fármacos , Autorreceptores/metabolismo , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Interneurônios/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Agonistas Muscarínicos/farmacologia , Oxotremorina/farmacologia , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia , Proteínas RGS/genética , Ratos , Receptor Muscarínico M4/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
18.
Mol Neurobiol ; 56(8): 5701-5714, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30671783

RESUMO

Local control of protein translation is a fundamental process for the regulation of synaptic plasticity. It has been demonstrated that the local protein synthesis occurring in axons and dendrites can be shaped by numerous mechanisms, including miRNA-mediated regulation. However, several aspects underlying this regulatory process have not been elucidated yet. Here, we analyze the differential miRNA profile in cell bodies and neurites of primary hippocampal neurons and find an enrichment of the precursor and mature forms of miR-218 in the neuritic projections. We show that miR-218 abundance is regulated during hippocampal development and by chronic silencing or activation of neuronal network. Overexpression and knockdown of miR-218 demonstrated that miR-218 targets the mRNA encoding the GluA2 subunit of AMPA receptors and modulates its expression. At the functional level, miR-218 overexpression increases glutamatergic synaptic transmission at both single neuron and network levels. Our data demonstrate that miR-218 may play a key role in the regulation of AMPA-mediated excitatory transmission and in the homeostatic regulation of synaptic plasticity.


Assuntos
MicroRNAs/metabolismo , Neuritos/metabolismo , Biossíntese de Proteínas , Subunidades Proteicas/metabolismo , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Animais , Sequência de Bases , Corpo Celular/metabolismo , Potenciais Pós-Sinápticos Excitadores , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Rede Nervosa/metabolismo
19.
J Neurosci ; 27(49): 13552-66, 2007 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18057213

RESUMO

Neurons in the external segment of the globus pallidus (GPe) are autonomous pacemakers that are capable of sustained fast spiking. The cellular and molecular determinants of pacemaking and fast spiking in GPe neurons are not fully understood, but voltage-dependent Na+ channels must play an important role. Electrophysiological studies of these neurons revealed that macroscopic activation and inactivation kinetics of their Na+ channels were similar to those found in neurons lacking either autonomous activity or the capacity for fast spiking. What was distinctive about GPe Na+ channels was a prominent resurgent gating mode. This mode was significantly reduced in GPe neurons lacking functional Nav1.6 channels. In these Nav1.6 null neurons, pacemaking and the capacity for fast spiking were impaired, as was the ability to follow stimulation frequencies used to treat Parkinson's disease (PD). Simulations incorporating Na+ channel models with and without prominent resurgent gating suggested that resurgence was critical to fast spiking but not to pacemaking, which appeared to be dependent on the positioning of Na+ channels in spike-initiating regions of the cell. These studies not only shed new light on the mechanisms underlying spiking in GPe neurons but also suggest that electrical stimulation therapies in PD are unlikely to functionally inactivate neurons possessing Nav1.6 Na+ channels with prominent resurgent gating.


Assuntos
Potenciais de Ação/fisiologia , Globo Pálido/fisiologia , Ativação do Canal Iônico/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Canais de Sódio/fisiologia , Potenciais de Ação/genética , Animais , Simulação por Computador , Ativação do Canal Iônico/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.6 , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Canais de Sódio/deficiência , Canais de Sódio/genética , Fatores de Tempo
20.
Nat Neurosci ; 6(3): 258-66, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12592408

RESUMO

Neurons with the capacity to discharge at high rates--'fast-spiking' (FS) neurons--are critical participants in central motor and sensory circuits. It is widely accepted that K+ channels with Kv3.1 or Kv3.2 subunits underlie fast, delayed-rectifier (DR) currents that endow neurons with this FS ability. Expression of these subunits in heterologous systems, however, yields channels that open at more depolarized potentials than do native Kv3 family channels, suggesting that they differ. One possibility is that native channels incorporate a subunit that modifies gating. Molecular, electrophysiological and pharmacological studies reported here suggest that a splice variant of the Kv3.4 subunit coassembles with Kv3.1 subunits in rat brain FS neurons. Coassembly enhances the spike repolarizing efficiency of the channels, thereby reducing spike duration and enabling higher repetitive spike rates. These results suggest that manipulation of K3.4 subunit expression could be a useful means of controlling the dynamic range of FS neurons.


Assuntos
Neuropeptídeos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Processamento Alternativo , Animais , Linhagem Celular , Separação Celular , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Rim/citologia , Rim/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/genética , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Ligação Proteica/fisiologia , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Ratos , Tempo de Reação/fisiologia , Canais de Potássio Shaw
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA