Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Glia ; 69(2): 310-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32865323

RESUMO

Astrocyte heterogeneity is an emerging concept in which astrocytes within or between brain regions show variable morphological and/or gene expression profiles that presumably reflect different functional roles. Recent evidence indicates that retrotrapezoid nucleus (RTN) astrocytes sense changes in tissue CO2/ H+ to regulate respiratory activity; however, mechanism(s) by which they do so remain unclear. Alterations in inward K+ currents represent a potential mechanism by which CO2 /H+ signals may be conveyed to neurons. Here, we use slice electrophysiology in rats of either sex to show that RTN astrocytes intrinsically respond to CO2 /H+ by inhibition of an inward rectifying potassium (Kir ) conductance and depolarization of the membrane, while cortical astrocytes do not exhibit such CO2 /H+ -sensitive properties. Application of Ba2+ mimics the effect of CO2 /H+ on RTN astrocytes as measured by reductions in astrocyte Kir -like currents and increased RTN neuronal firing. These CO2 /H+ -sensitive currents increase developmentally, in parallel to an increased expression in Kir 4.1 and Kir 5.1 in the brainstem. Finally, the involvement of Kir 5.1 in the CO2 /H+ -sensitive current was verified using a Kir5.1 KO rat. These data suggest that Kir inhibition by CO2 /H+ may govern the degree to which astrocytes mediate downstream chemoreceptive signaling events through cell-autonomous mechanisms. These results identify Kir channels as potentially important regional CO2 /H+ sensors early in development, thus expanding our understanding of how astrocyte heterogeneity may uniquely support specific neural circuits and behaviors.


Assuntos
Astrócitos , Animais , Encéfalo , Dióxido de Carbono/farmacologia , Células Quimiorreceptoras , Neurônios , Canais de Potássio Corretores do Fluxo de Internalização , Ratos , Canal Kir5.1
2.
J Physiol ; 596(5): 901-919, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29274075

RESUMO

KEY POINTS: An increase in the excitability of GABAergic cells has typically been assumed to decrease network activity, potentially producing overall anti-epileptic effects. Recent data suggest that inhibitory networks may actually play a role in initiating epileptiform activity. We show that activation of GABAergic interneurons can elicit synchronous long-lasting network activity. Specific interneuron subpopulations differentially contributed to GABA network synchrony, indicating cell type-specific contributions of interneurons to cortical network activity. Interneurons may critically contribute to the generation of aberrant network activity characteristic of epilepsy, warranting further investigation into the contribution of distinct cortical interneuron subpopulations to the propagation and rhythmicity of epileptiform activity. ABSTRACT: In the presence of the A-type K+ channel blocker 4-aminopyrdine, spontaneous synchronous network activity develops in the neocortex of mice of either sex. This aberrant synchrony persists in the presence of excitatory amino acid receptor antagonists (EAA blockers) and is considered to arise from synchronous firing of cortical interneurons (INs). Although much attention has been given to the mechanisms underlying this GABAergic synchrony, the contribution of specific IN subtypes to the generation of these long-lasting discharges (LLDs) is incompletely understood. We employed genetically-encoded channelrhodopsin and archaerhodopsin opsins to investigate the sufficiency and necessity, respectively, of activation of parvalbumin (PV), somatostatin (SST) and vasointestinal peptide (VIP)-expressing INs for the generation of synchronous neocortical GABAergic discharges. We found light-induced activation of PV or SST INs to be equally sufficient for the generation of LLDs, whereas activation of VIP INs was not. By contrast, light-induced inhibition of PV INs strongly reduced LLD initiation, whereas suppression of SST or VIP IN activity only partially attenuated LLD magnitude. These results suggest neocortical INs perform cell type-specific roles in the generation of aberrant GABAergic cortical network activity.


Assuntos
Neurônios GABAérgicos/fisiologia , Hormônios/farmacologia , Interneurônios/fisiologia , Neocórtex/fisiologia , Optogenética , Parvalbuminas/fisiologia , Somatostatina/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Potenciais de Ação , Animais , Animais Recém-Nascidos , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Neocórtex/efeitos dos fármacos
3.
J Neurosci ; 35(46): 15276-90, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26586816

RESUMO

Circuit dysfunction in complex brain disorders such as schizophrenia and autism is caused by imbalances between inhibitory and excitatory synaptic transmission (I/E). Short-term plasticity differentially alters responses from excitatory and inhibitory synapses, causing the I/E ratio to change as a function of frequency. However, little is known about I/E ratio dynamics in complex brain disorders. Transcriptional dysregulation in interneurons, particularly parvalbumin interneurons, is a consistent pathophysiological feature of schizophrenia. Peroxisome proliferator activated receptor γ coactivator 1α (PGC-1α) is a transcriptional coactivator that in hippocampus is highly concentrated in inhibitory interneurons and regulates parvalbumin transcription. Here, we used PGC-1α(-/-) mice to investigate effects of interneuron transcriptional dysregulation on the dynamics of the I/E ratio at the synaptic and circuit level in hippocampus. We find that loss of PGC-1α increases the I/E ratio onto CA1 pyramidal cells in response to Schaffer collateral stimulation in slices from young adult mice. The underlying mechanism is enhanced basal inhibition, including increased inhibition from parvalbumin interneurons. This decreases the spread of activation in CA1 and dramatically limits pyramidal cell spiking, reducing hippocampal output. The I/E ratio and CA1 output are partially restored by paired-pulse stimulation at short intervals, indicating frequency-dependent effects. However, circuit dysfunction persists, indicated by alterations in kainate-induced gamma oscillations and impaired nest building. Together, these results show that transcriptional dysregulation in hippocampal interneurons causes frequency-dependent alterations in I/E ratio and circuit function, suggesting that PGC-1α deficiency in psychiatric and neurological disorders contributes to disease by causing functionally relevant alterations in I/E balance. SIGNIFICANCE STATEMENT: Alteration in the inhibitory and excitatory synaptic transmission (I/E) balance is a fundamental principle underlying the circuit dysfunction observed in many neuropsychiatric and neurodevelopmental disorders. The I/E ratio is dynamic, continuously changing because of synaptic short-term plasticity. We show here that transcriptional dysregulation in interneurons, particularly parvalbumin interneurons, causes frequency-dependent alterations in the I/E ratio and in circuit function in hippocampus. Peroxisome proliferator activated receptor γ coactivator 1α (PGC-1α-deficient) mice have enhanced inhibition in CA1, the opposite of what is seen in cortex. This study fills an important gap in current understanding of how changes in inhibition in complex brain disorders affect I/E dynamics, leading to region-specific circuit dysfunction and behavioral impairment. This study also provides a conceptual framework for analyzing the effects of short-term plasticity on the I/E balance in disease models.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Interneurônios/fisiologia , Inibição Neural/fisiologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Comportamento de Nidação/fisiologia , Neurotransmissores/farmacologia , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética , Imagens com Corantes Sensíveis à Voltagem
4.
J Neurosci ; 34(43): 14375-87, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339750

RESUMO

Accumulating evidence strongly implicates the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in the pathophysiology of multiple neurological disorders, but the downstream gene targets of PGC-1α in the brain have remained enigmatic. Previous data demonstrate that PGC-1α is primarily concentrated in inhibitory neurons and that PGC-1α is required for the expression of the interneuron-specific Ca(2+)-binding protein parvalbumin (PV) throughout the cortex. To identify other possible transcriptional targets of PGC-1α in neural tissue, we conducted a microarray on neuroblastoma cells overexpressing PGC-1α, mined results for genes with physiological relevance to interneurons, and measured cortical gene and protein expression of these genes in mice with underexpression and overexpression of PGC-1α. We observed bidirectional regulation of novel PGC-1α-dependent transcripts spanning synaptic [synaptotagmin 2 (Syt2) and complexin 1 (Cplx1)], structural [neurofilament heavy chain (Nefh)], and metabolic [neutral cholesterol ester hydrolase 1 (Nceh1), adenylate kinase 1 (Ak1), inositol polyphosphate 5-phosphatase J (Inpp5j), ATP synthase mitochondrial F1 complex O subunit (Atp5o), phytanol-CoA-2hydroxylase (Phyh), and ATP synthase mitrochondrial F1 complex α subunit 1 (Atp5a1)] functions. The neuron-specific genes Syt2, Cplx1, and Nefh were developmentally upregulated in an expression pattern consistent with that of PGC-1α and were expressed in cortical interneurons. Conditional deletion of PGC-1α in PV-positive neurons significantly decreased cortical transcript expression of these genes, promoted asynchronous GABA release, and impaired long-term memory. Collectively, these data demonstrate that PGC-1α is required for normal PV-positive interneuron function and that loss of PGC-1α in this interneuron subpopulation could contribute to cortical dysfunction in disease states.


Assuntos
Interneurônios/metabolismo , Parvalbuminas/biossíntese , Fatores de Transcrição/biossíntese , Transcrição Gênica/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo
5.
J Neurophysiol ; 110(8): 1733-43, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23864381

RESUMO

Hyperpolarization-activated, cyclic nucleotide-gated, nonspecific cation (HCN) channels have a well-characterized role in regulation of cellular excitability and network activity. The role of these channels in control of epileptiform discharges is less thoroughly understood. This is especially pertinent given the altered HCN channel expression in epilepsy. We hypothesized that inhibition of HCN channels would enhance bicuculline-induced epileptiform discharges. Whole cell recordings were obtained from layer (L)2/3 and L5 pyramidal neurons and L1 and L5 GABAergic interneurons. In the presence of bicuculline (10 µM), HCN channel inhibition with ZD 7288 (20 µM) significantly increased the magnitude (defined as area) of evoked epileptiform events in both L2/3 and L5 neurons. We recorded activity associated with epileptiform discharges in L1 and L5 interneurons to test the hypothesis that HCN channels regulate excitatory synaptic inputs differently in interneurons versus pyramidal neurons. HCN channel inhibition increased the magnitude of epileptiform events in both L1 and L5 interneurons. The increased magnitude of epileptiform events in both pyramidal cells and interneurons was due to an increase in network activity, since holding cells at depolarized potentials under voltage-clamp conditions to minimize HCN channel opening did not prevent enhancement in the presence of ZD 7288. In neurons recorded with ZD 7288-containing pipettes, bath application of the noninactivating inward cationic current (Ih) antagonist still produced increases in epileptiform responses. These results show that epileptiform discharges in disinhibited rat neocortex are modulated by HCN channels.


Assuntos
Potenciais de Ação , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Epilepsia/fisiopatologia , Neocórtex/fisiopatologia , Animais , Bicuculina/farmacologia , Convulsivantes/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/fisiologia , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Potenciais da Membrana , Neocórtex/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Pirimidinas/farmacologia , Ratos , Transmissão Sináptica
6.
Neuroscience ; 513: 38-53, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36682446

RESUMO

N-methyl-D-aspartate receptor (NMDAR) hypofunction during brain development is likely to contribute to the manifestation of schizophrenia (SCZ) in young adulthood. The cellular targets of NMDAR hypofunction appear to be at least in part corticolimbic fast-spiking (FS) interneurons. However, functional alterations in parvalbumin (PV)-positive FS interneurons following NMDAR hypofunction are poorly understood. Paired patch-clamp recordings from murine cortical PV interneurons and pyramidal neurons revealed that genetic deletion of NMDAR subunit Grin1 in prospective PV interneurons before the second postnatal week impaired evoked- and synchronized-GABA release. Whereas intrinsic excitability and spiking characteristics were also disturbed by Grin1 deletion, neither restoring their excitability by K+ channel blockade nor increasing extracellular Ca2+ rescued the GABA release. GABA release was also insensitive to the Cav2.1 channel antagonist ω-agatoxin IVA. Heterozygous deletion of Cacna1a gene (encoding Cav2.1) in PV interneurons produced a similar GABA release phenotype as the Grin1 mutants. Treatment with the Cav2.1/2.2 channel agonist GV-58 augmented somatic Ca2+ currents and GABA release in Cacna1a-haploinsufficient PV interneurons, but failed to enhance GABA release in the Grin1-deleted PV interneurons. Taken together, our results suggest that Grin1 deletion in prospective PV interneurons impairs proper maturation of membrane excitability and Cav2.1-recruited evoked GABA release. This may increase synaptic excitatory/inhibitory ratio in principal neurons, contributing to the emergence of SCZ-like phenotypes.


Assuntos
Neocórtex , Receptores de N-Metil-D-Aspartato , Animais , Camundongos , Receptores de N-Metil-D-Aspartato/metabolismo , Parvalbuminas/metabolismo , Neocórtex/metabolismo , Estudos Prospectivos , Transmissão Sináptica/fisiologia , Interneurônios/metabolismo , Ácido gama-Aminobutírico
7.
Epilepsy Res ; 183: 106937, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35526331

RESUMO

4-Aminopyridine (4-AP) induces ictal-like epileptiform discharges in a variety of brain regions. These events are associated with enhanced inhibitory and excitatory synaptic neurotransmission. The relative contribution of specific subclasses of GABAergic interneurons (INs) to epileptiform activity in the 4-AP model is not well characterized. We have used genetically encoded channelrhodopsin (ChR) and Archaerhodopsin (Arch) expression in parvalbumin (PV), somatostatin (SST) and vasoactive intestinal polypeptide (VIP) INs to investigate the role of interneuron subclasses in 4-AP-induced epileptiform discharges. Whole-cell patch-clamp recordings were obtained from L5 pyramidal cells (PYRs) in somatosensory cortex of 30-to-70-day old mice. In the presence of 100 µM 4-AP, photostimulation of ChR in PV and SST, but not VIP INs, evoked epileptiform discharges similar to spontaneous and electrically evoked events. Light activation of Arch in PV INs was more effective in reducing epileptiform activity compared to SST and VIP INs. Epileptiform discharges were evoked at offset of Arch induced hyperpolarizations in PV and SST interneurons but not VIP INs. PV and SST INs could both initiate and inhibit 4-AP-induced epileptiform activity in L5 PYRs. VIP INs did not contribute significantly to eliciting or inhibiting epileptiform discharges. These results suggest that subclasses of INs contribute differently to the initiation and modulation of epileptiform discharges in cortical networks.


Assuntos
Interneurônios , Córtex Somatossensorial , Potenciais de Ação/fisiologia , Animais , Interneurônios/metabolismo , Camundongos , Parvalbuminas/metabolismo , Células Piramidais
8.
Transl Psychiatry ; 12(1): 168, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459266

RESUMO

Blockade of N-methyl-D-aspartate receptors (NMDAR) is known to augment cortical serotonin 2A receptors (5-HT2ARs), which is implicated in psychosis. However, the pathways from NMDAR hypofunction to 5-HT2AR up-regulation are unclear. Here we addressed in mice whether genetic deletion of the indispensable NMDAR-subunit Grin1 principally in corticolimbic parvalbumin-positive fast-spiking interneurons, could up-regulate 5-HT2ARs leading to cortical hyper-excitability. First, in vivo local-field potential recording revealed that auditory cortex in Grin1 mutant mice became hyper-excitable upon exposure to acoustic click-train stimuli that release 5-HT in the cortex. This excitability increase was reproduced ex vivo where it consisted of an increased frequency of action potential (AP) firing in layer 2/3 pyramidal neurons of mutant auditory cortex. Application of the 5-HT2AR agonist TCB-2 produced similar results. The effect of click-trains was reversed by the 5-HT2AR antagonist M100907 both in vivo and ex vivo. Increase in AP frequency of pyramidal neurons was also reversed by application of Gαq protein inhibitor BIM-46187 and G protein-gated inwardly-rectifying K+ (GIRK) channel activator ML297. In fast-spiking interneurons, 5-HT2AR activation normally promotes GABA release, contributing to decreased excitability of postsynaptic pyramidal neurons, which was missing in the mutants. Moreover, unlike the controls, the GABAA receptor antagonist (+)-bicuculline had little effect on AP frequency of mutant pyramidal neurons, indicating a disinhibition state. These results suggest that the auditory-induced hyper-excitable state is conferred via GABA release deficits from Grin1-lacking interneurons leading to 5-HT2AR dysregulation and GIRK channel suppression in cortical pyramidal neurons, which could be involved in auditory psychosis.


Assuntos
Receptores de N-Metil-D-Aspartato , Esquizofrenia , Animais , Modelos Animais de Doenças , Camundongos , Células Piramidais/metabolismo , Receptor 5-HT2A de Serotonina/genética , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Ácido gama-Aminobutírico/metabolismo
9.
J Neurophysiol ; 105(4): 1768-84, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21307327

RESUMO

Dysfunctions of neuronal and network excitability have emerged as common features in disorders associated with intellectual disabilities, autism, and seizure activity, all common clinical manifestations of Rett syndrome (RTT), a neurodevelopmental disorder caused by loss-of-function mutations in the transcriptional regulator methyl-CpG-binding protein 2 (MeCP2). Here, we evaluated the consequences of Mecp2 mutation on hippocampal network excitability, as well as synapse structure and function using a combination of imaging and electrophysiological approaches in acute slices. Imaging the amplitude and spatiotemporal spread of neuronal depolarizations with voltage-sensitive dyes (VSD) revealed that the CA1 and CA3 regions of hippocampal slices from symptomatic male Mecp2 mutant mice are highly hyperexcitable. However, only the density of docked synaptic vesicles and the rate of release from the readily releasable pool are impaired in Mecp2 mutant mice, while synapse density and morphology are unaffected. The differences in network excitability were not observed in surgically isolated CA1 minislices, and blockade of GABAergic inhibition enhanced VSD signals to the same extent in Mecp2 mutant and wild-type mice, suggesting that network excitability originates in area CA3. Indeed, extracellular multiunit recordings revealed a higher level of spontaneous firing of CA3 pyramidal neurons in slices from symptomatic Mecp2 mutant mice. The neuromodulator adenosine reduced the amplitude and spatiotemporal spread of VSD signals evoked in CA1 of Mecp2 mutant slices to wild-type levels, suggesting its potential use as an anticonvulsant in RTT individuals. The present results suggest that hyperactive CA3 pyramidal neurons contribute to hippocampal dysfunction and possibly to limbic seizures observed in Mecp2 mutant mice and RTT individuals.


Assuntos
Hipocampo/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/genética , Mutação/genética , Rede Nervosa/fisiopatologia , Imagens com Corantes Sensíveis à Voltagem/métodos , Animais , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Sistema Límbico/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Células Piramidais/fisiopatologia , Síndrome de Rett/fisiopatologia , Sinapses/fisiologia
10.
J Neurophysiol ; 106(5): 2189-200, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21795624

RESUMO

Focal cortical dysplasia is associated with the development of seizures in children and is present in up to 40% of intractable childhood epilepsies. Transcortical freeze lesions in newborn rats reproduce many of the anatomical and physiological characteristics of human cortical dysplasia. Rats with freeze lesions have increased seizure susceptibility and a region of hyperexcitable cortex adjacent to the lesion. Since alterations in hyperpolarization-activated nonspecific cation (HCN) channels are often associated with epilepsy, we used whole cell patch-clamp recording and voltage-sensitive dye imaging to examine alterations in HCN channels and inwardly rectifying hyperpolarization-activated currents (I(h)) in cortical dysplasia. (L5) pyramidal neurons in lesioned animals had hyperpolarized resting membrane potentials, increased input resistances and reduced voltage "sag" associated with I(h) activation. These differences became nonsignificant after application of the I(h) blocker ZD7288. Temporal excitatory postsynaptic potential (EPSP) summation and intrinsic excitability were increased in neurons near the freeze lesion. Using voltage-sensitive dye imaging of neocortical slices, we found that inhibiting I(h) with ZD7288 increased the half-width of dye signals. The anticonvulsant lamotrigine produced a significant decrease in spread of activity. The ability of lamotrigine to decrease network activity was reduced in the hyperexcitable cortex near the freeze lesion. These results suggest that I(h) serves to constrain network activity in addition to its role in regulating cellular excitability. Reduced I(h) may contribute to increased network excitability in cortical dysplasia.


Assuntos
Córtex Cerebral/fisiopatologia , Malformações do Desenvolvimento Cortical/fisiopatologia , Células Piramidais/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Anticonvulsivantes/farmacologia , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Epilepsia/tratamento farmacológico , Epilepsia/patologia , Epilepsia/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Lamotrigina , Malformações do Desenvolvimento Cortical/tratamento farmacológico , Malformações do Desenvolvimento Cortical/patologia , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Células Piramidais/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Triazinas/farmacologia , Imagens com Corantes Sensíveis à Voltagem/métodos
11.
Cell Mol Neurobiol ; 31(4): 587-96, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21274618

RESUMO

The metabotropic GABA(B) and adenosine A(1) receptors mediate presynaptic inhibition through regulation of voltage-dependent Ca(2+) channels, whereas K(+) channel regulation is believed to have no role at the CA3-CA1 synapse. We show here that the inhibitory effect of baclofen (20 µM) and adenosine (300 µM) on field EPSPs are differentially sensitive to Cs(+) (3.5 mM) and Ba(2+) (200 µM), but not 4-aminopyridine (100 µM). Barium had no effect on paired-pulse facilitation (PPF) in itself, but gave significant reduction (14 ± 5%) when applied in the presence of baclofen, but not adenosine, suggesting that the effect is presynaptic and selective on the GABA(B) receptor-mediated response. The effect of Ba(2+) on PPF was not mimicked by tertiapin (30 nM), indicating that the underlying mechanism does not involve GIRK channels. Barium did not affect PPF in slices from young rats (P7-P8), suggesting developmental regulation. The above effects of Ba(2+) on adult tissue were reproduced when measuring evoked whole-cell EPSCs from CA1 pyramidal neurons: PPF was reduced by 22 ± 3% in the presence of baclofen and unaltered in adenosine. In contrast, Ba(2+) caused no significant change in frequency or amplitude of miniature EPSCs. The Ba(2+)-induced reduction of PPF was antagonized by LY341495, suggesting metabotropic glutamate receptor involvement. We propose that these novel effects of Ba(2+) and Cs(+) are exerted through blockade of inwardly rectifying K(+) channels in glial cells, which are functionally interacting with the GABA(B) receptor-dependent glutamate release that generates heterosynaptic depression.


Assuntos
Adenosina/farmacologia , Baclofeno/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , 4-Aminopiridina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Aminoácidos/farmacologia , Animais , Bário/farmacologia , Venenos de Abelha/farmacologia , Região CA1 Hipocampal/fisiologia , Região CA3 Hipocampal/fisiologia , Césio/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas In Vitro , Masculino , Ratos , Ratos Wistar , Receptor A1 de Adenosina/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Sinapses/fisiologia , Teofilina/análogos & derivados , Teofilina/farmacologia , Xantenos/farmacologia
12.
Epilepsia ; 51 Suppl 3: 52-5, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20618401

RESUMO

Cortical dysplasia is often associated with intractable seizures. Studies in animal models have described changes in inhibitory and excitatory synaptic function that contribute to hyperexcitability. The role of changes in intrinsic excitability and abnormal dendritic properties has received less attention. Changes in hyperpolarization-activated nonselective cation (HCN) channels have been implicated in several models of epilepsy. Herein we review evidence for alterations in HCN channels and dendritic morphology in the rat freeze-lesion model of cortical dysplasia. Immunocytochemical HCN1 staining, typically seen in the apical dendrites of layer V pyramidal cells in normal cortex, was greatly reduced in the region adjacent to the freeze-induced microgyrus. Although staining was preserved in layer I, fewer dendrites were stained in upper cortical layers. Deeper cortical layers were virtually devoid of immunoreactivity. Examination of biocytin-labeled pyramidal cells revealed markedly altered dendritic trees in the lesioned animals. In addition, resting membrane properties were altered and a subpopulation of neurons with abnormal dendritic arbors was present. These changes are likely to interact with the previously reported synaptic changes in this model of cortical dysplasia. HCN channel alterations are a potentially important cellular mechanism underlying hyperexcitability in cortical dysplasia.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Epilepsia/etiologia , Malformações do Desenvolvimento Cortical/patologia , Canais de Potássio/fisiologia , Células Piramidais/patologia , Animais , Animais Recém-Nascidos , Canais de Cátion Regulados por Nucleotídeos Cíclicos/biossíntese , Modelos Animais de Doenças , Epilepsia/patologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Malformações do Desenvolvimento Cortical/complicações , Canais de Potássio/biossíntese , Células Piramidais/fisiopatologia , Ratos , Ratos Sprague-Dawley
13.
Neuropsychopharmacology ; 45(13): 2207-2218, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32859995

RESUMO

Cortical gamma oscillations are believed to be involved in mental processes which are disturbed in schizophrenia. For example, the magnitudes of sensory-evoked oscillations, as measured by auditory steady-state responses (ASSRs) at 40 Hz, are robustly diminished, whereas the baseline gamma power is enhanced in schizophrenia. Such dual gamma oscillation abnormalities are also present in a mouse model of N-methyl-D-aspartate receptor hypofunction (Ppp1r2cre/Grin1 knockout mice). However, it is unclear whether the abnormal gamma oscillations are associated with dysfunction in schizophrenia. We found that glycogen synthase kinase-3 (GSK3) is overactivated in corticolimbic parvalbumin-positive GABAergic interneurons in Grin1 mutant mice. Here we addressed whether GSK3ß inhibition reverses both abnormal gamma oscillations and behavioral deficits with high correlation by pharmacological and genetic approach. We demonstrated that the paralog selective-GSK3ß inhibitor, but not GSK3α inhibitor, normalizes the diminished ASSRs, excessive baseline gamma power, and deficits in spatial working memory and prepulse inhibition (PPI) of acoustic startle in Grin1 mutant mice. Cell-type specific GSK3B knockdown, but not GSK3A knockdown, also reversed abnormal gamma oscillations and behavioral deficits. Moreover, GSK3B knockdown, but not GSK3A knockdown, reverses the mutants' in vivo spike synchrony deficits. Finally, ex vivo patch-clamp recording from pairs of neighboring cortical pyramidal neurons showed a reduction of synchronous spontaneous inhibitory-postsynaptic-current events in mutants, which was reversed by GSK3ß inhibition genetically and pharmacologically. Together, GSK3ß inhibition in corticolimbic interneurons ameliorates the deficits in spatial working memory and PPI, presumably by restoration of synchronous GABA release, synchronous spike firing, and evoked-gamma power increase with lowered baseline power.


Assuntos
Receptores de N-Metil-D-Aspartato , Esquizofrenia , Animais , Cognição , Quinase 3 da Glicogênio Sintase , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Knockout , Esquizofrenia/tratamento farmacológico
14.
J Neurosci ; 28(3): 725-31, 2008 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-18199771

RESUMO

Inhibitory control of local neuronal circuits is critical for prefrontal cortical functioning. Modulation of inhibitory circuits by several neuromodulators has been demonstrated, but the underlying mechanisms are unclear. Neuromodulator effects on synaptic vesicle recycling have received little attention. Controversy also exists whether different pools of synaptic vesicles underlie spontaneous and activity-dependent vesicle recycling. We therefore investigated the effects of kainate receptor activation on GABA release in rat prefrontal neocortex using electrophysiological and styryl dye imaging techniques in acute neocortical slices. Electrophysiological studies demonstrated that activation of kainate receptors increased the frequency, but not the amplitude of miniature IPSCs, suggesting a presynaptic action. Using styryl dye staining and multiphoton excitation microscopy, we visualized vesicular release from inhibitory GABAergic terminals in prefrontal cortical slices and demonstrate that kainate facilitates GABA release from presynaptic terminals. Our findings also indicate the presence of two pools of GABA-containing vesicles within inhibitory terminals. Kainate modulates both pools but only when vesicles are endocytosed and exocytosed by matching protocols of dye loading, i.e., spontaneous or evoked afferent activity.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Caínico/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Potenciais Pós-Sinápticos Inibidores/efeitos da radiação , Neocórtex/citologia , Técnicas de Patch-Clamp/métodos , Potássio/farmacologia , Terminações Pré-Sinápticas/efeitos da radiação , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/efeitos da radiação , Compostos de Piridínio/metabolismo , Compostos de Amônio Quaternário/metabolismo , Ratos , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
15.
Neuroscientist ; 15(3): 218-24, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19436074

RESUMO

Fast synaptic inhibition in the neocortex is mediated by the neurotransmitter GABA, acting on GABA( A) receptors. Neurotransmitters, including GABA, are stored in synaptic vesicles at presynaptic nerve terminals. A long-held assumption has been that evoked and spontaneous neurotransmissions draw on the same pools of vesicles. We review the evidence from FM1-43 studies supporting the contention that at least 2 distinct pools of GABA vesicles are present at inhibitory synapses in the rat neocortex. FM1-43 uptake during spontaneous vesicle endocytosis labels a vesicle pool within neocortical inhibitory nerve terminals that is released much more slowly ("reluctant" pool) than those vesicles loaded by electrical stimulation of afferent fibers or hyperkalemic solutions. These multiple pools may play diverse roles in such processes as long-term depression and/or potentiating of inhibitory synaptic transmission, homeostatic plasticity of inhibitory activity, or developmental changes in inhibitory synaptic transmission.


Assuntos
Neurônios/citologia , Sinapses/fisiologia , Vesículas Sinápticas/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Dopamina/farmacologia , Neocórtex/citologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Compostos de Piridínio , Compostos de Amônio Quaternário , Sinapses/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos
16.
Neurobiol Dis ; 32(2): 254-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18674619

RESUMO

Glutamate transporters function to maintain low levels of extracellular glutamate and play an important role in synaptic transmission at many synapses. Disruption of glutamate transporter function or expression can result in increased extracellular glutamate levels. Alterations in glutamate transporter expression have been reported in human epilepsy and animal seizure models. Functional electrophysiological changes that occur when transporter expression is disrupted in chronic epilepsy models have not been examined. Here, we used a freeze-induced model of cortical dysplasia to test the role of glutamate transporters in synaptic hyperexcitability. We report that inhibiting glutamate transporters with the non-selective antagonist, DL-threo-beta-benzylozyaspartic acid (TBOA) preferentially prolongs postsynaptic currents (PSCs) and decreases the threshold for evoking epileptiform activity in lesioned compared to control cortex. The effect of inhibiting uptake is mediated primarily by the glia glutamate transporter (GLT-1) since the selective antagonist dihydrokainate (DHK) mimicked the effects of TBOA. The effect of uptake inhibition is mediated by activation of N-methyl-D-aspartate (NMDA) receptors since D-(-)-2-amino-5-phosphonovaleric acid (APV) prevents TBOA-induced effects. Neurons in lesioned cortex also have a larger tonic NMDA current. These results indicate that chronic changes in glutamate transporters and NMDA receptors contribute to hyperexcitability in cortical dysplasia.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Córtex Cerebral/patologia , Malformações do Desenvolvimento Cortical/metabolismo , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Ácido Aspártico/farmacologia , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Malformações do Desenvolvimento Cortical/etiologia , Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Ratos , Ratos Sprague-Dawley
17.
Neuropharmacology ; 55(1): 106-16, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18508095

RESUMO

We examined the mechanisms of kainate (KA) induced modulation of GABA release in rat prefrontal cortex. Pharmacologically isolated IPSCs were recorded from visually identified layer II/III pyramidal cells using whole-cell patch clamp techniques. KA produced an increase in evoked IPSC amplitude at low nanomolar concentrations (100-500 nM). The frequency but not the amplitude of miniature (m) IPSCs was also increased. The GluR5 subunit selective agonist (RS)-2-amino-3-(3-hydroxy-5-tert-butylisoxazol-4-yl) propanoic acid (ATPA) caused an increase in mIPSC frequency whereas (3S,4aR,6S,8aR)-6-(4-carboxyphenyl)methyl-1,2,3,4,4a,5,6,7,8,8a-decahydroisoquinoline-3-carboxylic acid (LY382884), a selective GluR5 subunit antagonist, inhibited this facilitation. Philanthotoxin-433 (PhTx) blocked the effect of KA, indicating involvement of Ca(2+)-permeable GluR5 receptors. No IPSC facilitation was seen when Ca(2+) was omitted from the bathing solution. Facilitation was observed when slices were preincubated in ruthenium red or high concentrations of ryanodine, but was inhibited with application of thapsigargin. The IP3 receptor (IP3R) antagonists diphenylboric acid 2-amino-ethyl ester (2-APB) (15 microM) and Xestospongin C (XeC) blocked IPSC facilitation. These results show that activation of KA receptors (KARs) on GABAergic nerve terminals results is linked to intracellular Ca(2+) release via activation of IP3, but not ryanodine, receptors. This represents a new mechanism of presynaptic modulation whereby Ca(2+) entry through Ca(2+)-permeable GluR5 subunit containing KARs activates IP3Rs receptors leading to an increase in GABA release.


Assuntos
Cálcio/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Ácido Caínico/farmacologia , Neocórtex/citologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Animais , Compostos de Boro/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/fisiologia , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Compostos Macrocíclicos/farmacologia , Neocórtex/efeitos dos fármacos , Neocórtex/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Antagonistas Nicotínicos/farmacologia , Oxazóis/farmacologia , Técnicas de Patch-Clamp , Poliaminas/farmacologia , Terminações Pré-Sinápticas/metabolismo , Ratos
18.
Front Cell Neurosci ; 12: 20, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29440994

RESUMO

Layer 1 (L1) interneurons (INs) play a key role in modulating the integration of inputs to pyramidal neurons (PNs) and controlling cortical network activity. Hyperpolarization-activated, cyclic nucleotide-gated, non-specific cation (HCN) channels are known to alter the intrinsic and synaptic excitability of principal components (PCs) as well as select populations of GABAergic INs. However, the developmental profile and functional role of HCN channels in diverse L1 IN populations is not completely understood. In the present study, we used electrophysiological characterization, in conjunction with unbiased hierarchical cluster analysis, to examine developmental modulation of L1 INs by HCN channels in the rat medial agranular cortex (AGm). We identified three physiologically discrete IN populations which were classified as regular spiking (RS), burst accommodating (BA) and non-accommodating (NA). A distinct developmental pattern of excitability modulation by HCN channels was observed for each group. RS and NA cells displayed distinct morphologies with modulation of EPSPs increasing in RS cells and decreasing in NA cells across development. The results indicate a possible role of HCN channels in the formation and maintenance of cortical circuits through alteration of the excitability of distinct AGm L1 INs.

19.
Neuropharmacology ; 53(1): 37-47, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17543353

RESUMO

Kainate receptors mediate both direct excitatory and indirect modulatory actions in the CNS. We report here that kainate has both pre- and postsynaptic actions in layer II/III pyramidal neurons of rat prefrontal cortex. Application of low concentration of kainate (50-500 nM) increased the amplitude of evoked excitatory postsynaptic currents (EPSCs) whereas higher concentrations (3 microM) caused a decrease. The frequency of spontaneous and miniature (action potential-independent) EPSCs was increased by low concentrations of kainate without affecting their amplitudes, suggesting a presynaptic mechanism of action. The facilitatory and inhibitory effects of kainate were mimicked by the GluR5 subunit selective agonist ATPA. In addition to decreasing EPSC amplitudes, high concentrations of kainate and ATPA induced an inward current which was not blocked by AMPA- or NMDA-receptor antagonists GYKI52466 and D-APV, respectively. The inward currents were blocked by the AMPA/KA receptor antagonist CNQX, indicating the presence of postsynaptic kainate receptors. Single shock stimulation in the presence of GYKI52466 and D-APV evoked an EPSC which was blocked by CNQX. The GluR5 antagonist LY382884 changed paired-pulse facilitation to paired pulse depression, indicating that synaptically released glutamate can activate presynaptic kainate receptors. These results suggest that kainate receptors containing GluR5 subunits play a major role in glutamatergic transmission in rat neocortex, having both presynaptic modulatory and direct postsynaptic excitatory actions.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Caínico/farmacologia , Neocórtex/citologia , Células Piramidais/citologia , Sinapses/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Técnicas In Vitro , Técnicas de Patch-Clamp/métodos , Ratos , Ratos Sprague-Dawley
20.
Front Cell Neurosci ; 11: 109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28469560

RESUMO

Cortical malformations are often associated with pharmaco-resistant epilepsy. Alterations in hyperpolarization-activated, cyclic nucleotide-gated, non-specific cation (HCN) channels have been shown to contribute to malformation associated hyperexcitability. We have recently demonstrated that expression of HCN channels and Ih current amplitudes are reduced in layer (L) 5 pyramidal neurons of rats with freeze lesion induced malformations. These changes were associated with an increased EPSP temporal summation. Here, we examine the effects of HCN channel inhibition on synaptic responses in fast spiking, presumptive basket cells and accommodating, presumptive Martinotti, GABAergic interneurons in slices from freeze lesioned animals. In control animals, fast spiking cells showed small sag responses which were reduced by the HCN channel antagonist ZD7288. Fast spiking cells in lesioned animals showed absent or reduced sag responses. The amplitude of single evoked EPSPs in fast spiking cells in the control group was not affected by HCN channel inhibition with ZD7288. EPSP ratios during short stimulus trains at 25 Hz were not significantly different between control and lesion groups. ZD7288 produced an increase in EPSP ratios in the control but not lesion groups. Under voltage clamp conditions, ZD7288 did not affect EPSC ratios. In the control group, accommodating interneurons showed robust sag responses which were significantly reduced by ZD7288. HCN channel inhibition increased EPSP ratios and area in controls but not the lesioned group. The results indicate that HCN channels differentially modulate EPSPs in different classes of GABAergic interneurons and that this control is reduced in malformed rat neocortex.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA