Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(20): e2120870119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35544691

RESUMO

Transient receptor potential canonical 4 (TRPC4) is a receptor-operated cation channel codependent on both the Gq/11­phospholipase C signaling pathway and Gi/o proteins for activation. This makes TRPC4 an excellent coincidence sensor of neurotransmission through Gq/11- and Gi/o-coupled receptors. In whole-cell slice recordings of lateral septal neurons, TRPC4 mediates a strong depolarizing plateau that shuts down action potential firing, which may or may not be followed by a hyperpolarization that extends the firing pause to varying durations depending on the strength of Gi/o stimulation. We show that the depolarizing plateau is codependent on Gq/11-coupled group I metabotropic glutamate receptors and on Gi/o-coupled γ-aminobutyric acid type B receptors. The hyperpolarization is mediated by Gi/o activation of G protein­activated inwardly rectifying K+ (GIRK) channels. Moreover, the firing patterns, elicited by either electrical stimulation or receptor agonists, encode information about the relative strengths of Gq/11 and Gi/o inputs in the following fashion. Pure Gq/11 input produces weak depolarization accompanied by firing acceleration, whereas pure Gi/o input causes hyperpolarization that pauses firing. Although coincident Gq/11­Gi/o inputs also pause firing, the pause is preceded by a burst, and both the pause duration and firing recovery patterns reflect the relative strengths of Gq/11 versus Gi/o inputs. Computer simulations demonstrate that different combinations of TRPC4 and GIRK conductances are sufficient to produce the range of firing patterns observed experimentally. Thus, concurrent neurotransmission through the Gq/11 and Gi/o pathways is converted to discernible electrical responses by the joint actions of TRPC4 and GIRK for communication to downstream neurons.


Assuntos
Potenciais de Ação , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Subunidades alfa de Proteínas de Ligação ao GTP , Neurônios , Transmissão Sináptica , Canais de Cátion TRPC , Animais , Comunicação Celular , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Camundongos , Neurônios/fisiologia , Canais de Cátion TRPC/fisiologia
2.
Neuropharmacology ; 209: 109021, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35245509

RESUMO

G protein-gated inwardly rectifying potassium (GIRK) channels are one of the main regulators of neuronal excitability. Activation of GIRK channels in the CNS usually leads to postsynaptic inhibition. However, the function of GIRK channels in the presynaptic processes, notably neurotransmitter release form motor nerve terminals, is yet to be comprehensively understood. Here, using electrophysiological and fluorescent approaches, the role of GIRK channels in neurotransmitter release from frog motor nerve terminals was studied. We found that the inhibition of GIRK channels with nanomolar tertiapin-Q synchronized exocytosis events with action potential but suppressed spontaneous and evoked neurotransmitter release, as well as Ca2+ transient and membrane permeability for K+. The action of GIRK channel inhibition on evoked neurotransmission was prevented by selective antagonist of voltage-gated Ca2+ channels of L-type. Furthermore, the effects of muscarinic acetylcholine receptor activation on neurotransmitter release, Ca2+ transient and K+ channel activity were markedly modulated by inhibition of GIRK channels. Thus, at the motor nerve terminals GIRK channels can regulate timing of neurotransmitter release and be a positive modulator of synaptic vesicle exocytosis acting partially via L-type Ca2+ channels. In addition, GIRK channels are key players in a feedback control of neurotransmitter release by muscarinic acetylcholine receptors.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Junção Neuromuscular , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Neurotransmissores/farmacologia , Receptores Muscarínicos , Transmissão Sináptica
3.
J Neurosci ; 41(33): 7086-7102, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34261700

RESUMO

The G-protein-gated inwardly rectifying potassium (Kir3/GIRK) channel is the effector of many G-protein-coupled receptors (GPCRs). Its dysfunction has been linked to the pathophysiology of Down syndrome, Alzheimer's and Parkinson's diseases, psychiatric disorders, epilepsy, drug addiction, or alcoholism. In the hippocampus, GIRK channels decrease excitability of the cells and contribute to resting membrane potential and inhibitory neurotransmission. Here, to elucidate the role of GIRK channels activity in the maintenance of hippocampal-dependent cognitive functions, their involvement in controlling neuronal excitability at different levels of complexity was examined in C57BL/6 male mice. For that purpose, GIRK activity in the dorsal hippocampus CA3-CA1 synapse was pharmacologically modulated by two drugs: ML297, a GIRK channel opener, and Tertiapin-Q (TQ), a GIRK channel blocker. Ex vivo, using dorsal hippocampal slices, we studied the effect of pharmacological GIRK modulation on synaptic plasticity processes induced in CA1 by Schaffer collateral stimulation. In vivo, we performed acute intracerebroventricular (i.c.v.) injections of the two GIRK modulators to study their contribution to electrophysiological properties and synaptic plasticity of dorsal hippocampal CA3-CA1 synapse, and to learning and memory capabilities during hippocampal-dependent tasks. We found that pharmacological disruption of GIRK channel activity by i.c.v. injections, causing either function gain or function loss, induced learning and memory deficits by a mechanism involving neural excitability impairments and alterations in the induction and maintenance of long-term synaptic plasticity processes. These results support the contention that an accurate control of GIRK activity must take place in the hippocampus to sustain cognitive functions.SIGNIFICANCE STATEMENT Cognitive processes of learning and memory that rely on hippocampal synaptic plasticity processes are critically ruled by a finely tuned neural excitability. G-protein-gated inwardly rectifying K+ (GIRK) channels play a key role in maintaining resting membrane potential, cell excitability and inhibitory neurotransmission. Here, we demonstrate that modulation of GIRK channels activity, causing either function gain or function loss, transforms high-frequency stimulation (HFS)-induced long-term potentiation (LTP) into long-term depression (LTD), inducing deficits in hippocampal-dependent learning and memory. Together, our data show a crucial GIRK-activity-mediated mechanism that governs synaptic plasticity direction and modulates subsequent hippocampal-dependent cognitive functions.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Hipocampo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Condicionamento Operante/fisiologia , Emoções/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Aprendizagem/fisiologia , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Desempenho Psicomotor/fisiologia
4.
Pharmacol Ther ; 223: 107808, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33476640

RESUMO

G protein-gated inwardly rectifying potassium channels (Kir3/GirK) are important for maintaining resting membrane potential, cell excitability and inhibitory neurotransmission. Coupled to numerous G protein-coupled receptors (GPCRs), they mediate the effects of many neurotransmitters, neuromodulators and hormones contributing to the general homeostasis and particular synaptic plasticity processes, learning, memory and pain signaling. A growing number of behavioral and genetic studies suggest a critical role for the appropriate functioning of the central nervous system, as well as their involvement in many neurologic and psychiatric conditions, such as neurodegenerative diseases, mood disorders, attention deficit hyperactivity disorder, schizophrenia, epilepsy, alcoholism and drug addiction. Hence, GirK channels emerge as a very promising tool to be targeted in the current scenario where these conditions already are or will become a global public health problem. This review examines recent findings on the physiology, function, dysfunction, and pharmacology of GirK channels in the central nervous system and highlights the relevance of GirK channels as a worthful potential target to improve therapies for related diseases.


Assuntos
Doenças do Sistema Nervoso Central , Sistema Nervoso Central , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Terapia de Alvo Molecular , Sistema Nervoso Central/fisiologia , Doenças do Sistema Nervoso Central/tratamento farmacológico , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Humanos
5.
J Neurosci ; 40(44): 8543-8555, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33020214

RESUMO

A rare mutation affecting the Forkhead-box protein P2 (FOXP2) transcription factor causes a severe monogenic speech and language disorder. Mice carrying an identical point mutation to that observed in affected patients (Foxp2+/R552H mice) display motor deficits and impaired synaptic plasticity in the striatum. However, the consequences of the mutation on neuronal function, in particular in the cerebral cortex, remain little studied. Foxp2 is expressed in a subset of Layer VI cortical neurons. Here, we used Ntsr1-EGFP mice to identify Foxp2+ neurons in the mouse auditory cortex ex vivo. We studied the functional impact of the R552H mutation on the morphologic and functional properties of Layer VI cortical neurons from Ntsr1-EGFP; Foxp2+/R552H male and female mice. The complexity of apical, but not basal dendrites was significantly lower in Foxp2+/R552H cortico-thalamic neurons than in control Foxp2+/+ neurons. Excitatory synaptic inputs, but not inhibitory synaptic inputs, were decreased in Foxp2+/R552H mice. In response, homeostatic mechanisms would be expected to increase neuronal gain, i.e., the conversion of a synaptic input into a firing output. However, the intrinsic excitability of Foxp2+ cortical neurons was lower in Foxp2+/R552H neurons. A-type and delayed-rectifier (DR) potassium currents, two putative transcriptional targets of Foxp2, were not affected by the mutation. In contrast, GABAB/GIRK signaling, another presumed target of Foxp2, was increased in mutant neurons. Blocking GIRK channels strongly attenuated the difference in intrinsic excitability between wild-type (WT) and Foxp2+/R552H neurons. Our results reveal a novel role for Foxp2 in the control of neuronal input/output homeostasis.SIGNIFICANCE STATEMENT Mutations of the Forkhead-box protein 2 (FOXP2) gene in humans are the first known monogenic cause of a speech and language disorder. The Foxp2 mutation may directly affect neuronal development and function in neocortex, where Foxp2 is expressed. Brain imaging studies in patients with a heterozygous mutation in FOXP2 showed abnormalities in cortical language-related regions relative to the unaffected members of the same family. However, the role of Foxp2 in neocortical neurons is poorly understood. Using mice with a Foxp2 mutation equivalent to that found in patients, we studied functional modifications in auditory cortex neurons ex vivo We found that mutant neurons exhibit alterations of synaptic input and GABAB/GIRK signaling, reflecting a loss of neuronal homeostasis.


Assuntos
Córtex Cerebral/fisiologia , Fatores de Transcrição Forkhead/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Neurônios/fisiologia , Receptores de GABA-B/fisiologia , Proteínas Repressoras/genética , Tálamo/fisiologia , Animais , Córtex Cerebral/citologia , Canais de Potássio de Retificação Tardia/fisiologia , Espinhas Dendríticas/fisiologia , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Antagonistas GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Vias Neurais/citologia , Vias Neurais/fisiologia , Sinapses/fisiologia , Tálamo/citologia
6.
Neuropharmacology ; 176: 108214, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32622786

RESUMO

Rett Syndrome (RTT) is an X-linked neurodevelopmental disorder caused mainly by mutations in the MECP2 gene. One of the major RTT features is breathing dysfunction characterized by periodic hypo- and hyperventilation. The breathing disorders are associated with increased brainstem neuronal excitability, which can be alleviated with GABA agonists. Since neuronal hypoexcitability occurs in the forebrain of RTT models, it is necessary to find pharmacological agents with a relative preference to brainstem neurons. Here we show evidence for the improvement of breathing disorders of Mecp2-disrupted mice with the brainstem-acting drug cloperastine (CPS) and its likely neuronal targets. CPS is an over-the-counter cough medicine that has an inhibitory effect on brainstem neuronal networks. In Mecp2-disrupted mice, CPS (30 mg/kg, i.p.) decreased the occurrence of apneas/h and breath frequency variation. GIRK currents expressed in HEK cells were inhibited by CPS with IC50 1 µM. Whole-cell patch clamp recordings in locus coeruleus (LC) and dorsal tegmental nucleus (DTN) neurons revealed an overall inhibitory effect of CPS (10 µM) on neuronal firing activity. Such an effect was reversed by the GABAA receptor antagonist bicuculline (20 µM). Voltage clamp studies showed that CPS increased GABAergic sIPSCs in LC cells, which was blocked by the GABAB receptor antagonist phaclofen. Functional GABAergic connections of DTN neurons with LC cells were shown. These results suggest that CPS improves breathing dysfunction in Mecp2-null mice by blocking GIRK channels in synaptic terminals and enhancing GABA release.


Assuntos
Antitussígenos/uso terapêutico , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Piperidinas/uso terapêutico , Respiração/efeitos dos fármacos , Síndrome de Rett/tratamento farmacológico , Ácido gama-Aminobutírico/metabolismo , Animais , Antitussígenos/farmacologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Relação Dose-Resposta a Droga , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Agonistas GABAérgicos/farmacologia , Agonistas GABAérgicos/uso terapêutico , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Ratos , Síndrome de Rett/genética , Síndrome de Rett/fisiopatologia
7.
Cerebellum ; 19(6): 751-761, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32617840

RESUMO

G-protein-coupled inwardly rectifying potassium (GIRK) channels contribute to the resting membrane potential of many neurons and play an important role in controlling neuronal excitability. Although previous studies have revealed a high expression of GIRK subunits in the cerebellum, their functional role has never been clearly described. Using patch-clamp recordings in mice cerebellar slices, we examined the properties of the GIRK currents in Purkinje cells (PCs) and investigated the effects of a selective agonist of GIRK1-containing channels, ML297 (ML), on PC firing and synaptic plasticity. We demonstrated that GIRK channel activation decreases the PC excitability by inhibiting both sodium and calcium spikes and, in addition, modulates the complex spike response evoked by climbing fiber stimulation. Our results indicate that GIRK channels have also a marked effect on synaptic plasticity of the parallel fiber-PC synapse, as the application of ML297 increased the expression of LTP while preventing LTD. We, therefore, propose that the recruitment of GIRK channels represents a crucial mechanism by which neuromodulators can control synaptic strength and membrane conductance for proper refinement of the neural network involved in memory storage and higher cognitive functions.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Plasticidade Neuronal/fisiologia , Neurotransmissores/farmacologia , Células de Purkinje/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/agonistas , Masculino , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Células de Purkinje/efeitos dos fármacos , Pirazóis/farmacologia
8.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 42(1): 108-116, 2020 Feb 28.
Artigo em Chinês | MEDLINE | ID: mdl-32131949

RESUMO

G protein-gated inward rectifier potassium(GIRK)channels are widely distributed in the central nervous system and play important roles in maintaining the resting membrane potential of neurons,adjusting neuronal excitability,and regulating the release of neurotransmitter.Studies have shown that addictive behavior is closely related to the expression and activity of the GIRK channels in the brain reward system and the GIRK channels may be a potential target for addiction treatment.This article summarizes the recent research advances in GIRK channels in terms of structure,intracranial tissue distribution,and especially substance addiction.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Neurônios/citologia , Transtornos Relacionados ao Uso de Substâncias , Encéfalo/fisiologia , Humanos , Potenciais da Membrana
9.
Naunyn Schmiedebergs Arch Pharmacol ; 393(4): 573-580, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31720798

RESUMO

Chloroform has been used over decades in anesthesia before it was replaced by other volatile anesthetics like halothane or sevoflurane. Some of the reasons were inadmissible side effects of chloroform like bradycardia or neural illness. In the present study, we identified members of the G protein-activated inwardly rectifying potassium channel family (Kir3) expressed in Xenopus oocytes as potential common molecular targets for both the neural and cardiac effects of chloroform. Millimolar concentration currents representing a 1:10000 dilution of commercially available chloroform were used in laboratories that augment neuronal Kir3.1/3.2 currents as well as cardiac Kir3.1/3.4. This effect was selective and only observed in currents from Kir3 subunits but not in currents from Kir2 subunits. Augmentation of atrial Kir3.1/3.4 currents leads to an effective drop of the heart rate and a reduction in contraction force in isolated mouse atria.


Assuntos
Função Atrial/efeitos dos fármacos , Bradicardia/induzido quimicamente , Clorofórmio/toxicidade , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Átrios do Coração/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Bradicardia/fisiopatologia , Células HEK293 , Humanos , Camundongos , Neurônios/fisiologia , Oócitos , Xenopus laevis
10.
J Gen Physiol ; 151(10): 1231-1246, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31511304

RESUMO

Inwardly rectifying potassium (Kir) channels play a key role in controlling membrane potentials in excitable and unexcitable cells, thereby regulating a plethora of physiological processes. G-protein-gated Kir channels control heart rate and neuronal excitability via small hyperpolarizing outward K+ currents near the resting membrane potential. Despite recent breakthroughs in x-ray crystallography and cryo-EM, the gating and conduction mechanisms of these channels are poorly understood. MD simulations have provided unprecedented details concerning the gating and conduction mechanisms of voltage-gated K+ and Na+ channels. Here, we use multi-microsecond-timescale MD simulations based on the crystal structures of GIRK2 (Kir3.2) bound to phosphatidylinositol-4,5-bisphosphate to provide detailed insights into the channel's gating dynamics, including insights into the behavior of the G-loop gate. The simulations also elucidate the elementary steps that underlie the movement of K+ ions through an inward-rectifier K+ channel under an applied electric field. Our simulations suggest that K+ permeation might occur via direct knock-on, similar to the mechanism recently shown for Kv channels.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Potássio/metabolismo , Simulação por Computador , Humanos , Simulação de Dinâmica Molecular , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ligação Proteica
11.
Br J Pharmacol ; 176(13): 2238-2249, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30924523

RESUMO

BACKGROUND AND PURPOSE: G protein-gated inwardly rectifying K+ (Kir 3) channels moderate the activity of excitable cells and have been implicated in neurological disorders and cardiac arrhythmias. Most neuronal Kir 3 channels consist of Kir 3.1 and Kir 3.2 subtypes, while cardiac Kir 3 channels consist of Kir 3.1 and Kir 3.4 subtypes. Previously, we identified a family of urea-containing Kir 3 channel activators, but these molecules exhibit suboptimal pharmacokinetic properties and modest selectivity for Kir 3.1/3.2 relative to Kir 3.1/3.4 channels. Here, we characterize a non-urea activator, VU0810464, which displays nanomolar potency as a Kir 3.1/3.2 activator, improved selectivity for neuronal Kir 3 channels, and improved brain penetration. EXPERIMENTAL APPROACH: We used whole-cell electrophysiology to measure the efficacy and potency of VU0810464 in neurons and the selectivity of VU0810464 for neuronal and cardiac Kir 3 channel subtypes. We tested VU0810464 in vivo in stress-induced hyperthermia and elevated plus maze paradigms. Parallel studies with ML297, the prototypical activator of Kir 3.1-containing Kir 3 channels, were performed to permit direct comparisons. KEY RESULTS: VU0810464 and ML297 exhibited comparable efficacy and potency as neuronal Kir 3 channel activators, but VU0810464 was more selective for neuronal Kir 3 channels. VU0810464, like ML297, reduced stress-induced hyperthermia in a Kir 3-dependent manner in mice. ML297, but not VU0810464, decreased anxiety-related behaviour as assessed with the elevated plus maze test. CONCLUSION AND IMPLICATIONS: VU0810464 represents a new class of Kir 3 channel activator with enhanced selectivity for Kir 3.1/3.2 channels. VU0810464 may be useful for examining Kir 3.1/3.2 channel contributions to complex behaviours and for probing the potential of Kir 3 channel-dependent manipulations to treat neurological disorders.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Neurônios/efeitos dos fármacos , Animais , Ansiedade/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Feminino , Febre/etiologia , Febre/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/fisiologia , Nó Sinoatrial/citologia , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia
12.
Brain Res ; 1710: 157-162, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30599137

RESUMO

The mechanisms underlying antiepileptic effects of deep brain stimulation (DBS) are complex and poorly understood. Studies on the effects of applied electric fields on epileptic nervous tissue could enable future advances in DBS treatments. Applied electric fields are known to inhibit or enhance epileptic activity in vitro through direct effects on local neurons, but it is unclear whether trans-synaptic effects participate in such actions. The present study investigates, in an epileptic brain slice model, the influence of GABAB receptor activation on excitatory and suppressive effects of a short-duration (10 ms) electric field in rat hippocampus. The results show that perfusion of the GABAB receptor antagonist, CGP 55845 (2 µM), could abolish applied-field induced suppression of orthodromic-stimulus evoked epileptiform afterdischarge activity in the CA1 region. GABAB receptor blockade was associated with an enhanced excitatory (proepileptic) effect of the applied field. However, the suppressive effect, observed in isolation using weak field stimuli, was left unchanged. The G-protein-activated inwardly rectifying K+ channel (GIRK) antagonist, tertiapin (30-50 nM), mimicked the effects of CGP 55845. The results suggest that the applied field activate (elements of) local interneurons to release GABA onto GABAB receptors. The resulting activation of postsynaptic GIRK channels inhibits neuronal activity thereby dampening the direct stimulatory effect of the applied field. The study indicates that local-stimulus induced GABAB receptor activation can serve a protective role under antiepileptic paradigms by preventing electrical stimulation from causing hyperexcitation.


Assuntos
Estimulação Elétrica , Epilepsia/fisiopatologia , Hipocampo/fisiopatologia , Neurônios/fisiologia , Receptores de GABA-B/fisiologia , Animais , Venenos de Abelha/administração & dosagem , Estimulação Encefálica Profunda , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Antagonistas de Receptores de GABA-B/administração & dosagem , Masculino , Ácidos Fosfínicos/administração & dosagem , Bloqueadores dos Canais de Potássio/administração & dosagem , Propanolaminas/administração & dosagem , Ratos Wistar
13.
J Neurosci ; 38(46): 10002-10015, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30315127

RESUMO

The neuromodulatory effects of GABA on pyramidal neurons are mediated by GABAB receptors (GABABRs) that signal via a conserved G-protein-coupled pathway. Two prominent effectors regulated by GABABRs include G-protein inwardly rectifying K+ (GIRK) and P/Q/N type voltage-gated Ca2+ (CaV2) ion channels that control excitability and synaptic output of these neurons, respectively. Regulator of G-protein signaling 7 (RGS7) has been shown to control GABAB effects, yet the specificity of its impacts on effector channels and underlying molecular mechanisms is poorly understood. In this study, we show that hippocampal RGS7 forms two distinct complexes with alternative subunit configuration bound to either membrane protein R7BP (RGS7 binding protein) or orphan receptor GPR158. Quantitative biochemical experiments show that both complexes account for targeting nearly the entire pool of RGS7 to the plasma membrane. We analyzed the effect of genetic elimination in mice of both sexes and overexpression of various components of RGS7 complex by patch-clamp electrophysiology in cultured neurons and brain slices. We report that RGS7 prominently regulates GABABR signaling to CaV2, in addition to its known involvement in modulating GIRK. Strikingly, only complexes containing R7BP, but not GPR158, accelerated the kinetics of both GIRK and CaV2 modulation by GABABRs. In contrast, GPR158 overexpression exerted the opposite effect and inhibited RGS7-assisted temporal modulation of GIRK and CaV2 by GABA. Collectively, our data reveal mechanisms by which distinctly composed macromolecular complexes modulate the activity of key ion channels that mediate the inhibitory effects of GABA on hippocampal CA1 pyramidal neurons.SIGNIFICANCE STATEMENT This study identifies the contributions of distinct macromolecular complexes containing a major G-protein regulator to controlling key ion channel function in hippocampal neurons with implications for understanding molecular mechanisms underlying synaptic plasticity, learning, and memory.


Assuntos
Caveolina 2/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Hipocampo/fisiologia , Neurônios/fisiologia , Proteínas RGS/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Feminino , Insetos , Canais Iônicos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Inibição Neural/fisiologia
14.
J Physiol ; 596(10): 1833-1845, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29063617

RESUMO

Ivermectin (IVM) is an antiparasitic drug that is used worldwide and rescues hundreds of millions of people from onchocerciasis and lymphatic filariasis. It was discovered by Satoshi Omura and William C. Campbell, to whom the 2015 Nobel Prize in Physiology or Medicine was awarded. It kills parasites by activating glutamate-gated Cl- channels, and it also targets several ligand-gated ion channels and receptors, including Cys-loop receptors, P2X4 receptors and fernesoid X receptors. Recently, we found that IVM also activates a novel target, the G-protein-gated inwardly rectifying K+ channel, and also identified the structural determinant for the activation. In this review, we aim to provide an update and summary of recent progress in the identification of IVM targets, as well as their modulation mechanisms, through molecular structures, chimeras and site-directed mutagenesis, and molecular docking and modelling studies.


Assuntos
Antiparasitários/farmacologia , Canais de Cloreto/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Ativação do Canal Iônico , Ivermectina/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores Purinérgicos P2X4/fisiologia , Animais , Canais de Cloreto/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Humanos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Purinérgicos P2X4/efeitos dos fármacos
15.
Nat Commun ; 8(1): 1135, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29074844

RESUMO

Early-life stress, including maternal separation (MS), increases the vulnerability to develop mood disorders later in life, but the underlying mechanisms remain elusive. We report that MS promotes depressive-like symptoms in mice at a mature stage of life. Along with this behavioral phenotype, MS drives reduction of GABAB-GIRK signaling and the subsequent lateral habenula (LHb) hyperexcitability-an anatomical substrate devoted to aversive encoding. Attenuating LHb hyperactivity using chemogenetic tools and deep-brain stimulation ameliorates MS depressive-like symptoms. This provides insights on mechanisms and strategies to alleviate stress-dependent affective behaviors.


Assuntos
Depressão/fisiopatologia , Habenula/fisiopatologia , Privação Materna , Estresse Psicológico/fisiopatologia , Animais , Animais Recém-Nascidos , Estimulação Encefálica Profunda , Depressão/psicologia , Depressão/terapia , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Receptores de GABA-B/fisiologia , Transdução de Sinais/fisiologia , Estresse Psicológico/psicologia , Estresse Psicológico/terapia , Transmissão Sináptica/fisiologia
16.
J Neurosci ; 37(36): 8667-8677, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28821664

RESUMO

Activation of somatic µ-opioid receptors (MORs) in hypothalamic proopiomelanocortin (POMC) neurons leads to the activation of G-protein-coupled inward rectifier potassium (GIRK) channels and hyperpolarization, but in response to continued signaling MORs undergo acute desensitization resulting in robust reduction in the peak GIRK current after minutes of agonist exposure. We hypothesized that the attenuation of the GIRK current would lead to a recovery of neuronal excitability whereby desensitization of the receptor would lead to a new steady state of POMC neuron activity reflecting the sustained GIRK current observed after the initial decline from peak with continued agonist exposure. However, electrophysiologic recordings and GCaMP6f Ca2+ imaging in POMC neurons in mouse brain slices indicate that maximal inhibition of cellular activity by these measures can be maintained after the GIRK current declines. Blockade of the GIRK current by Ba2+ or Tertiapin-Q did not disrupt the sustained inhibition of Ca2+ transients in the continued presence of agonist, indicating the activation of an effector other than GIRK channels. Use of an irreversible MOR antagonist and Furchgott analysis revealed a low receptor reserve for the activation of GIRK channels but a >90% receptor reserve for the inhibition of Ca2+ events. Altogether, the data show that somatodendritic MORs in POMC neurons inhibit neuronal activity through at least two effectors with distinct levels of receptor reserve and that differentially reflect receptor desensitization. Thus, in POMC cells, the decline in the GIRK current during prolonged MOR agonist exposure does not reflect an increase in cellular activity as expected.SIGNIFICANCE STATEMENT Desensitization of the µ-opioid receptor (MOR) is thought to underlie the development of cellular tolerance to opiate therapy. The present studies focused on MOR desensitization in hypothalamic proopiomelanocortin (POMC) neurons as these neurons produce the endogenous opioid ß-endorphin and are heavily regulated by opioids. Prolonged activation of somatic MORs in POMC neurons robustly inhibited action potential firing and Ca2+ activity despite desensitization of the MOR and reduced activation of a potassium current over the same time course. The data show that somatic MORs in POMC neurons couple to multiple effectors that have differential sensitivity to desensitization of the receptor. Thus, in these cells, the cellular consequence of MOR desensitization cannot be defined by the activity of a single effector system.


Assuntos
Potenciais de Ação/fisiologia , Hipotálamo/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Pró-Opiomelanocortina/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Animais , Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Sensibilização do Sistema Nervoso Central/fisiologia , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Hipotálamo/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos
17.
J Physiol ; 595(17): 5895-5912, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28715108

RESUMO

KEY POINTS: Ivermectin (IVM) is a widely used antiparasitic drug in humans and pets which activates glutamate-gated Cl- channel in parasites. It is known that IVM binds to the transmembrane domains (TMs) of several ligand-gated channels, such as Cys-loop receptors and P2X receptors. We found that the G-protein-gated inwardly rectifying K+ (GIRK) channel, especially GIRK2, is activated by IVM directly in a Gßγ -independent manner, but the activation is dependent on phosphatidylinositol-4,5-biphosphate (PIP2 ). We identified a critical amino acid residue of GIRK2 for activation by IVM, Ile82, located in the slide helix between the TM1 and the N-terminal cytoplasmic tail domain (CTD). The results demonstrate that the TM-CTD interface in GIRK channel, rather than the TMs, governs IVM-mediated activation and provide us with novel insights on the mode of action of IVM in ion channels. ABSTRACT: Ivermectin (IVM) is a widely used antiparasitic drug in humans and pets which activates glutamate-gated Cl- channel in parasites. It is also known that IVM binds to the transmembrane domains (TMs) of several ligand-gated channels, such as Cys-loop receptors and P2X receptors. In this study, we found that the G-protein-gated inwardly rectifying K+ (GIRK) channel is activated by IVM directly. Electrophysiological recordings in Xenopus oocytes revealed that IVM activates GIRK channel in a phosphatidylinositol-4,5-biphosphate (PIP2 )-dependent manner, and that the IVM-mediated GIRK activation is independent of Gßγ subunits. We found that IVM activates GIRK2 more efficiently than GIRK4. In cultured hippocampal neurons, we also observed that IVM activates native GIRK current. Chimeric and mutagenesis analyses identified an amino acid residue unique to GIRK2 among the GIRK family, Ile82, located in the slide helix between the TM1 and the N-terminal cytoplasmic tail domain (CTD), which is critical for the activation. The results demonstrate that the TM-CTD interface in GIRK channels, rather than the TMs, governs IVM-mediated activation. These findings provide us with novel insights on the mode of action of IVM in ion channels that could lead to identification of new pharmacophores which activate the GIRK channel.


Assuntos
Antiparasitários/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Ivermectina/farmacologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Ratos Wistar , Xenopus laevis
18.
J Neurosci ; 37(14): 3940-3955, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28280255

RESUMO

Studies comparing neuronal activity at the dorsal and ventral poles of the hippocampus have shown that the scale of spatial information increases and the precision with which space is represented declines from the dorsal to ventral end. These dorsoventral differences in neuronal output and spatial representation could arise due to differences in computations performed by dorsal and ventral CA1 neurons. In this study, we tested this hypothesis by quantifying the differences in dendritic integration and synaptic plasticity between dorsal and ventral CA1 pyramidal neurons of rat hippocampus. Using a combination of somatic and dendritic patch-clamp recordings, we show that the threshold for LTP induction is higher in dorsal CA1 neurons and that a G-protein-coupled inward-rectifying potassium channel mediated regulation of dendritic plateau potentials and dendritic excitability underlies this gating. By contrast, similar regulation of LTP is absent in ventral CA1 neurons. Additionally, we show that generation of plateau potentials and LTP induction in dorsal CA1 neurons depends on the coincident activation of Schaffer collateral and temporoammonic inputs at the distal apical dendrites. The ventral CA1 dendrites, however, can generate plateau potentials in response to temporally dispersed excitatory inputs. Overall, our results highlight the dorsoventral differences in dendritic computation that could account for the dorsoventral differences in spatial representation.SIGNIFICANCE STATEMENT The dorsal and ventral parts of the hippocampus encode spatial information at very different scales. Whereas the place-specific firing fields are small and precise at the dorsal end of the hippocampus, neurons at the ventral end have comparatively larger place fields. Here, we show that the dorsal CA1 neurons have a higher threshold for LTP induction and require coincident timing of excitatory synaptic inputs for the generation of dendritic plateau potentials. By contrast, ventral CA1 neurons can integrate temporally dispersed inputs and have a lower threshold for LTP. Together, these dorsoventral differences in the threshold for LTP induction could account for the differences in scale of spatial representation at the dorsal and ventral ends of the hippocampus.


Assuntos
Região CA1 Hipocampal/fisiologia , Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Compostos de Bário/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Cloretos/farmacologia , Dendritos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
19.
Trends Pharmacol Sci ; 38(4): 378-392, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28188005

RESUMO

G protein-gated inwardly rectifying potassium (GIRK) channels are regulators of neuronal excitability in the brain. Knockout mice lacking GIRK channels display altered behavioral responses to multiple addictive drugs, implicating GIRK channels in addictive behaviors. Here, we review the effects of GIRK subunit deletions on the behavioral response to psychostimulants, such as cocaine and methamphetamine. Additionally, exposure of mice to psychostimulants produces alterations in the surface expression of GIRK channels in multiple types of neurons within the reward system of the brain. Thus, we compare the subcellular mechanisms by which drug exposure appears to alter GIRK expression in multiple cell types and provide an outlook on future studies examining the role of GIRK channels in addiction. A greater understanding of how GIRK channels are regulated by addictive drugs may enable the development of therapies to prevent or treat drug abuse.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Humanos , Potenciação de Longa Duração , Receptores de Dopamina D2/fisiologia , Receptores de GABA-B/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Recompensa , Área Tegmentar Ventral/fisiologia
20.
Physiol Rep ; 5(3)2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28196855

RESUMO

The serotonergic tone of the dorsal raphe (DR) is regulated by 5-HT1A receptors, which negatively control serotonergic activity via the activation of G protein-coupled inwardly rectifying K+ (GIRK) channels. In addition, DR activity is modulated by local GABAergic transmission, which is believed to play a key role in the development of mood-related disorders. Here, we sought to characterize the role of GIRK2 subunit-containing channels on the basal electrophysiological properties of DR neurons and to investigate whether the presynaptic and postsynaptic activities of 5-HT1A, GABAB, and GABAA receptors are affected by Girk2 gene deletion. Whole-cell patch-clamp recordings in brain slices from GIRK2 knockout mice revealed that the GIRK2 subunit contributes to maintenance of the resting membrane potential and to the membrane input resistance of DR neurons. 5-HT1A and GABAB receptor-mediated postsynaptic currents were almost absent in the mutant mice. Spontaneous and evoked GABAA receptor-mediated transmissions were markedly reduced in GIRK2 KO mice, as the frequency and amplitude of spontaneous IPSCs were reduced, the paired-pulse ratio was increased and GABA-induced whole-cell currents were decreased. Similarly, the pharmacological blockade of GIRK channels with tertiapin-Q prevented the 5-HT1A and GABAB receptor-mediated postsynaptic currents and increased the paired-pulse ratio. Finally, deletion of the Girk2 gene also limited the presynaptic inhibition of GABA release exerted by 5-HT1A and GABAB receptors. These results indicate that the properties and inhibitory activity of DR neurons are highly regulated by GIRK2 subunit-containing channels, introducing GIRK channels as potential candidates for studying the pathophysiology and treatment of affective disorders.


Assuntos
Núcleo Dorsal da Rafe/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Potenciais Pós-Sinápticos Inibidores , Neurônios/fisiologia , Sinapses/fisiologia , Potenciais de Ação , Animais , Camundongos , Camundongos Knockout , Subunidades Proteicas , Receptor 5-HT1A de Serotonina/fisiologia , Receptores de GABA-A/fisiologia , Receptores de GABA-B/fisiologia , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA