Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-37090510

RESUMO

Use of prescription opioids continues to rise, especially in adolescent individuals. As adolescence is a critical development window for higher order cognitive functions, thus opioid exposure during this period may have significant long-lasting effects on cognitive function and predisposition individuals to be at greater risk of developing opioid use later in life. Here, we examine previously explored effects of opioid exposure during adolescence on affect-related behavior, motivation, and cognitive flexibility. We find that a two-week exposure to non-contingent morphine during adolescence (i.e., post-weaning) does not alter performance in an elevated plus maze, forced swim test, or motivation for appetitive reward in male or female mice when tested during adolescence or adulthood. Examination of how adolescent morphine impacts cognition revealed impairments in visual-based discriminative learning and cognitive flexibility in female but not male mice, as assessed using an operant-based attentional set-shifting task. Unexpectedly, deficits in discriminative learning are observed when testing occurred during adolescence but not adulthood, whereas impaired performance in the extradimensional shift remained impaired into adulthood. The data indicate that opioid exposure during adolescence has a greater impact on cognitive function in female mice and that these deficits may be more widespread during acute withdrawal periods, while deficits in flexibility more enduring.

2.
J Neurosci ; 42(6): 940-953, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34876469

RESUMO

Ghrelin receptor, also known as growth hormone secretagogue receptor (GHS-R1a), is coexpressed with its truncated isoform GHS-R1b, which does not bind ghrelin or signal, but oligomerizes with GHS-R1a, exerting a complex modulatory role that depends on its relative expression. D1 dopamine receptor (D1R) and D5R constitute the two D1-like receptor subtypes. Previous studies showed that GHS-R1b also facilitates oligomerization of GHS-R1a with D1R, conferring GHS-R1a distinctive pharmacological properties. Those include a switch in the preferred coupling of GHS-R1a from Gq to Gs and the ability of D1R/D5R agonists and antagonists to counteract GHS-R1a signaling. Activation of ghrelin receptors localized in the ventral tegmental area (VTA) seems to play a significant role in the contribution of ghrelin to motivated behavior. In view of the evidence indicating that dopaminergic cells of the VTA express ghrelin receptors and D5R, but not D1R, we investigated the possible existence of functional GHS-R1a:GHS-R1b:D5R oligomeric complexes in the VTA. GHS-R1a:GHS-R1b:D5R oligomers were first demonstrated in mammalian transfected cells, and their pharmacological properties were found to be different from those of GHS-R1a:GHS-R1b:D1R oligomers, including weak Gs coupling and the ability of D1R/D5R antagonists, but not agonists, to counteract the effects of ghrelin. However, analyzing the effect of ghrelin in the rodent VTA on MAPK activation with ex vivo experiments, on somatodendritic dopamine release with in vivo microdialysis and on the activation of dopaminergic cells with patch-clamp electrophysiology, provided evidence for a predominant role of GHS-R1a:GHS-R1b:D1R oligomers in the rodent VTA as main mediators of the dopaminergic effects of ghrelin.SIGNIFICANCE STATEMENT The activation of ghrelin receptors localized in the ventral tegmental area (VTA) plays a significant role in the contribution of ghrelin to motivated behavior. We present evidence that indicates these receptors form part of oligomeric complexes that include the functional ghrelin receptor GHS-R1a, its truncated nonsignaling isoform GHS-R1b, and the dopamine D1 receptor (D1R). The binding of ghrelin to these complexes promotes activation of the dopaminergic neurons of the VTA by activation of adenylyl cyclase-protein kinase A signaling, which can be counteracted by both GHS-R1a and D1R antagonists. Our study provides evidence for a predominant role of GHS-R1a:GHS-R1b:D1R oligomers in rodent VTA as main mediators of the dopaminergic effects of ghrelin.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Grelina/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Grelina/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Wistar
3.
Physiol Behav ; 242: 113597, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34536435

RESUMO

Dysfunction of the infralimbic cortical (ILC) region of the medial prefrontal cortex (mPFC) is thought to be an underlying factor in both affect- and cognition-related behavioral deficits that co-occur across neuropsychiatric disorders. Increasing evidence highlights pathological imbalances in prefrontal pyramidal neuron excitability and associated aberrant firing as an underlying factor in this dysfunction. G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels mediate excitability of mPFC pyramidal neurons, however the functional role of these channels in ILC-dependent regulation of behavior and pyramidal neuron excitation is unknown. The present study used a viral-cre approach in male mice harboring a 'floxed' version of the kcnj3 (Girk1) gene, to disrupt GIRK1-containing channel expression in pyramidal neurons within the ILC. Loss of GIRK1-dependent signaling increased excitability and spike firing of pyramidal neurons but did not alter affective behavior measured in an elevated plus maze, forced swim test, or progressive ratio test of motivation. Alternatively, ablation of GIRK1 impaired performance in an operant-based attentional set-shifting task designed to assess cognitive flexibility. These data highlight a unique role for GIRK1 signaling in ILC pyramidal neurons in the regulation of strategy shifting but not affect and suggest that these channels may represent a therapeutic target for treatment of cognitive deficits in neuropsychiatric disease.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Células Piramidais , Animais , Cognição , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Masculino , Camundongos , Córtex Pré-Frontal/metabolismo , Transdução de Sinais
4.
Neuropsychopharmacology ; 46(12): 2158-2169, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34158613

RESUMO

Imbalance in prefrontal cortical (PFC) pyramidal neuron excitation:inhibition is thought to underlie symptomologies shared across stress-related disorders and neuropsychiatric disease, including dysregulation of emotion and cognitive function. G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels mediate excitability of medial PFC pyramidal neurons, however, the functional role of these channels in mPFC-dependent regulation of affect, cognition, and cortical dynamics is unknown. We used a viral-cre approach in male and female mice harboring a "floxed" version of the kcnj3 (Girk1) gene, to disrupt GIRK1-containing channel expression in pyramidal neurons within the prelimbic cortex (PrL). In males, loss of pyramidal GIRK1-dependent signaling differentially impacted measures of affect and impaired working memory and cognitive flexibility. Unexpectedly, ablation of PrL GIRK1-dependent signaling did not impact affect or cognition in female mice. Additional studies used a model of chronic unpredictable stress (CUS) to determine the impact on PrL GIRK-dependent signaling and cognitive function. CUS exposure in male mice produced deficits in cognition that paralleled a reduction in PrL pyramidal GIRK-dependent signaling akin to viral approaches whereas CUS exposure in female mice did not alter cognitive flexibility performance. Stress-induced behavioral deficits in male mice were rescued by systemic injection of a novel, GIRK1-selective agonist, ML297. In conclusion, GIRK1-dependent signaling in male mice, but not females, is critical for maintaining optimal PrL function and behavioral control. Disruption of this inhibition may underlie stress-related dysfunction of the PrL and represent a therapeutic target for treating stress-induced deficits in affect regulation and impaired cognition that reduce quality of life.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Qualidade de Vida , Animais , Cognição , Feminino , Proteínas de Ligação ao GTP , Masculino , Camundongos , Células Piramidais
5.
Neuropsychopharmacology ; 46(10): 1734-1745, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34012018

RESUMO

Opioid-based drugs are frequently used for pain management in both males and females despite the known risk of prefrontal cortex dysfunction and cognitive impairments. Although poorly understood, loss of cognitive control following chronic drug use has been linked to decreased activation of frontal cortex regions. Here, we show that self-administration of the potent opioid, remifentanil, causes a long-lasting hypoactive basal state evidenced by a decrease in ex vivo excitability that is paralleled by an increase in firing capacity of layer 5/6 pyramidal neurons in the prelimbic, but not infralimbic region of the medial prefrontal cortex. This phenomenon was observed in females after as few as 5 days and up to 25-30 days of self-administration. In contrast, pyramidal neurons in males showed increased excitability following 10-16 days of self-administration, with hypoactive states arising only following 25-30 days of self-administration. The emergence of a hypoactive, but not hyperactive basal state following remifentanil self-administration aligned with deficits in cognitive flexibility as assessed using an operant-based attentional set-shifting task. In females, the hypoactive basal state is driven by a reduction in excitatory synaptic transmission mediated by AMPA-type glutamate receptors. Alternatively, hyper- and hypoactive states in males align selectively with decreased and increased GABAB signaling, respectively. Chemogenetic compensation for this hypoactive state prior to testing restored cognitive flexibility, basal hypoactive state, and remifentanil-induced plasticity. These data define cellular and synaptic mechanisms by which opioids impair prefrontal function and cognitive control; indicating that interventions aimed at targeting opioid-induced adaptations should be tailored based on biological sex.


Assuntos
Córtex Pré-Frontal , Células Piramidais , Animais , Cognição , Feminino , Masculino , Camundongos , Remifentanil , Transmissão Sináptica
6.
J Neurosci ; 41(24): 5303-5314, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33879537

RESUMO

Relapse susceptibility in women with substance use disorders (SUDs) has been linked to the estrogen, 17ß-estradiol (E2). Our previous findings in female rats suggest that the influence of E2 on cocaine seeking can be localized to the prelimbic prefrontal cortex (PrL-PFC). Here, we investigated the receptor mechanisms through which E2 regulates the reinstatement of extinguished cocaine seeking. Sexually mature female rats underwent intravenous cocaine self-administration (0.5 mg/inf; 14 × 2 h daily) and extinction, and then were ovariectomized before reinstatement testing. E2 (10 µg/kg, i.p.) alone did not reinstate cocaine seeking, but it potentiated reinstatement when combined with an otherwise subthreshold priming dose of cocaine. A similar effect was observed following intra-PrL-PFC microinfusions of E2 and by systemic or intra-PrL-PFC administration of the estrogen receptor (ER)ß agonist, DPN, but not agonists at ERα or the G-protein-coupled ER1 (GPER1). By contrast, E2-potentiated reinstatement was prevented by intra-PrL-PFC microinfusions of the ERß antagonist, MPP, or the GPER1 antagonist, G15, but not an ERα antagonist. Whole-cell recordings in PrL-PFC layer (L)5/6 pyramidal neurons revealed that E2 decreases the frequency, but not amplitude, of GABAA-dependent miniature IPSCs (mIPSC). As was the case with E2-potentiated reinstatement, E2 reductions in mIPSC frequency were prevented by ERß and GPER1, but not ERα, antagonists and mimicked by ERß, but not GPER1, agonists. Altogether, the findings suggest that E2 activates ERß and GPER1 in the PrL-PFC to attenuate the GABA-mediated constraint of key outputs that mediate cocaine seeking.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Comportamento de Procura de Droga/fisiologia , Estradiol/metabolismo , Córtex Pré-Frontal/metabolismo , Animais , Receptor beta de Estrogênio/metabolismo , Extinção Psicológica/fisiologia , Feminino , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
7.
Brain Struct Funct ; 224(7): 2311-2324, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31201496

RESUMO

Despite evidence that morphine-related pathologies reflect adaptations in NAc glutamate signaling, substantial gaps in basic information remain. The current study examines the impact of non-contingent acute, repeated, and withdrawal-inducing morphine dosing regimens on glutamate transmission in D1- or D2-MSNs in the nucleus accumbens shell (NAcSh) and core (NAcC) sub-regions in hopes of identifying excitatory plasticity that may contribute to unique facets of opioid addiction-related behavior. Following an acute morphine injection (10 mg/kg), average miniature excitatory postsynaptic current (mEPSC) amplitude mediated by AMPA-type glutamate receptors was increased at D1-MSNs in the both the NAcShl and NAcC, whereas only the frequency of events was elevated at D2-MSNs in the NAcSh. In contrast, spontaneous somatic withdrawal induced by escalating dose of repeated morphine twice per day (20, 40, 60, 80, 100 mg/kg) enhanced mEPSC frequency specifically at D2-MSNs in the NAcSh. Similar to previous findings, excitatory drive was elevated at NAcSh D1-MSNs after 10-14 days home cage abstinence. Following abstinence, an acute drug re-exposure produced a rapid and enduring endocytosis of GluA2-containing AMPARs at D1-MSNs in the shell, that when blocked by an intra-NAc shell infusion of the Tat-GluA23Y peptide, increased reinstatement of morphine place preference-a phenomenon distinctly different than effects previously found with cocaine. The present study is the first to directly identify unique circuit specific adaptations in NAc glutamate synaptic transmission associated with morphine-related acute reward and somatic withdrawal as well as post-abstinence short-term plasticity. Moreover, while differing classes of abused drugs (i.e., psychostimulants and opioids) produce seemingly similar bidirectional plasticity in the NAc following drug re-exposure, our findings indicate this plasticity has distinct behavioral consequences.


Assuntos
Morfina/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Animais , Cocaína/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Masculino , Camundongos Transgênicos , Neurônios/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de AMPA/metabolismo , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Recompensa
8.
J Neurosci ; 39(24): 4785-4796, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-30948476

RESUMO

Understanding the neurobiological processes that incite drug craving and drive relapse has the potential to help target efforts to treat addiction. The NAc serves as a critical substrate for reward and motivated behavior, in part due to alterations in excitatory synaptic strength within cortical-accumbens pathways. The present studies investigated a causal link between cocaine-induced reinstatement of conditioned place preference and rapid reductions of cocaine-dependent increases in NAc shell synaptic strength in male mice. Cocaine-conditioned place preference behavior and ex vivo whole-cell electrophysiology showed that cocaine-primed reinstatement and synaptic depotentiation were disrupted by inhibiting AMPAR internalization via intra-NAc shell infusion of a Tat-GluA23Y peptide. Furthermore, reinstatement was driven by an mGluR5-dependent reduction in AMPAR signaling. Intra-NAc shell infusion of the mGluR5 antagonist MTEP blocked cocaine-primed reinstatement and corresponding depotentiation, whereas infusion of the mGluR5 agonist CHPG itself promoted reinstatement and depotentiated synaptic strength in the NAc shell. Optogenetic examination of circuit-specific plasticity showed that inhibition of infralimbic cortical input to the NAc shell blocked cocaine-primed reinstatement, whereas low-frequency stimulation (10 Hz) of this pathway in the absence of cocaine triggered a reduction in synaptic strength akin to that observed with cocaine, and was sufficient to promote reinstatement in the absence of a cocaine challenge. These data support a model in which mGluR5-mediated reduction in GluA2-containing AMPARs at NAc shell synapses receiving input from the infralimbic cortex is a critical factor in triggering reinstatement of cocaine-primed conditioned approach behavior.SIGNIFICANCE STATEMENT These studies identified a sequence of neural events whereby reexposure to cocaine activates a signaling cascade that alters synaptic strength in the NAc shell and triggers a behavioral response driven by a drug-associated memory.


Assuntos
Cocaína/farmacologia , Condicionamento Operante/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de Ácido Caínico/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Animais , Cocaína/antagonistas & inibidores , Fenômenos Eletrofisiológicos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Optogenética , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Receptores de AMPA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
9.
Neuroscience ; 384: 340-349, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29885524

RESUMO

Experience-dependent synaptic plasticity is an important component of both learning and motivational disturbances found in addicted individuals. Here, we investigated the role of cocaine experience-dependent plasticity at excitatory synapses in the nucleus accumbens shell (NAcSh) in relapse-related behavior in mice with a history of volitional cocaine self-administration. Using an extinction/reinstatement paradigm of cocaine-seeking behavior, we demonstrate that cocaine-experienced mice with extinguished cocaine-seeking behavior show potentiation of synaptic strength at excitatory inputs onto NAcSh medium spiny neurons (MSNs). Conversely, we found that exposure to various distinct types of reinstating stimuli (cocaine, cocaine-associated cues, yohimbine "stress") after extinction can produce a relative depotentiation of NAcSh synapses that is strongly associated with the magnitude of cocaine-seeking behavior exhibited in response to these challenges. Furthermore, we show that these effects are due to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-specific mechanisms that differ depending on the nature and context of the reinstatement-inducing stimuli. Together, our findings identify common themes as well as differential mechanisms that are likely important for the ability of diverse environmental stimuli to drive relapse to addictive-like cocaine-seeking behavior.


Assuntos
Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento de Procura de Droga/fisiologia , Extinção Psicológica/fisiologia , Plasticidade Neuronal/fisiologia , Núcleo Accumbens/metabolismo , Receptores de AMPA/metabolismo , Animais , Comportamento de Procura de Droga/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Autoadministração , Ioimbina/farmacologia
10.
Neuropharmacology ; 131: 154-165, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29225042

RESUMO

Repeated exposure to drugs of abuse alters the structure and function of neural circuits mediating reward, generating maladaptive plasticity in circuits critical for motivated behavior. Within meso-corticolimbic dopamine circuitry, repeated exposure to cocaine induces progressive alterations in AMPAR-mediated glutamatergic synaptic transmission. During a 10-14 day period of abstinence from cocaine, AMPAR signaling is potentiated at synapses on nucleus accumbens (NAc) medium spiny neurons (MSNs), promoting a state of heightened synaptic excitability. Re-exposure to cocaine during abstinence, however, rapidly reverses and depotentiates enhanced AMPAR signaling. To understand how re-exposure to cocaine alters AMPAR synaptic transmission, we investigated the roles of dopamine and endocannabinoid (eCB) signaling in modifying synaptic strength in the NAc shell. Using patch-clamp recordings from NAc slices prepared after 10-14 days of abstinence from repeated cocaine, we found that AMPAR-mediated depotentiation is rapidly induced in the NAc shell within 20 min of cocaine re-exposure ex vivo, and persists for up to five days before synapses return to levels of potentiation observed during abstinence. In cocaine-treated animals, global dopamine receptor activation was both necessary and sufficient for the cocaine-evoked depotentiation of AMPAR synaptic function. Additionally, we identified that CB1 receptors are engaged by endogenous endocannabinoids (eCBs) during re-exposure to cocaine ex vivo. Overall, these results indicate the central role that dopamine and eCB signaling mechanisms play in modulating cocaine-induced AMPAR plasticity in the NAc shell.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Dopamina/metabolismo , Endocanabinoides/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Benzoxazinas/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Flupentixol/farmacologia , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Morfolinas/farmacologia , Naftalenos/farmacologia , Núcleo Accumbens/citologia , Núcleo Accumbens/metabolismo , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Pirazóis/farmacologia , Rimonabanto
11.
Proc Natl Acad Sci U S A ; 113(3): 757-62, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26739562

RESUMO

Drug-evoked plasticity at excitatory synapses on medium spiny neurons (MSNs) of the nucleus accumbens (NAc) drives behavioral adaptations in addiction. MSNs expressing dopamine D1 (D1R-MSN) vs. D2 receptors (D2R-MSN) can exert antagonistic effects in drug-related behaviors, and display distinct alterations in glutamate signaling following repeated exposure to psychostimulants; however, little is known of cell-type-specific plasticity induced by opiates. Here, we find that repeated morphine potentiates excitatory transmission and increases GluA2-lacking AMPA receptor expression in D1R-MSNs, while reducing signaling in D2-MSNs following 10-14 d of forced abstinence. In vivo reversal of this pathophysiology with optogenetic stimulation of infralimbic cortex-accumbens shell (ILC-NAc shell) inputs or treatment with the antibiotic, ceftriaxone, blocked reinstatement of morphine-evoked conditioned place preference. These findings confirm the presence of overlapping and distinct plasticity produced by classes of abused drugs within subpopulations of MSNs that may provide targetable molecular mechanisms for future pharmacotherapies.


Assuntos
Morfina/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Animais , Antibacterianos/farmacologia , Ceftriaxona/farmacologia , Genótipo , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Fenômenos Ópticos , Subunidades Proteicas/metabolismo , Receptores de AMPA/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Proc Natl Acad Sci U S A ; 111(29): 10755-60, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002517

RESUMO

ML297 was recently identified as a potent and selective small molecule agonist of G-protein-gated inwardly rectifying K(+) (GIRK/Kir3) channels. Here, we show ML297 selectively activates recombinant neuronal GIRK channels containing the GIRK1 subunit in a manner that requires phosphatidylinositol-4,5-bisphosphate (PIP2), but is otherwise distinct from receptor-induced, G-protein-dependent channel activation. Two amino acids unique to the pore helix (F137) and second membrane-spanning (D173) domain of GIRK1 were identified as necessary and sufficient for the selective activation of GIRK channels by ML297. Further investigation into the behavioral effects of ML297 revealed that in addition to its known antiseizure efficacy, ML297 decreases anxiety-related behavior without sedative or addictive liabilities. Importantly, the anxiolytic effect of ML297 was lost in mice lacking GIRK1. Thus, activation of GIRK1-containing channels by ML297 or derivatives may represent a new approach to the treatment of seizure and/or anxiety disorders.


Assuntos
Ansiolíticos/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Pirazóis/farmacologia , Sequência de Aminoácidos , Animais , Baclofeno/farmacologia , Comportamento Animal/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
13.
Cardiovasc Res ; 101(1): 175-84, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24148898

RESUMO

AIMS: The purpose of this study was to investigate the functional role of G-protein-coupled inward rectifier potassium (GIRK) channels in the cardiac ventricle. METHODS AND RESULTS: Immunofluorescence experiments demonstrated that GIRK4 was localized in outer sarcolemmas and t-tubules in GIRK1 knockout (KO) mice, whereas GIRK4 labelling was not detected in GIRK4 KO mice. GIRK4 was localized in intercalated discs in rat ventricle, whereas it was expressed in intercalated discs and outer sarcolemmas in rat atrium. GIRK4 was localized in t-tubules and intercalated discs in human ventricular endocardium and epicardium, but absent in mid-myocardium. Electrophysiological recordings in rat ventricular tissue ex vivo showed that the adenosine A1 receptor agonist N6-cyclopentyladenosine (CPA) and acetylcholine (ACh) shortened action potential duration (APD), and that the APD shortening was reversed by either the GIRK channel blocker tertiapin-Q, the adenosine A1 receptor antagonist DPCPX or by the muscarinic M2 receptor antagonist AF-DX 116. Tertiapin-Q prolonged APD in the absence of the exogenous receptor activation. Furthermore, CPA and ACh decreased the effective refractory period and the effect was reversed by either tertiapin-Q, DPCPX or AF-DX 116. Receptor activation also hyperpolarized the resting membrane potential, an effect that was reversed by tertiapin-Q. In contrast, tertiapin-Q depolarized the resting membrane potential in the absence of the exogenous receptor activation. CONCLUSION: Confocal microscopy shows that among species GIRK4 is differentially localized in the cardiac ventricle, and that it is heterogeneously expressed across human ventricular wall. Electrophysiological recordings reveal that GIRK current may contribute significantly to ventricular repolarization and thereby to cardiac electrical stability.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ventrículos do Coração/metabolismo , Animais , Humanos , Masculino , Potenciais da Membrana , Camundongos , Camundongos Knockout , Potássio/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
14.
Rev Neurosci ; 23(4): 325-51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22944653

RESUMO

The addictive properties of psychostimulants such as cocaine are rooted in their ability to activate the mesocorticolimbic dopamine (DA) system. This system consists primarily of dopaminergic projections arising from the ventral tegmental area (VTA) and projecting to the limbic and cortical brain regions, such as the nucleus accumbens (NAc) and prefrontal cortex (PFC). While the basic anatomy and functional relevance of the mesocorticolimbic DA system is relatively well-established, a key challenge remaining in addiction research is to understand where and how molecular adaptations and corresponding changes in function of this system facilitate a pathological desire to seek and take drugs. Several lines of evidence indicate that inhibitory signaling, particularly signaling mediated by the Gi/o class of heterotrimeric GTP-binding proteins (G proteins), plays a key role in the acute and persistent effects of drugs of abuse. Moreover, recent evidence argues that these signaling pathways are targets of drug-induced adaptations. In this review we discuss inhibitory signaling pathways involving DA and the inhibitory neurotransmitter GABA in two brain regions - the VTA and PFC - that are central to the effects of acute and repeated cocaine exposure and represent sites of adaptations linked to addiction-related behaviors including sensitization, craving, and relapse.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Sistema Límbico/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Córtex Cerebral/metabolismo , Cocaína/metabolismo , Humanos , Sistema Límbico/metabolismo , Sistema Límbico/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia
15.
Int J Neuropsychopharmacol ; 14(6): 784-95, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20942997

RESUMO

The caudate putamen (CPu) has been implicated in habit learning and neuroadaptive changes that mediate the compulsive nature of drug-seeking following chronic cocaine self-administration. Re-exposure to an operant chamber previously associated with cocaine, but not yoked-saline, increases activity-regulated cytoskeleton-associated (Arc) gene mRNA expression within the dorsolateral (dl) CPu following prolonged abstinence. In this study, we tested the hypothesis that antisense gene knockdown of Arc within the dlCPu would alter cocaine-seeking. Initial studies showed that a single infusion of Arc antisense oligodeoxynucleotide (ODN) into the dlCPu significantly attenuated the induction of Arc mRNA and Arc protein by a single cocaine exposure (20 mg/kg i.p.) compared to scrambled-ODN-infused controls. In cocaine self-administering rats, infusion of Arc antisense ODN into the dlCPu 3 h prior to a test of context-driven drug-seeking significantly attenuated Arc protein induction, but failed to alter responding during testing, suggesting striatal Arc does not facilitate context-induced drug-seeking following prolonged abstinence. However, Arc antisense ODN infusion blunted the decrease in responding during subsequent 1-h extinction tests 24 and 48 h later. Following re-exposure to a cocaine-paired context, surface expression of the AMPA-type glutamate receptor GluR1 was significantly reduced whereas GluR2 was significantly increased in the dlCPu, independent of Arc antisense ODN infusion. Together, these findings indicate an important role for Arc in neuroadaptations within brain regions responsible for drug-seeking after abstinence and direct attention to changes occurring within striatal circuitry that are necessary to break down the habitual behaviour that leads to relapse.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Corpo Estriado/metabolismo , Proteínas do Citoesqueleto/antagonistas & inibidores , Comportamento de Procura de Droga , Técnicas de Silenciamento de Genes , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/metabolismo , Animais , Comportamento Animal , Membrana Celular/metabolismo , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Transtornos Relacionados ao Uso de Cocaína/prevenção & controle , Corpo Estriado/patologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Extinção Psicológica , Masculino , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Putamen/metabolismo , Putamen/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Prevenção Secundária
16.
J Neurosci ; 28(35): 8801-9, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18753382

RESUMO

Delayed-rectifier Kv2.1 potassium channels regulate somatodendritic excitability during periods of repetitive, high-frequency activity. Recent evidence suggests that Kv2.1 channel modulation is linked to glutamatergic neurotransmission. Because NMDA-type glutamate receptors are critical regulators of synaptic plasticity, we investigated NMDA receptor modulation of Kv2.1 channels in rodent hippocampus and cortex. Bath application of NMDA potently unclustered and dephosphorylated Kv2.1 and produced a hyperpolarizing shift in voltage-dependent activation of voltage-sensitive potassium currents (I(K)). In contrast, driving synaptic activity in Mg2+-free media to hyperactivate synaptic NMDA receptors had no effect on Kv2.1 channels, and moderate pentylenetetrazole-induced seizure activity in adult mice did not dephosphorylate hippocampal Kv2.1 channels. Selective activation of extrasynaptic NMDA receptors unclustered and dephosphorylated Kv2.1 channels and produced a hyperpolarizing shift in neuronal I(K). In addition, inhibition of glutamate uptake rapidly activated NMDA receptors and dephosphorylated Kv2.1 channels. These observations demonstrate that regulation of intrinsic neuronal activity by Kv2.1 is coupled to extrasynaptic but not synaptic NMDA receptors. These data support a novel mechanism for glutamate transporters in regulation of neuronal excitability and plasticity through extrasynaptic NMDA receptor modulation of Kv2.1 channels.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Potássio Shab/fisiologia , Sinapses/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Proteína 4 Homóloga a Disks-Large , Maleato de Dizocilpina/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Potenciais da Membrana/efeitos da radiação , Proteínas de Membrana/metabolismo , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurônios/efeitos da radiação , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp/métodos , Pentilenotetrazol , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
17.
Neuroreport ; 18(12): 1261-5, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17632279

RESUMO

Neuroadaptations affecting dopamine transmission within the prefrontal cortex and striatum are thought to underlie relapse to cocaine seeking after extended periods of abstinence. Regulator of G-protein signaling 4 (RGS4) is a forebrain-enriched protein known to be dynamically regulated by dopamine receptors in response to acute psychostimulant administration. In this report, chronic noncontingent (cocaine binge) or response-contingent (self-administration) delivery of cocaine followed by 2-3 weeks of abstinence resulted in a decrease of RGS4 mRNA in the dorsal striatum and prefrontal cortex. Furthermore, re-exposure to the cocaine-associated context after abstinence renewed the drug seeking and restored the levels of RGS4 mRNA to control values. Changes in RGS4 mRNA levels might signal abnormal receptor G-protein coupling that impacts cocaine seeking.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cocaína/farmacologia , Neostriado/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Proteínas RGS/genética , Animais , Comportamento Aditivo/genética , Comportamento Aditivo/metabolismo , Comportamento Aditivo/fisiopatologia , Transtornos Relacionados ao Uso de Cocaína/genética , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Modelos Animais de Doenças , Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Esquema de Medicação , Masculino , Neostriado/metabolismo , Neostriado/fisiopatologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Dopaminérgicos/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Recidiva , Autoadministração , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...