Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 10(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553240

RESUMO

Expanding knowledge about the cellular composition of subcortical brain regions demonstrates large heterogeneity and differences from the cortical architecture. Previously we described three subtypes of somatostatin-expressing (Sst) neurons in the mouse ventral tegmental area (VTA) and showed their local inhibitory action on the neighboring dopaminergic neurons (Nagaeva et al., 2020). Here, we report that Sst+ neurons especially from the anterolateral part of the mouse VTA also project far outside the VTA and innervate forebrain regions that are mainly involved in the regulation of emotional behavior, including the ventral pallidum, lateral hypothalamus, the medial part of the central amygdala, anterolateral division of the bed nucleus of stria terminalis, and paraventricular thalamic nucleus. Deletion of these VTASst neurons in mice affected several behaviors, such as home cage activity, sensitization of locomotor activity to morphine, fear conditioning responses, and reactions to the inescapable stress of forced swimming, often in a sex-dependent manner. Together, these data demonstrate that VTASst neurons have selective projection targets distinct from the main targets of VTA dopamine neurons. VTASst neurons are involved in the regulation of behaviors primarily associated with the stress response, making them a relevant addition to the efferent VTA pathways and stress-related neuronal network.


Assuntos
Neurônios Dopaminérgicos , Área Tegmentar Ventral , Camundongos , Animais , Área Tegmentar Ventral/metabolismo , Vias Eferentes/metabolismo , Neurônios Dopaminérgicos/metabolismo , Região Hipotalâmica Lateral , Somatostatina/metabolismo
2.
Cells ; 11(24)2022 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-36552881

RESUMO

The PSEN1 ΔE9 mutation causes a familial form of Alzheimer's disease (AD) by shifting the processing of amyloid precursor protein (APP) towards the generation of highly amyloidogenic Aß42 peptide. We have previously shown that the PSEN1 ΔE9 mutation in human-induced pluripotent stem cell (iPSC)-derived astrocytes increases Aß42 production and impairs cellular responses. Here, we injected PSEN1 ΔE9 mutant astrosphere-derived glial progenitors into newborn mice and investigated mouse behavior at the ages of 8, 12, and 16 months. While we did not find significant behavioral changes in younger mice, spatial learning and memory were paradoxically improved in 16-month-old PSEN1 ΔE9 glia-transplanted male mice as compared to age-matched isogenic control-transplanted animals. Memory improvement was associated with lower levels of soluble, but not insoluble, human Aß42 in the mouse brain. We also found a decreased engraftment of PSEN1 ΔE9 mutant cells in the cingulate cortex and significant transcriptional changes in both human and mouse genes in the hippocampus, including the extracellular matrix-related genes. Overall, the presence of PSEN1 ΔE9 mutant glia exerted a more beneficial effect on aged mouse brain than the isogenic control human cells likely as a combination of several factors.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Humanos , Masculino , Camundongos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Astrócitos/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Aprendizagem Espacial , Envelhecimento
3.
J Psychopharmacol ; 36(7): 860-874, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35695174

RESUMO

BACKGROUND: Psychedelics, like lysergic acid diethylamide (LSD), are again being studied as potential therapies for many neuropsychiatric disorders, including addictions. At the same time, the acute effects of psychedelics on rewarding behaviours have been scarcely studied. AIMS: The current study aimed to clarify if LSD decreases binge-like ethanol drinking in mice, and whether the observed acute effects on ethanol consumption are generalizable to a natural reinforcer, sucrose, and if the effects resulted from aversive or reward-attenuating effects caused by LSD. METHODS: The effects of acute LSD were examined using 2-bottle choice intermittent ethanol (20%) and sucrose drinking (10%), discrete-trial current-intensity threshold method of intracranial self-stimulation and short-term feeding behaviour assay in C57BL/6 male mice. RESULTS: The results showed that acute 0.1 mg/kg, but not 0.05 mg/kg, dose (i.p.) of LSD reduced 2-h intermittent ethanol drinking transiently without any prolonged effects. No effects were seen in intermittent 2-h sucrose drinking. The tested LSD doses had neither effect on the intracranial self-stimulation current-intensity thresholds, nor did LSD affect the threshold-lowering, or rewarding, effects of simultaneous amphetamine treatment. Furthermore, LSD had small, acute diminishing effects on 2-h food and water intake. CONCLUSIONS: Based on these results, LSD decreases binge-like ethanol drinking in mice, but only acutely. This effect is not likely to stem from reward-attenuating effects but could be in part due to reduced consummatory behaviour.


Assuntos
Alucinógenos , Dietilamida do Ácido Lisérgico , Animais , Etanol/farmacologia , Alucinógenos/farmacologia , Dietilamida do Ácido Lisérgico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Autoestimulação , Sacarose/farmacologia
4.
Elife ; 92020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32749220

RESUMO

The cellular architecture of the ventral tegmental area (VTA), the main hub of the brain reward system, remains only partially characterized. To extend the characterization to inhibitory neurons, we have identified three distinct subtypes of somatostatin (Sst)-expressing neurons in the mouse VTA. These neurons differ in their electrophysiological and morphological properties, anatomical localization, as well as mRNA expression profiles. Importantly, similar to cortical Sst-containing interneurons, most VTA Sst neurons express GABAergic inhibitory markers, but some of them also express glutamatergic excitatory markers and a subpopulation even express dopaminergic markers. Furthermore, only some of the proposed marker genes for cortical Sst neurons were expressed in the VTA Sst neurons. Physiologically, one of the VTA Sst neuron subtypes locally inhibited neighboring dopamine neurons. Overall, our results demonstrate the remarkable complexity and heterogeneity of VTA Sst neurons and suggest that these cells are multifunctional players in the midbrain reward circuitry.


Assuntos
Neurônios/metabolismo , Somatostatina/biossíntese , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Animais , Fenômenos Eletrofisiológicos , Feminino , Perfilação da Expressão Gênica , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/classificação , Neurônios/citologia , Neurotransmissores/metabolismo
5.
Front Pharmacol ; 11: 594, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32435195

RESUMO

Histamine/gamma-aminobutyric acid (GABA) neurons of posterior hypothalamus send wide projections to many brain areas and participate in stabilizing the wake state. Recent research has suggested that GABA released from the histamine/GABA neurons acts on extrasynaptic GABAA receptors and balances the excitatory effect of histamine. In the current study, we show the presence of vesicular GABA transporter mRNA in a majority of quantified hypothalamic histaminergic neurons, which suggest vesicular release of GABA. As histamine/GABA neurons form conventional synapses infrequently, it is possible that GABA released from these neurons diffuses to target areas by volume transmission and acts on extrasynaptic GABA receptors. To investigate this hypothesis, mice lacking extrasynaptic GABAA receptor δ subunit (Gabrd KO) were used. A pharmacological approach was employed to activate histamine/GABA neurons and induce histamine and presumably, GABA, release. Control and Gabrd KO mice were treated with histamine receptor 3 (Hrh3) inverse agonists ciproxifan and pitolisant, which block Hrh3 autoreceptors on histamine/GABA neurons and histamine-dependently promote wakefulness. Low doses of ciproxifan (1 mg/kg) and pitolisant (5 mg/kg) reduced locomotion in Gabrd KO, but not in WT mice. EEG recording showed that Gabrd KO mice were also more sensitive to the wake-promoting effect of ciproxifan (3 mg/kg) than control mice. Low frequency delta waves, associated with NREM sleep, were significantly suppressed in Gabrd KO mice compared with the WT group. Ciproxifan-induced wakefulness was blocked by histamine synthesis inhibitor α-fluoromethylhistidine (αFMH). The findings indicate that both histamine and GABA, released from histamine/GABA neurons, are involved in regulation of brain arousal states and δ-containing subunit GABAA receptors are involved in mediating GABA response.

6.
Int J Obes (Lond) ; 44(8): 1691-1702, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32317752

RESUMO

OBJECTIVE: Human TNKS, encoding tankyrase 1 (TNKS1), localizes to a susceptibility locus for obesity and type 2 diabetes mellitus (T2DM). Here, we addressed the therapeutic potential of G007-LK, a TNKS-specific inhibitor, for obesity and T2DM. METHODS: We administered G007-LK to diabetic db/db mice and measured the impact on body weight, abdominal adiposity, and serum metabolites. Muscle, liver, and white adipose tissues were analyzed by quantitative RT-PCR and western blotting to determine TNKS inhibition, lipolysis, beiging, adiponectin level, mitochondrial oxidative metabolism and mass, and gluconeogenesis. Protein interaction and PARylation analyses were carried out by immunoprecipitation, pull-down and in situ proximity ligation assays. RESULTS: TNKS inhibition reduced body weight gain, abdominal fat content, serum cholesterol levels, steatosis, and proteins associated with lipolysis in diabetic db/db mice. We discovered that TNKS associates with PGC-1α and that TNKS inhibition attenuates PARylation of PGC-1α, contributing to increased PGC-1α level in WAT and muscle in db/db mice. PGC-1α upregulation apparently modulated transcriptional reprogramming to increase mitochondrial mass and fatty acid oxidative metabolism in muscle, beiging of WAT, and raised circulating adiponectin level in db/db mice. This was in sharp contrast to the liver, where TNKS inhibition in db/db mice had no effect on PGC-1α expression, lipid metabolism, or gluconeogenesis. CONCLUSION: Our study unravels a novel molecular mechanism whereby pharmacological inhibition of TNKS in obesity and diabetes enhances oxidative metabolism and ameliorates lipid disorder. This happens via tissue-specific PGC-1α-driven transcriptional reprogramming in muscle and WAT, without affecting liver. This highlights inhibition of TNKS as a potential pharmacotherapy for obesity and T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/tratamento farmacológico , Obesidade/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Tanquirases/antagonistas & inibidores , Gordura Abdominal , Tecido Adiposo Branco , Animais , Peso Corporal , Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Oxirredução , Poli ADP Ribosilação , Sulfonas/uso terapêutico , Tanquirases/metabolismo , Triazóis/uso terapêutico
7.
Pharmacol Biochem Behav ; 190: 172852, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31952939

RESUMO

Acute lipopolysaccharide (LPS) administration induces innate inflammatory signalling and produces sickness reaction characterized by reduced drinking, eating and reduced locomotor exploration, as well as emotional changes indicating increased helplessness/despair. LPS administration has been used to model behavioral and emotional responses to inflammatory reactions. Our aim was to find out whether the lack of metabotropic glutamate receptor 3 (mGluR3) in the knockout (KO) mice affects behavioral effects of LPS in vivo, as mGluR3 may have a role in inflammatory signalling. After LPS (1 mg/kg, i.p.) administration, we compared wild-type (WT) and mGluR3-KO mice for differences in gross appearance and locomotion at 3- and 6-h time points, anxiety-like behavior in the light-dark test at 24-h, depression-like behavior in the tail-suspension test at 25-h, and in the forced-swim test at 48-h time points. Body weight and water consumption were monitored. Based on behavioral scorings at the 3-h and 6-h time points, the mGluR3-KO mice reacted to LPS in a similar way as the WT mice. LPS-induced reductions in the body weight or water consumption did not differ between genotypes. Interestingly, LPS-induced reductions in the body temperature were significantly enhanced in male and female mGluR3-KO mice at 6-h and 3-h time points, respectively. In the light-dark anxiety-test the saline-treated mGluR3-KOs showed increased anxiolytic-like behaviors compared to the saline-treated WT mice. LPS treatment significantly reduced the KO entries to the light compartment to the same level as WT mice given saline. Total locomotion was significantly reduced in both genotypes by LPS. In the despair models, no genotype difference was observed after saline or LPS, neither had LPS treatment any significant effect on immobility. Although changes in glutamatergic neurotransmission may partly mediate effects of systemic LPS administration, mGluR3 appears not to be crucial in behavioral responses to acute activation of innate immune system.


Assuntos
Comportamento Animal/efeitos dos fármacos , Técnicas de Inativação de Genes , Lipopolissacarídeos/farmacologia , Receptores de Glutamato Metabotrópico/genética , Animais , Ansiedade/induzido quimicamente , Temperatura Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Depressão/induzido quimicamente , Ingestão de Líquidos/efeitos dos fármacos , Genótipo , Inflamação/induzido quimicamente , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Knockout
8.
J Psychopharmacol ; 33(7): 855-864, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31070489

RESUMO

BACKGROUND: Alcohol use associates with environmental cues that can later reinstate drinking patterns without any alcohol exposure. Alcohol-induced reward, when combined with contextual signals of various sensory modalities in the central synapses of mesolimbic reward circuitries, can lead to the formation of conditioned responses. AIMS: As the activation of glutamatergic synapses is pivotal in such processes, we aimed to investigate whether the metabotropic glutamate receptor subtype 3 plays a role in alcohol-induced behaviours including place preference. METHODS: The metabotropic glutamate receptor subtype 3 knockout (mGluR3-KO) mouse line was used to study alcohol-induced place preference, locomotor activating and ataxic effects, limited access alcohol drinking, and preference for sucrose and saccharin. RESULTS: Alcohol-induced horizontal locomotor stimulation and reduced rearing behaviour remained unchanged in the mGluR3-KO mice. However, alcohol-induced place conditioning in an unbiased paradigm setup was lacking in the mGluR3-KO mice, but clearly present in wildtype mice. Locomotor activity was not different between the mGluR3-KO and wildtype mice during the acquisition and expression trials. Alcohol consumption, studied through the 'drinking in the dark' model, remained unchanged in the mGluR3-KO mice, although low consumption in both wildtype and knockout mice hampers interpretation. The mGluR3-KO mice also showed normal sucrose and saccharin preference. CONCLUSIONS: These studies indicate a role for metabotropic glutamate receptor subtype 3 in the conditioned contextual alcohol responses, but not in stimulatory, and ataxic alcohol effects.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Condicionamento Psicológico/efeitos dos fármacos , Etanol/farmacologia , Receptores de Glutamato Metabotrópico/genética , Animais , Sinais (Psicologia) , Etanol/administração & dosagem , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recompensa
9.
Addict Biol ; 24(6): 1191-1203, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30421860

RESUMO

Drugs of abuse induce widespread synaptic adaptations in the mesolimbic dopamine (DA) neurons. Such drug-induced neuroadaptations may constitute an initial cellular mechanism eventually leading to compulsive drug-seeking behavior. To evaluate the impact of GABAB receptors on addiction-related persistent neuroplasticity, we tested the ability of orthosteric agonist baclofen and two positive allosteric modulators (PAMs) of GABAB receptors to suppress neuroadaptations in the ventral tegmental area (VTA) and reward-related behaviors induced by ethanol and cocaine. A novel compound (S)-1-(5-fluoro-2,3-dihydro-1H-inden-2-yl)-4-methyl-6,7,8,9-tetrahydro-[1,2,4]triazolo[4,3-a]quinazolin-5(4H)-one (ORM-27669) was found to be a GABAB PAM of low efficacy as agonist, whereas the reference compound (R,S)-5,7-di-tert-butyl-3-hydroxy-3-trifluoromethyl-3H-benzofuran-2-one (rac-BHFF) had a different allosteric profile being a more potent PAM in the calcium-based assay and an agonist, coupled with potent PAM activity, in the [35 S] GTPγS binding assay in rat and human recombinant receptors. Using autoradiography, the high-efficacy rac-BHFF and the low-efficacy ORM-27669 potentiated the effects of baclofen on [35 S] GTPγS binding with identical brain regional distribution. Treatment of mice with baclofen, rac-BHFF, or ORM-27669 failed to induce glutamate receptor neuroplasticity in the VTA DA neurons. Pretreatment with rac-BHFF at non-sedative doses effectively reversed both ethanol- and cocaine-induced plasticity and attenuated cocaine i.v. self-administration and ethanol drinking. Pretreatment with ORM-27669 only reversed ethanol-induced neuroplasticity and attenuated ethanol drinking but had no effects on cocaine-induced neuroplasticity or self-administration. These findings encourage further investigation of GABAB receptor PAMs with different efficacies in addiction models to develop novel treatment strategies for drug addiction.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Etanol/farmacologia , Moduladores GABAérgicos/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Receptores de GABA-B/efeitos dos fármacos , Regulação Alostérica , Animais , Baclofeno/farmacologia , Comportamento Animal/efeitos dos fármacos , Benzofuranos/farmacologia , Células CHO , Cricetulus , Agonistas dos Receptores de GABA-B/farmacologia , Humanos , Camundongos , Quinazolinonas/farmacologia , Ratos , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/metabolismo , Recompensa , Autoadministração , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos
10.
Basic Clin Pharmacol Toxicol ; 123(5): 558-566, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29781560

RESUMO

Extrasynaptic δ subunit-containing γ-aminobutyric acid type A receptors (δ-GABAA Rs) are emerging as targets for a number of neuropsychopharmacological drugs, including the direct GABA site agonist gaboxadol and neuroactive steroids. Among other regions, these δ-GABAA Rs are functionally expressed in the ventral tegmental area (VTA), the cell body region of mesocorticolimbic dopamine (DA) system important for motivated behaviours, and in the target region, the nucleus accumbens. Gaboxadol and neurosteroids induce VTA DA neuron plasticity ex vivo, by inhibiting the VTA GABA neurons, and aversive place conditioning, which are absent in the δ-GABAA R knockout mice (δ-KO). It is not known whether δ-GABAA Rs are important for the effects of other drugs, such as opioids (that also inhibit GABA neurons) and stimulants (that primarily elevate monoamine levels). Here, we used δ-KO mice and conditioned place preference (CPP) test to study the rewarding effects of morphine (20 mg/kg), methamphetamine (1 mg/kg) and mephedrone (5 mg/kg). Morphine-induced nociception was also assessed using tail-flick and hot-plate tests. We found that the δ-KO mice failed to express morphine-induced CPP, but that they were more sensitive to morphine-induced analgesia in the tail-flick test. In contrast, stimulant-induced CPP in the δ-KO mice was similar to that in the wild-type controls. Thus, the conditioned rewarding effect by opioids, but not that of stimulants, was impaired in the absence of δ-GABAA Rs. Further studies are warranted to assess the potential of δ-GABAA R antagonists as possible targets for reducing morphine reward and potentiating morphine analgesia.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Metanfetamina/análogos & derivados , Metanfetamina/farmacologia , Morfina/farmacologia , Motivação , Receptores de GABA-A , Analgésicos Opioides/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Camundongos , Camundongos Knockout , Motivação/efeitos dos fármacos , Motivação/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
11.
Alcohol ; 65: 1-10, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29084623

RESUMO

Neuroinflammation may play an important role in the development of alcohol addiction. Recent preclinical reports suggest that enhanced innate immune system signaling increases consumption of alcohol. Our aim was to study whether consequences of lipopolysaccharide (LPS)-induced sickness reaction increase long-term alcohol intake. Adult male C57BL/6J mice, housed in individually ventilated cages, were injected with LPS intraperitoneally (i.p.) and allowed to recover from an acute sickness reaction for 1 week before analysis of their alcohol intake in two different drinking models. Effects of LPS challenge were tested in a continuous two-bottle free choice test with increasing concentrations of alcohol and in a drinking in the dark (DID) binge model. In addition, the effect of repeatedly administered LPS during abstinence periods between binge drinking was analyzed in the DID model. In addition, the DID model was used to study the effects of the microglia inhibitor minocycline (50 mg/kg/day, 4 days) and purinergic P2X7 receptor antagonist Brilliant Blue G (75 mg/kg/day, 7 days) on alcohol intake. In contrast to previous findings, pretreatment with a 1-mg/kg dose of LPS did not significantly increase ethanol consumption in the continuous two-bottle choice test. As a novel finding, we report that increasing the LPS dose to 1.5 mg/kg reduced consumption of 18 and 21% (v/v) ethanol. In the DID model, pretreatment with LPS (0.2-1.5 mg/kg) did not significantly alter 15% or 20% ethanol consumption. Neither did repeated LPS injections affect binge alcohol drinking. Minocycline reduced alcohol, but also water, intake regardless of LPS pretreatment. No data on effects of P2X7 antagonists on alcohol consumption have been previously published; therefore, we report here that subchronic Brilliant Blue G had no effect on alcohol intake in the DID model. As a conclusion, further studies are needed to validate this LPS model of the interaction between immune system activation and alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Consumo Excessivo de Bebidas Alcoólicas/psicologia , Etanol/administração & dosagem , Lipopolissacarídeos/toxicidade , Recuperação de Função Fisiológica/fisiologia , Consumo de Bebidas Alcoólicas/tratamento farmacológico , Consumo de Bebidas Alcoólicas/fisiopatologia , Animais , Benzenossulfonatos/uso terapêutico , Consumo Excessivo de Bebidas Alcoólicas/tratamento farmacológico , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Minociclina/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos
12.
Addict Biol ; 22(4): 1022-1035, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26990998

RESUMO

Opioid antagonist treatments reduce alcohol drinking in rodent models and in alcohol-dependent patients, with variable efficacy across different studies. These treatments may suffer from the development of tolerance and opioid receptor supersensitivity, as suggested by preclinical models showing activation of these processes during and after subchronic high-dose administration of the short-acting opioid antagonist naloxone. In the present study, we compared equipotent low and moderate daily doses of naltrexone and nalmefene, two opioid antagonists in the clinical practice for treatment of alcoholism. The antagonists were given here subcutaneously for 7 days either as daily injections or continuous osmotic minipump-driven infusions to alcohol-preferring AA rats having trained to drink 10% alcohol in a limited access protocol. One day after stopping the antagonist treatment, [35 S]GTPγS autoradiography on brain cryostat sections was carried out to examine the coupling of receptors to G protein activation. The results prove the efficacy of repeated injections over infused opioid antagonists in reducing alcohol drinking. Tolerance to the reducing effect on alcohol drinking and to the enhancement of G protein coupling to µ-opioid receptors in various brain regions were consistently detected only after infused antagonists. Supersensitivity of κ-opioid receptors was seen in the ventral and dorsal striatal regions especially by infused nalmefene. Nalmefene showed no clear agonistic activity in rat brain sections or at human recombinant κ-opioid receptors. The findings support the as-needed dosing practice, rather than the standard continual dosing, in the treatment of alcoholism with opioid receptor antagonists.


Assuntos
Encéfalo/efeitos dos fármacos , Etanol/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides/efeitos dos fármacos , Alcoolismo/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Ratos
13.
Front Pharmacol ; 7: 403, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27833556

RESUMO

Endogenous neurosteroids and neuroactive steroids have potent and widespread actions on the brain via inhibitory GABAA receptors. In recombinant receptors and genetic mouse models their actions depend on the α, ß, and δ subunits of the receptor, especially on those that form extrasynaptic GABAA receptors responsible for non-synaptic (tonic) inhibition, but they also act on synaptically enriched γ2 subunit-containing receptors and even on αß binary receptors. Here we tested whether behavioral sensitivity to the neuroactive steroid agonist 5ß-pregnan-3α-ol-20-one is altered in genetically engineered mouse models that have deficient GABAA receptor-mediated synaptic inhibition in selected neuronal populations. Mouse lines with the GABAA receptor γ2 subunit gene selectively deleted either in parvalbumin-containing cells (including cerebellar Purkinje cells), cerebellar granule cells, or just in cerebellar Purkinje cells were trained on the accelerated rotating rod and then tested for motor impairment after cumulative intraperitoneal dosing of 5ß-pregnan-3α-ol-20-one. Motor-impairing effects of 5ß-pregnan-3α-ol-20-one were strongly increased in all three mouse models in which γ2 subunit-dependent synaptic GABAA responses in cerebellar neurons were genetically abolished. Furthermore, rescue of postsynaptic GABAA receptors in Purkinje cells normalized the effect of the steroid. Anxiolytic/explorative effects of the steroid in elevated plus maze and light:dark exploration tests in mice with Purkinje cell γ2 subunit inactivation were similar to those in control mice. The results suggest that, when the deletion of γ2 subunit has removed synaptic GABAA receptors from the specific cerebellar neuronal populations, the effects of neuroactive steroids solely on extrasynaptic αß or αßδ receptors lead to enhanced changes in the cerebellum-generated behavior.

14.
PLoS One ; 10(5): e0124931, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25970770

RESUMO

The adverse effects of alcohol consumption during pregnancy are known, but the molecular events that lead to the phenotypic characteristics are unclear. To unravel the molecular mechanisms, we have used a mouse model of gestational ethanol exposure, which is based on maternal ad libitum ingestion of 10% (v/v) ethanol for the first 8 days of gestation (GD 0.5-8.5). Early neurulation takes place by the end of this period, which is equivalent to the developmental stage early in the fourth week post-fertilization in human. During this exposure period, dynamic epigenetic reprogramming takes place and the embryo is vulnerable to the effects of environmental factors. Thus, we hypothesize that early ethanol exposure disrupts the epigenetic reprogramming of the embryo, which leads to alterations in gene regulation and life-long changes in brain structure and function. Genome-wide analysis of gene expression in the mouse hippocampus revealed altered expression of 23 genes and three miRNAs in ethanol-exposed, adolescent offspring at postnatal day (P) 28. We confirmed this result by using two other tissues, where three candidate genes are known to express actively. Interestingly, we found a similar trend of upregulated gene expression in bone marrow and main olfactory epithelium. In addition, we observed altered DNA methylation in the CpG islands upstream of the candidate genes in the hippocampus. Our MRI study revealed asymmetry of brain structures in ethanol-exposed adult offspring (P60): we detected ethanol-induced enlargement of the left hippocampus and decreased volume of the left olfactory bulb. Our study indicates that ethanol exposure in early gestation can cause changes in DNA methylation, gene expression, and brain structure of offspring. Furthermore, the results support our hypothesis of early epigenetic origin of alcohol-induced disorders: changes in gene regulation may have already taken place in embryonic stem cells and therefore can be seen in different tissue types later in life.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Metilação de DNA/efeitos dos fármacos , Epigênese Genética , Etanol/farmacologia , Hipocampo/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/patologia , Animais , Medula Óssea/efeitos dos fármacos , Ilhas de CpG , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Hipocampo/patologia , Histonas/genética , Histonas/metabolismo , Humanos , Cadeias Leves Substitutas da Imunoglobulina/genética , Cadeias Leves Substitutas da Imunoglobulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Neurulação/efeitos dos fármacos , Neurulação/genética , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/patologia , Receptores Odorantes/genética , Receptores Odorantes/metabolismo
15.
Behav Brain Res ; 266: 94-103, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24631392

RESUMO

Polymorphisms in the metabotropic glutamate receptor 3 (mGluR3) encoding gene GRM3 have been linked to schizophrenia and cognitive performance in humans. Our aim was to analyze the role of mGluR3 in basal working memory and attentional processes, and also when these functions were distracted by the psychotomimetic N-methyl-d-aspartate (NMDA) receptor antagonist dizocilpine (MK-801). mGluR3 knockout (KO) mice were used. Spontaneous alternation in a T-maze test was significantly reduced in mGluR3-KO mice compared to wildtype (WT) mice, particularly after a low dose of MK-801 (0.03 mg/kg, i.p., 30 min). In a Y-maze novelty discrimination test, the locomotor stimulatory effect of MK-801 (0.1mg/kg) was enhanced in mGluR3-KO mice. Interestingly, mGluR3-KO mice showed the significantly reduced alternation in the spontaneous alternation T-maze test and the significantly enhanced sensitivity to MK-801 in the Y-maze test only when forced to enter the right arm first, not when the forced arm was on the left. A side-biased response was also found in a rewarded alternation T-maze test, where mGluR3-KO mice made significantly more incorrect visits to the left arm than the right arm after a 25-s delay. No genotype difference was found in the novelty discrimination in the Y-maze test, rewarded alternation with a 5-s delay, preference for left or right when free to enter either arm or in MK-801-induced circling. Our findings indicate cognitive disturbance and left-right asymmetry in certain behavioral responses of mGluR3-KO mice. This novel observation warrants further elucidation, and should also be considered in other studies of mGluR3 in brain functions.


Assuntos
Maleato de Dizocilpina/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/genética , Memória de Curto Prazo/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/deficiência , Análise de Variância , Animais , Atenção/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Lateralidade Funcional , Aprendizagem em Labirinto/classificação , Aprendizagem em Labirinto/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Knockout
16.
Pharmacol Biochem Behav ; 103(3): 501-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23099177

RESUMO

INTRODUCTION/AIMS: The use of cathinone-derivative designer drugs methylone and mephedrone has increased rapidly in recent years. Our aim was to investigate the possible long-term effects of these drugs on a range of behavioral tests in mice. Further, we investigated the long-term effects of these drugs on brain neurochemistry in both rats and mice. METHODS: We treated animals with a binge-like regimen of methylone or mephedrone (30 mg/kg, twice daily for 4 days) and, starting 2 weeks later, we performed behavioral tests of memory, anxiety and depression and measured brain levels of dopamine (DA), serotonin (5-HT), their metabolites and norepinephrine (NE). 5-HT and DA transporter (5-HTT and DAT) levels were also measured in rats by [(3)H]paroxetine and [(3)H]mazindol binding. RESULTS: Mephedrone reduced working memory performance in the T-maze spontaneous alternation task but did not affect neurotransmitter levels aside from a 22% decrease in striatal homovanillic acid (HVA) levels in mice. Methylone had little effect on behavior or neurotransmitter levels in mice but produced a widespread depletion of 5-HT and 5-HTT levels in rats. CONCLUSIONS: Both methylone and mephedrone appeared to have a long-term effect on either behavioral or biochemical gauges of neurotoxicity in rodents.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Drogas Desenhadas/efeitos adversos , Transtornos da Memória/induzido quimicamente , Metanfetamina/análogos & derivados , Animais , Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Depressão/induzido quimicamente , Depressão/metabolismo , Drogas Desenhadas/farmacologia , Modelos Animais de Doenças , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Elevação dos Membros Posteriores , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/metabolismo , Metanfetamina/efeitos adversos , Metanfetamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Ensaio Radioligante , Ratos , Ratos Wistar , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
17.
PLoS One ; 6(9): e24159, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21912668

RESUMO

We investigated the behavioral significance of fast synaptic inhibition by αßγ2-type GABA(A) receptors on parvalbumin (Pv) cells. The GABA(A) receptor γ2 subunit gene was selectively inactivated in Pv-positive neurons by Cre/loxP recombination. The resulting Pv-Δγ2 mice were relatively healthy in the first postnatal weeks; but then as Cre started to be expressed, the mice progressively developed wide-ranging phenotypic alterations including low body weight, motor deficits and tremor, decreased anxiety levels, decreased pain sensitivity and deficient prepulse inhibition of the acoustic startle reflex and impaired spatial learning. Nevertheless, the deletion was not lethal, and mice did not show increased mortality even after one year. Autoradiography with t-butylbicyclophosphoro[(35)S]thionate suggested an increased amount of GABA(A) receptors with only α and ß subunits in central nervous system regions that contained high levels of parvalbumin neurons. Using BAC-transgenesis, we reduced some of the Pv-Δγ2 phenotype by selectively re-expressing the wild-type γ2 subunit back into some Pv cells (reticular thalamic neurons and cerebellar Pv-positive neurons). This produced less severe impairments of motor skills and spatial learning compared with Pv-Δγ2 mice, but all other deficits remained. Our results reveal the widespread significance of fast GABAergic inhibition onto Pv-positive neurons for diverse behavioral modalities, such as motor coordination, sensorimotor integration, emotional behavior and nociception.


Assuntos
Comportamento Animal , Inativação Gênica , Neurônios/metabolismo , Parvalbuminas/metabolismo , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética , Analgésicos/farmacologia , Animais , Ansiedade/genética , Ansiedade/metabolismo , Comportamento Animal/efeitos dos fármacos , Sítios de Ligação , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiologia , Carbolinas/farmacologia , Feminino , Aprendizagem/efeitos dos fármacos , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Fenótipo , Piridinas/farmacologia , Receptores de GABA-A/metabolismo , Comportamento Espacial/efeitos dos fármacos , Especificidade da Espécie , Temperatura , Zolpidem , Ácido gama-Aminobutírico/farmacologia
18.
Eur J Pharmacol ; 666(1-3): 111-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21616065

RESUMO

The potent sedative-hypnotic zolpidem and the convulsant methyl-6,7-dimethoxy-4-ethyl-ß-carboline-3-carboxylate (DMCM) act primarily by binding to the benzodiazepine site of the main inhibitory neurotransmitter receptor, the pentameric γ-aminobutyric acid type A receptor (GABA(A)). This binding depends critically on the wild-type F77 residue of the GABA(A) receptor γ2 subunit. Mice with γ2 subunit F77I point mutation (γ2I77 mouse line) lose the high-affinity nanomolar binding of these ligands as well as their most robust behavioral actions at low doses. Interestingly, the γ2I77 mice offer a tool to study the actions of these substances mediated via other possible binding sites of the GABA(A) receptor. In ligand autoradiographic experiments, we discovered in γ2I77 mouse brain sections a significant amount of residual non-γ2 subunit-dependent benzodiazepine site binding enriched to the striatum and septum. Zolpidem only weakly affected this residual binding at micromolar concentrations, and only a high zolpidem dose (≥ 40 mg/kg) caused sedation and deficits in motor coordination in γ2I77 mice. DMCM had an agonistic action through a secondary, low-affinity non-benzodiazepine binding site of the GABA(A) receptor in the forebrain of γ2I77 mice, and this drug also fully displaced the residual benzodiazepine-site labeling. In behavioral tests, a high dose (20mg/kg) of DMCM was sedative and modulated fear learning. DMCM, but not zolpidem, acted as an agonist in recombinant GABA(A) α1/6ß3 receptors studied using ligand binding and electrophysiological assays. Our results highlight the less well-known actions of high doses of DMCM and zolpidem that are not mediated via the γ2 subunit-containing benzodiazepine site of the GABA(A) receptor.


Assuntos
Benzodiazepinas/metabolismo , Carbolinas/metabolismo , Carbolinas/farmacologia , Piridinas/metabolismo , Piridinas/farmacologia , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Animais , Autorradiografia , Azidas/farmacologia , Comportamento Animal/efeitos dos fármacos , Benzodiazepinas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Convulsivantes/metabolismo , Convulsivantes/farmacologia , Feminino , Células HEK293 , Humanos , Hipnóticos e Sedativos/metabolismo , Hipnóticos e Sedativos/farmacologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Zolpidem
19.
Front Neurosci ; 5: 3, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21270945

RESUMO

Ethyl alcohol (ethanol) has many molecular targets in the nervous system, its potency at these sites being low compared to those of sedative drugs. This has made it difficult to discover ethanol's binding site(s). There are two putative binding sites at γ-aminobutyric acid (GABA) type A receptor subtypes for the proposed ethanol antagonist Ro 15-4513, the established γ2 subunit-dependent benzodiazepine site and the recently reported δ subunit-dependent Ro 15-4513/ethanol binding site. Here, we aimed at clarifying the in vivo role of Ro 15-4513 at these two sites. We found that the antagonism of ethanol actions by Ro 15-4513 in wildtype mice was dependent on the test: an open field test showed that light sedation induced by 1.5-1.8 g/kg ethanol was sensitive to Ro 15-4513, whereas several tests for ethanol-induced anxiolytic effects showed that the ethanol-induced effects were insensitive to Ro 15-4513. Antagonism of ethanol-induced sedation by Ro 15-4513 was unaffected in GABA(A) receptor δ subunit knockout mice. By contrast, when testing the GABA(A) receptor γ2 subunit F77I knock-in mouse line (γ2I77 mice) with its strongly reduced affinity of the benzodiazepine sites for Ro 15-4513, we found that the ethanol-induced sedation was no longer antagonized by Ro 15-4513. Indeed, γ2I77 mice had only a small proportion of high-affinity binding of [(3)H]Ro 15-4513 left as compared to wildtype mice, especially in the caudate-putamen and septal areas, but these residual sites are apparently not involved in ethanol antagonism. In conclusion, we found that Ro 15-4513 abolished the sedative effect of ethanol by an action on γ2 subunit-dependent benzodiazepine sites.

20.
Eur J Neurosci ; 33(1): 161-74, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21073553

RESUMO

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor GluA1 subunit-deficient (GluA1-/-) mice display novelty-induced hyperactivity, cognitive and social defects and may model psychiatric disorders, such as schizophrenia and depression/mania. We used c-Fos expression in GluA1-/- mice to identify brain regions responsible for novelty-induced hyperlocomotion. Exposure to a novel cage for 2 h significantly increased c-Fos expression in many brain regions in both wild-type and knockout mice. Interestingly, the clearest genotype effect was observed in the hippocampus and its main input region, the entorhinal cortex, where the novelty-induced c-Fos expression was more strongly enhanced in GluA1-/- mice. Their novelty-induced hyperlocomotion partly depended on the activity of AMPA receptors, as it was diminished by the AMPA receptor antagonist 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulphonamide (NBQX) and unaffected by the AMPA receptor potentiator 2,3-dihydro-1,4-benzodioxin-6-yl-1-piperidinylmethanone (CX546). The hyperlocomotion of GluA1-/- mice was normalised to the level of wild-type mice within 5-6 h, after which their locomotion followed normal circadian rhythm and was not affected by acute or chronic treatments with the selective serotonin reuptake inhibitor escitalopram. We propose that hippocampal dysfunction, as evidenced by the excessive c-Fos response to novelty, is the major contributor to novelty-induced hyperlocomotion in GluA1-/- mice. Hippocampal dysfunction was also indicated by changes in proliferation and survival of adult-born dentate gyrus cells in the knockout mice. These results suggest focusing on the functions of hippocampal formation, such as novelty detection, when using the GluA1-/- mouse line as a model for neuropsychiatric and cognitive disorders.


Assuntos
Comportamento Exploratório/fisiologia , Hipocampo/fisiologia , Atividade Motora/fisiologia , Neurogênese/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de AMPA/metabolismo , Animais , Comportamento Animal/fisiologia , Citalopram/metabolismo , Feminino , Genótipo , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças do Sistema Nervoso/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/genética , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/genética , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA