Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Brain Behav Immun ; 109: 271-284, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36746342

RESUMO

Prenatal infections can increase the risk of developing psychiatric disorders such as schizophrenia in the offspring, especially when combined with other postnatal insults. Here, we tested, in a rat model of prenatal immune challenge by the viral mimic polyriboinosinic-polyribocytidilic acid, whether maternal immune activation (MIA) affects the endocannabinoid system and endocannabinoid-mediated modulation of dopamine functions. Experiments were performed during adolescence to assess i) the behavioral endophenotype (locomotor activity, plus maze, prepulse inhibition of startle reflex); ii) the locomotor activity in response to Δ9-Tetrahydrocannabinol (THC) and iii) the properties of ventral tegmental area (VTA) dopamine neurons in vivo and their response to THC; iv) endocannabinoid-mediated synaptic plasticity in VTA dopamine neurons; v) the expression of cannabinoid receptors and enzymes involved in endocannabinoid synthesis and catabolism in mesolimbic structures and vi) MIA-induced neuroinflammatory scenario evaluated by measurements of levels of cytokine and neuroinflammation markers. We revealed that MIA offspring displayed an altered locomotor activity in response to THC, a higher bursting activity of VTA dopamine neurons and a lack of response to cumulative doses of THC. Consistently, MIA adolescence offspring showed an enhanced 2-arachidonoylglycerol-mediated synaptic plasticity and decreased monoacylglycerol lipase activity in mesolimbic structures. Moreover, they displayed a higher expression of cyclooxygenase 2 (COX-2) and ionized calcium-binding adaptor molecule 1 (IBA-1), associated with latent inflammation and persistent microglia activity. In conclusion, we unveiled neurobiological mechanisms whereby inflammation caused by MIA influences the proper development of endocannabinoid signaling that negatively impacts the dopamine system, eventually leading to psychotic-like symptoms in adulthood.


Assuntos
Efeitos Tardios da Exposição Pré-Natal , Esquizofrenia , Gravidez , Feminino , Ratos , Masculino , Animais , Humanos , Endocanabinoides/metabolismo , Dopamina/metabolismo , Transdução de Sinais , Neurônios Dopaminérgicos/metabolismo
2.
J Neurosci ; 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34083258

RESUMO

Background: About 5 million people die from diseases related to nicotine addiction and tobacco use each year. Nicotine-induced increase of corticomesolimbic dopaminergic (DAergic) transmission and hypodopaminergic conditions occurring during abstinence are important for maintaining drug-use habits. Methods: We examined the notion of re-equilibrating DAergic transmission by inhibiting phosphodiesterase 7 (PDE7), an intracellular enzyme highly expressed in the corticomesolimbic circuitry and responsible for the degradation of cyclic adenosine monophosphate (cAMP), the main second messenger modulated by DA receptor activation. Results: Using selective PDE7 inhibitors, we demonstrated in male rats that systemic PDE7 enzyme inhibition reduced nicotine self-administration and prevented reinstatement to nicotine seeking evoked by cues or by the pharmacological stressor yohimbine. The effect was also observed by direct application of the PDE7 inhibitors into the nucleus accumbens (NAc) shell but not into the core. Inhibition of PDE7 resulted in increased DA- and cAMP-regulated neuronal phosphoprotein (DARPP-32) and cAMP response element-binding protein (CREB) and their phosphorylated forms in the NAc. It also enhanced the DA D1 receptor agonism-mediated effects, indicating potentiation of protein kinase A (PKA)-dependent transmission downstream of D1 receptor activation. In electrophysiological recordings from DA neurons in the lateral posterior ventral tegmental area (VTA), the PDE7 inhibitors attenuated the spontaneous activity of DA neurons. This effect was exerted through the potentiation of D1 receptor signaling and the subsequent facilitation of γ-aminobutyric acid (GABA) transmission. The PDE7 inhibitors did not elicit conditioned place preference and did not induce intravenous self-administration, indicating lack of reinforcing properties. Conclusions: PDE7 inhibitors have the potential to treat nicotine abuse.SIGNIFICANCE STATEMENTThe World Health Organization (WHO) estimates that there are 1.25 billion smokers worldwide, representing one third of the global population over the age of 15. Nicotine-induced increase of corticomesolimbic dopaminergic (DAergic) transmission and hypodopaminergic conditions occurring during abstinence are critical for maintaining drug-use habits. Here we demonstrate that nicotine consumption and relapse to nicotine seeking are attenuated by re-equilibrating DAergic transmission through inhibition of phosphodiesterase 7 (PDE7), an intracellular enzyme responsible for the degradation of cyclic adenosine monophosphate (cAMP), the main second messenger modulated by DA receptor activation. PDE7 inhibition may represent a novel treatment approach to aid smoking cessation.

3.
Stroke ; 53(4): e176-e187, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35142225

RESUMO

Marijuana is perceived as a harmless drug, and its recreational use has gained popularity among young individuals. The concentration of active ingredients in recreational formulations has gradually increased over time, and high-potency illicit cannabinomimetics have become available. Thus, the consumption of cannabis in the general population is rising. Data from preclinical models demonstrate that cannabinoid receptors are expressed in high density in areas involved in cognition and behavior, particularly during periods of active neurodevelopment and maturation. In addition, growing evidence highlights the role of endogenous cannabinoid pathways in the regulation of neurotransmitter release, synaptic plasticity, and neurodevelopment. In animal models, exogenous cannabinoids disrupt these important processes and lead to cognitive and behavioral abnormalities. These data correlate with the higher risk of cognitive impairment reported in some observational studies done in humans. It is unclear whether the effect of cannabis on cognition reverts after abstinence. However, this evidence, along with the increased risk of stroke reported in marijuana users, raises concerns about its potential long-term effects on cognitive function. This scientific statement reviews the safety of cannabis use from the perspective of brain health, describes mechanistically how cannabis may cause cognitive dysfunction, and advocates for a more informed health care worker and consumer about the potential for cannabis to adversely affect the brain.


Assuntos
Canabinoides , Cannabis , American Heart Association , Animais , Encéfalo/metabolismo , Canabinoides/efeitos adversos , Cannabis/efeitos adversos , Cannabis/metabolismo , Endocanabinoides/metabolismo , Humanos
4.
Eur J Neurosci ; 55(4): 903-908, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35118747

RESUMO

The endocannabinoid system is widely expressed both in the brain and in the periphery. This system regulates a plethora of physiological functions and is composed of cannabinoid receptors, their endogenous ligands, and the enzymes involved in their metabolic processes. In the last few years, the development of new imaging and molecular tools has demonstrated that these receptors are distributed in many cell types (e.g., neuronal or glial cells) and intracellular compartments (e.g., mitochondria). Interestingly, cellular or molecular effects are differentially mediated by cannabinoid receptors according to their specific localization in different cell-types or in different subcellular locations. Moreover, the endocannabinoid system is also expressed throughout the body where it can serve to modulate the connection between the brain and the periphery. Finally, better understanding of the cannabinoid receptors structure and pharmacology has led researchers to propose interesting and new allosteric modulators of synaptic communication. The latest advances and innovative research in the cannabinoid field will provide new insights and better approaches to improve its interesting potential therapeutic profile. This special issue intends to bring together a series of empirical papers, targeted reviews and opinions from leaders in the field that will highlight the new advances in cannabinoid research.


Assuntos
Canabinoides , Endocanabinoides , Receptores de Canabinoides , Encéfalo/metabolismo , Moduladores de Receptores de Canabinoides/farmacologia , Moduladores de Receptores de Canabinoides/fisiologia , Canabinoides/metabolismo , Canabinoides/farmacologia , Endocanabinoides/metabolismo , Receptores de Canabinoides/metabolismo , Transdução de Sinais
5.
Nat Rev Neurosci ; 18(11): 685-693, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28951609

RESUMO

Substance use disorders (SUDs) are one of the leading causes of morbidity and mortality worldwide. In spite of considerable advances in understanding the neural underpinnings of SUDs, therapeutic options remain limited. Recent studies have highlighted the potential of transcranial magnetic stimulation (TMS) as an innovative, safe and cost-effective treatment for some SUDs. Repetitive TMS (rTMS) influences neural activity in the short and long term by mechanisms involving neuroplasticity both locally, under the stimulating coil, and at the network level, throughout the brain. The long-term neurophysiological changes induced by rTMS have the potential to affect behaviours relating to drug craving, intake and relapse. Here, we review TMS mechanisms and evidence that rTMS is opening new avenues in addiction treatments.


Assuntos
Comportamento Aditivo/terapia , Encéfalo/fisiologia , Plasticidade Neuronal/fisiologia , Transtornos Relacionados ao Uso de Substâncias/terapia , Estimulação Magnética Transcraniana/métodos , Comportamento Aditivo/diagnóstico , Comportamento Aditivo/fisiopatologia , Humanos , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Resultado do Tratamento
6.
Addict Biol ; 26(3): e12967, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33021007

RESUMO

Physical exercise, which can be addictogenic on its own, is considered a therapeutic alternative for drug craving. Exercise might thus share with drugs the ability to strengthen excitatory synapses onto ventral tegmental area (VTA) dopaminergic neurones, as assessed by the ratio of AMPA receptor (AMPAR)-mediated excitatory postsynaptic currents (EPSCs) to NMDA receptor (NMDAR)-mediated EPSCs. As did acute cocaine, amphetamine, or Δ9 -tetrahydrocannabinol (THC) pretreatments, an acute 1-h wheel-running session increased the AMPAR/NMDAR ratio in VTA dopaminergic neurones. To dissect the respective influences of wheel-running seeking and performance, mice went through an operant protocol wherein wheel-running was conditioned by nose poking under fixed ratio schedules of reinforcement. Conditioned wheel-running increased the AMPAR/NMDAR ratio to a higher extent than free wheel-running, doing so although running performance was lower in the former paradigm than in the latter. Thus, the cue-reward association, rather than reward consumption, played a major role in this increase. The AMPAR/NMDAR ratio returned to baseline levels in mice that had extinguished the cued-running motivated task, but it increased after a cue-induced reinstatement session. The amplitude of this increase correlated with the intensity of exercise craving, as assessed by individual nose poke scores. Finally, cue-induced reinstatement of running seeking proved insensitive to acute cocaine or THC pretreatments. Our study reveals for the first time that the drive for exercise bears synaptic influences on VTA dopaminergic neurones which are reminiscent of drug actions. Whether these influences play a role in the therapeutic effects of exercise in human drug craving remains to be established.


Assuntos
Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Neurônios Dopaminérgicos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Anfetamina/administração & dosagem , Animais , Fissura/efeitos dos fármacos , Sinais (Psicologia) , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/fisiologia , Dronabinol/administração & dosagem , Potenciais Pós-Sinápticos Excitadores/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reforço Psicológico , Recompensa , Sinapses/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/fisiologia
7.
Int J Mol Sci ; 22(4)2021 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-33562259

RESUMO

Cannabis use among pregnant women is increasing worldwide along with permissive sociocultural attitudes toward it. Prenatal cannabis exposure (PCE), however, is associated with adverse outcome among offspring, ranging from reduced birth weight to child psychopathology. We have previously shown that male rat offspring prenatally exposed to Δ9-tetrahydrocannabinol (THC), a rat model of PCE, exhibit extensive molecular, cellular, and synaptic changes in dopamine neurons of the ventral tegmental area (VTA), resulting in a susceptible mesolimbic dopamine system associated with a psychotic-like endophenotype. This phenotype only reveals itself upon a single exposure to THC in males but not females. Here, we characterized the impact of PCE on female behaviors and mesolimbic dopamine system function by combining in vivo single-unit extracellular recordings in anesthetized animals and ex vivo patch clamp recordings, along with neurochemical and behavioral analyses. We find that PCE female offspring do not show any spontaneous or THC-induced behavioral disease-relevant phenotypes. The THC-induced increase in dopamine levels in nucleus accumbens was reduced in PCE female offspring, even when VTA dopamine activity in vivo and ex vivo did not differ compared to control. These findings indicate that PCE impacts mesolimbic dopamine function and its related behavioral domains in a sex-dependent manner and warrant further investigations to decipher the mechanisms determining this sex-related protective effect from intrauterine THC exposure.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dopamina/metabolismo , Dronabinol/toxicidade , Sistema Límbico/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/patologia , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Feminino , Alucinógenos/toxicidade , Sistema Límbico/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Área Tegmentar Ventral/patologia
8.
J Neurosci ; 39(42): 8250-8258, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619494

RESUMO

The recent shift in sociopolitical debates and growing liberalization of cannabis use across the globe has raised concern regarding its impact on vulnerable populations, such as pregnant women and adolescents. Epidemiological studies have long demonstrated a relationship between developmental cannabis exposure and later mental health symptoms. This relationship is especially strong in people with particular genetic polymorphisms, suggesting that cannabis use interacts with genotype to increase mental health risk. Seminal animal research directly linked prenatal and adolescent exposure to delta-9-tetrahydrocannabinol, the major psychoactive component of cannabis, with protracted effects on adult neural systems relevant to psychiatric and substance use disorders. In this article, we discuss some recent advances in understanding the long-term molecular, epigenetic, electrophysiological, and behavioral consequences of prenatal, perinatal, and adolescent exposure to cannabis/delta-9-tetrahydrocannabinol. Insights are provided from both animal and human studies, including in vivo neuroimaging strategies.


Assuntos
Cannabis/efeitos adversos , Cognição/fisiologia , Uso da Maconha/efeitos adversos , Transtornos Mentais/etiologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Adolescente , Animais , Feminino , Humanos , Transtornos Mentais/psicologia , Gravidez
9.
Addict Biol ; 25(5): e12803, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31342609

RESUMO

Smoking during adolescence may increase the likelihood to develop nicotine dependence and to abuse other drugs such as cocaine. Despite great efforts to understand underlying neurobiological mechanisms of this progression, less attention has been paid to the role of genetic factors. Here, we investigated the influence of both genetic background and age at first nicotine exposure in the long-lasting effects on mesolimbic dopamine transmission including the increased cocaine-rewarding effect. Mid-adolescent and adult rats of inbred strains Lewis (addiction prone) and Fischer 344 (addiction resistant) were administered nicotine (0.4 mg/kg) or vehicle once daily for 5 days. Changes in dopamine transmission were investigated by in vivo microdialysis and electrophysiology after 30 days of withdrawal, whereas changes in cocaine-rewarding effect were assessed via conditioned place preference paradigm. Nicotine pre-exposure differentially changed mesolimbic dopamine transmission depending on strain and age of pre-exposure. A potentiation of dopamine response to nicotine was observed in nucleus accumbens (NAc) core of both strains and age groups, whereas dopamine response in NAc shell was enhanced exclusively in Lewis rats exposed to nicotine during adolescence. A similar response was observed following cocaine challenge at adulthood. Changes in VTA dopamine cell population and activity were observed only in adolescent nicotine-pretreated Lewis rats, which also showed an increased cocaine-rewarding effect at adulthood. These results highlight the influence of genetic background in the long-lasting effects of nicotine exposure and suggest that exposure during adolescence might increase nicotine and cocaine-rewarding properties in genetically vulnerable individuals, thereby facilitating progression toward dependence.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/genética , Dopamina/metabolismo , Patrimônio Genético , Nicotina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Fatores Etários , Animais , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Masculino , Microdiálise , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew
10.
Epilepsia ; 58(10): 1762-1770, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28766701

RESUMO

OBJECTIVE: Nocturnal frontal lobe epilepsy (NFLE) is an idiopathic partial epilepsy with a family history in about 25% of cases, with autosomal dominant inheritance (autosomal dominant NFLE [ADNFLE]). Traditional antiepileptic drugs are effective in about 55% of patients, whereas the rest remains refractory. One of the key pathogenetic mechanisms is a gain of function of neuronal nicotinic acetylcholine receptors (nAChRs) containing the mutated α4 or ß2 subunits. Fenofibrate, a common lipid-regulating drug, is an agonist at peroxisome proliferator-activated receptor alpha (PPARα) that is a ligand-activated transcription factor, which negatively modulates the function of ß2-containing nAChR. To test clinical efficacy of adjunctive therapy with fenofibrate in pharmacoresistant ADNFLE\NFLE patients, we first demonstrated the effectiveness of fenofibrate in a mutated mouse model displaying both disease genotype and phenotype. METHODS: We first tested the efficacy of fenofibrate in transgenic mice carrying the mutation in the α4-nAChR subunit (Chrna4S252F) homologous to that found in humans. Subsequently, an add-on protocol was implemented in a clinical setting and fenofibrate was administered to pharmacoresistant NFLE patients. RESULTS: Here, we show that a chronic fenofibrate diet markedly reduced the frequency of large inhibitory postsynaptic currents (IPSCs) recorded from cortical pyramidal neurons in Chrna4S252F mice, and prevented nicotine-induced increase of IPSC frequency. Moreover, fenofibrate abolished differences between genotypes in the frequency of sleep-related movements observed under basal conditions. Patients affected by NFLE, nonresponders to traditional therapy, by means of adjunctive therapy with fenofibrate displayed a reduction of seizure frequency. Furthermore, digital video-polysomnographic recordings acquired in NFLE subjects after 6 months of adjunctive fenofibrate substantiated the significant effects on control of motor-behavioral seizures. SIGNIFICANCE: Our preclinical and clinical studies suggest PPARα as a novel disease-modifying target for antiepileptic drugs due to its ability to regulate dysfunctional nAChRs.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia Resistente a Medicamentos/tratamento farmacológico , Epilepsia do Lobo Frontal/tratamento farmacológico , Fenofibrato/uso terapêutico , PPAR alfa/agonistas , Adulto , Animais , Benzodiazepinas/uso terapêutico , Carbamazepina/análogos & derivados , Carbamazepina/uso terapêutico , Clobazam , Modelos Animais de Doenças , Epilepsia Resistente a Medicamentos/genética , Quimioterapia Combinada , Eletroencefalografia , Epilepsia do Lobo Frontal/genética , Feminino , Fenofibrato/farmacologia , Humanos , Lamotrigina , Levetiracetam , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação , Oxcarbazepina , Piracetam/análogos & derivados , Piracetam/uso terapêutico , Polissonografia , Receptores Nicotínicos/genética , Triazinas/uso terapêutico , Ácido Valproico/uso terapêutico , Adulto Jovem
11.
Artigo em Inglês | MEDLINE | ID: mdl-26819283

RESUMO

BACKGROUND: In utero exposure to maternal viral infections is associated with a higher incidence of psychiatric disorders with a supposed neurodevelopmental origin, including schizophrenia. Hence, immune response factors exert a negative impact on brain maturation that predisposes the offspring to the emergence of pathological phenotypes later in life. Although ventral tegmental area dopamine neurons and their target regions play essential roles in the pathophysiology of psychoses, it remains to be fully elucidated how dopamine activity and functionality are disrupted in maternal immune activation models of schizophrenia. METHODS: Here, we used an immune-mediated neurodevelopmental disruption model based on prenatal administration of the polyriboinosinic-polyribocytidilic acid in rats, which mimics a viral infection and recapitulates behavioral abnormalities relevant to psychiatric disorders in the offspring. Extracellular dopamine levels were measured by brain microdialysis in both the nucleus accumbens shell and the medial prefrontal cortex, whereas dopamine neurons in ventral tegmental area were studied by in vivo electrophysiology. RESULTS: Polyriboinosinic-polyribocytidilic acid-treated animals, at adulthood, displayed deficits in sensorimotor gating, memory, and social interaction and increased baseline extracellular dopamine levels in the nucleus accumbens, but not in the prefrontal cortex. In polyriboinosinic-polyribocytidilic acid rats, dopamine neurons showed reduced spontaneously firing rate and population activity. CONCLUSIONS: These results confirm that maternal immune activation severely impairs dopamine system and that the polyriboinosinic-polyribocytidilic acid model can be considered a proper animal model of a psychiatric condition that fulfills a multidimensional set of validity criteria predictive of a human pathology.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Viroses/imunologia , Viroses/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Feminino , Masculino , Transtornos da Memória/induzido quimicamente , Microdiálise , Núcleo Accumbens/metabolismo , Poli I-C/imunologia , Poli I-C/farmacologia , Córtex Pré-Frontal/metabolismo , Gravidez , Ratos , Filtro Sensorial/efeitos dos fármacos , Transtornos do Comportamento Social/induzido quimicamente , Área Tegmentar Ventral/fisiopatologia
12.
Addict Biol ; 21(5): 1043-51, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26935237

RESUMO

Sex differences in inhibition and self-regulation at a behavioral level have been widely described. From an evolutionary point of view, the different selection pressures placed on male and female hominids led them to differ in their behavioral strategies that allowed our species to survive during natural selection processes. These differences reflect changes in neural and structural plasticity that might be the core of sex differences, and of the susceptibility towards one psychiatric condition rather than another. The goal of the present review is to summarize current evidence for such a dichotomy in impulsive and compulsive behavior with a focus on drug addiction. Sex-dependent differences in drug abuse and dependence will be examined in the context of pathophysiological regulation of impulse and motivation by neuromodulators (i.e. gonadal hormones) and neurotransmitters (i.e. dopamine). Advances in the understanding of the sex differences in the capability to control impulses and motivational states is key for the determination of efficacious biologically based intervention and prevention strategies for several neuropsychiatric disorders where loss of impulse control and compulsivity are the core symptoms.


Assuntos
Comportamento Compulsivo/fisiopatologia , Comportamento Impulsivo/fisiologia , Caracteres Sexuais , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Feminino , Humanos , Masculino , Fatores Sexuais
13.
J Neurosci ; 34(38): 12716-24, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25232109

RESUMO

The progressive predominance of rewarding effects of addictive drugs over their aversive properties likely contributes to the transition from drug use to drug dependence. By inhibiting the activity of DA neurons in the VTA, GABA projections from the rostromedial tegmental nucleus (RMTg) are well suited to shift the balance between drug-induced reward and aversion. Since cannabinoids suppress RMTg inputs to DA cells and CB1 receptors affect alcohol intake in rodents, we hypothesized that the endocannabinoid system, by modulating this pathway, might contribute to alcohol preference. Here we found that RMTg afferents onto VTA DA neurons express CB1 receptors and display a 2-arachidonoylglycerol (2-AG)-dependent form of short-term plasticity, that is, depolarization-induced suppression of inhibition (DSI). Next, we compared rodents with innate opposite alcohol preference, the Sardinian alcohol-preferring (sP) and alcohol-nonpreferring (sNP) rats. We found that DA cells from alcohol-naive sP rats displayed a decreased probability of GABA release and a larger DSI. This difference was due to the rate of 2-AG degradation. In vivo, we found a reduced RMTg-induced inhibition of putative DA neurons in sP rats that negatively correlated with an increased firing. Finally, alcohol failed to enhance RMTg spontaneous activity and to prolong RMTg-induced silencing of putative DA neurons in sP rats. Our results indicate functional modifications of RMTg projections to DA neurons that might impact the reward/aversion balance of alcohol attributes, which may contribute to the innate preference observed in sP rats and to their elevated alcohol intake.


Assuntos
Ácidos Araquidônicos/fisiologia , Comportamento Aditivo/fisiopatologia , Neurônios Dopaminérgicos/fisiologia , Endocanabinoides/fisiologia , Etanol/farmacologia , Glicerídeos/fisiologia , Núcleo Tegmental Pedunculopontino/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Endogâmicos , Ácidos Araquidônicos/metabolismo , Comportamento Aditivo/induzido quimicamente , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Camundongos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Núcleo Tegmental Pedunculopontino/efeitos dos fármacos , Ratos , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia , Ácido gama-Aminobutírico/metabolismo
14.
Neurobiol Dis ; 73: 60-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25281318

RESUMO

Current concepts suggest that exposure to THC during adolescence may act as a risk factor for the development of psychiatric disorders later in life. However, the molecular underpinnings of this vulnerability are still poorly understood. To analyze this, we investigated whether and how THC exposure in female rats interferes with different maturational events occurring in the prefrontal cortex during adolescence through biochemical, pharmacological and electrophysiological means. We found that the endocannabinoid system undergoes maturational processes during adolescence and that THC exposure disrupts them, leading to impairment of both endocannabinoid signaling and endocannabinoid-mediated LTD in the adult prefrontal cortex. THC also altered the maturational fluctuations of NMDA subunits, leading to larger amounts of gluN2B at adulthood. Adult animals exposed to THC during adolescence also showed increased AMPA gluA1 with no changes in gluA2 subunits. Finally, adolescent THC exposure altered cognition at adulthood. All these effects seem to be triggered by the disruption of the physiological role played by the endocannabinoid system during adolescence. Indeed, blockade of CB1 receptors from early to late adolescence seems to prevent the occurrence of pruning at glutamatergic synapses. These results suggest that vulnerability of adolescent female rats to long-lasting THC adverse effects might partly reside in disruption of the pivotal role played by the endocannabinoid system in the prefrontal cortex maturation.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Deficiências do Desenvolvimento/induzido quimicamente , Dronabinol/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Fatores Etários , Animais , Cicloexanóis/farmacocinética , Maleato de Dizocilpina/farmacocinética , Estradiol/sangue , Ciclo Estral/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacocinética , Feminino , Técnicas In Vitro , Neuritos/efeitos dos fármacos , Piperidinas/farmacologia , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/ultraestrutura , Pirazóis/farmacologia , Cintilografia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Trítio/farmacocinética
15.
Front Neuroendocrinol ; 35(3): 272-84, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24769267

RESUMO

Gender-dependent differences in the rate of initiation and frequency of misuse of addicting drugs have been widely described. Yet, men and women also differ in their propensity to become addicted to other rewarding stimuli (e.g., sex, food) or activities (e.g., gambling, exercising). The goal of the present review is to summarize current evidence for gender differences not only in drug addiction, but also in other forms of addictive behaviours. Thus, we first reviewed studies showing gender-dependent differences in drug addiction, food addiction, compulsive sexual activity, pathological gambling, Internet addiction and physical exercise addiction. Potential risk factors and underlying brain mechanisms are also examined, with particular emphasis given to the role of sex hormones in modulating addictive behaviours. Investigations on factors allowing the pursuit of non-drug rewards to become pathological in men and women are crucial for designing gender-appropriate treatments of both substance and non-substance addictions.


Assuntos
Comportamento Aditivo , Jogo de Azar , Transtornos Relacionados ao Uso de Substâncias , Animais , Comportamento Aditivo/psicologia , Jogo de Azar/psicologia , Humanos , Internet , Recompensa , Caracteres Sexuais , Transtornos Relacionados ao Uso de Substâncias/etiologia , Transtornos Relacionados ao Uso de Substâncias/psicologia
16.
Addict Biol ; 20(1): 182-93, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24103023

RESUMO

Ethanol excites dopamine (DA) neurons in the posterior ventral tegmental area (pVTA). This effect is responsible for ethanol's motivational properties and may contribute to alcoholism. Evidence indicates that catalase-mediated conversion of ethanol into acetaldehyde in pVTA plays a critical role in this effect. Acetaldehyde, in the presence of DA, condensates with it to generate salsolinol. Salsolinol, when administered in pVTA, excites pVTA DA cells, elicits DA transmission in nucleus accumbens and sustains its self-administration in pVTA. Here we show, by using ex vivo electrophysiology, that ethanol and acetaldehyde, but not salsolinol, failed to stimulate pVTA DA cell activity in mice administered α-methyl-p-tyrosine, a DA biosynthesis inhibitor that reduces somatodendritic DA release. This effect was specific for ethanol and acetaldehyde since morphine, similarly to salsolinol, was able to excite pVTA DA cells in α-methyl-p-tyrosine-treated mice. However, when DA was bath applied in slices from α-methyl-p-tyrosine-treated mice, ethanol-induced excitation of pVTA DA neurons was restored. This effect requires ethanol oxidation into acetaldehyde given that, when H2 O2 -catalase system was impaired by either 3-amino-1,2,4-triazole or in vivo administration of α-lipoic acid, ethanol did not enhance DA cell activity. Finally, high performance liquid chromatography-tandem mass spectrometry analysis of bath medium detected salsolinol only after co-application of ethanol and DA in α-methyl-p-tyrosine-treated mice. These results demonstrate the relationship between ethanol and salsolinol effects on pVTA DA neurons, help to untangle the mechanism(s) of action of ethanol in this area and contribute to an exciting research avenue prosperous of theoretical and practical consequences.


Assuntos
Acetaldeído/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Etanol/farmacologia , Isoquinolinas/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos , Amitrol (Herbicida)/farmacologia , Animais , Antioxidantes/farmacologia , Catalase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Ácido Tióctico/farmacologia , alfa-Metiltirosina/farmacologia
17.
J Neurosci ; 33(14): 6203-11, 2013 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-23554501

RESUMO

Ventral tegmental area dopamine neurons control reward-driven learning, and their dysregulation can lead to psychiatric disorders. Tonic and phasic activity of these dopaminergic neurons depends on cholinergic tone and activation of nicotinic acetylcholine receptors (nAChRs), particularly those containing the ß2 subunit (ß2*-nAChRs). Nuclear peroxisome proliferator-activated receptors type-α (PPARα) tonically regulate ß2*-nAChRs and thereby control dopamine neuron firing activity. However, it is unknown how and when PPARα endogenous ligands are synthesized by dopamine cells. Using ex vivo and in vivo electrophysiological techniques combined with biochemical and behavioral analysis, we show that activation of α7-nAChRs increases in the rat VTA both the tyrosine phosphorylation of the ß2 subunit of nAChRs and the levels of two PPARα endogenous ligands in a Ca(2+)-dependent manner. Accordingly, in vivo production of endogenous PPARα ligands, triggered by α7-nAChR activation, blocks in rats nicotine-induced increased firing activity of dopamine neurons and displays antidepressant-like properties. These data demonstrate that endogenous PPARα ligands are effectors of α7-nAChRs and that their neuromodulatory properties depend on phosphorylation of ß2*-nAChRs on VTA dopamine cells. This reveals an autoinhibitory mechanism aimed at reducing dopamine cell overexcitation engaged during hypercholinergic drive. Our results unveil important physiological functions of nAChR/PPARα signaling in dopamine neurons and how behavioral output can change after modifications of this signaling pathway. Overall, the present study suggests PPARα as new therapeutic targets for disorders associated with unbalanced dopamine-acetylcholine systems.


Assuntos
Colinérgicos/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , PPAR alfa/metabolismo , Receptores Nicotínicos/metabolismo , Área Tegmentar Ventral/citologia , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Benzamidas/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Carbamatos/farmacologia , Di-Hidro-beta-Eritroidina/farmacologia , Neurônios Dopaminérgicos/fisiologia , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Etanolaminas/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Ligantes , Masculino , PPAR alfa/agonistas , Técnicas de Patch-Clamp , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Natação/psicologia , Tirosina 3-Mono-Oxigenase/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
18.
Neurobiol Dis ; 65: 133-41, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24491965

RESUMO

Synchronized activity is common during various physiological operations but can culminate in seizures and consequently in epilepsy in pathological hyperexcitable conditions in the brain. Many types of seizures are not possible to control and impose significant disability for patients with epilepsy. Such intractable epilepsy cases are often associated with degeneration of inhibitory interneurons in the cortical areas resulting in impaired inhibitory drive onto the principal neurons. Recently emerging optogenetic technique has been proposed as an alternative approach to control such seizures but whether it may be effective in situations where inhibitory processes in the brain are compromised has not been addressed. Here we used pharmacological and optogenetic techniques to block inhibitory neurotransmission and induce epileptiform activity in vitro and in vivo. We demonstrate that NpHR-based optogenetic hyperpolarization and thereby inactivation of a principal neuronal population in the hippocampus is effectively attenuating seizure activity caused by disconnected network inhibition both in vitro and in vivo. Our data suggest that epileptiform activity in the hippocampus caused by impaired inhibition may be controlled by optogenetic silencing of principal neurons and potentially can be developed as an alternative treatment for epilepsy.


Assuntos
Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Optogenética , Estado Epiléptico/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Aminopiridinas/farmacologia , Análise de Variância , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Técnicas In Vitro , Ácido Caínico/toxicidade , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Picrotoxina/farmacologia , Estado Epiléptico/induzido quimicamente , Transdução Genética
19.
Eur J Neurosci ; 39(7): 1189-201, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24712998

RESUMO

Synaptic neuromodulation acts across different functional domains to regulate cognitive processing and behavior. Recent challenges are related to elucidating the molecular and cellular mechanisms through which neuromodulatory pathways act on multiple time scales to signal state-dependent contingencies at the synaptic level or to stabilise synaptic connections during behavior. Here, we present a framework with the synaptic neuromodulators endocannabinoids (eCBs) as key players in dynamic synaptic changes. Modulation of various molecular components of the eCB pathway yields interconnected functional activation states of eCB signaling (prior, tonic, and persistent), which may contribute to metaplastic control of synaptic and behavioral functions in health and disease. The emerging picture supports aberrant metaplasticity as a contributor to cognitive dysfunction associated with several pathological states in which eCB signaling, or other neuromodulatory pathways, are deregulated.


Assuntos
Endocanabinoides/metabolismo , Plasticidade Neuronal , Sinapses/metabolismo , Transmissão Sináptica , Animais , Humanos , Sinapses/fisiologia
20.
Pharmacol Res ; 86: 42-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24704146

RESUMO

Nicotine is one of the drugs of abuse that frequently causes addiction and relapse during abstinence. Nicotine's strong addicting properties reside in its ability to enhance dopamine transmission, and to induce specific changes in synaptic plasticity. Currently, approved therapies for smoking cessation increase the chances of remaining abstinent, but lack high levels of efficacy and are associated with significant adverse side effects. As a result, there is an urgent need for more effective antismoking medications. Studies have revealed that drugs targeting the peroxisome proliferator-activated-receptor-α (PPARα) show promise for the treatment of nicotine addiction. These drugs include synthetic PPARα ligands, such as the clinically available hypolipidemic fibrates, and drugs that increase levels of endogenous endocannabinoid-like fatty acid ethanolamides (FAEs) that act as PPARα agonists. In this review, we will discuss the specific interaction between PPARα and nicotine, and the molecular mechanisms whereby these intracellular receptors regulate nicotinic acetylcholine receptor functions in neurons. Modulation of neurophysiological, neurochemical and behavioral effects of nicotine by PPARα will be also reviewed. Indeed, a picture is emerging where FAEs are endogenous regulators of acetylcholine transmission. Notably, the implications of this specific cross talk extend beyond nicotine addiction, and might bear relevance for psychiatric disorders and epilepsy.


Assuntos
Endocanabinoides/metabolismo , Etanolaminas/metabolismo , Doenças do Sistema Nervoso/metabolismo , Ácidos Oleicos/metabolismo , Ácidos Palmíticos/metabolismo , Receptores Nicotínicos/metabolismo , Tabagismo/metabolismo , Amidas , Animais , Humanos , Terapia de Alvo Molecular , Doenças do Sistema Nervoso/tratamento farmacológico , Nicotina/metabolismo , PPAR alfa/agonistas , PPAR alfa/metabolismo , Tabagismo/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA