Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 552
Filtrar
1.
J Neural Transm (Vienna) ; 131(5): 509-523, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38630190

RESUMO

Substance use disorders (SUDs) are the most costly and prevalent psychiatric conditions. Recent calls emphasize a need for biomarkers-measurable, stable indicators of normal and abnormal processes and response to treatment or environmental agents-and, in particular, brain-based neuromarkers that will advance understanding of the neurobiological basis of SUDs and clinical practice. To develop neuromarkers, researchers must be grounded in evidence that a putative marker (i) is sensitive and specific to the psychological phenomenon of interest, (ii) constitutes a predictive model, and (iii) generalizes to novel observations (e.g., through internal cross-validation and external application to novel data). These neuromarkers may be used to index risk of developing SUDs (susceptibility), classify individuals with SUDs (diagnostic), assess risk for progression to more severe pathology (prognostic) or index current severity of pathology (monitoring), detect response to treatment (response), and predict individualized treatment outcomes (predictive). Here, we outline guidelines for developing and assessing neuromarkers, we then review recent advances toward neuromarkers in addiction neuroscience centering our discussion around neuromarkers of craving-a core feature of SUDs. In doing so, we specifically focus on the Neurobiological Craving Signature (NCS), which show great promise for meeting the demand of neuromarkers.


Assuntos
Biomarcadores , Transtornos Relacionados ao Uso de Substâncias , Humanos , Biomarcadores/metabolismo , Transtornos Relacionados ao Uso de Substâncias/diagnóstico , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Encéfalo/metabolismo , Comportamento Aditivo/diagnóstico , Comportamento Aditivo/metabolismo
2.
Int J Mol Sci ; 24(23)2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-38069445

RESUMO

Substance addiction is a chronic and relapsing brain disorder characterized by compulsive seeking and continued substance use, despite adverse consequences. The high prevalence and social burden of addiction are indisputable; however, the available intervention is insufficient. The modulation of gene expression and aberrant adaptation of neural networks are attributed to the changes in brain functions under repeated exposure to addictive substances. Considerable studies have demonstrated that miRNAs are strong modulators of post-transcriptional gene expression in substance addiction. The emerging role of microRNA (miRNA) provides new insights into many biological and pathological processes in the central nervous system: their variable expression in different regions of the brain and tissues may play a key role in regulating the pathophysiological events of addiction. This work provides an overview of the current literature on miRNAs involved in addiction, evaluating their impaired expression and regulatory role in neuroadaptation and synaptic plasticity. Clinical implications of such modulatory capacities will be estimated. Specifically, it will evaluate the potential diagnostic role of miRNAs in the various stages of drug and substance addiction. Future perspectives about miRNAs as potential novel therapeutic targets for substance addiction and abuse will also be provided.


Assuntos
Comportamento Aditivo , MicroRNAs , Transtornos Relacionados ao Uso de Substâncias , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Comportamento Aditivo/metabolismo , Encéfalo/metabolismo
3.
Biol Psychiatry ; 92(11): 836-844, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36328706

RESUMO

In 2014, we proposed that orexin signaling transformed motivationally relevant states into adaptive behavior directed toward exploiting an opportunity or managing a threat, a process we referred to as motivational activation. Advancements in animal models since then have permitted higher-resolution measurements of motivational states; in particular, the behavioral economics approach for studying drug demand characterizes conditions that lead to the enhanced motivation that underlies addiction. This motivational plasticity is paralleled by persistently increased orexin expression in a topographically specific manner-a finding confirmed across species, including in humans. Normalization of orexin levels also reduces drug motivation in addiction models. These new advancements lead us to update our proposed framework for the orexin function. We now propose that the capacity of orexin neurons to exhibit dynamic shifts in peptide production contributes to their role in adaptive motivational regulation and that this is achieved via a pool of reserve orexin neurons. This reserve is normally bidirectionally recruited to permit motivational plasticity that promotes flexible, adaptive behavior. In pathological states such as addiction, however, we propose that the orexin system loses capacity to adaptively adjust peptide production, resulting in focused hypermotivation for drug, driven by aberrantly and persistently high expression in the orexin reserve pool. This mechanistic framework has implications for the understanding and treatment of several psychiatric disorders beyond addiction, particularly those characterized by motivational dysfunction.


Assuntos
Comportamento Aditivo , Neuropeptídeos , Animais , Humanos , Orexinas , Neuropeptídeos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Comportamento Aditivo/metabolismo , Motivação , Receptores de Orexina
4.
Neurosci Biobehav Rev ; 141: 104832, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35988803

RESUMO

Reducing sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric disorders. Furthermore, previous studies have shown that reduction in sleep time is a factor that favors relapse in addicted patients. Additionally, animal models have demonstrated that both sleep restriction and sleep deprivation increase the preference for alcohol, methylphenidate, and the self-administration of cocaine. Therefore, the present review discusses current knowledge about the influence of sleep hours reduction on addictivebehaviors; likewise, we discuss the neuronal basis underlying the sleep reduction-addiction relationship, like the role of the orexin and dopaminergic system and neuronal plasticity (i.e., delta FosB expression). Potentially, chronic sleep restriction could increase brain vulnerability and promote addictive behavior.


Assuntos
Comportamento Aditivo , Cocaína , Metilfenidato , Distúrbios do Início e da Manutenção do Sono , Animais , Comportamento Aditivo/metabolismo , Humanos , Orexinas , Sono
5.
Chaos ; 32(2): 021102, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35232044

RESUMO

Substances of abuse are known to activate and disrupt neuronal circuits in the brain reward system. We propose a simple and easily interpretable dynamical systems model to describe the neurobiology of drug addiction that incorporates the psychiatric concepts of reward prediction error, drug-induced incentive salience, and opponent process theory. Drug-induced dopamine releases activate a biphasic reward response with pleasurable, positive "a-processes" (euphoria, rush) followed by unpleasant, negative "b-processes" (cravings, withdrawal). Neuroadaptive processes triggered by successive intakes enhance the negative component of the reward response, which the user compensates for by increasing drug dose and/or intake frequency. This positive feedback between physiological changes and drug self-administration leads to habituation, tolerance, and, eventually, to full addiction. Our model gives rise to qualitatively different pathways to addiction that can represent a diverse set of user profiles (genetics, age) and drug potencies. We find that users who have, or neuroadaptively develop, a strong b-process response to drug consumption are most at risk for addiction. Finally, we include possible mechanisms to mitigate withdrawal symptoms, such as through the use of methadone or other auxiliary drugs used in detoxification.


Assuntos
Comportamento Aditivo , Transtornos Relacionados ao Uso de Substâncias , Comportamento Aditivo/metabolismo , Comportamento Aditivo/psicologia , Encéfalo/metabolismo , Humanos , Modelos Teóricos , Motivação , Recompensa , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/psicologia
6.
Int J Mol Sci ; 22(24)2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34948106

RESUMO

Nicotine, the main psychoactive component in tobacco smoke, plays a major role in tobacco addiction, producing a high morbidity and mortality in the world. A great amount of research has been developed to elucidate the neural pathways and neurotransmitter systems involved in such a complex addictive behavior. The endocannabinoid system, which has been reported to participate in the addictive properties of most of the prototypical drugs of abuse, is also implicated in nicotine dependence. This review summarizes and updates the main behavioral and biochemical data involving the endocannabinoid system in the rewarding properties of nicotine as well as in nicotine withdrawal and relapse to nicotine-seeking behavior. Promising results from preclinical studies suggest that manipulation of the endocannabinoid system could be a potential therapeutic strategy for treating nicotine addiction.


Assuntos
Comportamento Aditivo , Canabinoides/uso terapêutico , Endocanabinoides/metabolismo , Tabagismo , Animais , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/metabolismo , Humanos , Tabagismo/tratamento farmacológico , Tabagismo/metabolismo
7.
Neuron ; 109(18): 2943-2966.e8, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34480866

RESUMO

Neuronal alternative splicing is a key gene regulatory mechanism in the brain. However, the spliceosome machinery is insufficient to fully specify splicing complexity. In considering the role of the epigenome in activity-dependent alternative splicing, we and others find the histone modification H3K36me3 to be a putative splicing regulator. In this study, we found that mouse cocaine self-administration caused widespread differential alternative splicing, concomitant with the enrichment of H3K36me3 at differentially spliced junctions. Importantly, only targeted epigenetic editing can distinguish between a direct role of H3K36me3 in splicing and an indirect role via regulation of splice factor expression elsewhere on the genome. We targeted Srsf11, which was both alternatively spliced and H3K36me3 enriched in the brain following cocaine self-administration. Epigenetic editing of H3K36me3 at Srsf11 was sufficient to drive its alternative splicing and enhanced cocaine self-administration, establishing the direct causal relevance of H3K36me3 to alternative splicing of Srsf11 and to reward behavior.


Assuntos
Processamento Alternativo/fisiologia , Comportamento Aditivo/metabolismo , Cromatina/metabolismo , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Recompensa , Processamento Alternativo/efeitos dos fármacos , Animais , Comportamento Aditivo/genética , Comportamento Aditivo/psicologia , Cromatina/genética , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Autoadministração
8.
Alcohol Clin Exp Res ; 45(11): 2231-2245, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34585391

RESUMO

BACKGROUND: Few studies have examined the association between APOE genotype and alcohol use. Although some of these studies have reported outcomes associated with a history of drinking, none have examined alcohol-seeking behavior. In addition, no preclinical studies have examined alcohol use as a function of APOE genotype with or without traumatic brain injury. METHODS: Male and female human APOE3- and APOE4-targeted replacement (TR) mice were used to assess voluntary alcohol seeking longitudinally using a 2-bottle choice paradigm conducted within the automated IntelliCage system prior to and following repeated mild TBI (rmTBI). Following an acquisition phase in which the concentration of ethanol (EtOH) was increased to 12%, a variety of drinking paradigms that included extended alcohol access (EAA1 and EAA2), alcohol deprivation effect (ADE), limited access drinking in the dark (DID), and progressive ratio (PR) were used to assess alcohol-seeking behavior. Additional behavioral tasks were performed to measure cognitive function and anxiety-like behavior. RESULTS: All groups readily consumed increasing concentrations of EtOH (4-12%) during the acquisition phase. During the EAA1 period (12% EtOH), there was a significant genotype effect in both males and females for EtOH preference. Following a 3-week abstinence period, mice received sham or rmTBI resulting in a genotype- and sex-independent main effect of rmTBI on the recovery of righting reflex and a main effect of rmTBI on spontaneous home-cage activity in females only. Reintroduction of 12% EtOH (EAA2) resulted in a significant effect genotype for alcohol preference in males with APOE4 mice displaying increased preference and motivation for alcohol compared with APOE3 mice independent of TBI while in females, there was a significant genotype × TBI interaction under the ADE and DID paradigms. Finally, there was a main effect of rmTBI on increased risk-seeking behavior in both sexes, but no effect on spatial learning or cognitive flexibility. CONCLUSION: These results suggest that sex and APOE genotype play a significant role in alcohol consumption and may subsequently influence long-term recovery following traumatic brain insults.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Apolipoproteínas E/metabolismo , Comportamento Aditivo/metabolismo , Genótipo , Consumo de Bebidas Alcoólicas/genética , Animais , Apolipoproteínas E/genética , Comportamento Aditivo/genética , Condicionamento Clássico/fisiologia , Feminino , Humanos , Masculino , Camundongos
9.
PLoS One ; 16(6): e0252516, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34061895

RESUMO

Gambling Disorder (GD) has recently been reclassified from an impulse-control disorder to a behavioural addiction and, as in other addictive disorders, the dopaminergic reward system is involved. According to neuroimaging studies, alterations within the striatal dopaminergic signalling can occur in GD. However, the findings to date are controversial and there has been no agreement yet on how the reward system is affected on a molecular basis. Within the last 20 years, there has been growing evidence for a higher risk to develop GD in response to certain dopaminergic medication. Especially the dopamine agonists pramipexole and ropinirole, and the dopamine modulator aripiprazole seem to increase the likelihood for GD. The goal of this study was to examine the association between a prescription for either of the three pharmaceuticals and a GD diagnosis in a large cross-sectional study of the Swedish population. Compared to patients with any other dopaminergic drug prescription (38.7% with GD), the diagnosis was more common in patients with a dopamine agonist prescription (69.8% with GD), resulting in an odds ratio of 3.2. A similar association was found between aripiprazole prescriptions and GD diagnoses, which were analysed within the subgroup of all patients with schizophrenia or a schizotypal, delusional, or another non-mood psychotic disorder. An aripiprazole prescription increased the likelihood of GD (88.8%) in comparison to patients without an aripiprazole prescription (71.2%) with an odds ratio of 3.4. This study contributes to the increasingly reliable evidence for an association between several dopaminergic drugs and a higher risk for developing GD. Therefore, one future research goal should be a better understanding of the neurobiology in GD to be able to design more selective dopaminergic medication with less severe side effects. Additionally, this knowledge could enable the development of pharmacotherapy in GD and other addictive disorders.


Assuntos
Aripiprazol/efeitos adversos , Comportamento Aditivo/induzido quimicamente , Agonistas de Dopamina/efeitos adversos , Jogo de Azar/induzido quimicamente , Indóis/efeitos adversos , Pramipexol/efeitos adversos , Sistema de Registros , Adulto , Idoso , Idoso de 80 Anos ou mais , Comportamento Aditivo/diagnóstico , Comportamento Aditivo/epidemiologia , Comportamento Aditivo/metabolismo , Estudos Transversais , Dopamina/metabolismo , Feminino , Jogo de Azar/diagnóstico , Jogo de Azar/epidemiologia , Jogo de Azar/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Recompensa , Fatores de Risco , Suécia/epidemiologia , Adulto Jovem
10.
Pharmacol Biochem Behav ; 206: 173208, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34022293

RESUMO

Methylphenidate (MET) has a putative cognitive enhancer effect that has led adolescents and young adults to increase and indiscriminate its use aiming to ameliorate their productivity. However, the impacts of MET on addiction-related behaviors, emotional levels, and cognition are still not fully understood. To investigate the influence of chronic treatment with MET during adolescence on addiction-like behaviors, memory, and anxiety in adult mice. Thirty-day-old female mice received i.p. 10 mg/kg MET or Veh injections for 10 consecutive days. Forty days after the treatment (mice were 70-days-old), animals were submitted to the behavioral evaluation under the effects of MET, which included: MET-induced conditioned place preference (CPP), behavioral sensitization, and plus-maze discriminative avoidance task. Pre-exposure to MET during adolescence promoted an early expression of CPP and also facilitated the development of MET-induced behavioral sensitization during adulthood. These addictive-like behaviors were accompanied by anxiogenic effects of MET but not by any memory-enhancing effect. We demonstrated that exposure to MET during adolescence can increase the vulnerability to addiction-like behaviors and anxiety during adulthood. Our results reinforce the necessity of a more efficient system to control MET indiscriminate use, thus avoiding its potential tardive addictive effects.


Assuntos
Ansiedade/metabolismo , Comportamento Aditivo/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Memória/efeitos dos fármacos , Metilfenidato/farmacologia , Animais , Ansiedade/psicologia , Comportamento Aditivo/psicologia , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Feminino , Humanos , Camundongos , Modelos Animais , Atividade Motora/efeitos dos fármacos
11.
Neuropharmacology ; 192: 108599, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33965397

RESUMO

There has been a dramatic increase in illicit fentanyl use in the United States over the last decade. In 2018, more than 31,000 overdose deaths involved fentanyl or fentanyl analogs, highlighting an urgent need to identify effective treatments for fentanyl use disorder. An emerging literature shows that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate the reinforcing efficacy of drugs of abuse. However, the effects of GLP-1R agonists on fentanyl-mediated behaviors are unknown. The first goal of this study was to determine if the GLP-1R agonist exendin-4 reduced fentanyl self-administration and the reinstatement of fentanyl-seeking behavior, an animal model of relapse, in rats. We found that systemic exendin-4 attenuated fentanyl taking and seeking at doses that also produced malaise-like effects in rats. To overcome these adverse effects and enhance the clinical potential of GLP-1R agonists, we recently developed a novel dual agonist of GLP-1Rs and neuropeptide Y2 receptors (Y2Rs), GEP44, that does not produce nausea-like behavior in drug-naïve rats or emesis in drug-naïve shrews. The second goal of this study was to determine if GEP44 reduced fentanyl self-administration and reinstatement with fewer adverse effects compared to exendin-4 alone. In contrast to exendin-4, GEP44 attenuated opioid taking and seeking at a dose that did not suppress food intake or produce adverse malaise-like effects in fentanyl-experienced rats. Taken together, these findings indicate a novel role for GLP-1Rs and Y2Rs in fentanyl reinforcement and highlight a potential new therapeutic approach to treating opioid use disorders.


Assuntos
Analgésicos Opioides/administração & dosagem , Comportamento Aditivo/tratamento farmacológico , Comportamento de Procura de Droga/efeitos dos fármacos , Fentanila/administração & dosagem , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptores de Neuropeptídeo Y/agonistas , Animais , Comportamento Aditivo/metabolismo , Comportamento Aditivo/psicologia , Relação Dose-Resposta a Droga , Comportamento de Procura de Droga/fisiologia , Exenatida/farmacologia , Exenatida/uso terapêutico , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/metabolismo , Autoadministração
12.
Neuropharmacology ; 192: 108598, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33965398

RESUMO

Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.


Assuntos
Eixo Encéfalo-Intestino/fisiologia , Encéfalo/imunologia , Microbioma Gastrointestinal/fisiologia , Neuroimunomodulação/fisiologia , Transtornos Relacionados ao Uso de Substâncias/imunologia , Comportamento Aditivo/imunologia , Comportamento Aditivo/metabolismo , Comportamento Aditivo/psicologia , Encéfalo/metabolismo , Disbiose/imunologia , Disbiose/psicologia , Humanos , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/psicologia
13.
Front Neurol Neurosci ; 45: 117-127, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34052815

RESUMO

Significant sleep impairments often accompany substance use disorders (SUDs). Sleep disturbances in SUD patients are associated with poor clinical outcomes and treatment adherence, emphasizing the importance of normalizing sleep when treating SUDs. Orexins (hypocretins) are neuropeptides exclusively produced by neurons in the posterior hypothalamus that regulate various behavioral and physiological processes, including sleep-wakefulness and motivated drug taking. Given its dual role in sleep and addiction, the orexin system represents a promising therapeutic target for treating SUDs and their comorbid sleep deficits. Here, we review the literature on the role of the orexin system in sleep and drug addiction and discuss the therapeutic potential of orexin receptor antagonists for SUDs. We argue that orexin receptor antagonists may be effective therapeutics for treating addiction because they target orexin's regulation of sleep (top-down) and motivation (bottom-up) pathways.


Assuntos
Comportamento Aditivo/metabolismo , Motivação/fisiologia , Antagonistas dos Receptores de Orexina/farmacologia , Orexinas/metabolismo , Recompensa , Distúrbios do Início e da Manutenção do Sono/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Comportamento Aditivo/tratamento farmacológico , Humanos , Motivação/efeitos dos fármacos , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico
14.
Neuropharmacology ; 192: 108617, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34019906

RESUMO

The epithalamic lateral habenula (LHb) regulates monoaminergic systems and contributes to the expression of both appetitive and aversive behaviours. Over the past years, the LHb has emerged as a vulnerable brain structure in mental illnesses including addiction. Behavioural and functional evidence in humans and rodents provide substantial support for a role of LHb in the negative affective symptoms emerging during withdrawal from addictive substances. Multiple forms of cellular and synaptic adaptations that take hold during drug withdrawal within the LHb are causally linked with the emergence of negative affective symptoms. These results indicate that targeting drug withdrawal-driven adaptations in the LHb may represent a potential strategy to normalize drug-related behavioural adaptations. In the current review we describe the mechanisms leading to functional alterations in the LHb, as well as the existing interventions used to counteract addictive behaviours. Finally, closing this loop we discuss and propose new avenues to potentially target the LHb in humans in light of the mechanistic understanding stemming from pre-clinical studies. Altogether, we provide an overview on how to leverage cellular-level understanding to envision clinically-relevant approaches for the treatment of specific aspects in drug addiction.


Assuntos
Adaptação Fisiológica/fisiologia , Comportamento Aditivo/metabolismo , Habenula/metabolismo , Neurônios/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Comportamento Aditivo/genética , Comportamento Aditivo/terapia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/terapia , Transtornos Relacionados ao Uso de Substâncias/genética , Transtornos Relacionados ao Uso de Substâncias/terapia
15.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673694

RESUMO

Recently, oxytocin (OXT) has been investigated for its potential therapeutic role in addiction. OXT has been found to diminish various drug-seeking and drug-induced behaviors. Although its behavioral effects are well-established, there is not much consensus on how this neuropeptide exerts its effects. Previous research has given thought to how dopamine (DA) may be involved in oxytocinergic mechanisms, but there has not been as strong of a focus on the role that glutamate (Glu) has. The glutamatergic system is critical for the processing of rewards and the disruption of glutamatergic projections produces the behaviors seen in drug addicts. We introduce the idea that OXT has direct effects on Glu transmission within the reward processing pathway. Thus, OXT may reduce addictive behaviors by restoring abnormal drug-induced changes in the glutamatergic system and in its interactions with other neurotransmitters. This review offers insight into the mechanisms through which a potentially viable therapeutic target, OXT, could be used to reduce addiction-related behaviors.


Assuntos
Comportamento Aditivo/prevenção & controle , Comportamento de Procura de Droga/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Ocitócicos/farmacologia , Ocitocina/farmacologia , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle , Animais , Comportamento Aditivo/metabolismo , Humanos , Transtornos Relacionados ao Uso de Substâncias/metabolismo
16.
Adv Pharmacol ; 90: 89-115, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33706940

RESUMO

Paul Greengard brought to neuroscience the idea of, and evidence for, the role of second messenger systems in neuronal signaling. The fundamental nature of his contributions is evident in the far reach of his work, relevant to various subfields and topics in neuroscience. In this review, we discuss some of Greengard's work from the perspective of nicotinic acetylcholine receptors and their relevance to nicotine addiction. Specifically, we review the roles of dopamine- and cAMP-regulated phospho-protein of 32kDa (DARPP-32) and Ca2+/calmodulin-dependent kinase II (CaMKII) in nicotine-dependent behaviors. For each protein, we discuss the historical context of their discovery and initial characterization, focusing on the extensive biochemical and immunohistochemical work conducted by Greengard and colleagues. We then briefly summarize contemporary understanding of each protein in key intracellular signaling cascades and evidence for the role of each protein with respect to systems and behaviors relevant to nicotine addiction.


Assuntos
Comportamento Aditivo/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Nicotina/farmacologia , Transdução de Sinais , Animais , Humanos , Receptores Nicotínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Pharmacol Biochem Behav ; 204: 173155, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33631255

RESUMO

Alcohol tolerance refers to a lower effect of alcohol with repeated exposure. Although alcohol tolerance has been historically included in diagnostic manuals as one of the key criteria for a diagnosis of alcohol use disorder (AUD), understanding its neurobiological mechanisms has been neglected in preclinical studies. In this mini-review, we provide a theoretical framework for alcohol tolerance. We then briefly describe chronic tolerance, followed by a longer discussion of behavioral and neurobiological aspects that underlie rapid tolerance in rodent models. Glutamate/nitric oxide, γ-aminobutyric acid, opioids, serotonin, dopamine, adenosine, cannabinoids, norepinephrine, vasopressin, neuropeptide Y, neurosteroids, and protein kinase C all modulate rapid tolerance. Most studies have evaluated the ability of pharmacological manipulations to block the development of rapid tolerance, but only a few studies have assessed their ability to reverse already established tolerance. Notably, only a few studies analyzed sex differences. Neglected areas of study include the incorporation of a key element of tolerance that involves opponent process-like neuroadaptations. Compared with alcohol drinking models, models of rapid tolerance are relatively shorter in duration and are temporally defined, which make them suitable for combining with a wide range of classic and modern research tools, such as pharmacology, optogenetics, calcium imaging, in vivo electrophysiology, and DREADDs, for in-depth studies of tolerance. We conclude that studies of the neurobiology of alcohol tolerance should be revisited with modern conceptualizations of addiction and modern neurobiological tools. This may contribute to our understanding of AUD and uncover potential targets that can attenuate hazardous alcohol drinking.


Assuntos
Alcoolismo/metabolismo , Encéfalo/efeitos dos fármacos , Tolerância a Medicamentos , Etanol/farmacologia , Neurobiologia/métodos , Consumo de Bebidas Alcoólicas/metabolismo , Alcoolismo/patologia , Animais , Comportamento Aditivo/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Etanol/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Humanos , Masculino , Camundongos , Norepinefrina/metabolismo , Ratos , Serotonina/metabolismo , Caracteres Sexuais
18.
Brain Res Bull ; 170: 98-105, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33592274

RESUMO

Prenatal opioids exposure negatively affects the neurobehavioral abilities of children born from dependence dams. Adolescent housing conditions can buffer the detrimental impacts of early life experiences or contradictory can worsen individual psychosocial functions. The present study investigated the effects of maternal morphine dependence and different rearing conditions on behaviors and protein expression in brain reward circuits of male pups. Female Wistar rats a week before conception, during pregnancy and lactation were injected twice daily with escalating doses of morphine or saline. On a postnatal day 21, male pups were weaned and subjected to three different environments for two months: standard (STD), isolated (ISO), or enriched environment (EE). The anxiety and drug-related reward were measured using elevated plus maze, open field test, and conditioned place preference. Western blotting was used to determine the protein level of ΔFosB and µ-opioid receptor proteins in the striatum and the midbrain of male offspring, respectively. Results showed that maternal morphine administration dramatically increased anxiety-like and morphine place preference behaviors in offspring. Also, ISO condition aggravated these behavioral outcomes. While, rearing in EE could attenuate anxiety and morphine conditioning in pups. At molecular levels, maternal morphine exposure and social isolation markedly increased both of ΔFosB and µ-opioid receptor proteins expression. However, rearing in the EE declined ΔFosB protein expression. Together, these findings help to elucidate long lasting impacts of early life morphine exposure and rearing environment on the behavioral and molecular profile of addicted individuals.


Assuntos
Comportamento Aditivo/metabolismo , Dependência de Morfina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Opioides mu/metabolismo , Recompensa , Meio Social , Isolamento Social , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Abrigo para Animais , Morfina/farmacologia , Ratos Wistar
19.
J Neuroinflammation ; 18(1): 56, 2021 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-33612110

RESUMO

Chronic use of drugs of abuse affects neuroimmune signaling; however, there are still many open questions regarding the interactions between neuroimmune mechanisms and substance use disorders (SUDs). Further, chronic use of drugs of abuse can induce glutamatergic changes in the brain, but the relationship between the glutamate system and neuroimmune signaling in addiction is not well understood. Therefore, the purpose of this review is to bring into focus the role of neuroimmune signaling and its interactions with the glutamate system following chronic drug use, and how this may guide pharmacotherapeutic treatment strategies for SUDs. In this review, we first describe neuroimmune mechanisms that may be linked to aberrant glutamate signaling in addiction. We focus specifically on the nuclear factor-kappa B (NF-κB) pathway, a potentially important neuroimmune mechanism that may be a key player in driving drug-seeking behavior. We highlight the importance of astroglial-microglial crosstalk, and how this interacts with known glutamatergic dysregulations in addiction. Then, we describe the importance of studying non-neuronal cells with unprecedented precision because understanding structure-function relationships in these cells is critical in understanding their role in addiction neurobiology. Here we propose a working model of neuroimmune-glutamate interactions that underlie drug use motivation, which we argue may aid strategies for small molecule drug development to treat substance use disorders. Together, the synthesis of this review shows that interactions between glutamate and neuroimmune signaling may play an important and understudied role in addiction processes and may be critical in developing more efficacious pharmacotherapies to treat SUDs.


Assuntos
Comportamento Aditivo/metabolismo , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Neuroimunomodulação/fisiologia , Plasticidade Neuronal/fisiologia , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Comportamento Aditivo/imunologia , Encéfalo/imunologia , Ácido Glutâmico/imunologia , Humanos , Microglia/imunologia , Microglia/metabolismo , Transdução de Sinais/fisiologia , Transtornos Relacionados ao Uso de Substâncias/imunologia
20.
Pharmacol Biochem Behav ; 203: 173117, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33561479

RESUMO

Preferences for novel environments (novelty-seeking) is a risk factor for addiction, with little known about its underlying circuitry. Exposure to drug cues facilitates addiction maintenance, leading us to hypothesize that exposure to a novel environment activates a shared neural circuitry. Stimulation of the D1 receptor in the prelimbic cortex increases responsivity to drug-associated environments. Here, we use D1 receptor overexpression in the prelimbic cortex to probe brain responses to novelty-preferences (in a free-choice paradigm) and cocaine-associated odors following place conditioning. These same cocaine-conditioned odors were used to study neural circuitry with Blood Oxygen Level Dependent (BOLD) activity. D1 overexpressing females had deactivated BOLD signals related to novelty-preferences within the insula cortex and amygdala and activation in the frontal cortex and dopamine cell bodies. BOLD responses to cocaine cues were also sensitive to D1. Control females demonstrated a place preference for cocaine environments with no significant BOLD response, while D1 overexpressing females demonstrated a place aversion and weak BOLD responses to cocaine-conditioned odor cues within the insula cortex. For comparison, we provide data from an earlier study with juvenile males overexpressing D1 that show a strong preference for cocaine and elevated BOLD responses. The results support the use of a pharmacological manipulation (e.g., D1 overexpression) to probe the neural circuitry downstream from the prelimbic cortex.


Assuntos
Cocaína/administração & dosagem , Sinais (Psicologia) , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento Exploratório/efeitos dos fármacos , Odorantes , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D1/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/psicologia , Animais , Comportamento Aditivo/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Feminino , Vetores Genéticos/administração & dosagem , Lentivirus/genética , Imageamento por Ressonância Magnética/métodos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/genética , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...