Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 431
Filtrar
1.
Int J Obes (Lond) ; 43(2): 344-354, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29453460

RESUMO

BACKGROUND/OBJECTIVES: Ghrelin, a stomach-derived hormone implicated in numerous behaviors including feeding, reward, stress, and addictive behaviors, acts by binding to the growth hormone secretagogue receptor (GHSR). Here, we present the development, verification, and initial characterization of a novel GHSR knockout (KO) Wistar rat model created with CRISPR genome editing. METHODS: Using CRISPR/Cas9, we developed a GHSR KO in a Wistar background. Loss of GHSR mRNA expression was histologically verified using RNAscope in wild-type (WT; n = 2) and KO (n = 2) rats. We tested the effects of intraperitoneal acyl-ghrelin administration on food consumption and plasma growth hormone (GH) concentrations in WT (n = 8) and KO (n = 8) rats. We also analyzed locomotion, food consumption, and body fat composition in these animals. Body weight was monitored from early development to adulthood. RESULTS: The RNAscope analysis revealed an abundance of GHSR mRNA expression in the hypothalamus, midbrain, and hippocampus in WTs, and no observed probe binding in KOs. Ghrelin administration increased plasma GH levels (p = 0.0067) and food consumption (p = 0.0448) in WT rats but not KOs. KO rats consumed less food overall at basal conditions and weighed significantly less compared with WTs throughout development (p = 0.0001). Compared with WTs, KOs presented higher concentrations of brown adipose tissue (BAT; p = 0.0322). CONCLUSIONS: We have verified GHSR deletion in our KO model using histological, physiological, neuroendocrinological, and behavioral measures. Our findings indicate that GHSR deletion in rats is not only associated with a lack of response to ghrelin, but also associated with decreases in daily food consumption and body growth, and increases in BAT. This GHSR KO Wistar rat model provides a novel tool for studying the role of the ghrelin system in obesity and in a wide range of medical and neuropsychiatric disorders.


Assuntos
Sistemas CRISPR-Cas/genética , Técnicas de Inativação de Genes/métodos , Receptores de Grelina/genética , Animais , Peso Corporal/genética , Química Encefálica/genética , Grelina/análise , Masculino , Ratos , Ratos Wistar
2.
Mol Psychiatry ; 23(6): 1466-1473, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28461696

RESUMO

Aldosterone regulates electrolyte and fluid homeostasis through binding to the mineralocorticoid receptors (MRs). Previous work provides evidence for a role of aldosterone in alcohol use disorders (AUDs). We tested the hypothesis that high functional activity of the mineralocorticoid endocrine pathway contributes to vulnerability for AUDs. In Study 1, we investigated the relationship between plasma aldosterone levels, ethanol self-administration and the expression of CYP11B2 and MR (NR3C2) genes in the prefrontal cortex area (PFC) and central nucleus of the amygdala (CeA) in monkeys. Aldosterone significantly increased after 6- and 12-month ethanol self-administration. NR3C2 expression in the CeA was negatively correlated to average ethanol intake during the 12 months. In Study 2, we measured Nr3c2 mRNA levels in the PFC and CeA of dependent and nondependent rats and the correlates with ethanol drinking during acute withdrawal. Low Nr3c2 expression levels in the CeA were significantly associated with increased anxiety-like behavior and compulsive-like drinking in dependent rats. In Study 3, the relationship between plasma aldosterone levels, alcohol drinking and craving was investigated in alcohol-dependent patients. Non-abstinent patients had significantly higher aldosterone levels than abstinent patients. Aldosterone levels positively correlated with the number of drinks consumed, craving and anxiety scores. These findings support a relationship between ethanol drinking and the aldosterone/MR pathway in three different species.


Assuntos
Alcoolismo/metabolismo , Aldosterona/metabolismo , Receptores de Mineralocorticoides/metabolismo , Adulto , Consumo de Bebidas Alcoólicas/genética , Alcoolismo/genética , Tonsila do Cerebelo/metabolismo , Animais , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Modelos Animais de Doenças , Etanol/metabolismo , Humanos , Macaca mulatta/metabolismo , Masculino , Mineralocorticoides/metabolismo , Córtex Pré-Frontal/metabolismo , Dados Preliminares , Ratos , Ratos Wistar , Receptores de Mineralocorticoides/genética , Autoadministração
3.
Mol Psychiatry ; 23(3): 629-638, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28348387

RESUMO

Adult neurogenesis in the dentate gyrus (DG) is strongly influenced by drug-taking behavior and may have a role in the etiology of drug-seeking behavior. However, mechanistic studies on the relationship of neurogenesis on drug seeking are limited. Outbred Wistar rats experienced extended access methamphetamine self-administration and individual differences in drug taking defined animals with higher preferred and lower preferred levels of drug intake. Forced abstinence from higher preferred levels of drug taking enhanced neurogenesis and neuronal activation of granule cell neurons (GCNs) in the DG and produced compulsive-like drug reinstatement. Systemic treatment with the drug Isoxazole-9 (a synthetic small molecule known to modulate neurogenesis in the adult rodent brain) during abstinence blocked compulsive-like context-driven methamphetamine reinstatement. Isoxazole-9 modulated neurogenesis, neuronal activation and structural plasticity of GCNs, and expression of synaptic proteins associated with learning and memory in the DG. These findings identify a subset of newly born GCNs within the DG that could directly contribute to drug-seeking behavior. Taken together, these results support a direct role for the importance of adult neurogenesis during abstinence in compulsive-like drug reinstatement.


Assuntos
Comportamento de Procura de Droga/efeitos dos fármacos , Isoxazóis/farmacologia , Neurogênese/efeitos dos fármacos , Tiofenos/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Individualidade , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Metanfetamina/efeitos adversos , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Recidiva , Autoadministração , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico
4.
Mol Psychiatry ; 23(9): 1900-1910, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28848234

RESUMO

Alcohol use disorder (AUD) is a common and chronic disorder with substantial effects on personal and public health. The underlying pathophysiology is poorly understood but strong evidence suggests significant roles of both genetic and epigenetic components. Given that alcohol affects many organ systems, we performed a cross-tissue and cross-phenotypic analysis of genome-wide methylomic variation in AUD using samples from 3 discovery, 4 replication, and 2 translational cohorts. We identified a differentially methylated region in the promoter of the proprotein convertase subtilisin/kexin 9 (PCSK9) gene that was associated with disease phenotypes. Biological validation showed that PCSK9 promoter methylation is conserved across tissues and positively correlated with expression. Replication in AUD datasets confirmed PCSK9 hypomethylation and a translational mouse model of AUD showed that alcohol exposure leads to PCSK9 downregulation. PCSK9 is primarily expressed in the liver and regulates low-density lipoprotein cholesterol (LDL-C). Our finding of alcohol-induced epigenetic regulation of PCSK9 represents one of the underlying mechanisms between the well-known effects of alcohol on lipid metabolism and cardiovascular risk, with light alcohol use generally being protective while chronic heavy use has detrimental health outcomes.


Assuntos
Alcoolismo/genética , Pró-Proteína Convertase 9/efeitos dos fármacos , Pró-Proteína Convertase 9/genética , Adulto , Alcoolismo/fisiopatologia , Animais , LDL-Colesterol/metabolismo , Metilação de DNA/genética , Epigênese Genética/genética , Epigenômica/métodos , Etanol/efeitos adversos , Etanol/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Humanos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Camundongos , Fenótipo , Regiões Promotoras Genéticas/genética , Pró-Proteína Convertase 9/fisiologia , Ratos , Ratos Wistar
5.
Nat Genet ; 24(4): 410-4, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742108

RESUMO

Corticotropin-releasing hormone (Crh) is a critical coordinator of the hypothalamic-pituitary-adrenal (HPA) axis. In response to stress, Crh released from the paraventricular nucleus (PVN) of the hypothalamus activates Crh receptors on anterior pituitary corticotropes, resulting in release of adrenocorticotropic hormone (Acth) into the bloodstream. Acth in turn activates Acth receptors in the adrenal cortex to increase synthesis and release of glucocorticoids. The receptors for Crh, Crhr1 and Crhr2, are found throughout the central nervous system and periphery. Crh has a higher affinity for Crhr1 than for Crhr2, and urocortin (Ucn), a Crh-related peptide, is thought to be the endogenous ligand for Crhr2 because it binds with almost 40-fold higher affinity than does Crh. Crhr1 and Crhr2 share approximately 71% amino acid sequence similarity and are distinct in their localization within the brain and peripheral tissues. We generated mice deficient for Crhr2 to determine the physiological role of this receptor. Crhr2-mutant mice are hypersensitive to stress and display increased anxiety-like behaviour. Mutant mice have normal basal feeding and weight gain, but decreased food intake following food deprivation. Intravenous Ucn produces no effect on mean arterial pressure in the mutant mice.


Assuntos
Ansiedade/genética , Comportamento Animal/fisiologia , Receptores de Hormônio Liberador da Corticotropina/deficiência , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Fisiológico/genética , Animais , Transtornos de Ansiedade/genética , Pressão Sanguínea/efeitos dos fármacos , Hormônio Liberador da Corticotropina/administração & dosagem , Hormônio Liberador da Corticotropina/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Privação de Alimentos/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Infusões Intravenosas , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Camundongos Mutantes , Nitroprussiato/farmacologia , Sistema Hipófise-Suprarrenal/fisiologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Estresse Fisiológico/fisiopatologia , Urocortinas , Vasodilatadores/farmacologia , Aumento de Peso/fisiologia
6.
Addict Neurosci ; 12022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37323812

RESUMO

Opioid use disorder (OUD) is defined as a compulsion to seek and take opioids, loss of control over intake and the development of a negative emotional state when access to opioids is denied. Using functional magnetic resonance imaging (fMRI) data in a rat model of OUD, we demonstrate that the escalation of heroin self-administration (SA) and the increased heroin SA following an injection of an opioid receptor antagonist (naloxone) are associated with changes in distinct brain circuits, centered on the cingulate cortex (Cg). Here, SA escalation score was negatively associated with changes in resting state functional connectivity (rsFC) between the Cg and the dorsal striatum. Conversely, increased heroin SA following naloxone injection, was associated with increased connectivity between the Cg and the extended amygdala and hypothalamus. Naloxone-induced increased SA was also positively associated with changes in the amplitude of low frequency fluctuations within the Cg, a measure of spontaneous neuronal activity. Characterizing the distinct brain circuit and behavior changes associated with different facets of addiction increases our understanding of OUD and may provide insight into addiction prevention and treatment.

7.
Sci Adv ; 6(32): eabc0413, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32821843

RESUMO

Intravenous drug self-administration is considered the "gold standard" model to investigate the neurobiology of drug addiction in rodents. However, its use in mice is limited by frequent complications of intravenous catheterization. Given the many advantages of using mice in biomedical research, we developed a noninvasive mouse model of opioid self-administration using vaporized fentanyl. Mice readily self-administered fentanyl vapor, titrated their drug intake, and exhibited addiction-like behaviors, including escalation of drug intake, somatic signs of withdrawal, drug intake despite punishment, and reinstatement of drug seeking. Electrophysiological recordings from ventral tegmental area dopamine neurons showed a lower amplitude of GABAB receptor-dependent currents during protracted abstinence from fentanyl vapor self-administration. This mouse model of fentanyl self-administration recapitulates key features of opioid addiction, overcomes limitations of the intravenous model, and allows investigation of the neurobiology of opioid addiction in unprecedented ways.


Assuntos
Fentanila , Transtornos Relacionados ao Uso de Opioides , Analgésicos Opioides , Animais , Comportamento de Procura de Droga , Camundongos , Autoadministração
8.
Science ; 282(5387): 298-300, 1998 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-9765157

RESUMO

Differential access to cocaine self-administration produced two patterns of drug intake in rats. With 1 hour of access per session, drug intake remained low and stable. In contrast, with 6 hours of access, drug intake gradually escalated over days. After escalation, drug consumption was characterized by an increased early drug loading and an upward shift in the cocaine dose-response function, suggesting an increase in hedonic set point. After 1 month of abstinence, escalation of cocaine intake was reinstated to a higher level than before. These findings may provide an animal model for studying the development of excessive drug intake and the basis of addiction.


Assuntos
Comportamento Aditivo , Transtornos Relacionados ao Uso de Cocaína/etiologia , Cocaína/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Masculino , Ratos , Ratos Wistar , Reforço Psicológico , Fatores de Tempo
9.
Science ; 213(4505): 357-9, 1981 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-7244622

RESUMO

Systemic administration of the neuroleptic drug alpha-flupenthixol attenuated lever-pressing behavior in rats responding for rewarding brain stimulation. The magnitude of this attenuation was dose-dependent and resembled the effects of reward reduction and termination. However, when the operant response requirements of the same rats were changed to nose poking, identical drug treatments produced relatively little attenuation in performance. These data do not support the belief that neuroleptics produce a general state of anhedonia. Rather, the apparent suppression of reinforced behaviors depends at least in part on the kinetic requirements of the response.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Flupentixol/farmacologia , Recompensa , Tioxantenos/farmacologia , Animais , Encéfalo/fisiologia , Relação Dose-Resposta a Droga , Eletrochoque , Masculino , Ratos
10.
Science ; 242(4879): 715-23, 1988 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-2903550

RESUMO

The molecular and cellular actions of three classes of abused drugs--opiates, psychostimulants, and ethanol--are reviewed in the context of behavioral studies of drug dependence. The immediate effects of drugs are compared to those observed after long-term exposure. A neurobiological basis for drug dependence is proposed from the linkage between the cellular and behavioral effects of these drugs.


Assuntos
Encéfalo/fisiopatologia , Cocaína/farmacologia , Etanol/farmacologia , Ópio/farmacologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Animais , Comportamento Animal/efeitos dos fármacos , Tolerância a Medicamentos , Humanos , Neurotransmissores/fisiologia , Reforço Psicológico
11.
Science ; 260(5115): 1814-6, 1993 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-8099761

RESUMO

The reinforcing properties of cocaine are probably mediated by the mesocorticolimbic dopamine pathways in the central nervous system, but not all of the dopamine receptor subtypes involved in cocaine's reinforcing actions have been clearly identified. Recently, the D-3 receptor has been cloned, and its distribution in the brain has been found to be relatively restricted to limbic projections of the midbrain dopamine system. The D-3-selective compounds 7-hydroxy-N,N-di-n-propyl-2-aminotetralin (7-OHDPAT) and quinpirole potently decreased cocaine self-administration in the rat at doses that were not by themselves reinforcing. Moreover, three dopamine receptor agonists had affinities for binding to the D-3 receptor that correlated highly with their relative potencies in decreasing cocaine self-administration. The D-3 receptor may be involved in the reinforcing effects of cocaine and may be a useful target for the development of new pharmacotherapies for cocaine abuse.


Assuntos
Cocaína/administração & dosagem , Dopaminérgicos/farmacologia , Receptores de Dopamina D2 , Receptores Dopaminérgicos/metabolismo , Animais , Apomorfina/farmacologia , Relação Dose-Resposta a Droga , Ergolinas/farmacologia , Masculino , Quimpirol , Ratos , Ratos Wistar , Receptores de Dopamina D3 , Reforço Psicológico , Autoadministração , Tetra-Hidronaftalenos/farmacologia
12.
Science ; 278(5335): 52-8, 1997 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-9311926

RESUMO

Understanding the neurobiological mechanisms of addiction requires an integration of basic neuroscience with social psychology, experimental psychology, and psychiatry. Addiction is presented as a cycle of spiralling dysregulation of brain reward systems that progressively increases, resulting in compulsive drug use and a loss of control over drug-taking. Sensitization and counteradaptation are hypothesized to contribute to this hedonic homeostatic dysregulation, and the neurobiological mechanisms involved, such as the mesolimbic dopamine system, opioid peptidergic systems, and brain and hormonal stress systems, are beginning to be characterized. This framework provides a realistic approach to identifying the neurobiological factors that produce vulnerability to addiction and to relapse in individuals with a history of addiction.


Assuntos
Comportamento Aditivo , Drogas Ilícitas/efeitos adversos , Transtornos Relacionados ao Uso de Substâncias , Adaptação Fisiológica , Tonsila do Cerebelo/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Dopamina/fisiologia , Homeostase , Humanos , Neurotransmissores/fisiologia , Recidiva , Reforço Psicológico , Recompensa , Síndrome de Abstinência a Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Transtornos Relacionados ao Uso de Substâncias/terapia
13.
Science ; 194(4265): 637-9, 1976 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-982032

RESUMO

A paper clip applied to the tails of rats induced gnawing and eating, which decreased in latency and increased in duration with experience. With sustained pressure to the tail, rats learned a new habit in order to gain access to wood chips on which to gnaw. That these are also properties of behavior elicited by electrical brain stimulation suggests that both manipulations may act through the same mechanism. These results support the hypothesis that a nonspecific arousing stimulus can be a sufficient condition for establishing learned habits.


Assuntos
Nível de Alerta/fisiologia , Comportamento Animal/fisiologia , Aprendizagem/fisiologia , Dor/fisiopatologia , Animais , Feminino , Hipotálamo/fisiologia , Ratos , Cauda
14.
Science ; 259(5094): 528-31, 1993 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-8380941

RESUMO

The function of neuropeptide Y, one of the most abundant peptide transmitters of the mammalian brain, remains unclear because of a lack of specific receptor antagonists. An antisense oligodeoxynucleotide corresponding to the NH2-terminus of the rat Y1 receptor was constructed and added to cultures of rat cortical neurons. This treatment resulted in a reduced density of Y1 (but not Y2) receptors and diminished the decrease in adenosine 3',5'-monophosphate (cAMP) usually seen after Y1 receptor activation. Repeated injection of the same oligodeoxynucleotide into the lateral cerebral ventricle of rats was followed by a similar reduction of cortical Y1 (but not Y2) receptors. Such antisense-treated animals displayed behavioral signs of anxiety. Thus, specific inhibition of neurotransmitter receptor expression can be accomplished in the living brain and demonstrates that altered central neuropeptide Y transmission produces an anxiety-like state.


Assuntos
Ansiedade , Córtex Cerebral/fisiologia , Neurônios/fisiologia , Neuropeptídeo Y/fisiologia , Oligonucleotídeos Antissenso/farmacologia , Receptores de Neuropeptídeo Y/efeitos dos fármacos , Receptores de Neuropeptídeo Y/genética , Animais , Sequência de Bases , Células Cultivadas , AMP Cíclico/metabolismo , Regulação para Baixo , Embrião de Mamíferos , Aprendizagem , Masculino , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Oligodesoxirribonucleotídeos , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Receptores de Neuropeptídeo Y/metabolismo
15.
Science ; 276(5321): 2050-4, 1997 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-9197270

RESUMO

Corticotropin-releasing factor (CRF) has been implicated in the mediation of the stress-like and negative affective consequences of withdrawal from drugs of abuse, such as alcohol, cocaine, and opiates. This study sought to determine whether brain CRF systems also have a role in cannabinoid dependence. Rats were treated daily for 2 weeks with the potent synthetic cannabinoid HU-210. Withdrawal, induced by the cannabinoid antagonist SR 141716A, was accompanied by a marked elevation in extracellular CRF concentration and a distinct pattern of Fos activation in the central nucleus of the amygdala. Maximal increases in CRF corresponded to the time when behavioral signs resulting from cannabinoid withdrawal were at a maximum. These data suggest that long-term cannabinoid administration alters CRF function in the limbic system of the brain, in a manner similar to that observed with other drugs of abuse, and also induces neuroadaptive processes that may result in future vulnerability to drug dependence.


Assuntos
Tonsila do Cerebelo/metabolismo , Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Dronabinol/análogos & derivados , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Ansiedade/induzido quimicamente , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Corticosterona/sangue , Dronabinol/efeitos adversos , Dronabinol/antagonistas & inibidores , Dronabinol/farmacologia , Masculino , Microdiálise , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-fos/análise , Pirazóis/farmacologia , Ratos , Ratos Wistar , Receptores de Canabinoides , Receptores de Droga/antagonistas & inibidores , Rimonabanto , Síndrome de Abstinência a Substâncias/metabolismo
16.
Science ; 273(5281): 1561-4, 1996 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-8703220

RESUMO

The neuropeptide corticotropin-releasing factor (CRF) is well known to act on the central nervous system in ways that mimic stress and result in decreases in exploration, increases in sympathetic activity, decreases in parasympathetic outflow, and decreases in appetitive behavior. Urocortin, a neuropeptide related to CRF, binds with high affinity to the CRF2 receptor, is more potent than CRF in suppressing appetite, but is less potent than CRF in producing anxiety-like effects and activation. Doses as low as 10 nanograms injected intracerebroventricularly were effective in decreasing food intake in food-deprived and free-feeding rats. These results suggest that urocortin may be an endogenous CRF-like factor in the brain responsible for the effects of stress on appetite.


Assuntos
Depressores do Apetite/farmacologia , Apetite/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Animais , Depressores do Apetite/administração & dosagem , Depressores do Apetite/metabolismo , Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Hormônio Liberador da Corticotropina/administração & dosagem , Hormônio Liberador da Corticotropina/metabolismo , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Jejum , Injeções Intraventriculares , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Urocortinas , Urotensinas/farmacologia
17.
Pharmacopsychiatry ; 42 Suppl 1: S32-41, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19434554

RESUMO

Drug addiction is conceptualized as chronic, relapsing compulsive use of drugs with significant dysregulation of brain hedonic systems. Compulsive drug use is accompanied by decreased function of brain substrates for drug positive reinforcement and recruitment of brain substrates mediating the negative reinforcement of motivational withdrawal. The neural substrates for motivational withdrawal ("dark side" of addiction) involve recruitment of elements of the extended amygdala and the brain stress systems, including corticotropin-releasing factor and norepinephrine. These changes, combined with decreased reward function, are hypothesized to persist in the form of an allostatic state that forms a powerful motivational background for relapse. Relapse also involves a key role for the basolateral amygdala in mediating the motivational effects of stimuli previously paired with drug seeking and drug motivational withdrawal. The basolateral amygdala has a key role in mediating emotional memories in general. The hypothesis argued here is that brain stress systems activated by the motivational consequences of drug withdrawal can not only form the basis for negative reinforcement that drives drug seeking, but also potentiate associative mechanisms that perpetuate the emotional state and help drive the allostatic state of addiction.


Assuntos
Drogas Ilícitas/farmacologia , Memória/fisiologia , Vias Neurais/fisiopatologia , Reforço Psicológico , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Biologia de Sistemas , Alostase/fisiologia , Animais , Medo/fisiologia , Humanos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Dor/tratamento farmacológico , Recompensa , Síndrome de Abstinência a Substâncias/fisiopatologia , Síndrome de Abstinência a Substâncias/psicologia
18.
Neuron ; 20(6): 1093-102, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9655498

RESUMO

Corticotropin releasing factor (CRF) is a major integrator of adaptive responses to stress. Two biochemically and pharmacologically distinct CRF receptor subtypes (CRFR1 and CRFR2) have been described. We have generated mice null for the CRFR1 gene to elucidate the specific developmental and physiological roles of CRF receptor mediated pathways. Behavioral analyses revealed that mice lacking CRFR1 displayed markedly reduced anxiety. Mutant mice also failed to exhibit the characteristic hormonal response to stress due to a disruption of the hypothalamic-pituitary-adrenal (HPA) axis. Homozygous mutant mice derived from crossing heterozygotes displayed low plasma corticosterone concentrations resulting from a marked agenesis of the zona fasciculata region of the adrenal gland. The offspring from homozygote crosses died within 48 hr after birth due to a pronounced lung dysplasia. The adrenal agenesis in mutant animals was attributed to insufficient adrenocorticotropic hormone (ACTH) production during the neonatal period and was rescued by ACTH replacement. These results suggest that CRFR1 plays an important role both in the development of a functional HPA axis and in mediating behavioral changes associated with anxiety.


Assuntos
Ansiedade/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Sistemas Neurossecretores/crescimento & desenvolvimento , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Fisiológico/genética , Adaptação Fisiológica/fisiologia , Doenças das Glândulas Suprarrenais/tratamento farmacológico , Doenças das Glândulas Suprarrenais/genética , Doenças das Glândulas Suprarrenais/mortalidade , Glândulas Suprarrenais/crescimento & desenvolvimento , Glândulas Suprarrenais/patologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Ansiedade/metabolismo , Comportamento Animal/fisiologia , Quimera , Corticosterona/farmacologia , Feminino , Homozigoto , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Mutação/fisiologia , Sistemas Neurossecretores/patologia , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/metabolismo , Hipófise/crescimento & desenvolvimento , Estresse Fisiológico/metabolismo , Análise de Sobrevida
19.
Alcohol Clin Exp Res ; 32(9): 1688-96, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18482158

RESUMO

BACKGROUND: Chronic intermittent alcohol vapor exposure and selective breeding procedures have been used separately for many years to model specific aspects of alcohol dependence. The purpose of the present investigation was to combine these 2 approaches by exposing alcohol-preferring (P) rats to chronic intermittent alcohol vapor for extended periods of time and then testing them for operant alcohol responding in parallel with a group of outbred Wistar rats at multiple time points following the termination of vapor exposure. METHODS: P rats (n = 20) and Wistar rats (n = 18) were trained to respond for 10% (w/v) ethanol in an operant situation, then divided into groups matched for intake levels. Animals were then exposed to chronic intermittent alcohol vapor (14 hours ON/10 hours OFF) or air for 8 weeks. Rats were then tested for operant alcohol responding under various conditions and at multiple time points during alcohol withdrawal (6 hours) and protracted abstinence (1 to 15 days). RESULTS: Chronic alcohol vapor exposure produced similar increases in operant alcohol responding in P rats and Wistar rats during acute withdrawal and protracted abstinence. CONCLUSIONS: These results illustrate the separate and combined effects of genetic selection for high alcohol preference and dependence on alcohol drinking behavior. Furthermore, these results confirm past findings that dependent rats consume more alcohol than nondependent controls well into abstinence following extended periods of alcohol vapor exposure.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Alcoolismo/genética , Alcoolismo/fisiopatologia , Comportamento Animal/fisiologia , Comportamento de Ingestão de Líquido/fisiologia , Animais , Modelos Animais de Doenças , Etanol/administração & dosagem , Etanol/sangue , Masculino , Nebulizadores e Vaporizadores , Ratos , Ratos Endogâmicos , Ratos Wistar , Autoadministração , Temperança
20.
Trends Neurosci ; 15(5): 186-91, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1377426

RESUMO

Drug withdrawal is an integral part of most types of dependence and, to a large extent, opiate withdrawal has been considered the prototypic, classic measure of opiate dependence. The opiate withdrawal syndrome is characterized by multiple behavioral and physiological signs such as behavioral activation, ptosis, diarrhea, 'wet dog' shakes and motivational dysfunction, which may be represented in the CNS at multiple sites. It seems that the activating effects associated with the opiate withdrawal syndrome may be mediated by the nucleus locus coeruleus. Other signs such as wet dog shakes may involve sites in the hypothalamus important for temperature regulation. Certain other signs such as diarrhea and lacrimation may be dependent on peripheral opiate receptors. The motivational aspects of opiate withdrawal as demonstrated by the aversive stimulus effects or negative reinforcing effects (e.g. disrupted lever-pressing for food and place aversions) may involve those elements of the nucleus accumbens that are known to be important for the acute reinforcing effects of opiates in nondependent rats. Evidence exists at the cellular and molecular level for both 'within-system' and 'between-system' adaptations to dependence. Elucidation of the neural networks, cellular mechanisms and molecular elements involved in opiate withdrawal may provide not only a model for our understanding of the adaptive processes associated with drug dependence but also of those associated with other chronic insults to CNS function.


Assuntos
Entorpecentes/farmacologia , Neurônios/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA