Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Psychiatry ; 86(9): 657-668, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31255250

RESUMO

BACKGROUND: Despite the twofold higher prevalence of major depressive and posttraumatic stress disorders in women compared with men, most clinical and preclinical studies have focused on male subjects. We used an ethological murine model to study several cardinal symptoms of affective disorders in the female targets of female aggression. METHODS: Intact Swiss Webster (CFW) female resident mice were housed with castrated male mice and tested for aggression toward female intruders. For 10 days, aggressive CFW female residents defeated C57BL/6J (B6) female intruders during 5-minute encounters. Measures of corticosterone, c-Fos activation in hypothalamic and limbic structures, and species-typical behaviors were collected from defeated and control females. Ketamine (20 mg/kg) was tested for its potential to reverse stress-induced social deficits. RESULTS: Housed with a castrated male mouse, most intact resident CFW females readily attacked unfamiliar B6 female intruders, inflicting >40 bites in a 5-minute encounter. Compared with controls, defeated B6 females exhibited elevated plasma corticosterone and increased c-Fos activation in the medial amygdala, ventral lateral septum, ventromedial hypothalamus, and hypothalamic paraventricular nucleus. Chronically defeated females also showed vigilance-like behavior and deficits in social interactions, novel object investigation, and nesting. The duration of social interactions increased 24 hours after chronically defeated female mice received a systemic dose of ketamine. CONCLUSIONS: These findings demonstrate that CFW female mice living with male conspecifics can be used as aggressive residents in an ethological model of female social defeat stress. These novel behavioral methods will encourage further studies of sex-specific neural, physiological, and behavioral adaptations to chronic stress and the biological bases for interfemale aggression.


Assuntos
Agressão , Comportamento Animal , Encéfalo/fisiologia , Genes fos , Estresse Psicológico , Animais , Corticosterona/sangue , Modelos Animais de Doenças , Feminino , Genes Precoces , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais , Comportamento Social
2.
Psychopharmacology (Berl) ; 236(7): 2027-2037, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30798402

RESUMO

RATIONALE: Stressful life experiences can persistently increase the motivation for, and consumption of, intensely rewarding stimuli, like cocaine, over time. In rodents, intermittent versus continuous exposure to social stress engenders opposing changes to reward-related behavior, as measured by consumption of sucrose and cocaine. OBJECTIVE: The present study examines if the effects of intermittent versus continuous social stress on cocaine self-administration in mice parallel those seen in rats. METHODS: Both forms of social stress involve a brief daily physical confrontation with an aggressive resident for 10 consecutive days. Continuous social stress involves constant visual and olfactory exposure to an aggressive resident via habitation in a protected portion of the resident's home cage, while exposure to an aggressive resident during intermittent social stress is limited to a single, physical encounter per day. Implementing a femoral vein catheterization method for the first time in mice, we determined divergent changes to intravenous cocaine self-administration. RESULTS: Modestly increased cocaine self-administration after intermittent social stress was confirmed. In a subset of animals, continuous social stress in mice substantially increased cocaine self-administration and sucrose intake. By stark contrast, another subpopulation had substantial attenuation of cocaine self-administration and sucrose intake after continuous social stress. CONCLUSIONS: Bimodal divergence in responding for rewarding stimuli including cocaine after social stress experience likely reflects two opposing forms of coping to continuous social stress that promote either a sensitization or attenuation of reward-seeking.


Assuntos
Agressão/psicologia , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Relações Interpessoais , Estresse Psicológico/psicologia , Agressão/efeitos dos fármacos , Agressão/fisiologia , Animais , Transtornos Relacionados ao Uso de Cocaína/psicologia , Feminino , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Autoadministração , Sacarose/administração & dosagem
3.
Front Behav Neurosci ; 12: 206, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271332

RESUMO

Alcohol drinking, in some individuals, culminates in pathologically aggressive and violent behaviors. Alcohol can escalate the urge to fight, despite causing disruptions in fighting performance. When orally administered under several dosing conditions the current study examined in a mouse model if repeated alcohol escalates the motivation to fight, the execution of fighting performance, or both. Specifically, seven daily administrations of alcohol (0, 1.8, or 2.2 g/kg) determined if changes in the motivation to initiate aggressive acts occur with, or without, shifts in the severity of fighting behavior. Responding under the control of a fixed interval (FI) schedule for aggression reinforcements across the initial daily sessions indicated the development of tolerance to alcohol's sedative effect. By day 7, alcohol augmented FI response rates for aggression rewards. While alcohol escalated the motivation to fight, fighting performance remained suppressed across the entire 7 days. Augmented FI responding for aggression rewards in response to a low dose of alcohol (1.0 g/kg) proved to be persistent, as we observed sensitized rates of responding for more than a month after alcohol pretreatment. In addition, this sensitization of motivated aggression did not occur with a general enhancement of motor activity. Antagonism of NMDA or AMPA receptors with ketamine, dizocilpine, or NBQX during later challenges with alcohol were largely serenic without having any notable impact on the expression of alcohol-escalated rates of FI responding. The current dissociation of appetitive and performance measures indicates that discrete neural mechanisms controlling aggressive arousal can be distinctly sensitized by alcohol.

4.
Psychopharmacology (Berl) ; 235(6): 1807-1820, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29696309

RESUMO

RATIONALE: Episodic bouts of social stress can precede the initiation, escalation, or relapse to disordered alcohol intake. Social stress may engender neuroadaptations in the hypothalamic-pituitary-adrenal (HPA) axis and in extrahypothalamic stress circuitry to promote the escalation of alcohol intake. OBJECTIVES: We aimed to (1) confirm a pattern of escalated drinking in socially defeated mice and to (2) test drugs that target distinct aspects of the HPA axis and extrahypothalamic neural substrates for their effectiveness in reducing murine, stress-escalated drinking. METHODS: Male C57BL/6J (B6) mice were socially defeated by resident Swiss-derived males for ten consecutive days receiving 30 bites/day. Ten days after the final defeat, cohorts of B6 mice received continuous or intermittent access to 20% EtOH (w/v) and water. After 4 weeks of drinking, mice were injected with weekly, systemic doses of the CRF-R1 antagonist, CP376395; the glucocorticoid receptor antagonist, mifepristone; the 11-beta-hydroxylase inhibitor, metyrapone; or the 5-alpha-reductase inhibitor, finasteride. RESULTS: Prior to drug treatments, defeated mice reliably consumed more EtOH than non-defeated controls, and mice given alcohol intermittently consumed more EtOH than those with continuous access. CP376395 (17-30 mg/kg) reduced continuous, but not intermittent EtOH intake (g/kg) in socially defeated mice. Mifepristone (100 mg/kg), however, increased drinking by defeated mice with intermittent access to alcohol while reducing drinking during continuous access. When administered finasteride (100 mg/kg) or metyrapone (50 mg/kg), all mice reduced their EtOH intake while increasing their water consumption. CONCLUSIONS: Mice with a history of episodic social defeat stress were selectively sensitive to the effects of CRF-R1 antagonism, suggesting that CRF-R1 may be a potential target for treating alcohol use disorders in individuals who escalate their drinking after exposure to repeated bouts of psychosocial stress. Future studies will clarify how social defeat stress may alter the expression of extrahypothalamic CRF-R1 and glucocorticoid receptors.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Consumo de Bebidas Alcoólicas/psicologia , Aminopiridinas/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/psicologia , Animais , Relação Dose-Resposta a Droga , Etanol/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Comportamento Social
5.
Neuropsychopharmacology ; 43(6): 1224-1234, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29052618

RESUMO

Alcohol is associated with nearly half of all violent crimes committed in the United States; yet, a potential neural basis for this type of pathological aggression remains elusive. Alcohol may act on N-methyl-d-aspartate receptors (NMDARs) within cortical circuits to impede processing and to promote aggression. Here, male mice were characterized as alcohol-heightened (AHAs) or alcohol non-heightened aggressors (ANAs) during resident-intruder confrontations after self-administering 1.0 g/kg alcohol (6% w/v) or water. Alcohol produced a pathological-like pattern of aggression in AHAs; these mice shifted their bites to more vulnerable locations on the body of a submissive animal, including the anterior back and ventrum after consuming alcohol. In addition, through immunoblotting, we found that AHAs overexpressed the NMDAR GluN2D subunit in the prefrontal cortex (PFC) as compared to ANAs while the two phenotypes expressed similar levels of GluN1, GluN2A and GluN2B. After identifying several behavioral and molecular characteristics that distinguish AHAs from ANAs, we tested additional mice for their aggression following preferential antagonism of GluN2D-containing NMDARs. In these experiments, groups of AHAs and ANAs self-administered 1.0 g/kg alcohol (6% w/v) or water before receiving intraperitoneal (i.p.) doses of ketamine or memantine, or infusions of memantine directly into the prelimbic (PLmPFC) or infralimbic medial PFC (ILmPFC). Moderate doses of IP ketamine, IP memantine, or intra-PLmPFC memantine increased aggression in AHAs, but only in the absence of alcohol. Prior alcohol intake blocked the pro-aggressive effects of ketamine or memantine. In contrast, only memantine, administered systemically or intra-PLmPFC, interacted with prior alcohol intake to escalate aggression in ANAs. Intra-ILmPFC memantine had no effect on aggression in either AHAs or ANAs. In sum, this work illustrates a potential role of GluN2D-containing NMDARs in the PLmPFC in alcohol-heightened aggression. GluN2D-containing NMDARs are highly expressed on cortical parvalbumin-containing interneurons, suggesting that, in a subset of individuals, alcohol may functionally alter signal integration within cortical microcircuits to dysregulate threat reactivity and promote aggression. This work suggests that targeting GluN2D-NMDARs may be of use in reducing the impact of alcohol-related violence in the human population.


Assuntos
Agressão/efeitos dos fármacos , Transtornos Relacionados ao Uso de Álcool/metabolismo , Depressores do Sistema Nervoso Central/efeitos adversos , Etanol/efeitos adversos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Agressão/fisiologia , Transtornos Relacionados ao Uso de Álcool/psicologia , Animais , Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Feminino , Masculino , Camundongos , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Autoadministração , Territorialidade
6.
Psychopharmacology (Berl) ; 234(18): 2823-2836, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28725939

RESUMO

RATIONALE: Exposure to intermittent social defeat stress elicits corticotropin releasing factor (CRF) release into the VTA and induces long-term modulation of mesocorticolimbic dopamine activity in rats. These adaptations are associated with an intense cocaine-taking phenotype, which is prevented by CRF receptor antagonists. OBJECTIVE: The present studies examine whether infusion of CRF into the VTA is sufficient to escalate cocaine-taking behavior, in the absence of social defeat experience. Additionally, we aimed to characterize changes in cocaine valuation that may promote binge-like cocaine intake. METHODS: Male Long-Evans rats were microinjected into the VTA with CRF (50 or 500 ng/side), vehicle, or subjected to social defeat stress, intermittently over 10 days. Animals were then trained to self-administer IV cocaine (FR5). Economic demand for cocaine was evaluated using a within-session behavioral-economics threshold procedure, which was followed by a 24-h extended access "binge." RESULTS: Rats that experienced social defeat or received intra-VTA CRF microinfusions (50 ng) both took significantly more cocaine than controls over the 24-h binge but showed distinct patterns of intake. Behavioral economic analysis revealed that individual demand for cocaine strongly predicts binge-like consumption, and demand elasticity (i.e. α) is augmented by intra-VTA CRF, but not by social defeat. The effects of CRF on cocaine-taking were also prevented by intra-VTA pretreatment with CP376395, but not Astressin-2B. CONCLUSIONS: Repeated infusion of CRF into the VTA persistently alters cocaine valuation and intensifies binge-like drug intake in a CRF-R1-dependent manner. Conversely, the persistent pattern of cocaine bingeing induced by social defeat stress may suggest impaired inhibitory control, independent of reward valuation.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/prevenção & controle , Transtornos Relacionados ao Uso de Cocaína/psicologia , Cocaína/administração & dosagem , Hormônio Liberador da Corticotropina/administração & dosagem , Estresse Psicológico/psicologia , Área Tegmentar Ventral/efeitos dos fármacos , Aminopiridinas/administração & dosagem , Animais , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/metabolismo , Infusões Intraventriculares , Masculino , Ratos , Ratos Long-Evans , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Autoadministração , Estresse Psicológico/metabolismo , Área Tegmentar Ventral/metabolismo
7.
Psychopharmacology (Berl) ; 234(18): 2813-2821, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28698920

RESUMO

BACKGROUND: A history of brief intermittent social defeat stress can escalate cocaine self-administration and induce long-term adaptations in the mesolimbic dopamine system. Extra-hypothalamic corticotrophin releasing factor (CRF) has been shown to be closely associated with stress-induced escalation of drug use. How repeated stress modulates CRF release in the ventral tegmental area (VTA) and the roles of CRF receptors during different phases of stress-induced cocaine self-administration remain to be defined. OBJECTIVE: The current study examines the roles of CRF and CRF receptor 1 (CRFR1) in escalated intravenous cocaine self-administration after exposure to social defeat stress in mice. METHODS AND RESULTS: First, CRFR1 antagonist (CP 376,395, 15 mg/kg, i.p.) given 30 min prior to each social defeat episode prevented later escalated cocaine self-administration. When CP 376,395 (5 and 15 mg/kg, i.p.) was administered 10 days after the last episode of social stress, the escalation of cocaine intake was dose-dependently reversed. Moreover, socially defeated mice showed increased CRF release in the VTA compared to controls. To further explore the role of CRFR1, CP 376,395 (0.5 and 1 µg/0.2 µl) was infused directly into the VTA before the cocaine self-administration session. Intra-VTA antagonism of CRFR1 was sufficient to reverse social defeat stress-escalated cocaine self-administration. CONCLUSION: These findings suggest that CRF and CRFR1 exert multiple roles in the response to social stress that are relevant to escalated cocaine self-administration.


Assuntos
Cocaína/administração & dosagem , Relações Interpessoais , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Estresse Psicológico/psicologia , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/metabolismo , Masculino , Camundongos , Microdiálise/métodos , Distribuição Aleatória , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Autoadministração , Área Tegmentar Ventral/metabolismo
8.
Alcohol Clin Exp Res ; 40(11): 2445-2455, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27717041

RESUMO

BACKGROUND: Alcohol use disorders are associated with single-nucleotide polymorphisms in GABRA2, the gene encoding the GABAA receptor α2-subunit in humans. Deficient GABAergic functioning is linked to impulse control disorders, intermittent explosive disorder, and to drug abuse and dependence, yet it remains unclear whether α2-containing GABAA receptor sensitivity to endogenous ligands is involved in excessive alcohol drinking. METHODS: Male wild-type (Wt) C57BL/6J and point-mutated mice rendered insensitive to GABAergic modulation by benzodiazepines (BZD; H101R), allopregnanolone (ALLO) or tetrahydrodeoxycorticosterone (THDOC; Q241M), or high concentrations of ethanol (EtOH) (S270H/L277A) at α2-containing GABAA receptors were assessed for their binge-like, moderate, or escalated chronic drinking using drinking in the dark, continuous access (CA) and intermittent access (IA) to alcohol protocols, respectively. Social approach by mutant and Wt mice in forced alcohol abstinence was compared to approach by EtOH-naïve controls. Social deficits in forced abstinence were treated with allopregnanolone (0, 3.0, 10.0 mg/kg, intraperitoneal [i.p.]) or midazolam (0, 0.56, 1.0 mg/kg, i.p.). RESULTS: Mice with BZD-insensitive α2-containing GABAA receptors (H101R) escalated their binge-like drinking. Mutants harboring the Q241M point substitution in Gabra2 showed blunted chronic intake in the CA and IA protocols. S270H/L277A mutants consumed excessive amounts of alcohol but, unlike wild-types, they did not show forced abstinence-induced social deficits. CONCLUSIONS: These findings suggest a role for: (i) H101 in species-typical binge-like drinking, (ii) Q241 in escalated chronic drinking, and (iii) S270 and/or L277 in the development of forced abstinence-associated social deficits. Clinical findings report reduced BZD-binding sites in the cortex of dependent patients; the present findings suggest a specific role for BZD-sensitive α2-containing receptors. In addition, amino acid residue 241 in Gabra2 is necessary for positive modulation and activation of GABAA receptors by ALLO and THDOC; we postulate that neurosteroid action on α2-containing receptor may be necessary for escalated chronic EtOH intake.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/genética , Receptores de GABA-A/genética , Abstinência de Álcool/psicologia , Animais , Feminino , Masculino , Camundongos Endogâmicos C57BL , Mutação Puntual , Quinina , Distribuição Aleatória , Comportamento Social , Sacarose
9.
Psychopharmacology (Berl) ; 233(17): 3173-86, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27376946

RESUMO

RATIONALE: Intermittent social defeat stress engenders persistent neuroadaptations and can result in later increased cocaine taking and seeking. However, there are individual differences in stress-escalated cocaine self-administration behavior, which may be a direct result of individual differences in the manner in which rats experience social defeat stress. OBJECTIVE: The present study dissected the discrete behavioral phases of social defeat and analyzed which behavioral characteristics may be predictive of subsequent cocaine self-administration. METHODS: Male Long-Evans rats underwent nine intermittent social defeat episodes over 21 days in a three-compartment apparatus permitting approach to and escape from a confrontation with an aggressive resident rat. Rats then self-administered intravenous cocaine, which culminated in a 24-h unlimited access "binge." Behaviors during social defeat and cocaine self-administration were evaluated by principal component analysis (PCA). RESULTS: PCA revealed that the latency to enter the threatening environment was highly predictive of later cocaine self-administration during the 24-h binge. This behavior was not associated with other cocaine-predictive traits, such as reactivity to novelty in an open field, saccharin preference, and motor impulsivity. Additionally, there was no effect of latency to enter a threatening environment on physiological measures of stress, including plasma corticosterone and corticotropin releasing factor (CRF) in the extended amygdala. However, latency to enter the threatening environment was negatively correlated with brain-derived neurotropic factor (BDNF) and its receptor, tyrosine kinase B (TrkB) in the hippocampus. CONCLUSION: These data suggest that latency to enter a threatening environment is a novel behavioral characteristic predictive of later cocaine self-administration.


Assuntos
Comportamento Animal , Comportamento de Escolha , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento Social , Estresse Psicológico/psicologia , Agressão , Animais , Transtornos Relacionados ao Uso de Cocaína , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Individualidade , Masculino , Análise de Componente Principal , Ratos , Ratos Long-Evans , Sacarina/administração & dosagem , Autoadministração , Estresse Psicológico/metabolismo , Edulcorantes/administração & dosagem
10.
Psychopharmacology (Berl) ; 233(14): 2727-36, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27251131

RESUMO

BACKGROUND: Activation of corticotropin-releasing factor type 1 receptors (CRF-R1) in the ventral tegmental area (VTA) represents a critical mechanism for social defeat to escalate cocaine self-administration in adult rats. OBJECTIVE: We determined the acute effect of a CRF-R1 antagonist (CP376395) microinfusion into the VTA prior to each episode of social defeat in adolescent rats and determined whether this drug treatment could prevent later escalation of cocaine taking in early adulthood. METHODS: Rats were implanted with bilateral cannulae aimed at the VTA 5 days before the first social defeat. Bilateral microinfusion of CP376395 (500 ng/side) or vehicle occurred 20 min before each episode of social defeat on postnatal days (P) 35, 38, 41, and 44. Behavior was quantified on P35 and P44. On P57, rats were implanted with intra-jugular catheters, and subsequent cocaine self-administration was analyzed. RESULTS: CP376395-treated adolescent rats walked less and were attacked more slowly but were socially investigated more than vehicle-treated adolescents. Vehicle-treated rats showed increased social and decreased non-social exploration from P35 to P44, while CP376395-treated rats did not. Socially defeated, vehicle-treated adolescents took more cocaine during a 24-h unlimited access binge during adulthood. The latency to supine posture on P44 was inversely correlated with later cocaine self-administration during fixed and progressive ratio schedules of reinforcement and during the binge. CONCLUSIONS: CP376395 treatment in adolescence blocked escalation of cocaine taking in adulthood. Episodes of social defeat stress engender neuroadaptation in CRF-R1s in the VTA that alter coping with social stress and that persist into adulthood.


Assuntos
Aminopiridinas/farmacologia , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Cocaína/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Comportamento Social , Área Tegmentar Ventral/efeitos dos fármacos , Adaptação Psicológica , Animais , Comportamento Aditivo/tratamento farmacológico , Hormônio Liberador da Corticotropina/metabolismo , Modelos Animais de Doenças , Locomoção/efeitos dos fármacos , Masculino , Ratos , Ratos Long-Evans , Reforço Psicológico , Autoadministração , Estresse Psicológico , Área Tegmentar Ventral/metabolismo
11.
J Neurosci ; 36(14): 4093-105, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-27053215

RESUMO

Intermittent social defeat stress escalates later cocaine self-administration. Reward and stress both activate ventral tegmental area (VTA) dopamine neurons, increasing downstream extracellular dopamine concentration in the medial prefrontal cortex and nucleus accumbens. The stress neuropeptide corticotropin releasing factor (CRF) and its receptors (CRF-R1, CRF-R2) are located in the VTA and influence dopaminergic activity. These experiments explore how CRF release and the activation of its receptors within the VTA both during and after stress influence later cocaine self-administration in rats.In vivo microdialysis of CRF in the VTA demonstrated that CRF is phasically released in the posterior VTA (pVTA) during acute defeat, but, with repeated defeat, CRF is recruited into the anterior VTA (aVTA) and CRF tone is increased in both subregions. Intra-VTA antagonism of CRF-R1 in the pVTA and CRF-R2 in the aVTA during each social defeat prevented escalated cocaine self-administration in a 24 h "binge." VTA CRF continues to influence cocaine seeking in stressed animals long after social defeat exposure. Unlike nonstressed controls, previously stressed rats show significant cocaine seeking after 15 d of forced abstinence. Previously stressed rats continue to express elevated CRF tone within the VTA and antagonism of pVTA CRF-R1 or aVTA CRF-R2 reverses cocaine seeking. In conclusion, these experiments demonstrate neuroadaptive changes in tonic and phasic CRF with repeated stress, that CRF release during stress may contribute to later escalated cocaine taking, and that persistently elevated CRF tone in the VTA may drive later cocaine seeking through increased activation of pVTA CRF-R1 and aVTA CRF-R2. SIGNIFICANCE STATEMENT: Corticotropin releasing factor (CRF) within the ventral tegmental area (VTA) has emerged as a likely candidate molecule underlying the fundamental link between stress history and escalated drug self-administration. However, the nature of CRF release in the VTA during acute and repeated stress, as well as its role in enduring neuroadaptations driving later drug taking and seeking, are poorly understood. These experiments explore how CRF is released and interacts with its receptors in specific regions of the VTA both during and after stress to fuel later escalated cocaine taking and seeking behavior. Understanding these acute and persistent changes to the VTA CRF system may lead to better therapeutic interventions for addiction.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/psicologia , Hormônio Liberador da Corticotropina/metabolismo , Meio Social , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Área Tegmentar Ventral/metabolismo , Animais , Comportamento de Procura de Droga , Masculino , Microdiálise , Ratos , Ratos Long-Evans , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Autoadministração , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/psicologia
12.
Addict Biol ; 21(1): 111-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25262980

RESUMO

Both the opioid antagonist naltrexone and corticotropin-releasing factor type-1 receptor (CRF-R1) antagonists have been investigated for the treatment of alcoholism. The current study examines the combination of naltrexone and CP154526 to reduce intermittent access ethanol drinking [intermittent access to alcohol (IAA)] in C57BL/6J male mice, and if these compounds reduce drinking via serotonergic mechanisms in the dorsal raphe nucleus (DRN). Systemic injections and chronic intracerebroventricular infusions of naltrexone, CP154526 or CP376395 transiently decreased IAA drinking. Immunohistochemistry revealed CRF-R1 or µ-opioid receptor immunoreactivity was co-localized in tryptophan hydroxylase (TPH)-immunoreactive neurons as well as non-TPH neurons in the DRN. Mice with a history of IAA or continuous access to alcohol were microinjected with artificial cerebral spinal fluid, naltrexone, CP154526 or the combination into the DRN or the median raphe nucleus (MRN). Either intra-DRN naltrexone or CP154526 reduced IAA in the initial 2 hours of fluid access, but the combination did not additively suppress IAA, suggesting a common mechanism via which these two compounds affect intermittent drinking. These alcohol-reducing effects were localized to the DRN of IAA drinkers, as intra-MRN injections only significantly suppressed water drinking, and continuous access drinkers were not affected by CRF-R1 antagonism. Extracellular serotonin was measured in the medial prefrontal cortex (mPFC) using in vivo microdialysis after intra-DRN microinjections in another group of mice. Intra-DRN CP154526 increased serotonin impulse flow to the mPFC while naltrexone did not. This suggests the mPFC may not be an essential location to intermittent drinking, as evidenced by different effects on serotonin signaling to the forebrain yet similar behavioral findings.


Assuntos
Consumo de Bebidas Alcoólicas , Comportamento Animal/efeitos dos fármacos , Depressores do Sistema Nervoso Central/administração & dosagem , Núcleo Dorsal da Rafe , Etanol/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Aminopiridinas/farmacologia , Animais , Imuno-Histoquímica , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Naltrexona/farmacologia , Córtex Pré-Frontal/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores Opioides mu/metabolismo , Autoadministração , Serotonina/metabolismo , Triptofano Hidroxilase/metabolismo
13.
Psychopharmacology (Berl) ; 233(4): 681-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26576941

RESUMO

RATIONALE: Excessive alcohol (EtOH) drinking is difficult to model in animals despite the extensive human literature demonstrating that stress increases EtOH consumption. OBJECTIVE: The current experiments show escalations in voluntary EtOH drinking caused by a history of social defeat stress and intermittent access to EtOH in C57BL/6J mice compared to non-stressed mice given intermittent EtOH or continuous EtOH. To explore a mechanistic link between stress and drinking, we studied the role of corticotropin-releasing factor type-1 receptors (CRF-R1) in the dopamine-rich ventral tegmental area (VTA). RESULTS: Intra-VTA infusions of a CRF-R1 antagonist, CP376395, infused into the VTA dose-dependently and selectively reduced intermittent EtOH intake in stressed and non-stressed mice, but not in mice given continuous EtOH. In contrast, intra-VTA infusions of the CRF-R2 antagonist astressin2B non-specifically suppressed both EtOH and H2O drinking in the stressed group without effects in the non-stressed mice. Using in vivo microdialysis in the nucleus accumbens (NAc) shell, we observed that stressed mice drinking EtOH intermittently had elevated levels of tonic dopamine concentrations compared to non-stressed drinking mice. Also, VTA CP376395 potentiated dopamine output to the NAc only in the stressed group causing further elevations of dopamine post-infusion. CONCLUSIONS: These findings illustrate a role for extrahypothalamic CRF-R1 as especially important for stress-escalated EtOH drinking beyond schedule-escalated EtOH drinking. CRF-R1 may be a mechanism for balancing the dysregulation of stress and reward in alcohol use disorders.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Núcleo Accumbens/metabolismo , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Estresse Psicológico/metabolismo , Área Tegmentar Ventral/metabolismo , Consumo de Bebidas Alcoólicas/psicologia , Animais , Hormônio Liberador da Corticotropina/administração & dosagem , Dopamina/metabolismo , Relações Interpessoais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Microinjeções , Núcleo Accumbens/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Recompensa , Estresse Psicológico/psicologia , Área Tegmentar Ventral/efeitos dos fármacos
14.
Psychopharmacology (Berl) ; 232(23): 4359-69, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26381154

RESUMO

RATIONALE: Benzodiazepines (BZDs) are prescribed to reduce anxiety, agitation, and muscle spasms and for their sedative-hypnotic and anticonvulsant effects. Under specific conditions, BZDs escalate aggression in some individuals. Specific effects of BZDs have been linked to the α-subunit subtype composition of GABAA receptors. OBJECTIVES: Point-mutated mice rendered selectively insensitive to BZDs at α1-, α2-, or α3-containing GABAA receptors were used to determine which α-subunit subtypes are necessary for BZDs to escalate aggression and social approach and to reduce fear-motivated behavior. METHODS: During resident-intruder confrontations, male wild-type (WT) and point-mutated α1(H101R), α2(H101R), and α3(H126R) mice were treated with midazolam (0-1.7 mg/kg, i.p.) and evaluated for aggression in an unfamiliar environment. Separate midazolam-treated WT and point-mutated mice were assessed for social approach toward a female or investigated in a 6-day fear-potentiated startle procedure. RESULTS: Moderate doses of midazolam (0.3-0.56 mg/kg, i.p.) escalated aggression in WT and α3(H126R) mutants and increased social approach in WT and α1(H101R) mice. The highest dose of midazolam (1.0 mg/kg) reduced fear-potentiated startle responding. All mice were sensitive to the sedative effect of midazolam (1.7 mg/kg) except α1(H101R) mutants. CONCLUSIONS: Midazolam requires BZD-sensitive α1- and α2-containing GABAA receptors in order to escalate aggression and α2- and α3-containing receptors to reduce social anxiety-like behavior. GABAA receptors containing the α1-subunit are crucial for BZD-induced sedation, while α2-containing GABAA receptors may be a shared site of action for the pro-aggressive and anxiolytic effects of BZDs.


Assuntos
Agressão/efeitos dos fármacos , Agressão/psicologia , Midazolam/farmacologia , Receptores de GABA-A/fisiologia , Comportamento Social , Agressão/fisiologia , Animais , Ansiolíticos/farmacologia , Ansiolíticos/uso terapêutico , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Benzodiazepinas/farmacologia , Benzodiazepinas/uso terapêutico , Medo/efeitos dos fármacos , Medo/fisiologia , Medo/psicologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Midazolam/uso terapêutico , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia
15.
Psychopharmacology (Berl) ; 232(24): 4469-79, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26403083

RESUMO

RATIONALE: Stress activates a subset of dopamine neurons in the ventral tegmental area (VTA), increasing extracellular dopamine in the medial prefrontal cortex (mPFC) and nucleus accumbens shell (NAcSh). The stress neuropeptide corticotropin releasing factor (CRF) and its receptors (CRF-R1 and CRF-R2) are located within the VTA and directly and indirectly influence dopaminergic activity. However, it has yet to be shown in vivo whether VTA CRF receptor activation is necessary for acute and repeated stress-induced dopamine efflux. OBJECTIVE: With intra-VTA CRF-R1 and CRF-R2 antagonism during social defeat, we assessed whether blockade of these receptors could prevent stress-induced dopamine increases in the mPFC and NAcSh using in vivo microdialysis. METHODS: Rats were microinjected with a CRF-R1 or CRF-R2 antagonist into the VTA prior to social defeat stress on days 1, 4, 7, and 10. In vivo microdialysis for dopamine in the mPFC and NAcSh was performed during stress on days 1 and 10. RESULTS: During the first social defeat, extracellular dopamine was significantly elevated in both the mPFC and NAcSh, and this increase in the NAcSh was blocked by intra-VTA CRF-R2, but not CRF-R1, antagonism. During the final social defeat, the dopaminergic increase was neither sensitized nor habituated in the mPFC and NAcSh, and intra-VTA CRF-R2, but not CRF-R1, antagonism prevented the dopamine increase in both brain regions. CONCLUSION: These findings show that CRF-R2 in the VTA is necessary for acute and repeated stress-induced dopamine efflux in the NAcSh, but is only recruited into mPFC-projecting dopamine neurons with repeated stress exposure.


Assuntos
Dopamina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Predomínio Social , Estresse Psicológico/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Hormônio Liberador da Corticotropina/metabolismo , Masculino , Microdiálise , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Long-Evans , Área Tegmentar Ventral/efeitos dos fármacos
16.
Alcohol Clin Exp Res ; 39(9): 1609-18, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26247973

RESUMO

BACKGROUND: Most studies with corticotropin releasing factor (CRF) and ethanol (EtOH) consumption have focused on CRF type 1 (CRF1 ) receptors; less is known about other components of the CRF system, such as the CRF type 2 (CRF2 ) receptors and the CRF binding protein (CRFBP). In humans, several nucleotide polymorphisms in the CRFBP gene have been associated with EtOH abuse. METHODS: The role of the CRFBP within the ventral tegmental area (VTA) and the central nucleus of the amygdala (CeA) was investigated in C57BL/6J mice exposed to an EtOH binge drinking paradigm (drinking in the dark [DID]), or to a dependence-producing drinking protocol (2-bottle choice, intermittent access to alcohol [IAA]) for 4 weeks. Potential interactions between VTA CRFBP and CRF2 receptors on EtOH binge drinking were also assessed. Mice were microinjected with the CRFBP antagonist CRF fragment 6-33 (CRF6-33 ) into the VTA or CeA, or with the CRF2 antagonist astressin-2B (A2B) alone or in combination with CRF6-33 into the VTA, and had access to 20% (w/v) EtOH for 4 hours (DID). Separate cohorts of mice received vehicle and doses of CRF6-33 into the VTA or CeA and had access to EtOH/water for 24 hours (IAA). Blood EtOH concentrations (BECs) were measured, and signs of withdrawal by handling-induced convulsions were determined. RESULTS: Intra-VTA CRF6-33 and A2B reduced EtOH intake dose dependently in mice during DID. Furthermore, a combination of a subeffective dose of CRF6-33 and a lower dose of A2B promoted additive effects in attenuating EtOH binge drinking. Intra-VTA CRF6-33 did not affect EtOH consumption in mice given IAA, and intra-CeA CRF6-33 did not change alcohol consumption in both models of drinking. DID and IAA promoted pharmacologically relevant BECs; however, only mice given IAA exhibited convulsive events during withdrawal. CONCLUSIONS: These findings suggest that VTA CRFBP is involved in the initial stages of escalated EtOH drinking by mechanisms that may involve CRF2 receptors.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Proteínas de Transporte/metabolismo , Etanol/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Área Tegmentar Ventral/efeitos dos fármacos
17.
Ann N Y Acad Sci ; 1349: 96-118, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26285061

RESUMO

Neurobiological processes underlying the epidemiologically established link between alcohol and several types of social, aggressive, and violent behavior remain poorly understood. Acute low doses of alcohol, as well as withdrawal from long-term alcohol use, may lead to escalated aggressive behavior in a subset of individuals. An urgent task will be to disentangle the host of interacting genetic and environmental risk factors in individuals who are predisposed to engage in escalated aggressive behavior. The modulation of 5-hydroxytryptamine impulse flow by gamma-aminobutyric acid (GABA) and glutamate, acting via distinct ionotropic and metabotropic receptor subtypes in the dorsal raphe nucleus during alcohol consumption, is of critical significance in the suppression and escalation of aggressive behavior. In anticipation and reaction to aggressive behavior, neuropeptides such as corticotropin-releasing factor, neuropeptide Y, opioid peptides, and vasopressin interact with monoamines, GABA, and glutamate to attenuate and amplify aggressive behavior in alcohol-consuming individuals. These neuromodulators represent novel molecular targets for intervention that await clinical validation. Intermittent episodes of brief social defeat during aggressive confrontations are sufficient to cause long-lasting neuroadaptations that can lead to the escalation of alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Monoaminas Biogênicas/metabolismo , Etanol/efeitos adversos , Neuropeptídeos/farmacologia , Violência/psicologia , Agressão/efeitos dos fármacos , Agressão/psicologia , Consumo de Bebidas Alcoólicas/efeitos adversos , Consumo de Bebidas Alcoólicas/epidemiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Crime/psicologia , Crime/estatística & dados numéricos , Humanos , Neuropeptídeos/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/epidemiologia , Estresse Psicológico/psicologia
18.
Psychopharmacology (Berl) ; 232(16): 2889-902, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25899790

RESUMO

RATIONALE: Disrupted social behavior, including occasional aggressive outbursts, is characteristic of withdrawal from long-term alcohol (EtOH) use. Heavy EtOH use and exaggerated responses during withdrawal may be treated using glutamatergic N-methyl-D-aspartate receptor (NMDAR) antagonists. OBJECTIVES: The current experiments explore aggression and medial prefrontal cortex (mPFC) glutamate as consequences of withdrawal from intermittent access to EtOH and changes in aggression and mPFC glutamate caused by NMDAR antagonists memantine and ketamine. METHODS: Swiss male mice underwent withdrawal following 1-8 weeks of intermittent access to 20 % EtOH. Aggressive and nonaggressive behaviors with a conspecific were measured 6-8 h into EtOH withdrawal after memantine or ketamine (0-30 mg/kg, i.p.) administration. In separate mice, extracellular mPFC glutamate after memantine was measured during withdrawal using in vivo microdialysis. RESULTS: At 6-8 h withdrawal from EtOH, mice exhibited more convulsions and aggression and decreased social contact compared to age-matched water controls. Memantine, but not ketamine, increased withdrawal aggression at the 5-mg/kg dose in mice with a history of 8 weeks of EtOH but not 1 or 4 weeks of EtOH or in water drinkers. Tonic mPFC glutamate was higher during withdrawal after 8 weeks of EtOH compared to 1 week of EtOH or 8 weeks of water. Five milligrams per kilogram of memantine increased glutamate in 8-week EtOH mice, but also in 1-week EtOH and water drinkers. CONCLUSIONS: These studies reveal aggressive behavior as a novel symptom of EtOH withdrawal in outbred mice and confirm a role of NMDARs during withdrawal aggression and for disrupted social behavior.


Assuntos
Agressão/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Etanol/efeitos adversos , Ácido Glutâmico/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/metabolismo , Agressão/fisiologia , Animais , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Masculino , Memantina/farmacologia , Camundongos , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Comportamento Social , Síndrome de Abstinência a Substâncias/psicologia
19.
Pharmacol Biochem Behav ; 130: 22-33, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25560941

RESUMO

Creatine is an antioxidant, neuromodulator and key regulator of energy metabolism shown to improve depressive symptoms in humans and animals, especially in females. To better understand the pharmacological effects of creatine, we examined its influence on depression-related hippocampal gene expression and behaviors in the presence and absence of sex steroids. Sham-operated and gonadectomized male and female rats were fed chow alone or chow blended with either 2% or 4% w/w creatine monohydrate for five weeks before forced swim, open field, and wire suspension tests, or seven weeks total. Before supplementation, males were chronically implanted with an empty or a testosterone-filled (T) capsule (10-mm surface release), and females were administered progesterone (P, 250 µg), estradiol benzoate (EB, 2.5 µg), EB+P, or sesame oil vehicle weekly. Relative to non-supplemented shams, all hippocampal plasticity-related mRNAs measured, including brain-derived neurotrophic factor (BDNF), tyrosine kinase B, doublecortin, calretinin, and calbindin, were downregulated in sham males given 4% creatine, and BDNF, doublecortin, and calbindin mRNAs were downregulated in sham females given 4% creatine. In contrast, combined 4% creatine+T in castrates prevented downregulation of BDNF, doublecortin, and calretinin mRNAs. Similarly, combined 4% creatine+EB+P in ovariectomized females attenuated downregulation of BDNF and calbindin mRNA levels. Moderate antidepressant and anxiolytic-like behaviors were observed in EB+P-treated ovariectomized females fed creatine, with similar trends in T-treated castrates fed creatine. Altogether, these data show that chronic, high-dose creatine has opposing effects on neuroplasticity-related genes and depressive behavior in intact and gonadectomized male and female rats. The dose and schedule of creatine used negatively impacted hippocampal neuronal integrity in otherwise healthy brains, possibly through negative compensatory changes in energy metabolism, whereas combined creatine and sex steroids acted in a neuroprotective manner in gonadectomized rats, potentially by reducing metabolic complications associated with castration or ovariectomy.


Assuntos
Comportamento Animal/efeitos dos fármacos , Creatina/farmacologia , Depressão/genética , Estradiol/análogos & derivados , Expressão Gênica/efeitos dos fármacos , Progesterona/farmacologia , Testosterona/farmacologia , Animais , Proteína Duplacortina , Sinergismo Farmacológico , Estradiol/farmacologia , Feminino , Hipocampo/metabolismo , Masculino , Plasticidade Neuronal/genética , Orquiectomia , Ovariectomia , RNA Mensageiro/genética , Ratos
20.
Psychopharmacology (Berl) ; 232(6): 1003-10, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25216798

RESUMO

RATIONALE: Social defeat stress results in escalation of cocaine taking and long-term neural adaptations in rats. How the intensity and timing of social defeat stress determine these effects, particularly in mice, have not been well characterized. OBJECTIVE: This study investigated the effects of mild vs. moderate intensities and durations of social stress on intravenous cocaine self-administration as well as on dopamine (DA) release in the nucleus accumbens shell (NAcSh) by using in vivo microdialysis. METHODS: Adult male Swiss Webster (CFW) mice experienced 10 days of social defeat stress, either mild (15 attack bites in ca. 1.8 min) or moderate (30 attack bites in ca. 3.6 min), and compared to controls that were handled daily. Subsequently, the socially stressed mice were assessed for either (1) intravenous cocaine self-administration, using several unit doses (0, 0.3, 0.6, 1.0 mg/kg/infusion) under limited access conditions, or (2) neural sensitization, as determined by in vivo microdialysis of DA in the NAcSh in response to acute d-amphetamine challenge. RESULTS: Social defeat stress resulted in escalated cocaine self-administration in both mild and moderate socially stressed groups. In addition, social defeat stress led to increased DA release after d-amphetamine challenge. CONCLUSIONS: These data suggest that both mild and moderate socially stressed mice exhibit increased cocaine taking compared to controls, and this increase is associated with escalated dopaminergic responses in the NAcSh.


Assuntos
Cocaína/administração & dosagem , Dominação-Subordinação , Inibidores da Captação de Dopamina/administração & dosagem , Dopamina/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Estresse Psicológico/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Dextroanfetamina/farmacologia , Masculino , Camundongos , Microdiálise , Núcleo Accumbens/metabolismo , Ratos , Autoadministração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...