Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Psychiatry ; 27(11): 4642-4652, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36123420

RESUMEN

Evidence suggests that spironolactone, a nonselective mineralocorticoid receptor (MR) antagonist, modulates alcohol seeking and consumption. Therefore, spironolactone may represent a novel pharmacotherapy for alcohol use disorder (AUD). In this study, we tested the effects of spironolactone in a mouse model of alcohol drinking (drinking-in-the-dark) and in a rat model of alcohol dependence (vapor exposure). We also investigated the association between spironolactone receipt for at least 60 continuous days and change in self-reported alcohol consumption, using the Alcohol Use Disorders Identification Test-Consumption (AUDIT-C), in a pharmacoepidemiologic cohort study in the largest integrated healthcare system in the US. Spironolactone dose-dependently reduced the intake of sweetened or unsweetened alcohol solutions in male and female mice. No effects of spironolactone were observed on drinking of a sweet solution without alcohol, food or water intake, motor coordination, alcohol-induced ataxia, or blood alcohol levels. Spironolactone dose-dependently reduced operant alcohol self-administration in dependent and nondependent male and female rats. In humans, a greater reduction in alcohol consumption was observed among those who received spironolactone, compared to propensity score-matched individuals who did not receive spironolactone. The largest effects were among those who reported hazardous/heavy episodic alcohol consumption at baseline (AUDIT-C ≥ 8) and those exposed to ≥ 50 mg/day of spironolactone. These convergent findings across rodent and human studies demonstrate that spironolactone reduces alcohol use and support the hypothesis that this medication may be further studied as a novel pharmacotherapy for AUD.


Asunto(s)
Alcoholismo , Humanos , Masculino , Femenino , Ratas , Animales , Ratones , Alcoholismo/tratamiento farmacológico , Espironolactona/uso terapéutico , Espironolactona/farmacología , Roedores , Estudios de Cohortes , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Etanol
2.
Addict Biol ; 26(3): e12978, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33142367

RESUMEN

Genetically-selected Marchigian Sardinian alcohol-preferring (msP) rats display comorbid symptoms of increased alcohol preference and elevated anxiety-like behavior. Heightened stress sensitivity in msPs is influenced by genetic polymorphisms of the corticotropin-releasing factor receptor in the central nucleus of the amygdala (CeA), as well as reduced influence of anti-stress mechanisms that normally constrain the stress response. Given this propensity for stress dysregulation, in this study, we expand on the possibility that msPs may display differences in neuroendocrine processes that normally terminate the stress response. We utilized behavioral, biochemical, and molecular assays to compare basal and restraint stress-induced changes in the hypothalamic-pituitary-adrenal (HPA) axis of male and female msPs relative to their nonselected Wistar counterparts. The results showed that msPs display deficits in marble-burying behavior influenced by environmental factors and procedures that modulate arousal states in a sex-dependent manner. Whereas male msPs display evidence of dysregulated neuroendocrine function (higher adrenocorticotropic hormone levels and subthreshold reductions in corticosterone), females display restraint-induced elevations in corticosterone levels that were persistently higher in msPs. A dexamethasone challenge reduced the circulation of these stress hormones, although the reduction in corticosterone was generally attenuated in msP versus Wistar rats. Finally, we found evidence of diminished stress-induced glucocorticoid receptor (GR) phosphorylation in the hypothalamic paraventricular nucleus of msPs, as well as innate increases in phosphorylated GR levels in the CeA of male msPs. Collectively, these findings suggest that negative feedback processes regulating HPA responsiveness are diminished in msP rats, possibly underlying differences in the expression of anxiety-like behaviors.


Asunto(s)
Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Restricción Física , Consumo de Bebidas Alcohólicas/genética , Animales , Ansiedad/genética , Corticosterona/sangre , Retroalimentación Fisiológica , Femenino , Glucocorticoides/genética , Hipotálamo/metabolismo , Masculino , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
3.
J Neurophysiol ; 117(2): 477-479, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27169510

RESUMEN

An important new study by Kvarta, Bradbrook, Dantrassy, Bailey, and Thompson (J Neurophysiol 114: 1713-1724, 2015) examined the effects of persistent stress and excessive glucocorticoid levels on hippocampal function and emotional behavior in rodents. The authors specifically implicate the temporoammonic pathway as being susceptible to reductions in excitatory function in the context of chronic stress. We discuss the importance of this new finding in the broader context of medication development for major depressive disorder.


Asunto(s)
Hipocampo/fisiopatología , Hormonas/metabolismo , Trastornos del Humor , Animales , Humanos , Trastornos del Humor/metabolismo , Trastornos del Humor/patología , Trastornos del Humor/fisiopatología , Estrés Psicológico/fisiopatología
4.
Alcohol Clin Exp Res ; 40(12): 2537-2547, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27796078

RESUMEN

BACKGROUND: Nicotine use increases alcohol drinking, suggesting that the combination of these drugs may produce synergistic effects in activating reward circuitry. Alternatively, use of either of these drugs may facilitate the development of cross-tolerance to the other to promote intake escalation. METHODS: In this study, adult male Wistar rats were chronically exposed to room air or chronic, intermittent nicotine vapor, which has been shown to produce symptoms of nicotine dependence as evidenced by elevated nicotine self-administration and a host of somatic and motivational withdrawal symptoms. We examined regional neuroadaptations in nicotine-experienced versus nonexperienced animals, focusing on changes in phosphorylation of the AMPA glutamate channel subunit GluA1 in reward-related brain regions as excitatory neuroadaptations are heavily implicated in both alcohol and nicotine addiction. RESULTS: During withdrawal, nicotine exposure and alcohol challenge (1 g/kg) interactively produced neuroadaptations in GluA1 phosphorylation in a brain region-dependent manner. Alcohol robustly increased protein kinase A-mediated phosphorylation of GluA1 at serine 845 in multiple regions. However, this neuroadaptation was largely absent in 3 areas (dorsomedial prefrontal cortex, dorsal striatum, and central amygdala) in nicotine-experienced animals. This interactive effect suggests a molecular tolerance to alcohol-stimulated phosphorylation of GluA1 in the context of nicotine dependence. CONCLUSIONS: Nicotine may modify the rewarding or reinforcing effects of alcohol by altering glutamate signaling in a region-specific manner, thereby leading to increased drinking in heavy smokers.


Asunto(s)
Tolerancia a Medicamentos , Etanol/farmacología , Nicotina/efectos adversos , Receptores AMPA/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Administración por Inhalación , Animales , Encéfalo/metabolismo , Interacciones Farmacológicas , Masculino , Nicotina/administración & dosificación , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
5.
ACS Chem Neurosci ; 13(13): 1832-1834, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35748762

RESUMEN

Despite the high prevalence and negative consequences associated with alcohol use disorder (AUD), currently available pharmacotherapies are limited in number and efficacy. Several neuroendocrine pathways have been identified and are under investigation as potential pharmacotherapeutic targets for AUD. Here, we present the promise of the mineralocorticoid receptor (MR) as a novel target and discuss associations between the aldosterone/MR system and AUD, the effects of MR antagonism on alcohol consumption, and the underlying neurobiology of these effects.


Asunto(s)
Alcoholismo , Receptores de Mineralocorticoides , Consumo de Bebidas Alcohólicas , Alcoholismo/tratamiento farmacológico , Aldosterona/metabolismo , Aldosterona/uso terapéutico , Humanos , Antagonistas de Receptores de Mineralocorticoides/farmacología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico
6.
Pharmacol Biochem Behav ; 204: 173155, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33631255

RESUMEN

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.


Asunto(s)
Alcoholismo/metabolismo , Encéfalo/efectos de los fármacos , Tolerancia a Medicamentos , Etanol/farmacología , Neurobiología/métodos , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/patología , Animales , Conducta Adictiva/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Etanol/metabolismo , Femenino , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Norepinefrina/metabolismo , Ratas , Serotonina/metabolismo , Caracteres Sexuales
7.
Neuropharmacology ; 188: 108510, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33647278

RESUMEN

Alcohol use disorder (AUD) is associated with the dysregulation of brain stress and reward systems, including glucocorticoid receptors (GRs). The mixed glucocorticoid/progesterone receptor antagonist mifepristone and selective GR antagonist CORT113176 have been shown to selectively reduce alcohol consumption in alcohol-dependent rats. Mifepristone has also been shown to decrease alcohol consumption and craving for alcohol in humans with AUD. The present study tested the effects of the GR modulators CORT118335, CORT122928, CORT108297, and CORT125134 on alcohol self-administration in nondependent (air-exposed) and alcohol-dependent (alcohol vapor-exposed) adult male rats. Different GR modulators recruit different GR-associated transcriptional cofactors. Thus, we hypothesized that these GR modulators would vary in their effects on alcohol drinking. CORT118335, CORT122928, and CORT125134 significantly reduced alcohol self-administration in both alcohol-dependent and nondependent rats. CORT108297 had no effect on alcohol self-administration in either group. The present results support the potential of GR modulators for the development of treatments for AUD. Future studies that characterize genomic and nongenomic effects of these GR modulators will elucidate potential molecular mechanisms that underlie alcohol drinking in alcohol-dependent and nondependent states.


Asunto(s)
Compuestos Aza/farmacología , Etanol/administración & dosificación , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Isoquinolinas/farmacología , Mifepristona/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Receptores de Glucocorticoides/efectos de los fármacos , Autoadministración , Timina/análogos & derivados , Animales , Masculino , Ratas , Ratas Wistar , Timina/farmacología
8.
Int Rev Neurobiol ; 157: 409-472, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33648675

RESUMEN

Addiction is a chronic disorder that consists of a three-stage cycle of binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. These stages involve, respectively, neuroadaptations in brain circuits involved in incentive salience and habit formation, stress surfeit and reward deficit, and executive function. Much research on addiction focuses on the neurobiology underlying single drug use. However, alcohol use disorder (AUD) can be co-morbid with substance use disorder (SUD), called dual dependence. The limited epidemiological data on dual dependence indicates that there is a large population of individuals suffering from addiction who are dependent on more than one drug and/or alcohol, yet dual dependence remains understudied in addiction research. Here, we review neurobiological data on neurotransmitter and neuropeptide systems that are known to contribute to addiction pathology and how the involvement of these systems is consistent or divergent across drug classes. In particular, we highlight the dopamine, opioid, corticotropin-releasing factor, norepinephrine, hypocretin/orexin, glucocorticoid, neuroimmune signaling, endocannabinoid, glutamate, and GABA systems. We also discuss the limited research on these systems in dual dependence. Collectively, these studies demonstrate that the use of multiple drugs can produce neuroadaptations that are distinct from single drug use. Further investigation into the neurobiology of dual dependence is necessary to develop effective treatments for addiction to multiple drugs.


Asunto(s)
Alcoholismo , Trastornos Relacionados con Sustancias , Alcoholismo/epidemiología , Alcoholismo/fisiopatología , Comorbilidad , Humanos , Trastornos Relacionados con Sustancias/epidemiología , Trastornos Relacionados con Sustancias/fisiopatología
9.
Neuropharmacology ; 144: 82-90, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30336151

RESUMEN

Alcohol use disorder is highly co-morbid with traumatic stress disorders in humans, and dually diagnosed individuals cite negative affective symptoms as a primary reason for drinking alcohol. Therefore, it is reasonable to hypothesize that traumatic stress history increases the rewarding properties and/or blunts the aversive properties of alcohol. We used a place conditioning procedure to test the rewarding/aversive properties of alcohol in adult male Wistar rats with or without a traumatic stress (i.e., predator odor exposure) history, and with or without an alcohol drinking history. Because extended amygdala regions have documented roles in stress, reward, and stress-induced changes in reward, we also tested the effect of acute alcohol on CREB phosphorylation (pCREB) and striatal-enriched protein tyrosine phosphatase (STEP) expression in central amygdala (CeA) and bed nucleus of stria terminalis (BNST). Our results show that a moderate alcohol dose (1.0 g/kg) produces conditioned place aversion (CPA) that is blunted by stress history but is not affected by alcohol drinking history, and this effect differed in pair-housed versus single-housed rats. Stress history reduced pCREB expression in BNST of rats with and without an alcohol drinking history. Finally, acute alcohol effects on pCREB and STEP expression in CeA were positively associated with preference for the alcohol-paired chamber. These data suggest that stress history reduces the aversive properties of moderate alcohol doses, and that alcohol aversion is associated with acute alcohol effects on pCREB and STEP expression in the extended amygdala.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Reacción de Prevención/fisiología , Condicionamiento Psicológico/fisiología , Percepción Olfatoria/fisiología , Conducta Predatoria , Estrés Psicológico/fisiopatología , Consumo de Bebidas Alcohólicas/psicología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Condicionamiento Psicológico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Etanol/administración & dosificación , Expresión Génica/efectos de los fármacos , Vivienda para Animales , Masculino , Odorantes , Percepción Olfatoria/efectos de los fármacos , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Ratas Wistar , Recompensa , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología , Estrés Psicológico/etiología
10.
Alcohol ; 75: 11-18, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30321699

RESUMEN

Alcohol use disorder (AUD) is a chronic, relapsing psychiatric disease characterized by the emergence of negative emotional states and the development of motivational deficits that manifest during alcohol withdrawal. Accordingly, alcohol may be sought after and taken in excessive amounts to alleviate withdrawal-related symptoms. To develop more effective treatments for AUD, it is necessary to identify potential molecular targets that underlie the transition from initial alcohol use to alcohol dependence, and our previous work has implicated a role for potentiated glucocorticoid receptor (GR) signaling in this regard. As a key negative regulator of GR-mediated signaling, the current study first measured c-Jun N-terminal kinase (JNK) phosphorylation in animals following an acute alcohol challenge. We found that JNK phosphorylation (pJNK) was significantly increased in the hippocampus, frontal cortical regions, and striatum of adult male Wistar rats following alcohol challenge, indicating that initial alcohol exposure increases JNK activity across several brain regions. A separate group of adult male Wistar rats were made dependent via chronic, intermittent ethanol vapor exposure and were trained to self-administer alcohol. We found that alcohol-dependent animals consumed significantly more alcohol and escalated their drinking over time compared to non-dependent animals. We then measured alterations in JNK phosphorylation in this alcohol-dependent group during acute withdrawal and found that pJNK was selectively decreased in the dorsal hippocampus, dorsomedial prefrontal cortex, and cingulate cortex. These findings demonstrate that withdrawal from chronic alcohol exposure leads to region-specific deficits in JNK phosphorylation. JNK signaling dysregulation may foster long-lasting behavioral and motivational impairments in alcohol dependence, either as a result of increased GR-mediated stress signaling or via other downstream mechanisms.


Asunto(s)
Alcoholismo/metabolismo , Etanol/toxicidad , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Administración por Inhalación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Etanol/administración & dosificación , Masculino , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Wistar , Autoadministración
11.
J Neurotrauma ; 36(11): 1847-1855, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30638118

RESUMEN

Traumatic brain injury (TBI) is associated with psychiatric dysfunction-including pain, cognitive impairment, anxiety, and increased alcohol use. We previously demonstrated that inhibiting endocannabinoid degradation post-TBI with JZL184 attenuates neuroinflammation and neuronal hyperexcitability at the site of injury and improves neurobehavioral recovery. This study aimed to determine the effect of JZL184 on post-TBI behavioral changes related to psychiatric dysfunction and post-TBI neuroadaptations in brain regions associated with these behaviors. We hypothesized that JZL184 would attenuate post-TBI behavioral and neural changes in alcohol-drinking rats. Adult male Wistar rats were trained to operantly self-administer alcohol before receiving lateral fluid percussion injury. Thirty minutes post-TBI, rats received JZL184 (16 mg/kg, i.p.) or vehicle. Spatial memory (Y-maze), anxiety-like behavior (open field), alcohol motivation (progressive ratio responding), and mechanosensitivity (Von Frey) were measured 3-10 days post-injury, and ventral striatum (VS) and central amygdala (CeA) tissue were collected for western blot analysis of phosphorylated glutamate receptor subunit 1 (GluR1) and glucocorticoid receptor (GR). TBI impaired spatial memory, increased anxiety-like behavior, and increased motivated alcohol drinking. JZL184 prevented these changes. TBI also increased phosphorylated GluR1 and GR in the CeA (but not the VS) compared with sham controls. JZL184 attenuated post-TBI GR phosphorylation in the CeA. These findings suggest that TBI produces comorbid cognitive dysfunction, increased alcohol motivation, and anxiety-like behavior, possibly related to amygdala dysfunction, and these changes are prevented by systemic post-TBI endocannabinoid degradation inhibition. Thus, boosting endocannabinoid tone post-TBI may represent a viable therapeutic strategy for TBI-related psychiatric comorbidities such as alcohol use disorder and anxiety.


Asunto(s)
Afecto/fisiología , Consumo de Bebidas Alcohólicas/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Motivación/fisiología , Receptores de Glucocorticoides/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Benzodioxoles/farmacología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/metabolismo , Núcleo Amigdalino Central/fisiopatología , Masculino , Piperidinas/farmacología , Ratas , Ratas Wistar , Receptores AMPA/metabolismo
12.
Curr Opin Behav Sci ; 13: 139-143, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28111628

RESUMEN

The prefrontal cortex (PFC) represents and executes the highest forms of goal-directed behavior, and has thereby attained a central neuroanatomical position in most pathophysiological conceptualizations of motivational disorders, including alcohol use disorder (AUD). Excessive, intermittent exposure to alcohol produces an allostatic dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis along with heightened forebrain glucocorticoid signaling that can damage PFC architecture and function. Negative affective states intimately associated with the transition to alcohol dependence result not only from a dysregulated HPA axis, but also from the inability of a damaged PFC to regulate subcortical stress and reinforcement centers, including the ventral striatum and amygdala. Several cognitive symptoms commonly associated with severe AUD, ranging from poor risk management to the cognitive/affective dimension of pain, are likely mediated by altered function of key anatomical elements that modulate PFC executive function, including contributions from the cingulate cortex and insula. Future therapeutic strategies for severe AUD should focus on attenuating the deleterious effects of excessive stress hormone activity on cognitive/affective and motivational behaviors gated by the PFC.

13.
Neuroscience ; 366: 1-14, 2017 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-29024786

RESUMEN

Repeated use of opioids can lead to the development of analgesic tolerance and dependence. Additionally, chronic opioid exposure can cause a paradoxical emergence of heightened pain sensitivity to noxious stimuli, termed hyperalgesia, which may drive continued or escalated use of opioids to manage worsening pain symptoms. Opioid-induced hyperalgesia has traditionally been measured in rodents via reflex-based assays, including the von Frey method. To better model the cognitive/motivational dimension of pain in a state of opioid dependence and withdrawal, we employed a recently developed non-reflex-based method for measuring pain avoidance-like behavior in animals (mechanical conflict avoidance test). Adult male Wistar rats were administered an escalating dose regimen of morphine (opioid-dependent group) or repeated saline (control group). Morphine-dependent rats exhibited significantly greater avoidance of noxious stimuli during withdrawal. We next investigated individual relationships between pain avoidance-like behavior and alterations in protein phosphorylation in central motivation-related brain areas. We discovered that pain avoidance-like behavior was significantly correlated with alterations in phosphorylation status of protein kinases (ERK, CaMKII), transcription factors (CREB), presynaptic markers of neurotransmitter release (Synapsin), and the rate-limiting enzyme for dopamine synthesis (TH) across specific brain regions. Our findings suggest that alterations in phosphorylation events in specific brain centers may support cognitive/motivational responses to avoid pain.


Asunto(s)
Reacción de Prevención , Encéfalo/metabolismo , Dependencia de Morfina/metabolismo , Dependencia de Morfina/psicología , Dolor/metabolismo , Dolor/psicología , Analgésicos Opioides/administración & dosificación , Animales , Masculino , Morfina/administración & dosificación , Motivación , Ratas Wistar , Recompensa , Síndrome de Abstinencia a Sustancias
14.
Alcohol ; 49(8): 803-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26008713

RESUMEN

Addiction, or substance use disorder (SUD), is a devastating psychiatric disease composed of multiple elemental features. As a biobehavioral disorder, escalation of drug and/or alcohol intake is both a cause and consequence of molecular neuroadaptations in central brain reinforcement circuitry. Multiple mesolimbic areas mediate a host of negative affective and motivational symptoms that appear to be central to the addiction process. Brain stress- and reinforcement-related regions such as the central amygdala (CeA), prefrontal cortex (PFC), and nucleus accumbens (NAc) also serve as central processors of ascending nociceptive input. We hypothesize that a sensitization of brain mechanisms underlying the processing of persistent and maladaptive pain contributes to a composite negative affective state to drive the enduring, relapsing nature of addiction, particularly in the case of alcohol and opioid use disorder. At the neurochemical level, pain activates central stress-related neuropeptide signaling, including the dynorphin and corticotropin-releasing factor (CRF) systems, and by this process may facilitate negative affect and escalated drug and alcohol use over time. Importantly, the widespread prevalence of unresolved pain and associated affective dysregulation in clinical populations highlights the need for more effective analgesic medications with reduced potential for tolerance and dependence. The burgeoning epidemic of prescription opioid abuse also demands a closer investigation into the neurobiological mechanisms of how pain treatment could potentially represent a significant risk factor for addiction in vulnerable populations. Finally, the continuing convergence of sensory and affective neuroscience fields is expected to generate insight into the critical balance between pain relief and addiction liability, as well as provide more effective therapeutic strategies for chronic pain and addiction.


Asunto(s)
Afecto , Alcoholismo/psicología , Encéfalo/metabolismo , Dolor Crónico/psicología , Trastornos del Humor/psicología , Alcoholismo/epidemiología , Alcoholismo/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Dolor Crónico/epidemiología , Dolor Crónico/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Dinorfinas/metabolismo , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/psicología , Trastornos del Humor/epidemiología , Neuropéptidos/metabolismo , Núcleo Accumbens/metabolismo , Dolor/epidemiología , Dolor/metabolismo , Dolor/psicología , Corteza Prefrontal/metabolismo , Factores de Riesgo , Trastornos Relacionados con Sustancias/epidemiología , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/psicología
15.
J Clin Invest ; 125(8): 3193-7, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26121746

RESUMEN

Alcoholism, or alcohol use disorder, is a major public health concern that is a considerable risk factor for morbidity and disability; therefore, effective treatments are urgently needed. Here, we demonstrated that the glucocorticoid receptor (GR) antagonist mifepristone reduces alcohol intake in alcohol-dependent rats but not in nondependent animals. Both systemic delivery and direct administration into the central nucleus of the amygdala, a critical stress-related brain region, were sufficient to reduce alcohol consumption in dependent animals. We also tested the use of mifepristone in 56 alcohol-dependent human subjects as part of a double-blind clinical and laboratory-based study. Relative to placebo, individuals who received mifepristone (600 mg daily taken orally for 1 week) exhibited a substantial reduction in alcohol-cued craving in the laboratory, and naturalistic measures revealed reduced alcohol consumption during the 1-week treatment phase and 1-week post-treatment phase in mifepristone-treated individuals. Mifepristone was well tolerated and improved liver-function markers. Together, these results support further exploration of GR antagonism via mifepristone as a therapeutic strategy for alcoholism.


Asunto(s)
Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Antagonistas de Hormonas/administración & dosificación , Mifepristona/administración & dosificación , Receptores de Glucocorticoides/antagonistas & inhibidores , Administración Oral , Adulto , Consumo de Bebidas Alcohólicas/fisiopatología , Alcoholismo/fisiopatología , Animales , Femenino , Antagonistas de Hormonas/efectos adversos , Humanos , Masculino , Mifepristona/efectos adversos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA