Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 160(4): 454-468, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34919270

RESUMEN

There is increasing evidence for a daily rhythm of µ-opioid receptor (MOR) efficacy and the development of alcohol dependence. Previous studies show that ß-arrestin 2 (bArr2) has an impact on alcohol intake, at least partially mediated via modulation of MOR signaling, which in turn mediates the alcohol rewarding effects. Considering the interplay of circadian rhythms on MOR and alcohol dependence, we aimed to investigate bArr2 in alcohol dependence at different time points of the day/light cycle on the level of bArr2 mRNA (in situ hybridization), MOR availability (receptor autoradiography), and MOR signaling (Damgo-stimulated G-protein coupling) in the nucleus accumbens of alcohol-dependent and non-dependent Wistar rats. Using a microarray data set we found that bArr2, but not bArr1, shows a diurnal transcription pattern in the accumbens of naïve rats with higher expression levels during the active cycle. In 3-week abstinent rats, bArr2 is up-regulated in the accumbens at the beginning of the active cycle (ZT15), whereas no differences were found at the beginning of the inactive cycle (ZT3) compared with controls. This effect was accompanied by a specific down-regulation of MOR binding in the active cycle. Additionally, we detect a higher receptor coupling during the inactive cycle compared with the active cycle in alcohol-dependent animals. Together, we report daily rhythmicity for bArr2 expression linked to an inverse pattern of MOR, suggesting an involvement for bArr2 on circadian regulation of G-protein coupled receptors in alcohol dependence. The presented data may have implications for the development of novel bArr2-related treatment targets for alcoholism.


Asunto(s)
Alcoholismo/genética , Ritmo Circadiano/genética , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/genética , Arrestina beta 2/genética , Alcoholismo/tratamiento farmacológico , Animales , Regulación hacia Abajo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Masculino , Análisis por Micromatrices , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Recompensa
2.
Alcohol Clin Exp Res ; 45(7): 1468-1478, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34273113

RESUMEN

BACKGROUND: Accumulating clinical evidence suggests that women with prior exposure to adverse childhood experiences are more susceptible to heavy drinking and other health-related behaviors. Yet, preclinical studies investigating sex-dependent effects of adolescent adverse social experiences (ASEs) on later alcohol-seeking behavior are lacking. This is mainly due to the unavailability of valid animal models and a shortage of studies that compare effects in males and females. Therefore, we sought to investigate the sex-dependent effects of ASE on adult alcohol-seeking behavior, locomotion, and reward sensitivity in male and female rats. METHODS: We recently developed a rat model for childhood/adolescent peer rejection that allows us to study the long-term consequences of ASEs. Adolescent Wistar rats were reared from postnatal day (pd) 21 to pd 50 either within a group of Fischer 344 rats (ASE) or within a group of Wistar rats (control). Wistar rats housed with Fischer 344 rats do not reciprocate social play in adolescence. This reduced play across adolescence mimics peer rejection and results in chronic dysregulation of social and pain-related behaviors. We tested adult male and female rats in the reinstatement paradigm for cue-induced alcohol-seeking behavior, circadian locomotor activity, and sucrose consumption long after the termination of the peer rejection condition. RESULTS: Peer rejection induced persistent sex-dependent changes in alcohol cue-induced reinstatement. Females showed an increased reinstatement effect while peer-rejected males demonstrated a decrease. Sex differences were observed in locomotor activity or reward sensitivity to sucrose. CONCLUSIONS: Peer rejection has long-lasting sex-dependent consequences on alcohol-seeking behavior without affecting locomotion or sweet reward sensitivity. Our results suggest that peer-rejected female rats represent a vulnerable population in which to study relapse-like behaviors that are similar to clinical findings, while males seem to buffer the peer rejection effect and demonstrate resilience to later life alcohol-seeking behaviors, as measured by the reinstatement effect. Finally, we provide a novel approach to investigate the molecular and neurobiological underpinnings of ASEs on alcohol and other drug-seeking behaviors.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Conducta Animal , Comportamiento de Búsqueda de Drogas , Etanol/administración & dosificación , Distancia Psicológica , Factores de Edad , Animales , Ritmo Circadiano , Femenino , Masculino , Actividad Motora , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Factores Sexuales , Sacarosa/administración & dosificación
3.
Addict Biol ; 26(2): e12945, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32761675

RESUMEN

A pronounced decrease of oxytocin and increase of oxytocin receptor binding sites were recently reported in male alcohol dependent rats and male alcohol dependent patients. Here we comment on this and emphasize that in female alcohol dependent rats and humans no changes occur in the oxytocin system. We therefore suggest specific intervention with oxytocin only in male subjects.


Asunto(s)
Alcoholismo/patología , Etanol/farmacología , Oxitocina/efectos de los fármacos , Receptores de Oxitocina/efectos de los fármacos , Animales , Femenino , Humanos , Masculino , Psicofarmacología , Ratas , Factores Sexuales
4.
Addict Biol ; 25(2): e12866, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31859437

RESUMEN

One of the major risk factors for global death and disability is alcohol, tobacco, and illicit drug use. While there is increasing knowledge with respect to individual factors promoting the initiation and maintenance of substance use disorders (SUDs), disease trajectories involved in losing and regaining control over drug intake (ReCoDe) are still not well described. Our newly formed German Collaborative Research Centre (CRC) on ReCoDe has an interdisciplinary approach funded by the German Research Foundation (DFG) with a 12-year perspective. The main goals of our research consortium are (i) to identify triggers and modifying factors that longitudinally modulate the trajectories of losing and regaining control over drug consumption in real life, (ii) to study underlying behavioral, cognitive, and neurobiological mechanisms, and (iii) to implicate mechanism-based interventions. These goals will be achieved by: (i) using mobile health (m-health) tools to longitudinally monitor the effects of triggers (drug cues, stressors, and priming doses) and modify factors (eg, age, gender, physical activity, and cognitive control) on drug consumption patterns in real-life conditions and in animal models of addiction; (ii) the identification and computational modeling of key mechanisms mediating the effects of such triggers and modifying factors on goal-directed, habitual, and compulsive aspects of behavior from human studies and animal models; and (iii) developing and testing interventions that specifically target the underlying mechanisms for regaining control over drug intake.


Asunto(s)
Terapia Conductista/métodos , Investigación Biomédica/métodos , Señales (Psicología) , Trastornos Relacionados con Sustancias/fisiopatología , Trastornos Relacionados con Sustancias/terapia , Telemedicina/métodos , Animales , Conducta Cooperativa , Modelos Animales de Enfermedad , Alemania , Humanos , Recurrencia , Trastornos Relacionados con Sustancias/psicología
5.
J Neurosci ; 38(14): 3507-3519, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29483279

RESUMEN

Cue-reward associations form distinct memories that can drive appetitive behaviors and are involved in craving for both drugs and natural rewards. Distinct sets of neurons, so-called neuronal ensembles, in the infralimbic area (IL) of the medial prefrontal cortex (mPFC) play a key role in alcohol seeking. Whether this ensemble is specific for alcohol or controls reward seeking in general remains unclear. Here, we compared IL ensembles formed upon recall of drug (alcohol) or natural reward (saccharin) memories in male Wistar rats. Using an experimental framework that allows identification of two distinct reward-associated ensembles within the same animal, we found that cue-induced seeking of either alcohol or saccharin activated ensembles of similar size and organization, whereby these ensembles consist of largely overlapping neuronal populations. Thus, the IL seems to act as a general integration hub for reward seeking behavior, but also contains subsets of neurons that encode for the different rewards.SIGNIFICANCE STATEMENT Cue-reward associations form distinct memories that can act as drivers of appetitive behaviors and are involved in craving for natural rewards as well as for drugs. Distinct sets of neurons, so-called neuronal ensembles, in the infralimbic area of the mPFC play a key role in cue-triggered reward seeking. However, it is unclear whether these ensembles act as broadly tuned controllers of approach behavior or represent the learned associations between specific cues and rewards. Using an experimental framework that allows identification of two distinct reward-associated ensembles within the same animal we find largely overlapping neuronal populations. Repeated activation by two distinct events could reflect the linking of the two memory traces within the same neuron.


Asunto(s)
Conducta de Elección , Comportamiento de Búsqueda de Drogas , Corteza Prefrontal/fisiología , Recompensa , Animales , Masculino , Neuronas/fisiología , Corteza Prefrontal/citología , Ratas , Ratas Wistar
6.
Proc Natl Acad Sci U S A ; 113(11): 3024-9, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26903621

RESUMEN

A major hypothesis in addiction research is that alcohol induces neuroadaptations in the mesolimbic dopamine (DA) system and that these neuroadaptations represent a key neurochemical event in compulsive drug use and relapse. Whether these neuroadaptations lead to a hypo- or hyperdopaminergic state during abstinence is a long-standing, unresolved debate among addiction researchers. The answer is of critical importance for understanding the neurobiological mechanism of addictive behavior. Here we set out to study systematically the neuroadaptive changes in the DA system during the addiction cycle in alcohol-dependent patients and rats. In postmortem brain samples from human alcoholics we found a strong down-regulation of the D1 receptor- and DA transporter (DAT)-binding sites, but D2-like receptor binding was unaffected. To gain insight into the time course of these neuroadaptations, we compared the human data with that from alcohol-dependent rats at several time points during abstinence. We found a dynamic regulation of D1 and DAT during 3 wk of abstinence. After the third week the rat data mirrored our human data. This time point was characterized by elevated extracellular DA levels, lack of synaptic response to D1 stimulation, and augmented motor activity. Further functional evidence is given by a genetic rat model for hyperdopaminergia that resembles a phenocopy of alcohol-dependent rats during protracted abstinence. In summary, we provide a new dynamic model of abstinence-related changes in the striatal DA system; in this model a hyperdopaminergic state during protracted abstinence is associated with vulnerability for relapse.


Asunto(s)
Abstinencia de Alcohol , Alcoholismo/metabolismo , Dopamina/fisiología , Etanol/efectos adversos , Síndrome de Abstinencia a Sustancias/metabolismo , Ácido 3,4-Dihidroxifenilacético/análisis , Adulto , Anciano , Animales , Benzazepinas/farmacología , Química Encefálica , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Etanol/toxicidad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Ácido Homovanílico/análisis , Humanos , Masculino , Persona de Mediana Edad , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Ratas Transgénicas , Ratas Wistar , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Recurrencia , Transcripción Genética
7.
J Neurosci ; 36(50): 12611-12623, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27810934

RESUMEN

PPARγ is one of the three isoforms of the Peroxisome Proliferator-Activated Receptors (PPARs). PPARγ is activated by thiazolidinediones such as pioglitazone and is targeted to treat insulin resistance. PPARγ is densely expressed in brain areas involved in regulation of motivational and emotional processes. Here, we investigated the role of PPARγ in the brain and explored its role in anxiety and stress responses in mice. The results show that stimulation of PPARγ by pioglitazone did not affect basal anxiety, but fully prevented the anxiogenic effect of acute stress. Using mice with genetic ablation of neuronal PPARγ (PPARγNestinCre), we demonstrated that a lack of receptors, specifically in neurons, exacerbated basal anxiety and enhanced stress sensitivity. The administration of GW9662, a selective PPARγ antagonist, elicited a marked anxiogenic response in PPARγ wild-type (WT), but not in PPARγNestinCre knock-out (KO) mice. Using c-Fos immunohistochemistry, we observed that acute stress exposure resulted in a different pattern of neuronal activation in the amygdala (AMY) and the hippocampus (HIPP) of PPARγNestinCre KO mice compared with WT mice. No differences were found between WT and KO mice in hypothalamic regions responsible for hormonal response to stress or in blood corticosterone levels. Microinjection of pioglitazone into the AMY, but not into the HIPP, abolished the anxiogenic response elicited by acute stress. Results also showed that, in both regions, PPARγ colocalizes with GABAergic cells. These findings demonstrate that neuronal PPARγ is involved the regulation of the stress response and that the AMY is a key substrate for the anxiolytic effect of PPARγ. SIGNIFICANCE STATEMENT: Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) is a classical target for antidiabetic therapies with thiazolidinedione compounds. PPARγ agonists such as rosiglitazone and pioglitazone are in clinical use for the treatment of insulin resistance. PPARγ has recently attracted attention for its involvement in the regulation of CNS immune response and functions. Here, we demonstrate that neuronal PPARγ activation prevented the negative emotional effects of stress and exerted anxiolytic actions without influencing hypothalamic-pituitary-adrenal axis function. Conversely, pharmacological blockade or genetic deletion of PPARγ enhanced anxiogenic responses and increased vulnerability to stress. These effects appear to be controlled by PPARγ neuronal-mediated mechanisms in the amygdala.


Asunto(s)
Amígdala del Cerebelo/fisiología , Ansiedad/genética , Ansiedad/psicología , Emociones/fisiología , PPAR gamma/genética , Estrés Psicológico/genética , Estrés Psicológico/psicología , Anilidas/farmacología , Animales , Corticosterona/sangre , Conducta Exploratoria/efectos de los fármacos , Glutamato Descarboxilasa/genética , Hipocampo/citología , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Microinyecciones , Neuronas/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Pioglitazona , Tiazolidinedionas/administración & dosificación , Tiazolidinedionas/farmacología
9.
J Neurosci ; 35(30): 10750-61, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26224858

RESUMEN

Loss of control over drinking is a key deficit in alcoholism causally associated with malfunction of the medial prefrontal cortex (mPFC), but underlying molecular and cellular mechanisms remain unclear. Cue-induced reinstatement of alcohol seeking activates a subset of mPFC neurons in rats, identified by their common expression of the activity marker cFos and comprised of both principal and interneurons. Here, we used cFos-lacZ and pCAG-lacZ transgenic rats for activity-dependent or nonselective inactivation of neurons, respectively, which by their lacZ encoded ß-galactosidase activity convert the inactive prodrug Daun02 into the neurotoxin daunorubicin. We report that activity-dependent ablation of a neuronal ensemble in the infralimbic but not the prelimbic subregion induced excessive alcohol seeking. The targeted neuronal ensemble was specific for the cue-induced response because stress-induced reinstatement was not affected in these animals. Importantly, nonselective inactivation of infralimbic neurons, using pCAG-lacZ rats, was without functional consequence on the cue-induced reinstatement task. Thus, inhibitory control over alcohol seeking is exerted by distinct functional ensembles within the infralimbic cortex rather than by a general inhibitory tone of this region on the behavioral output. This indicates a high level of functional compartmentation within the rat mPFC whereat many functional ensembles could coexist and interact within the same subregion. SIGNIFICANCE STATEMENT: Hebb's (1949) idea of memories as being represented in local neuronal networks is supported by identification of transiently stable activity patterns within subgroups of neurons. However, it is difficult to link individual networks to specific memory tasks, for example a learned behavior. By a novel approach of activity-dependent ablation, here we identify a specific neuronal ensemble located in the infralimbic subregion of the medial prefrontal cortex that controls a seeking response for alcohol in rats. Our data demonstrate that functional output depends on specific neuronal ensembles within a given brain region rather than on the global activity of that region, which raises important questions about the interpretation of numerous earlier experiments using site-directed silencing or stimulation for elucidating brain function.


Asunto(s)
Señales (Psicología) , Comportamiento de Búsqueda de Drogas/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Alcoholismo/fisiopatología , Animales , Extinción Psicológica , Inmunohistoquímica , Masculino , Ratas , Ratas Transgénicas
10.
J Neurosci ; 34(5): 1781-90, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24478360

RESUMEN

The incubation of cocaine craving describes the time-dependent augmentation of cue-induced cocaine seeking during withdrawal from prolonged cocaine self-administration and requires time-dependent changes in neuroplasticity at the level of glutamatergic synapses in the nucleus accumbens (NAc). In contrast to most studies that use multiple cocaine-cue conditioning sessions, the present study tested mice with limited cocaine experience (i.e., a single conditioning session) in the incubation of cue-mediated cocaine seeking and its associated changes in the glutamate system. Mice that self-administered cocaine during a single session exhibited a time-dependent increase in their response for the drug-associated cue as compared to mice that self-administered saline. This behavior was associated with changes in AMPA and NMDA receptor binding characteristics. Furthermore, Group I metabotropic glutamate receptor (mGluR1) mRNA levels were altered in several brain regions, including the NAc. Because of the pivotal role of mGluR1 in the control of cocaine-induced plasticity, we investigated the role of mGluR1 in the formation of drug cue-mediated cocaine seeking. After prolonged withdrawal, mice in which an mGluR1 antagonist was administered following cocaine self-administration displayed increased cocaine seeking compared to vehicle-treated mice. These results suggest that limited cocaine experience is sufficient to induce neurobiological changes that enable an initially neutral cue to acquire motivational value that increases over time, an effect that likely involves glutamate signaling through mGluR1.


Asunto(s)
Cocaína/administración & dosificación , Condicionamiento Operante/fisiología , Inhibidores de Captación de Dopamina/administración & dosificación , Comportamiento de Búsqueda de Drogas/fisiología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacocinética , Antagonistas de Aminoácidos Excitadores/farmacocinética , Extinción Psicológica/efectos de los fármacos , Alimentos , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Autoadministración , Factores de Tiempo , Tritio/farmacocinética , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacocinética
11.
J Neurosci ; 34(2): 363-72, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24403138

RESUMEN

Corticotropin releasing factor (CRF) is the primary mediator of stress responses, and nociceptin/orphanin FQ (N/OFQ) plays an important role in the modulation of these stress responses. Thus, in this multidisciplinary study, we explored the relationship between the N/OFQ and the CRF systems in response to stress. Using in situ hybridization (ISH), we assessed the effect of body restraint stress on the gene expression of CRF and N/OFQ-related genes in various subdivisions of the amygdala, a critical brain structure involved in the modulation of stress response and anxiety-like behaviors. We found a selective upregulation of the NOP and downregulation of the CRF1 receptor transcripts in the CeA and in the BLA after body restraint. Thus, we performed intracellular electrophysiological recordings of GABAA-mediated IPSPs in the central nucleus of the amygdala (CeA) to explore functional interactions between CRF and N/OFQ systems in this brain region. Acute application of CRF significantly increased IPSPs in the CeA, and this enhancement was blocked by N/OFQ. Importantly, in stress-restraint rats, baseline CeA GABAergic responses were elevated and N/OFQ exerted a larger inhibition of IPSPs compared with unrestraint rats. The NOP antagonist [Nphe1]-nociceptin(1-13)NH2 increased the IPSP amplitudes in restraint rats but not in unrestraint rats, suggesting a functional recruitment of the N/OFQ system after acute stress. Finally, we evaluated the anxiety-like response in rats subjected to restraint stress and nonrestraint rats after N/OFQ microinjection into the CeA. Intra-CeA injections of N/OFQ significantly and selectively reduced anxiety-like behavior in restraint rats in the elevated plus maze. These combined results demonstrate that acute stress increases N/OFQ systems in the CeA and that N/OFQ has antistress properties.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Conducta Animal/fisiología , Hormona Liberadora de Corticotropina/metabolismo , Péptidos Opioides/metabolismo , Animales , Modelos Animales de Enfermedad , Hibridación in Situ , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Restricción Física , Estrés Psicológico/metabolismo , Transcriptoma , Nociceptina
12.
Alcohol Clin Exp Res ; 39(4): 752-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25833034

RESUMEN

BACKGROUND: One of the most commonly used approaches to induce ethanol (EtOH) dependence in rodents is EtOH vapor inhalation. This procedure requires the co-administration of pyrazole-an inhibitor of the alcohol dehydrogenase-to obtain stable blood EtOH concentrations (BECs) during the entire induction course. However, pyrazole can produce unwanted side effects. Our goal was to obtain EtOH-dependent mice without pyrazole and to study their behavioral and molecular postdependent phenotype. In particular, we were interested in alterations in the corticotrophin-releasing hormone (CRH) and receptor (CRHR1 and CRHR2) system as a prominent role of CRH in driving the postdependent state via actions in the central extended amygdala (CeA) has been demonstrated in rats but not in postdependent mice. METHODS: We established an alternative model of chronic intermittent EtOH (CIE) inhalation without the use of pyrazole in C57BL/6N mice. Our CIE exposure protocol involved 8 cycles. One cycle consisted of 8 hours with EtOH inhalation and 8 hours without EtOH. We then examined withdrawal symptoms. After 2 weeks of abstinence, we studied relapse, reinstatement of EtOH-seeking, and stress-induced EtOH self-administration. We also did transcriptional analysis of components of the CRH system during CIE, protracted abstinence, and after stress-induced EtOH self-administration. RESULTS: CIE exposure without pyrazole resulted in reproducible BECs during the induction procedure. Mice showed strong withdrawal scores during 4 to 12 hours after the last CIE cycle and enhanced stress-induced EtOH self-administration. This postdependent phenotype during abstinence was accompanied by enhanced Crh and Crhr1 transcripts but no change in Crhr2 transcripts in the CeA. Cue-induced EtOH-seeking behavior and relapse (alcohol deprivation effect) were not affected by the inhalation procedure. CONCLUSIONS: We have established a CIE inhalation protocol without pyrazole in mice and showed excessive EtOH self-administration under mild stress and enhanced CRH/CRHR1 signaling in the CeA.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Etanol/administración & dosificación , Etanol/farmacología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Administración por Inhalación , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Modelos Animales de Enfermedad , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Masculino , Ratones , Recurrencia , Síndrome de Abstinencia a Sustancias/metabolismo
13.
J Neurosci ; 33(7): 2794-806, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407939

RESUMEN

A key deficit in alcohol dependence is disrupted prefrontal function leading to excessive alcohol seeking, but the molecular events underlying the emergence of addictive responses remain unknown. Here we show by convergent transcriptome analysis that the pyramidal neurons of the infralimbic cortex are particularly vulnerable for the long-term effects of chronic intermittent ethanol intoxication. These neurons exhibit a pronounced deficit in metabotropic glutamate receptor subtype 2 (mGluR(2)). Also, alcohol-dependent rats do not respond to mGluR(2/3) agonist treatment with reducing extracellular glutamate levels in the nucleus accumbens. Together these data imply a loss of autoreceptor feedback control. Alcohol-dependent rats show escalation of ethanol seeking, which was abolished by restoring mGluR(2) expression in the infralimbic cortex via viral-mediated gene transfer. Human anterior cingulate cortex from alcoholic patients shows a significant reduction in mGluR(2) transcripts compared to control subjects, suggesting that mGluR(2) loss in the rodent and human corticoaccumbal neurocircuitry may be a major consequence of alcohol dependence and a key pathophysiological mechanism mediating increased propensity to relapse. Normalization of mGluR(2) function within this brain circuit may be of therapeutic value.


Asunto(s)
Alcoholismo/psicología , Comportamiento de Búsqueda de Drogas/fisiología , Sistema Límbico/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Alcoholismo/genética , Alcoholismo/fisiopatología , Animales , Conducta Animal/fisiología , Encéfalo/patología , Depresores del Sistema Nervioso Central/farmacología , Manual Diagnóstico y Estadístico de los Trastornos Mentales , Regulación hacia Abajo/fisiología , Etanol/farmacología , Expresión Génica , Vectores Genéticos , Humanos , Inmunohistoquímica , Hibridación in Situ , Lentivirus/genética , Masculino , Análisis por Micromatrices , ARN/biosíntesis , ARN/genética , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glutamato Metabotrópico/deficiencia , Receptores de Glutamato Metabotrópico/genética , Síndrome de Abstinencia a Sustancias/psicología
14.
Nicotine Tob Res ; 16(6): 774-85, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24470632

RESUMEN

INTRODUCTION: L-type calcium channel (LTCC) activity in the brain is mediated by 2 subtypes, Ca(v)1.2 and Ca(v)1.3. The individual contributions of these LTCC subtypes to the long-term pharmacological and behavioral effects of nicotine are unknown. METHODS: Using quantitative in situ hybridization, we examined expression levels of Ca(v)1.2 and Ca(v)1.3 in forebrain regions of mice treated with nicotine (0.175 mg/kg) or saline for 1 or 14 days and sacrificed 24 hr or 7 days following the last injection. Additionally, we treated mice with nicotine for 14 days and then administered the nonspecific LTCC antagonist nifedipine twice daily during a 7-day abstinence period prior to testing for nicotine sensitization to determine the effect of LTCC blockade on sensitization. RESULTS: Ca(v)1.2 mRNA was unaffected 24 hr following a single nicotine exposure, whereas Ca(v)1.3 mRNA was upregulated in several brain regions. Following 14 days of nicotine treatment and 24 hr of abstinence, Ca(v)1.2 mRNA was downregulated throughout the areas examined, whereas Ca(v)1.3 mRNA had mostly returned to control values. Following 7 days of abstinence, a strong upregulation of Ca(v)1.2 transcripts was observed, whereas Ca(v)1.3 mRNA was largely unaffected. In our sensitization study, nifedipine administered during nicotine abstinence impaired subsequent nicotine sensitization. CONCLUSIONS: Our data suggest a differential involvement of Ca(v)1.2 and Ca(v)1.3 in nicotine-related processes. Ca(v)1.3 seems to be involved primarily during early exposure to nicotine. Ca(v)1.2 appears to play a role in the long-term molecular and behavioral changes that occur following chronic nicotine and abstinence. Nifedipine may counteract those nicotine-induced alterations in LTCC activity to impair nicotine sensitization.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Nicotina/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Nifedipino/farmacología , Prosencéfalo/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
15.
Alcohol Clin Exp Res (Hoboken) ; 48(2): 250-259, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38276909

RESUMEN

BACKGROUND: Alcohol use disorder (AUD) is associated with increased mortality and morbidity risk. A reason for this could be accelerated biological aging, which is strongly influenced by disease processes such as inflammation. As recent studies of AUD show changes in DNA methylation and gene expression in neuroinflammation-related pathways in the brain, biological aging represents a potentially important construct for understanding the adverse effects of substance use disorders. Epigenetic clocks have shown accelerated aging in blood samples from individuals with AUD. However, no systematic evaluation of biological age measures in AUD across different tissues and brain regions has been undertaken. METHODS: As markers of biological aging (BioAge markers), we assessed Levine's and Horvath's epigenetic clocks, DNA methylation telomere length (DNAmTL), telomere length (TL), and mitochondrial DNA copy number (mtDNAcn) in postmortem brain samples from Brodmann Area 9 (BA9), caudate nucleus, and ventral striatum (N = 63-94), and in whole blood samples (N = 179) of individuals with and without AUD. To evaluate the association between AUD status and BioAge markers, we performed linear regression analyses while adjusting for covariates. RESULTS: The majority of BioAge markers were significantly associated with chronological age in all samples. Levine's epigenetic clock and DNAmTL were indicative of accelerated biological aging in AUD in BA9 and whole blood samples, while Horvath's showed the opposite effect in BA9. No significant association of AUD with TL and mtDNAcn was detected. Measured TL and DNAmTL showed only small correlations in blood and none in brain. CONCLUSIONS: The present study is the first to simultaneously investigate epigenetic clocks, telomere length, and mtDNAcn in postmortem brain and whole blood samples in individuals with AUD. We found evidence for accelerated biological aging in AUD in blood and brain, as measured by Levine's epigenetic clock, and DNAmTL. Additional studies of different tissues from the same individuals are needed to draw valid conclusions about the congruence of biological aging in blood and brain.

16.
Alcohol Clin Exp Res (Hoboken) ; 47(1): 76-86, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36774629

RESUMEN

BACKGROUND: Chronic alcohol consumption and alcohol use disorder have a tremendous impact on the patient's psychological and physiological health. There is evidence that chronic alcohol consumption influences SARS-CoV2 infection risk, but so far, the molecular mechanism underlying such an effect is unknown. METHODS: We generated the expression data of SARS-CoV2 infection-relevant genes (Ace2, Tmprss2, and Mas) in different organs in rat models of chronic alcohol exposure and alcohol dependence. Ace2 and Tmprss2 represent the virus entry point, whereas Mas activates the anti-inflammatory response once the cells are infected. RESULTS: Across three different chronic alcohol test conditions, we found a consistent upregulation of Ace2 gene expression in the lung, which has been shown to be the most affected organ in COVID-19 patients. Other organs such as liver, ileum, kidney, heart, and brain also showed upregulation of Ace2 and Mas gene expression but less consistently across the different animal models, while Tmprss2 expression was unaffected in all conditions. CONCLUSIONS: We conclude that alcohol-induced upregulation of Ace2 gene expression can lead to an elevated stochastic probability of virus entry into cells and may thus confer a molecular risk for SARS-CoV2 infection.


Asunto(s)
COVID-19 , Ratas , Animales , Enzima Convertidora de Angiotensina 2 , ARN Viral , SARS-CoV-2 , Consumo de Bebidas Alcohólicas
17.
Behav Brain Res ; 438: 114180, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36349601

RESUMEN

Adolescence is a critical time of social learning in which both the quantity and quality of social interactions shape adult behavior and social function. During adolescence, social instability such as disrupting or limiting social interactions can lead to negative life-long effects on mental health and well-being in humans. Animal models on social instability are critically important in understanding those underlying neurobiological mechanisms. However, studies in rats using these models have produced partly inconsistent results and can be difficult to generalize. Here we assessed in a sex and age consistent manner the long-term behavioural consequences of social instability stress (SIS - 1-hr daily isolation and change in cage mate between postnatal day (PD30-45)) in Wistar rats. Female and male rats underwent a battery of tests for anxiety-like, exploratory, and social behaviour over five days beginning either in adolescence (PD46) or in adulthood (PD70). Social instability led to reduced anxiety-like behaviour in the elevated plus maze in both sexes in adolescence and in adulthood. Social interactions were also reduced in rats that underwent SIS - an effect that was independent of sex and age when tested. SIS improved social recognition memory in both sexes whereas a sex-dependent effect was seen in the social novelty preference test where male rats that underwent SIS spent more time in social approach toward a novel peer than toward their cage mate. In comparison, control male and female groups did not differ in this test, in time spent with novel versus the cage mate. Thus, overall, social instability stress in Wistar rats altered the behavioural repertoire, with enduring alterations in social behaviour, enhanced exploratory behaviour, and reduced anxiety-like behaviour. In conclusion, the social instability stress paradigm may better be interpreted as a form of enrichment in Wistar rats than as a stressor.


Asunto(s)
Ansiedad , Estrés Psicológico , Humanos , Ratas , Masculino , Femenino , Animales , Adulto , Ratas Wistar , Conducta Social , Conducta Exploratoria , Conducta Animal
18.
Front Psychiatry ; 14: 1075250, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36865068

RESUMEN

Background: Cocaine use disorder (CUD) is characterized by a loss of control over cocaine intake and is associated with structural, functional, and molecular alterations in the human brain. At the molecular level, epigenetic alterations are hypothesized to contribute to the higher-level functional and structural brain changes observed in CUD. Most evidence of cocaine-associated epigenetic changes comes from animal studies while only a few studies have been performed using human tissue. Methods: We investigated epigenome-wide DNA methylation (DNAm) signatures of CUD in human post-mortem brain tissue of Brodmann area 9 (BA9). A total of N = 42 BA9 brain samples were obtained from N = 21 individuals with CUD and N = 21 individuals without a CUD diagnosis. We performed an epigenome-wide association study (EWAS) and analyzed CUD-associated differentially methylated regions (DMRs). To assess the functional role of CUD-associated differential methylation, we performed Gene Ontology (GO) enrichment analyses and characterized co-methylation networks using a weighted correlation network analysis. We further investigated epigenetic age in CUD using epigenetic clocks for the assessment of biological age. Results: While no cytosine-phosphate-guanine (CpG) site was associated with CUD at epigenome-wide significance in BA9, we detected a total of 20 CUD-associated DMRs. After annotation of DMRs to genes, we identified Neuropeptide FF Receptor 2 (NPFFR2) and Kalirin RhoGEF Kinase (KALRN) for which a previous role in the behavioral response to cocaine in rodents is known. Three of the four identified CUD-associated co-methylation modules were functionally related to neurotransmission and neuroplasticity. Protein-protein interaction (PPI) networks derived from module hub genes revealed several addiction-related genes as highly connected nodes such as Calcium Voltage-Gated Channel Subunit Alpha1 C (CACNA1C), Nuclear Receptor Subfamily 3 Group C Member 1 (NR3C1), and Jun Proto-Oncogene, AP-1 Transcription Factor Subunit (JUN). In BA9, we observed a trend toward epigenetic age acceleration (EAA) in individuals with CUD remaining stable even after adjustment for covariates. Conclusion: Results from our study highlight that CUD is associated with epigenome-wide differences in DNAm levels in BA9 particularly related to synaptic signaling and neuroplasticity. This supports findings from previous studies that report on the strong impact of cocaine on neurocircuits in the human prefrontal cortex (PFC). Further studies are needed to follow up on the role of epigenetic alterations in CUD focusing on the integration of epigenetic signatures with transcriptomic and proteomic data.

19.
Addict Biol ; 17(5): 827-64, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22978651

RESUMEN

The identification and functional understanding of the neurocircuitry that mediates alcohol and drug effects that are relevant for the development of addictive behavior is a fundamental challenge in addiction research. Here we introduce an assumption-free construction of a neurocircuitry that mediates acute and chronic drug effects on neurotransmitter dynamics that is solely based on rodent neuroanatomy. Two types of data were considered for constructing the neurocircuitry: (1) information on the cytoarchitecture and neurochemical connectivity of each brain region of interest obtained from different neuroanatomical techniques; (2) information on the functional relevance of each region of interest with respect to alcohol and drug effects. We used mathematical data mining and hierarchical clustering methods to achieve the highest standards in the preprocessing of these data. Using this approach, a dynamical network of high molecular and spatial resolution containing 19 brain regions and seven neurotransmitter systems was obtained. Further graph theoretical analysis suggests that the neurocircuitry is connected and cannot be separated into further components. Our analysis also reveals the existence of a principal core subcircuit comprised of nine brain regions: the prefrontal cortex, insular cortex, nucleus accumbens, hypothalamus, amygdala, thalamus, substantia nigra, ventral tegmental area and raphe nuclei. Finally, by means of algebraic criteria for synchronizability of the neurocircuitry, the suitability for in silico modeling of acute and chronic drug effects is indicated. Indeed, we introduced as an example a dynamical system for modeling the effects of acute ethanol administration in rats and obtained an increase in dopamine release in the nucleus accumbens-a hallmark of drug reinforcement-to an extent similar to that seen in numerous microdialysis studies. We conclude that the present neurocircuitry provides a structural and dynamical framework for large-scale mathematical models and will help to predict chronic drug effects on brain function.


Asunto(s)
Conducta Adictiva/fisiopatología , Encéfalo/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Neurotransmisores/fisiología , Trastornos Relacionados con Sustancias/fisiopatología , Algoritmos , Animales , Encéfalo/anatomía & histología , Encéfalo/fisiología , Hormona Liberadora de Corticotropina/fisiología , Modelos Animales de Enfermedad , Glutamina/fisiología , Humanos , Vías Nerviosas/fisiología , Norepinefrina/fisiología , Ratas , Receptores de GABA/efectos de los fármacos , Refuerzo en Psicología , Serotonina/fisiología , Transmisión Sináptica/fisiología
20.
J Pers Med ; 12(4)2022 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-35455681

RESUMEN

(1) Background: Epigenome-wide association studies (EWAS) in peripheral blood have repeatedly found associations between tobacco smoking and aberrant DNA methylation (DNAm), but little is known about DNAm signatures of smoking in the human brain, which may contribute to the pathophysiology of addictive behavior observed in chronic smokers. (2) Methods: We investigated the similarity of DNAm signatures in matched blood and postmortem brain samples (n = 10). In addition, we performed EWASs in five brain regions belonging to the neurocircuitry of addiction: anterior cingulate cortex (ACC), Brodmann Area 9, caudate nucleus, putamen, and ventral striatum (n = 38-72). (3) Results: cg15925993 within the LOC339975 gene was epigenome-wide significant in the ACC. Of 16 identified differentially methylated regions, two (PRSS50 and LINC00612/A2M-AS1) overlapped between multiple brain regions. Functional enrichment was detected for biological processes related to neuronal development, inflammatory signaling and immune cell migration. Additionally, our results indicate the association of the well-known AHRR CpG site cg05575921 with smoking in the brain. (4) Conclusion: The present study provides further evidence of the strong relationship between aberrant DNAm and smoking.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA