Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(35): 9469-9474, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808012

RESUMEN

Chronic cocaine use is associated with prominent morphological changes in nucleus accumbens shell (NACsh) neurons, including increases in dendritic spine density along with enhanced motivation for cocaine, but a functional relationship between these morphological and behavioral phenomena has not been shown. Here we show that brain-derived neurotrophic factor (BDNF) signaling through tyrosine kinase B (TrkB) receptors in NACsh neurons is necessary for cocaine-induced dendritic spine formation by using either localized TrkB knockout or viral-mediated expression of a dominant negative, kinase-dead TrkB mutant. Interestingly, augmenting wild-type TrkB expression after chronic cocaine self-administration reverses the sustained increase in dendritic spine density, an effect mediated by TrkB signaling pathways that converge on extracellular regulated kinase. Loss of TrkB function after cocaine self-administration, however, leaves spine density intact but markedly enhances the motivation for cocaine, an effect mediated by specific loss of TrkB signaling through phospholipase Cgamma1 (PLCγ1). Conversely, overexpression of PLCγ1 both reduces the motivation for cocaine and reverses dendritic spine density, suggesting a potential target for the treatment of addiction in chronic users. Together, these findings indicate that BDNF-TrkB signaling both mediates and reverses cocaine-induced increases in dendritic spine density in NACsh neurons, and these morphological changes are entirely dissociable from changes in addictive behavior.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos Relacionados con Cocaína , Cocaína/farmacología , Espinas Dendríticas/efectos de los fármacos , Núcleo Accumbens/fisiología , Receptor trkB/metabolismo , Animales , Antralina , Células HEK293 , Humanos , Masculino , Neuronas/fisiología , Núcleo Accumbens/citología , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética , Transducción de Señal
2.
J Neurosci ; 38(4): 803-813, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29217682

RESUMEN

Repeated exposure to cocaine induces lasting epigenetic changes in neurons that promote the development and persistence of addiction. One epigenetic alteration involves reductions in levels of the histone dimethyltransferase G9a in nucleus accumbens (NAc) after chronic cocaine administration. This reduction in G9a may enhance cocaine reward because overexpressing G9a in the NAc decreases cocaine-conditioned place preference. Therefore, we hypothesized that HSV-mediated G9a overexpression in the NAc shell (NAcSh) would attenuate cocaine self-administration (SA) and cocaine-seeking behavior. Instead, we found that G9a overexpression, and the resulting increase in histone 3 lysine 9 dimethylation (H3K9me2), increases sensitivity to cocaine reinforcement and enhances motivation for cocaine in self-administering male rats. Moreover, when G9a overexpression is limited to the initial 15 d of cocaine SA training, it produces an enduring postexpression enhancement in cocaine SA and prolonged (over 5 weeks) increases in reinstatement of cocaine seeking induced by foot-shock stress, but in the absence of continued global elevations in H3K9me2. The increase in stress-induced reinstatement is paralleled by heightened anxiety measures, suggesting that countering the cocaine-induced decreases in endogenous G9a with ectopic G9a overexpression leads to lasting anxiogenic effects. Finally, we found an enduring reduction in phosphorylated cAMP-response element binding protein levels in the NAcSh that could account for the increased anxiety. These data demonstrate a novel role for G9a in promoting comorbid cocaine addiction and anxiety and suggest that increased epigenetic repression of transcription through H3K9 during cocaine use can have long-lasting and unexpected negative consequences on behavior.SIGNIFICANCE STATEMENT Cocaine addiction is a neuropsychiatric disorder that is detrimental to society and currently has no effective treatments. The difficulty in treating drug addiction is compounded by the high comorbidity with other psychiatric illnesses, including anxiety disorders. Here, we demonstrate that G9a, an epigenetic repressor of gene expression, acting in the nucleus accumbens, a brain reward region, is capable of increasing both addiction- and anxiety-like behaviors in rats. These findings are intriguing because repeated cocaine exposure decreases G9a in this region and thereby enhances expression of certain addiction-promoting genes. However, our results suggest that countering this cocaine-induced decrease in G9a activity actually exacerbates addiction and sensitivity to relapse under stressful situations.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/biosíntesis , Núcleo Accumbens/metabolismo , Animales , Ansiedad/etiología , Ansiedad/metabolismo , Cocaína/farmacología , Condicionamiento Operante , Inhibidores de Captación de Dopamina/farmacología , Comportamiento de Búsqueda de Drogas/fisiología , Epigénesis Genética/fisiología , Extinción Psicológica , Regulación de la Expresión Génica/fisiología , Histonas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Autoadministración
3.
J Neurosci ; 38(3): 575-585, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29196318

RESUMEN

Cocaine self-administration increases expression of GluA1 subunits in ventral tegmental area (VTA) dopamine neurons, which subsequently enhance the motivation for cocaine. This increase in GluA1 may be dependent on concomitant NMDA receptor (NMDAR) activation during self-administration, similar to cocaine-induced long-term potentiation in the VTA. In this study, we used viral-mediated expression of a dominant-negative GluN1 subunit (HSV-dnGluN1) in VTA neurons to study the effect of transient NMDAR inactivation on the GluA1 increases induced by chronic cocaine self-administration in male rats. We found that dnGluN1 expression in the VTA limited to the 3 weeks of cocaine self-administration prevents the subsequent increase in tissue GluA1 levels when compared with control infusions of HSV-LacZ. Surprisingly, dnGluN1 expression led to an enhancement in the motivation to self-administer cocaine as measured using a progressive ratio reinforcement schedule and to enhanced cocaine seeking measured in extinction/reinstatement tests following an extended 3 week withdrawal period. Despite blocking tissue GluA1 increases in cocaine self-administering animals, the HSV-dnGluN1 treatment resulted in increased membrane levels of GluA1 and GluN2B, along with markedly higher locomotor responses to intra-VTA infusions of AMPA, suggesting a paradoxical increase in VTA AMPA receptor responsiveness. Together, these data suggest that NMDARs mediate cocaine-induced increases in VTA GluA1 expression, but such transient NMDAR inactivation also leads to compensatory scaling of synaptic AMPA receptors that enhance the motivational for cocaine.SIGNIFICANCE STATEMENT Dopamine neurons in the ventral tegmental area (VTA) are critical substrates of drug rewards. Animal models indicate that chronic cocaine use enhances excitatory glutamatergic input to these neurons, making them more susceptible to environmental stimuli that trigger drug craving and relapse. We previously found that self-administration of cocaine increases AMPA glutamate receptors in the VTA, and this effect enhances motivation for cocaine. Here we report that the mechanism for this upregulation involves NMDA receptor activity during cocaine use. While interference with NMDA receptor function blocks AMPA receptor upregulation, it also produces a paradoxical enhancement in membrane AMPA receptor subunits, AMPA responsiveness, and the motivation for cocaine. Thus, pharmacotherapy targeting NMDA receptors may inadvertently produce substantial adverse consequences for cocaine addiction.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Cocaína/farmacología , Trastornos Relacionados con Cocaína/fisiopatología , Inhibidores de Captación de Dopamina/farmacología , Comportamiento de Búsqueda de Drogas/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Autoadministración , Regulación hacia Arriba , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiopatología
4.
J Neurosci ; 35(8): 3537-43, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716852

RESUMEN

Previous studies suggest that pharmacological or molecular activation of the nucleus accumbens shell (AcbSh) facilitates extinction of cocaine-seeking behavior. However, overexpression of CREB, which increases excitability of AcbSh neurons, enhances cocaine-seeking behavior while producing depression-like behavior in tests of mood. These discrepancies may reflect activity in differential AcbSh outputs, including those to the lateral hypothalamus (LH), a target region known to influence addictive behavior and mood. Presently, it is unknown whether there is a causal link between altered activity in the AcbSh-LH pathway and changes in the motivation for cocaine. In this study, we used an optogenetics approach to either globally stimulate AcbSh neurons or to selectively stimulate AcbSh terminal projections in the LH, in rats self-administering cocaine. We found that stimulation of the AcbSh-LH pathway enhanced the motivation to self-administer cocaine in progressive ratio testing, and led to long-lasting facilitation of cocaine-seeking behavior during extinction tests conducted after withdrawal from cocaine self-administration. In contrast, global AcbSh stimulation reduced extinction responding. We compared these opposing motivational effects with effects on mood using the forced swim test, where both global AcbSh neuron and selective AcbSh-LH terminal stimulation facilitated depression-like behavioral despair. Together, these findings suggest that the AcbSh neurons convey complex, pathway-specific modulation of addiction and depression-like behavior, and that these motivation and mood phenomenon are dissociable.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Comportamiento de Búsqueda de Drogas , Área Hipotalámica Lateral/fisiopatología , Motivación , Núcleo Accumbens/fisiopatología , Afecto , Animales , Cocaína/farmacología , Extinción Psicológica , Área Hipotalámica Lateral/citología , Masculino , Vías Nerviosas/citología , Vías Nerviosas/fisiopatología , Neuronas/fisiología , Núcleo Accumbens/citología , Optogenética , Ratas , Ratas Sprague-Dawley
5.
J Neurosci ; 33(47): 18381-95, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24259563

RESUMEN

The transcription factor, ΔFosB, is robustly and persistently induced in striatum by several chronic stimuli, such as drugs of abuse, antipsychotic drugs, natural rewards, and stress. However, very few studies have examined the degree of ΔFosB induction in the two striatal medium spiny neuron (MSN) subtypes. We make use of fluorescent reporter BAC transgenic mice to evaluate induction of ΔFosB in dopamine receptor 1 (D1) enriched and dopamine receptor 2 (D2) enriched MSNs in ventral striatum, nucleus accumbens (NAc) shell and core, and in dorsal striatum (dStr) after chronic exposure to several drugs of abuse including cocaine, ethanol, Δ(9)-tetrahydrocannabinol, and opiates; the antipsychotic drug, haloperidol; juvenile enrichment; sucrose drinking; calorie restriction; the serotonin selective reuptake inhibitor antidepressant, fluoxetine; and social defeat stress. Our findings demonstrate that chronic exposure to many stimuli induces ΔFosB in an MSN-subtype selective pattern across all three striatal regions. To explore the circuit-mediated induction of ΔFosB in striatum, we use optogenetics to enhance activity in limbic brain regions that send synaptic inputs to NAc; these regions include the ventral tegmental area and several glutamatergic afferent regions: medial prefrontal cortex, amygdala, and ventral hippocampus. These optogenetic conditions lead to highly distinct patterns of ΔFosB induction in MSN subtypes in NAc core and shell. Together, these findings establish selective patterns of ΔFosB induction in striatal MSN subtypes in response to chronic stimuli and provide novel insight into the circuit-level mechanisms of ΔFosB induction in striatum.


Asunto(s)
Cuerpo Estriado/citología , Dopaminérgicos/farmacología , Emociones/efectos de los fármacos , Optogenética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Antidepresivos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Ambiente , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/clasificación , Neuronas/efectos de los fármacos , Fosfopiruvato Hidratasa/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética
6.
J Neurosci ; 31(45): 16447-57, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22072694

RESUMEN

Chronic exposure to addictive drugs enhances cAMP response element binding protein (CREB)-regulated gene expression in nucleus accumbens (NAc), and these effects are thought to reduce the positive hedonic effects of passive cocaine administration. Here, we used viral-mediated gene transfer to produce short- and long-term regulation of CREB activity in NAc shell of rats engaging in volitional cocaine self-administration. Increasing CREB expression in NAc shell markedly enhanced cocaine reinforcement of self-administration behavior, as indicated by leftward (long-term) and upward (short-term) shifts in fixed ratio dose-response curves. CREB also increased the effort exerted by rats to obtain cocaine on more demanding progressive ratio schedules, an effect highly correlated with viral-induced modulation of BDNF protein in the NAc shell. CREB enhanced cocaine reinforcement when expressed either throughout acquisition of self-administration or when expression was limited to postacquisition tests, indicating a direct effect of CREB independent of reinforcement-related learning. Downregulating endogenous CREB in NAc shell by expressing a short hairpin RNA reduced cocaine reinforcement in similar tests, while overexpression of a dominant-negative CREB(S133A) mutant had no significant effect on cocaine self-administration. Finally, increasing CREB expression after withdrawal from self-administration enhanced cocaine-primed relapse, while reducing CREB levels facilitated extinction of cocaine seeking, but neither altered relapse induced by cocaine cues or footshock stress. Together, these findings indicate that CREB activity in NAc shell increases the motivation for cocaine during active self-administration or after withdrawal from cocaine. Our results also highlight that volitional and passive drug administration can lead to substantially different behavioral outcomes.


Asunto(s)
Anestésicos Locales/administración & dosificación , Proteína de Unión a CREB/metabolismo , Cocaína/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Refuerzo en Psicología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a CREB/genética , Cocaína/efectos adversos , Condicionamiento Operante/efectos de los fármacos , Vías de Administración de Medicamentos , Extinción Psicológica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Mutación/genética , Núcleo Accumbens/metabolismo , Interferencia de ARN/fisiología , Ratas , Ratas Sprague-Dawley , Esquema de Refuerzo , Autoadministración , Estadística como Asunto , Síndrome de Abstinencia a Sustancias/fisiopatología , Transfección/métodos
7.
J Neurosci ; 31(21): 7927-37, 2011 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-21613507

RESUMEN

Chronic cocaine use produces numerous biological changes in brain, but relatively few are functionally associated with cocaine reinforcement. Here we show that daily intravenous cocaine self-administration, but not passive cocaine administration, induces dynamic upregulation of the AMPA glutamate receptor subunits GluR1 and GluR2 in the ventral tegmental area (VTA) of rats. Increases in GluR1 protein and GluR1(S845) phosphorylation are associated with increased GluR1 mRNA in self-administering animals, whereas increased GluR2 protein levels occurred despite substantial decreases in GluR2 mRNA. We investigated the functional significance of GluR1 upregulation in the VTA on cocaine self-administration using localized viral-mediated gene transfer. Overexpression of GluR1(WT) in rat VTA primarily infected dopamine neurons (75%) and increased AMPA receptor-mediated membrane rectification in these neurons with AMPA application. Similar GluR1(WT) overexpression potentiated locomotor responses to intra-VTA AMPA, but not NMDA, infusions. In cocaine self-administering animals, overexpression of GluR1(WT) in the VTA markedly increased the motivation for cocaine injections on a progressive ratio schedule of cocaine reinforcement. In contrast, overexpression of protein kinase A-resistant GluR1(S845A) in the VTA reduced peak rates of cocaine self-administration on a fixed ratio reinforcement schedule. Neither viral vector altered sucrose self-administration, and overexpression of GluR1(WT) or GluR1(S845A) in the adjacent substantia nigra had no effect on cocaine self-administration. Together, these results suggest that dynamic regulation of AMPA receptors in the VTA during cocaine self-administration contributes to cocaine addiction by acting to facilitate subsequent cocaine use.


Asunto(s)
Conducta Adictiva , Cocaína/administración & dosificación , Motivación/fisiología , Receptores AMPA/fisiología , Refuerzo en Psicología , Área Tegmental Ventral/fisiología , Animales , Conducta Adictiva/psicología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Masculino , Motivación/efectos de los fármacos , Células PC12 , Subunidades de Proteína/fisiología , Ratas , Ratas Sprague-Dawley , Autoadministración , Área Tegmental Ventral/efectos de los fármacos
8.
Addict Biol ; 16(3): 450-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21309958

RESUMEN

Reexposure to cocaine-associated environments promotes relapse to cocaine seeking and represents a persistent impediment to successful abstinence. Neurobiological adaptations are thought to underlie the preservation of drug-seeking behavior during protracted withdrawal periods, possibly including changes associated specifically with cocaine-paired contexts. We measured GluR(1) (S845) and extracellular signal-regulated kinase (ERK) phosphorylation in rat striatal subregions in an animal model of cocaine relapse. Animals with cocaine self-administration experience and their yoked partners were exposed to extinction conditions for one hour in the drug-paired environmental context after one day or three weeks withdrawal to measure protein phosphorylation induced by the cocaine-paired context in the absence of cocaine reinforcement. GluR(1) (S845) (an index of protein kinase A (PKA) activity) and ERK phosphorylation increased in the nucleus accumbens core of self-administering but not yoked animals after three weeks (but not one day) withdrawal, indicating a time-dependent emergence of context-associated protein phosphorylation in this accumbens subregion. In comparison, animals trained to self-administer sucrose displayed a similar increase in ERK, but not GluR(1) (S845) , phosphorylation following reexposure to a sucrose-paired environment three weeks later, indicating that GluR(1) (S845) phosphorylation did not result solely from lever press behavior per se. In contrast, basal (home cage) GluR(1) (S845) phosphorylation was elevated in the nucleus accumbens shell and caudate-putamen after one day or three weeks cocaine withdrawal regardless of context exposure. These results suggest that time-dependent emergence of context-associated GluR(1) (S845) phosphorylation in the nucleus accumbens core may contribute to the persistence of cocaine-seeking behavior, whereas ERK phosphorylation may be a consequence of this behavior.


Asunto(s)
Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Estimulantes del Sistema Nervioso Central/efectos adversos , Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/efectos adversos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neostriado/fisiopatología , Núcleo Accumbens/fisiopatología , Receptores AMPA/fisiología , Medio Social , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Autoadministración
9.
Nat Neurosci ; 10(8): 1029-37, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17618281

RESUMEN

A single exposure to cocaine rapidly induces the brief activation of several immediate early genes, but the role of such short-term regulation in the enduring consequences of cocaine use is poorly understood. We found that 4 h of intravenous cocaine self-administration in rats induced a transient increase in brain-derived neurotrophic factor (BDNF) and activation of TrkB-mediated signaling in the nucleus accumbens (NAc). Augmenting this dynamic regulation with five daily NAc BDNF infusions caused enduring increases in cocaine self-administration, and facilitated relapse to cocaine seeking in withdrawal. In contrast, neutralizing endogenous BDNF regulation with intra-NAc infusions of antibody to BDNF subsequently reduced cocaine self-administration and attenuated relapse. Using localized inducible BDNF knockout in mice, we found that BDNF originating from NAc neurons was necessary for maintaining increased cocaine self-administration. These findings suggest that dynamic induction and release of BDNF from NAc neurons during cocaine use promotes the development and persistence of addictive behavior.


Asunto(s)
Anestésicos Locales/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cocaína/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Refuerzo en Psicología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/genética , Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Cocaína/fisiopatología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/citología , Ratas , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Autoadministración/métodos , Factores de Tiempo
10.
Nat Neurosci ; 10(1): 93-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143271

RESUMEN

Chronic morphine administration (via subcutaneous pellet) decreases the size of dopamine neurons in the ventral tegmental area (VTA), a key reward region in the brain, yet the molecular basis and functional consequences of this effect are unknown. In this study, we used viral-mediated gene transfer in rat to show that chronic morphine-induced downregulation of the insulin receptor substrate 2 (IRS2)-thymoma viral proto-oncogene (Akt) signaling pathway in the VTA mediates the decrease in dopamine cell size seen after morphine exposure and that this downregulation diminishes morphine reward, as measured by conditioned place preference. We further show that the reduction in size of VTA dopamine neurons persists up to 2 weeks after morphine withdrawal, which parallels the tolerance to morphine's rewarding effects caused by previous chronic morphine exposure. These findings directly implicate the IRS2-Akt signaling pathway as a critical regulator of dopamine cell morphology and opiate reward.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Dopamina/metabolismo , Mesencéfalo/citología , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Neuronas/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Western Blotting , Tamaño de la Célula/efectos de los fármacos , Condicionamiento Operante/fisiología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/fisiología , Proteína Oncogénica v-akt/fisiología , Fosfoproteínas/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
J Neurochem ; 115(1): 112-22, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20633205

RESUMEN

Chronic drug exposure induces alterations in gene expression profiles that are thought to underlie the development of drug addiction. The present study examined regulation of the Fos-family of transcription factors, specifically cFos, FosB, and ΔFosB, in striatal subregions during and after chronic intravenous cocaine administration in self-administering and yoked rats. We found that cFos, FosB, and ΔFosB exhibit regionally and temporally distinct expression patterns, with greater accumulation of ΔFosB protein in the nucleus accumbens (NAc) shell and core after chronic cocaine administration, whereas ΔFosB increases in the caudate-putamen (CPu) remained similar with either acute or chronic administration. In contrast, tolerance developed to cocaine-induced mRNA for ΔFosB in all three striatal subregions with chronic administration. Tolerance also developed to FosB expression, most notably in the NAc shell and CPu. Interestingly, tolerance to cocaine-induced cFos induction was dependent on volitional control of cocaine intake in ventral but not dorsal striatal regions, whereas regulation of FosB and ΔFosB was similar in cocaine self-administering and yoked animals. Thus, ΔFosB-mediated neuroadaptations in the CPu may occur earlier than previously thought with the initiation of intravenous cocaine use and, together with greater accumulation of ΔFosB in the NAc, could contribute to addiction-related increases in cocaine-seeking behavior.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Cocaína/efectos adversos , Cocaína/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Neostriado/fisiología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Inyecciones Intravenosas , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Refuerzo en Psicología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Autoadministración , Abuso de Sustancias por Vía Intravenosa
12.
Cereb Cortex ; 19(2): 435-44, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18539927

RESUMEN

Increased impulsivity caused by addictive drugs is believed to contribute to the maintenance of addiction and has been linked to hypofunction within the orbitofrontal cortex (OFC). Recent data indicate that cocaine "self-administration" induces the transcription factor DeltaFosB in the OFC that alters the effects of investigator-administered cocaine on impulsivity. Here, using viral-mediated gene transfer, the effects of overexpressing DeltaFosB within the OFC were assessed on the cognitive sequelae of chronic cocaine self-administration as measured by the 5-choice serial reaction time task (5CSRT). Cognitive testing occurred in the mornings, and self-administration sessions in the evenings, to enable the progressive assessment of repeated volitional drug intake on performance. Animals self-administering cocaine initially made more omissions and premature or impulsive responses on the 5CSRT but quickly developed tolerance to these disruptive effects. However, withdrawal from cocaine dramatically increased premature responding. When access to cocaine was increased, animals overexpressing DeltaFosB failed to regulate their intake as effectively and were more impulsive during withdrawal. In summary, rats develop tolerance to the cognitive disruption caused by cocaine self-administration and show a deficit in impulse control that is unmasked during withdrawal. Our findings suggest that induction of DeltaFosB within the OFC is one mediator of these effects and, thereby, increases vulnerability to addiction.


Asunto(s)
Cocaína/efectos adversos , Conducta Impulsiva/genética , Conducta Impulsiva/psicología , Corteza Prefrontal/fisiología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/fisiología , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/psicología , Adenoviridae/genética , Animales , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/psicología , Cognición/efectos de los fármacos , Cognición/fisiología , Relación Dosis-Respuesta a Droga , Extinción Psicológica/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos , Inmunohistoquímica , Masculino , Ratas , Ratas Long-Evans , Tiempo de Reacción/fisiología , Autoadministración
13.
Front Neurosci ; 14: 698, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32760242

RESUMEN

The consequence of repeated cocaine exposure and prolonged abstinence on glutamate receptor expression in the nucleus accumbens has been extensively studied. However, the early effects of cocaine on NMDAR signaling remain unknown. NMDAR signaling depends on the subunit composition, subcellular localization, and the interaction with proteins at the postsynaptic density (PSD), where NMDARs and other proteins form supercomplexes that are responsible for the signaling pathways activated by NMDAR-induced Ca2+ influx. Here, we investigated the effect of cocaine on NMDAR subunit composition and subcellular localization after both intraperitoneal non-contingent cocaine and response-contingent intravenous cocaine self-administration in mice. We found that repeated cocaine exposure, regardless of the route or contingency of drug administration, decreases NMDAR interactions with the PSD and synaptic lipid rafts in the accumbens shell and dorsal striatum. We provide evidence that cocaine triggers an early redistribution of NMDARs from synaptic to extrasynaptic sites, and that this adaptation has implications in the activation of downstream signaling pathways. Thus, consistent with a loss of NMDAR function, cocaine-induced ERK phosphorylation is attenuated. Because early NMDAR activity contributes to the initiation of lasting addiction-relevant neuroadaptations, these data may hold clues into cellular mechanisms responsible for the development of cocaine addiction.

14.
Neuron ; 48(2): 303-14, 2005 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16242410

RESUMEN

Given that cocaine induces neuroadaptations through regulation of gene expression, we investigated whether chromatin remodeling at specific gene promoters may be a key mechanism. We show that cocaine induces specific histone modifications at different gene promoters in striatum, a major neural substrate for cocaine's behavioral effects. At the cFos promoter, H4 hyperacetylation is seen within 30 min of a single cocaine injection, whereas no histone modifications were seen with chronic cocaine, consistent with cocaine's ability to induce cFos acutely, but not chronically. In contrast, at the BDNF and Cdk5 promoters, genes that are induced by chronic, but not acute, cocaine, H3 hyperacetylation was observed with chronic cocaine only. DeltaFosB, a cocaine-induced transcription factor, appears to mediate this regulation of the Cdk5 gene. Furthermore, modulating histone deacetylase activity alters locomotor and rewarding responses to cocaine. Thus, chromatin remodeling is an important regulatory mechanism underlying cocaine-induced neural and behavioral plasticity.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Cocaína/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Inhibidores de Captación de Dopamina/administración & dosificación , Plasticidad Neuronal/efectos de los fármacos , Acetilación , Animales , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Butiratos/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Cuerpo Estriado/fisiología , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Esquema de Medicación , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen/psicología , Histona Desacetilasas/metabolismo , Histonas/clasificación , Histonas/metabolismo , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Células PC12/metabolismo , Regiones Promotoras Genéticas/fisiología , Subunidades de Proteína , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo
15.
J Neurosci ; 28(48): 12808-14, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19036973

RESUMEN

Withdrawal from repeated cocaine is associated with increased synaptic and extrasynaptic AMPA receptor (AMPAR) expression in nucleus accumbens (NAc) neurons and enhanced behavioral sensitivity to AMPAR stimulation. Recent studies found that increased membrane expression of AMPARs is reversed or normalized on cocaine reexposure in withdrawal, but the mechanism for this AMPAR plasticity and the behavioral implications are unknown. Here, we examine the effects of renewed cocaine exposure during withdrawal on enhanced NAc AMPAR sensitivity and investigate the underlying mechanisms. Cocaine reexposure transiently reversed enhanced NAc AMPAR-mediated locomotion 1 d later, while enhancing cocaine-induced locomotion. Reversal in AMPAR sensitivity was prohibited by NAc AMPAR blockade with CNQX during cocaine reexposure and mimicked by intra-NAc infusions of AMPA, suggesting that cocaine-induced glutamate stimulation of NAc AMPARs is necessary for reversing AMPAR responsiveness. Similarly, systemic treatment with the dopamine D(1)-like agonist SKF 81297 [(+/-)-6-chloro-7,8-dihydroxy-l-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide] reversed AMPAR responsiveness in cocaine withdrawal, but the effect was prevented by local NAc AMPAR blockade in the NAc, and not local D(1)-like receptor blockade, suggesting a role for glutamate afferents in the reversal of enhanced AMPAR sensitivity. Together, these findings suggest that cocaine-induced glutamate release in sensitized animals is responsible for dynamic alterations in AMPAR function that contribute to enhanced cocaine sensitivity.


Asunto(s)
Conducta Animal/efectos de los fármacos , Trastornos Relacionados con Cocaína/metabolismo , Cocaína/farmacología , Núcleo Accumbens/efectos de los fármacos , Receptores AMPA/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Conducta Animal/fisiología , Trastornos Relacionados con Cocaína/fisiopatología , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatología , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
16.
J Neurosci ; 28(10): 2516-26, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18322096

RESUMEN

Relapse, a major problem in the treatment of cocaine addiction, is proposed to result in part from neuroadaptations in the hippocampus. We examined how a mediator of hippocampal neuroplasticity, adult neurogenesis in the subgranular zone (SGZ), was regulated by cocaine self-administration (CSA), and whether these changes were reversed by 4 weeks of withdrawal (CSA-WD) versus continued cocaine self-administration (CSA-CONT). Rats self-administered intravenous cocaine or saline for 3 weeks and were killed 2 h (CSA) or 4 weeks (CSA-WD, CSA-CONT) after injection with the S-phase marker bromodeoxyuridine (BrdU). Cells in several stages of adult neurogenesis were quantified: proliferating cells labeled by BrdU (2 h) or Ki-67; immature neurons labeled by doublecortin; and adult-generated neurons labeled with BrdU (4 weeks) and the mature neuronal marker NeuN. CSA decreased proliferation in both the SGZ and the subventricular zone (SVZ), a source of adult-generated olfactory neurons, changes reversed by CSA-WD. Unexpectedly, CSA-WD and CSA-CONT resulted in more immature doublecortin-immunopositive (+) neurons in the posterior SGZ and a normal number of adult-generated BrdU+ neurons in the SGZ, suggesting an enduring impact of CSA regardless of whether cocaine intake was stopped or continued. However, only CSA-WD rats had more adult-generated neurons with punctate BrdU staining, an indicator of enhanced maturity. These data suggest a mechanism for the cognitive and olfactory deficits seen in cocaine addicts, and further suggest that adult-generated neurons should be considered for their potential role in cocaine addiction and hippocampal-mediated relapse after cocaine withdrawal.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Senescencia Celular/fisiología , Cocaína/administración & dosificación , Hipocampo/citología , Hipocampo/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Síndrome de Abstinencia a Sustancias , Factores de Edad , Animales , Proteína Doblecortina , Masculino , Ratas , Ratas Sprague-Dawley , Autoadministración , Síndrome de Abstinencia a Sustancias/patología
17.
Synapse ; 63(3): 224-35, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19084907

RESUMEN

Negative motivational symptoms are observed soon after withdrawal from chronic opiate administration, and are thought to mediate dependence. Examination of brain region-specific signaling changes that accompany early withdrawal may shed light on neural mechanisms underlying negative reinforcement and dependence. Thus, we measured alterations in protein phosphorylation in multiple limbic brain regions in rats undergoing 24 h spontaneous or naltrexone-precipitated withdrawal from chronic (6 h/day) i.v. heroin self-administration. Region-specific increases in cyclic AMP-dependent GluR(1) (S845) phosphorylation were found in the nucleus accumbens shell, basolateral amygdala, hippocampal CA1 and CA3 subregions, and premotor cortex from 12 to 24 h of spontaneous withdrawal, and there were no changes in prefrontal cortex, nucleus accumbens core or caudate-putamen. Increased GluR(1) (S845) phosphorylation was detected earlier (12 h withdrawal) in the central amygdala and ventral tegmental area. In contrast, prominent increases in extracellular signal-regulated kinase phosphorylation were found in both prefrontal and premotor cortex, and CA1 and CA3 between 12 and 24 h withdrawal. Phosphorylation of striatal cyclic AMP response element binding protein increased in the caudate-putamen but not in the nucleus accumbens. Naltrexone administration after 24 h withdrawal increased extracellular signal-regulated kinase phosphorylation in the central amygdala, and nucleus accumbens core and shell. Thus, spontaneous withdrawal from heroin self-administration produces region- and time-dependent changes in cyclic AMP and extracellular signal-regulated kinase activity that could contribute to the behavioral manifestation of opiate dependence.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Dependencia de Heroína/metabolismo , Heroína/administración & dosificación , Narcóticos/administración & dosificación , Receptores AMPA/metabolismo , Análisis de Varianza , Animales , Encéfalo/metabolismo , Dependencia de Heroína/patología , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Autoadministración , Serina/metabolismo
18.
Neuropsychopharmacology ; 44(8): 1370-1376, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30587852

RESUMEN

Comorbid neuropsychiatric disorders such as addiction and anxiety could involve common underlying mechanisms. One potential mechanism involves epigenetic regulation of histone 3 dimethylation at lysine 9 residues (H3K9me2) by the histone dimethyltransferase G9a. Here we provide evidence that local AAV-RNAi-mediated knockdown of G9a expression in nucleus accumbens shell (NAcSh) of male rats reduces both addictive-related and anxiety-related behaviors. Specifically, G9a knockdown reduces sensitivity to low dose cocaine reinforcement when cocaine is freely available (fixed ratio schedule). Similarly, G9a knockdown reduces motivation for cocaine under higher effort demands (progressive ratio schedule). Following several weeks of forced abstinence, G9a knockdown attenuates extinction responding and reinstatement triggered by either cocaine-priming injections or footshock stress. This decrease in addictive behavior is associated with a long-term reduction in anxiety-like behavior as measured by the elevated plus maze (EPM). G9a knockdown also reduces basal anxiety-like behavior in EPM and marble burying tests in drug-naïve rats. These results complement our previous work showing that increased G9a expression in NAcSh enhances addictive-related and anxiety-related behaviors, indicating that G9a bi-directionally controls these responses. These results also suggest that regulation of G9a-influenced gene expression could be a common epigenetic mechanism for co-morbid anxiety and psychostimulant addiction.


Asunto(s)
Ansiedad/metabolismo , Conducta Adictiva/fisiopatología , Cocaína/farmacología , Extinción Psicológica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/fisiología , Núcleo Accumbens/metabolismo , Animales , Conducta Animal/fisiología , Estimulación Eléctrica , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Ratas , Recurrencia , Esquema de Refuerzo , Autoadministración
19.
J Neurosci ; 27(39): 10497-507, 2007 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-17898221

RESUMEN

Current cocaine users show little evidence of cognitive impairment and may perform better when using cocaine, yet withdrawal from prolonged cocaine use unmasks dramatic cognitive deficits. It has been suggested that such impairments arise in part through drug-induced dysfunction within the orbitofrontal cortex (OFC), yet the neurobiological mechanisms remain unknown. We observed that chronic cocaine self-administration increased expression of the transcription factor deltaFosB within both medial and orbitofrontal regions of the rat prefrontal cortex. However, the increase in OFC deltaFosB levels was more pronounced after self-administered rather than experimenter-administered cocaine, a pattern that was not observed in other regions. We then used rodent tests of attention and decision making to determine whether deltaFosB within the OFC contributes to drug-induced alterations in cognition. Chronic cocaine treatment produced tolerance to the cognitive impairments caused by acute cocaine. Overexpression of a dominant-negative antagonist of deltaFosB, deltaJunD, in the OFC prevented this behavioral adaptation, whereas locally overexpressing deltaFosB mimicked the effects of chronic cocaine. Gene microarray analyses identified potential molecular mechanisms underlying this behavioral change, including an increase in transcription of metabotropic glutamate receptor subunit 5 and GABA(A) receptors as well as substance P. Identification of deltaFosB in the OFC as a mediator of tolerance to the effects of cocaine on cognition provides fundamentally new insight into the transcriptional modifications associated with addiction.


Asunto(s)
Estimulantes del Sistema Nervioso Central/administración & dosificación , Cocaína/administración & dosificación , Trastornos del Conocimiento/fisiopatología , Tolerancia a Medicamentos/fisiología , Lóbulo Frontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Animales , Estimulantes del Sistema Nervioso Central/efectos adversos , Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/efectos adversos , Cocaína/farmacología , Trastornos Relacionados con Cocaína/etiología , Trastornos Relacionados con Cocaína/fisiopatología , Trastornos del Conocimiento/inducido químicamente , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Masculino , Pruebas Neuropsicológicas , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley
20.
Eur J Neurosci ; 27(9): 2229-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18430032

RESUMEN

Chronic cocaine use reduces glutamate levels in the nucleus accumbens (NAc), and is associated with experience-dependent changes in (+/-)-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) glutamate receptor membrane expression in NAc neurons. These changes accompany behavioral sensitization to cocaine and increased susceptibility to cocaine relapse. The functional relationship between neuroplasticity in AMPA receptors and the behavioral manifestation of cocaine addiction remains unclear. Thus, we examined the behavioral effects of up- and downregulating basal AMPA receptor function in the NAc core and shell using viral-mediated gene transfer of wild-type glutamate receptor 1 (wt-GluR1) or a dominant-negative pore-dead GluR1 (pd-GluR1), respectively. Transient increases in wt-GluR1 during or after cocaine treatments diminished the development of cocaine sensitization, while pd-GluR1 expression exacerbated cocaine sensitization. Parallel changes were found in D2, but not D1, receptor-mediated behavioral responses. As a correlate of the sensitization experiments, we overexpressed wt- or pd-GluR1 in the NAc core during cocaine self-administration, and tested the effects on subsequent drug-seeking behavior 3 weeks after overexpression declined. wt-GluR1 overexpression during self-administration had no effect on cocaine intake, but subsequently reduced cocaine seeking in extinction and cocaine-induced reinstatement, whereas pd-GluR1 facilitated cocaine-induced reinstatement. When overexpressed during reinstatement tests, wt-GluR1 directly attenuated cocaine- and D2 agonist-induced reinstatement, while pd-GluR1 enhanced reinstatement. In both experimental procedures, neither wt- nor pd-GluR1 expression affected cue-induced reinstatement. Together, these results suggest that degrading basal AMPA receptor function in NAc neurons is sufficient to facilitate relapse via sensitization in D2 receptor responses, whereas elevating basal AMPA receptor function attenuates these behaviors.


Asunto(s)
Conducta Adictiva/fisiopatología , Trastornos Relacionados con Cocaína/fisiopatología , Núcleo Accumbens/metabolismo , Receptores AMPA/biosíntesis , Animales , Conducta Adictiva/metabolismo , Conducta Animal/fisiología , Trastornos Relacionados con Cocaína/metabolismo , Vectores Genéticos , Masculino , Actividad Motora/fisiología , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA