Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Neurosci ; 38(6): 1462-1471, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29317484

RESUMEN

Abuse rates for inhalants among adolescents continue to be high, yet preclinical models for studying mechanisms underlying inhalant abuse remain limited. Our laboratory has previously shown that, in male rats, an acute binge-like exposure to toluene vapor that mimics human solvent abuse modifies the intrinsic excitability of mPFC pyramidal neurons projecting to the NAc. These changes showed region (infralimbic; IL vs prelimbic; PRL), layer (shallow; 2/3 vs deep; 5/6), target (core vs shell), and age (adolescent vs adult) dependent differences (Wayman and Woodward, 2017). To expand these findings using reward-based models that may better mimic human drug abuse, we used whole-cell electrophysiology and drug receptors exclusively activated by designer drugs to examine changes in neuronal function and behavior in rats showing a conditioned place preference (CPP) to toluene. Repeated pairings of adolescent rats to binge concentrations of toluene vapor previously shown to enhance dopamine release in reward-sensitive areas of the brain produced CPP that persisted for 7 but not 30 d. Toluene-induced CPP was associated with increased excitability of IL5/6 mPFC neurons projecting to the core of the NAc and reduced excitability of those projecting to the NAc shell. No changes in PRL-NAc-projecting neurons were found in toluene-CPP rats. Chemogenetic reversal of the toluene-induced decrease in IL5/6-NAc shell neurons blocked the expression of toluene-induced CPP while manipulating IL5/6-NAc core neuron activity had no effect. These data reveal that alterations in selective mPFC-NAc pathways are required for expression of toluene-induced CPP.SIGNIFICANCE STATEMENT Disturbed physiology of pyramidal neurons projecting from the mPFC to the NAc has been shown to have different roles in drug-seeking behaviors for a number of drugs (e.g., methamphetamine, cocaine, ecstasy, alcohol, heroin). Here, we report that rats repeatedly exposed to the volatile organic solvent toluene, a member of the class of abused inhalants often used for intoxicating purposes by adolescents, induces a preference for the drug-paired environment that is accompanied by altered physiology of a specific population of NAc-projecting mPFC neurons. Chemogenetic correction of this deficit before testing prevented expression of drug preference. Overall, these findings highlight the importance of corticolimbic circuitry in mediating the rewarding properties of abused inhalants.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Abuso de Inhalantes/psicología , Sistema Límbico/citología , Sistema Límbico/efectos de los fármacos , Neuronas/efectos de los fármacos , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Tolueno/farmacología , Administración por Inhalación , Envejecimiento , Animales , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Interleucinas/fisiología , Masculino , Células Piramidales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recompensa
2.
Eur J Neurosci ; 41(9): 1195-206, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25707266

RESUMEN

The medial prefrontal cortex (mPFC) plays a critical role in reward-motivated behaviors. Repeated cocaine exposure dysregulates the dorsal mPFC, and this is thought to contribute to cocaine-seeking and relapse of abstinent abusers. Neuropathology of the mPFC also occurs in human immunodeficiency virus (HIV)-positive individuals, and this is exaggerated in those who also abuse cocaine. The impact of the comorbid condition on mPFC neuronal function is unknown. To fill this knowledge gap, we performed a behavioral and electrophysiological study utilising adult male rats that self-administered cocaine by pressing a lever for 14 once-daily operant sessions. Saline-yoked (SAL-yoked) rats served as controls. Cue reactivity (CR) was used to indicate drug-seeking, assessed by re-exposing the rats to cocaine-paired cues wherein non-reinforced lever pressing was quantified 1 day (CR1) and 18-21 days (CR2) after the 14th operant session. Only cocaine self-administration (COC-SA) rats showed CR. One day after CR2, brain slices were prepared for electrophysiological assessment. Whole-cell patch-clamp recordings of dorsal (prelimbic) mPFC pyramidal neurons from COC-SA rats showed a significant increase in firing evoked by depolarizing currents as compared with those from SAL-yoked control rats. Bath application of the toxic HIV-1 protein transactivator of transcription (Tat) also depolarized neuronal membranes and increased evoked firing. The Tat-induced excitation was greater in the neurons from withdrawn COC-SA rats than in controls. Tat also reduced spike amplitude, and this co-varied with cocaine-seeking during CR2. Taken together, these novel findings provide support at the neuronal level for the concept that the increased excitability of mPFC pyramidal neurons following cocaine self-administration drives drug-seeking and augments the neuropathophysiology caused by HIV-1 Tat.


Asunto(s)
Potenciales de Acción , Cocaína/farmacología , Corteza Prefrontal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Cocaína/administración & dosificación , Señales (Psicología) , Comportamiento de Búsqueda de Drogas , Masculino , Potenciales de la Membrana , Corteza Prefrontal/citología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Autoadministración
3.
Neuropsychopharmacology ; 48(10): 1455-1464, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37221326

RESUMEN

The rostromedial tegmental nucleus (RMTg) encodes negative reward prediction error (RPE) and plays an important role in guiding behavioral responding to aversive stimuli. Previous research has focused on regulation of RMTg activity by the lateral habenula despite studies revealing RMTg afferents from other regions including the frontal cortex. The current study provides a detailed anatomical and functional analysis of cortical input to the RMTg of male rats. Retrograde tracing uncovered dense cortical input to the RMTg spanning the medial prefrontal cortex, the orbitofrontal cortex and anterior insular cortex. Afferents were most dense in the dorsomedial subregion of the PFC (dmPFC), an area that is also implicated in both RPE signaling and aversive responding. RMTg-projecting dmPFC neurons originate in layer V, are glutamatergic, and collateralize to select brain regions. In-situ mRNA hybridization revealed that neurons in this circuit are predominantly D1 receptor-expressing with a high degree of D2 receptor colocalization. Consistent with cFos induction in this neural circuit during exposure to foot shock and shock-predictive cues, optogenetic stimulation of dmPFC terminals in the RMTg drove avoidance. Lastly, acute slice electrophysiology and morphological studies revealed that exposure to repeated foot shock resulted in significant physiological and structural changes consistent with a loss of top-down modulation of RMTg-mediated signaling. Altogether, these data reveal the presence of a prominent cortico-subcortical projection involved in adaptive behavioral responding to aversive stimuli such as foot shock and provide a foundation for future work aimed at exploring alterations in circuit function in diseases characterized by deficits in cognitive control over reward and aversion.


Asunto(s)
Neuronas , Tegmento Mesencefálico , Ratas , Masculino , Animales , Tegmento Mesencefálico/fisiología , Neuronas/fisiología , Núcleo Celular , Área Tegmental Ventral/fisiología
4.
Front Neurosci ; 14: 880, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973434

RESUMEN

Inhalants, including volatile organic solvents such as toluene, continue to be one of the most prevalent, and often first substances abused by adolescents. Like other drugs of abuse, toluene affects the function of neurons within key brain reward circuits including the prefrontal cortex, ventral tegmental area, and nucleus accumbens. However, preclinical models used to study these toluene-induced adaptations generally employ passive exposure paradigms that do not mirror voluntary patterns of solvent exposure observed in humans. To address this shortcoming, we developed an inhalation chamber containing active and inactive nose pokes, cue lights, flow-through vaporizers, and software-controlled valves to test the hypothesis that rats will voluntarily self-administer toluene vapor. Following habituation and self-administration (SA) training rats achieve vapor concentrations associated with rewarding effects of toluene, and maintain responding for toluene vapor, but not for air. During extinction trials, rats showed an initial burst of drug-seeking behavior similar to that of other addictive drugs and then reduced responding to Air SA levels. Responding on the active nose poke recovered during cue-induced reinstatement but not following a single passive exposure to toluene vapor. The results from these studies establish a viable toluene SA protocol that will be useful in assessing toluene-induced changes in addiction neurocircuitry.

5.
Neuropsychopharmacology ; 43(4): 912-924, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28589963

RESUMEN

Inhalants, including toluene, target the addiction neurocircuitry and are often one of the first drugs of abuse tried by adolescents. The medial prefrontal cortex (mPFC) is involved in regulating goal-directed/reward-motivated behaviors and different mPFC sub-regions have been proposed to promote (prelimbic, PRL) or inhibit (infralimbic, IL) these behaviors. While this dichotomy has been studied in the context of other drugs of abuse, it is not known whether toluene exposure differentially affects neurons within PRL and IL regions. To address this question, we used whole-cell electrophysiology and determined the intrinsic excitability of PRL and IL pyramidal neurons in adolescent rats 24 h following a brief exposure to air or toluene vapor (10 500 p.p.m.). Prior to exposure, fluorescent retrobeads were injected into the NAc core (NAcc) or shell (NAcs) sub-regions to identify projection-specific mPFC neurons. In toluene treated adolescent rats, layer 5/6 NAcc projecting PRL (PRL5/6) neurons fired fewer action potentials and this was associated with increased rheobase, increased spike duration, and reductions in membrane resistance and amplitude of the Ih current. No changes in excitability were observed in layer 2/3 NAcc projecting PRL (PRL2/3) neurons. In contrast to PRL neurons, layer 5 IL (IL5) and layer 2/3 (IL2/3) NAcc projecting neurons showed enhanced firing in toluene-exposed animals and in IL5 neurons, this was associated with a reduction in rheobase and AHP. For NAcs projecting neurons, toluene exposure significantly decreased firing of IL5 neurons and this was accompanied by an increased rheobase, increased spike duration, and reduced Ih amplitude. The intrinsic excitability of PRL5, PRL2/3, and IL2/3 neurons projecting to the NAcs was not affected by exposure to toluene. The changes in excitability observed 24 h after toluene exposure were not observed when recordings were performed 7 days after the exposure. Finally, there were no changes in intrinsic excitability of any region in rats exposed to toluene as adults. These findings demonstrate that specific projections of the reward circuitry are uniquely susceptible to the effects of toluene during adolescence supporting the idea that adolescence is a critical period of the development that is vulnerable to drugs of abuse.


Asunto(s)
Abuso de Inhalantes/fisiopatología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Solventes/administración & dosificación , Tolueno/administración & dosificación , Factores de Edad , Animales , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
6.
Neuropharmacology ; 135: 316-327, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29580953

RESUMEN

Channelrhodopsins are light activated ion channels used extensively over the past decade to probe the function of genetically defined neuronal populations and distinct neural circuits with high temporal and spatial precision. The widely used Channelrhodopsin-2 variant (ChR2) is an excitatory opsin that undergoes conformational changes in response to blue light, allowing non-selective passage of protons and cations across the plasma membrane thus leading to depolarization. In the addiction neuroscience field, opsins such as ChR2 provide a means to disambiguate the overlapping circuitry involved in mediating the reinforcing and aversive effects of drugs of abuse as well as to determine the plasticity that can occur in these circuits during the development of dependence. Although ChR2 has been widely used in animal models of drug and alcohol self-administration, direct effects of drugs of abuse on ChR2 function may confound its use and lead to misinterpretation of data. As a variety of neuronal ion channels are primary targets of various drugs of abuse, it is critical to determine whether ChR2-mediated currents are modulated by these drugs. In this study, we performed whole-cell electrophysiological recordings in HEK293 cells expressing the commonly used ChR2(H134R) variant and examined the effects of various drugs of abuse and other commonly used agents on light-induced currents. We found no differences in ChR2-mediated currents in the presence of 30 µM nicotine, 30 µM cocaine, 100 µM methamphetamine or 3 mM toluene. Similarly, ChR2 currents were insensitive to 30 mM ethanol but higher concentrations (100-300 mM) produced significant effects on the desensitization and amplitude of light-evoked currents. Tetrahydrocannabinol (1-10 µM) and morphine (30-100 µM) significantly inhibited ChR2 currents while the cannabinoid receptor antagonist AM-251 had no effect. The sodium channel blocker tetrodotoxin (5 µM) and the generic channel blocker/contrast agent gadolinium chloride (10 mM) also reduced ChR2 currents while the divalent ion magnesium (10 mM) had no effect. Together, the results from this study highlight the importance of conducting appropriate control experiments when testing new compounds in combination with optogenetic approaches.


Asunto(s)
Channelrhodopsins/metabolismo , Drogas Ilícitas/farmacología , Moduladores del Transporte de Membrana/farmacología , Cationes/farmacología , Channelrhodopsins/genética , Cocaína/farmacología , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Etanol/farmacología , Gadolinio/farmacología , Células HEK293 , Humanos , Magnesio/farmacología , Potenciales de la Membrana/efectos de los fármacos , Metanfetamina/farmacología , Morfina/farmacología , Nicotina/farmacología , Técnicas de Placa-Clamp , Tetrodotoxina/farmacología , Tolueno/farmacología , Transfección
7.
Neuropsychopharmacology ; 41(8): 1965-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26677947

RESUMEN

The medial prefrontal cortex (mPFC) is dysregulated in HIV-1-infected humans and the dysregulation is enhanced by cocaine abuse. Understanding mPFC pathophysiology in this comorbid state has been hampered by the dearth of relevant animal models. To help fill this knowledge gap, electrophysiological assessments were made of mPFC pyramidal neurons (PN) from adult male HIV-1 transgenic (Tg) F344 rats (which express seven of the nine HIV-1 toxic proteins) and non-Tg F344 rats that self-administered cocaine for 14 days (COC-SA), as well as saline-yoked controls (SAL-Yoked) and experimentally naive Tg and non-Tg rats. Forebrain slices were harvested and prepared for whole-cell patch-clamp recording, and in treated rats, this occurred after 14-18 days of forced abstinence. Aged-matched rats were used for immunohistochemical detection of the L-channel protein, Cav1.2-α1c. We determined that: (i) the two genotypes acquired the operant task and maintained similar levels of COC-SA, (ii) forced abstinence from COC-SA enhanced mPFC PN excitability in both genotypes, and neurons from Tg rats exhibited the greatest pathophysiology, (iii) neurons from SAL-Yoked Tg rats were more excitable than those from SAL-Yoked non-Tg rats, and in Tg rats (iv) blockade of L-type Ca(2+) channels reduced the enhanced excitability, and (v) Cav1.2-immunoreactivity was increased. These findings provide the first assessment of the mPFC pathophysiology in a rodent model of HIV-1-mediated neuropathology with and without cocaine self-administration. Outcomes reveal an enhanced cortical excitability during chronic exposure to HIV-1 proteins that is excessively exacerbated with cocaine abuse. Such neuropathophysiology may underlie the cognitive dysregulation reported for comorbid humans.


Asunto(s)
Cocaína/administración & dosificación , Excitabilidad Cortical/efectos de los fármacos , VIH-1/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/virología , Potenciales de Acción/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , VIH-1/genética , Masculino , Corteza Prefrontal/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Células Piramidales/virología , Ratas Endogámicas F344 , Ratas Transgénicas , Autoadministración
8.
Curr HIV Res ; 13(1): 80-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25760043

RESUMEN

The life span of individuals that are sero-positive for human immunodeficiency virus (HIV) has greatly improved; however, complications involving the central nervous system (CNS) remain a concern. While HIV does not directly infect neurons, the proteins produced by the virus, including HIV transactivator of transcription (Tat), are released from infected glia; these proteins can be neurotoxic. This neurotoxicity is thought to mediate the pathology underlying HIVassociated neurological impairments. Cocaine abuse is common among HIV infected individuals, and this abuse augments HIV-associated neurological deficits. The brain regions and pathophysiological mechanisms that are dysregulated by both chronic cocaine and Tat are the focus of the current review.


Asunto(s)
Encéfalo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Trastornos Relacionados con Cocaína/complicaciones , Cocaína/efectos adversos , Infecciones por VIH/complicaciones , VIH-1 , Trastornos Neurocognitivos/etiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Animales , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Trastornos Neurocognitivos/fisiopatología , Ratas
9.
Neuroreport ; 23(14): 825-9, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22828409

RESUMEN

HIV-1 proteins, including the transactivator of transcription (Tat), are believed to be involved in HIV-associated neurocognitive disorders by disrupting Ca²âº homeostasis, which leads to progressive dysregulation, damage, or death of neurons in the brain. We have found previously that bath-applied Tat abnormally increased Ca²âº influx through overactivated, voltage-sensitive L-type Ca²âº channels in pyramidal neurons within the rat medial prefrontal cortex (mPFC). However, it is unknown whether the Tat-induced Ca²âº dysregulation was mediated by increased activity and/or the number of the L-channels. This study tested the hypothesis that transient/early exposure to Tat in vivo promoted enduring L-channel dysregulation in the mPFC without neuron loss. Accordingly, rats were administered a single intracerebroventricular injection of recombinant Tat (80 µg/20 µl; diluted by cerebrospinal fluids to pathophysiological concentrations) or vehicle. Rats were killed 14 days after injection for immunohistochemical assessments of the mPFC, motor cortex, caudate-putamen, and nucleus accumbens. Stereological estimates for positively stained cells indicated a significant increase in the number of cells expressing the pore-forming Ca(v)1.2-α1c subunit of L-channels in the mPFC compared with other regions in Tat-treated or vehicle-treated rat brains. Optical density measurements showed a Tat-induced increase in glial fibrillary acidic protein expression, indicating astrogliosis in the cortical regions. There was no significant loss of neurons in any brain region investigated. These findings indicate that transient Tat exposure in vivo induced enduring L-channel dysregulation and astrogliosis in the mPFC without neuron loss. Such maladaptations may contribute toward dysregulated Ca²âº homeostasis and neuropathology in the PFC in the early stages of HIV infection.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , VIH-1 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/toxicidad , Animales , Corteza Cerebral/metabolismo , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA