Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 210
Filtrar
1.
Nature ; 548(7668): 476-479, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28813419

RESUMEN

Fenethylline, also known by the trade name Captagon, is a synthetic psychoactive stimulant that has recently been linked to a substance-use disorder and 'pharmacoterrorism' in the Middle East. Although fenethylline shares a common phenethylamine core with other amphetamine-type stimulants, it also incorporates a covalently linked xanthine moiety into its parent structure. These independently active pharmacophores are liberated during metabolism, resulting in the release of a structurally diverse chemical mixture into the central nervous system. Although the psychoactive properties of fenethylline have been reported to differ from those of other synthetic stimulants, the in vivo chemical complexity it manifests upon ingestion has impeded efforts to unambiguously identify the specific species responsible for these effects. Here we develop a 'dissection through vaccination' approach, called DISSECTIV, to mitigate the psychoactive effects of fenethylline and show that its rapid-onset and distinct psychoactive properties are facilitated by functional synergy between theophylline and amphetamine. Our results demonstrate that incremental vaccination against a single chemical species within a multi-component mixture can be used to uncover emergent properties arising from polypharmacological activity. We anticipate that DISSECTIV will be used to expose unidentified active chemical species and resolve pharmacodynamic interactions within other chemically complex systems, such as those found in counterfeit or illegal drug preparations, post-metabolic tissue samples and natural product extracts.


Asunto(s)
Anfetamina/farmacología , Anfetaminas/inmunología , Anfetaminas/farmacología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Fraccionamiento Químico/métodos , Teofilina/análogos & derivados , Teofilina/farmacología , Vacunas/inmunología , Anfetamina/química , Anfetamina/inmunología , Anfetamina/metabolismo , Anfetaminas/antagonistas & inhibidores , Anfetaminas/metabolismo , Animales , Productos Biológicos/química , Productos Biológicos/inmunología , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Estimulantes del Sistema Nervioso Central/inmunología , Estimulantes del Sistema Nervioso Central/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Sinergismo Farmacológico , Haptenos/química , Haptenos/inmunología , Haptenos/farmacología , Hemocianinas/química , Hemocianinas/inmunología , Drogas Ilícitas/química , Drogas Ilícitas/inmunología , Drogas Ilícitas/metabolismo , Drogas Ilícitas/farmacología , Masculino , Ratones , Fenetilaminas/análisis , Fenetilaminas/química , Teofilina/antagonistas & inhibidores , Teofilina/química , Teofilina/inmunología , Teofilina/metabolismo , Vacunas/farmacología
2.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34281286

RESUMEN

Norepinephrine (NE) neurons and extracellular NE exert some protective effects against a variety of insults, including methamphetamine (Meth)-induced cell damage. The intimate mechanism of protection remains difficult to be analyzed in vivo. In fact, this may occur directly on target neurons or as the indirect consequence of NE-induced alterations in the activity of trans-synaptic loops. Therefore, to elude neuronal networks, which may contribute to these effects in vivo, the present study investigates whether NE still protects when directly applied to Meth-treated PC12 cells. Meth was selected based on its detrimental effects along various specific brain areas. The study shows that NE directly protects in vitro against Meth-induced cell damage. The present study indicates that such an effect fully depends on the activation of plasma membrane ß2-adrenergic receptors (ARs). Evidence indicates that ß2-ARs activation restores autophagy, which is impaired by Meth administration. This occurs via restoration of the autophagy flux and, as assessed by ultrastructural morphometry, by preventing the dissipation of microtubule-associated protein 1 light chain 3 (LC3) from autophagy vacuoles to the cytosol, which is produced instead during Meth toxicity. These findings may have an impact in a variety of degenerative conditions characterized by NE deficiency along with autophagy impairment.


Asunto(s)
Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Adrenérgicos/farmacología , Animales , Autofagia/efectos de los fármacos , Compartimento Celular/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Desipramina/farmacología , Relación Dosis-Respuesta a Droga , Metanfetamina/administración & dosificación , Microscopía Electrónica de Transmisión , Modelos Neurológicos , Neuronas/ultraestructura , Fármacos Neuroprotectores/farmacología , Norepinefrina/metabolismo , Células PC12 , Ratas , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vacuolas/ultraestructura
3.
J Neurosci ; 36(18): 5160-9, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147666

RESUMEN

UNLABELLED: Schizophrenia-related psychosis is associated with disturbances in mesolimbic dopamine (DA) transmission, characterized by hyperdopaminergic activity in the mesolimbic pathway. Currently, the only clinically effective treatment for schizophrenia involves the use of antipsychotic medications that block DA receptor transmission. However, these medications produce serious side effects leading to poor compliance and treatment outcomes. Emerging evidence points to the involvement of a specific phytochemical component of marijuana called cannabidiol (CBD), which possesses promising therapeutic properties for the treatment of schizophrenia-related psychoses. However, the neuronal and molecular mechanisms through which CBD may exert these effects are entirely unknown. We used amphetamine (AMPH)-induced sensitization and sensorimotor gating in rats, two preclinical procedures relevant to schizophrenia-related psychopathology, combined with in vivo single-unit neuronal electrophysiology recordings in the ventral tegmental area, and molecular analyses to characterize the actions of CBD directly in the nucleus accumbens shell (NASh), a brain region that is the current target of most effective antipsychotics. We demonstrate that Intra-NASh CBD attenuates AMPH-induced sensitization, both in terms of DAergic neuronal activity measured in the ventral tegmental area and psychotomimetic behavioral analyses. We further report that CBD controls downstream phosphorylation of the mTOR/p70S6 kinase signaling pathways directly within the NASh. Our findings demonstrate a novel mechanism for the putative antipsychotic-like properties of CBD in the mesolimbic circuitry. We identify the molecular signaling pathways through which CBD may functionally reduce schizophrenia-like neuropsychopathology. SIGNIFICANCE STATEMENT: The cannabis-derived phytochemical, cannabidiol (CBD), has been shown to have pharmacotherapeutic efficacy for the treatment of schizophrenia. However, the mechanisms by which CBD may produce antipsychotic effects are entirely unknown. Using preclinical behavioral procedures combined with molecular analyses and in vivo neuronal electrophysiology, our findings identify a functional role for the nucleus accumbens as a critical brain region whereby CBD can produce effects similar to antipsychotic medications by triggering molecular signaling pathways associated with the effects of classic antipsychotic medications. Specifically, we report that CBD can attenuate both behavioral and dopaminergic neuronal correlates of mesolimbic dopaminergic sensitization, via a direct interaction with mTOR/p70S6 kinase signaling within the mesolimbic pathway.


Asunto(s)
Anfetamina/antagonistas & inhibidores , Conducta Animal/efectos de los fármacos , Cannabidiol/farmacología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Inhibidores de Captación de Dopamina/antagonistas & inhibidores , Neuronas Dopaminérgicas/efectos de los fármacos , Sistema Límbico/fisiología , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Anfetamina/farmacología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Inhibidores de Captación de Dopamina/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos
4.
Pak J Pharm Sci ; 30(2): 369-374, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28649058

RESUMEN

Methylphenidate is effective in the treatment of attention deficit hyperactivity disorder (ADHD) in children and adults, but its long term use can cause potential adverse effect on growth rate and variable effects on appetite. Previous studies have shown that long term administration of psychostimulant drugs increases the effectiveness of somatodendritic 5-hydroxytryptamine (5-HT)-1A receptors. Repeated administration of buspirone attenuates the effectiveness of somatodendritic 5-HT1A receptors. The present study was designed to test the hypothesis that co-administration of buspirone may attenuate methylphenidate-induced effects on growth rate and food intake. Growth rate was calculated weekly in terms of change in body weight as percentage of preceding week's body weight and food intake was calculated weekly by subtracting the amount of food left in the hopper from the amount of food placed in the hopper as % in preceding week mg/gm of body weight after long-term administration of methylphenidate, buspirone and their co-administration. Long term oral administration of methylphenidate at a dose of 2.0 mg/kg/day decrease growth rate, but co-administration of buspirone at a dose of 10 mg/kg/day attenuates effect of methylphenidate on growth rate however food intake was significantly greater in all treated groups after 3 weeks of treatment. It is suggested that buspirone may oppose methylphenidate-induced growth inhibition by decreasing the sensitivity of somatodendritic 5-HT1A receptors. These findings may help to extend future therapeutics in ADHD.


Asunto(s)
Buspirona/farmacología , Ingestión de Alimentos/efectos de los fármacos , Crecimiento/efectos de los fármacos , Metilfenidato/antagonistas & inhibidores , Animales , Estimulantes del Sistema Nervioso Central/efectos adversos , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Interacciones Farmacológicas , Metilfenidato/efectos adversos , Ratas
5.
Artículo en Inglés | MEDLINE | ID: mdl-27207905

RESUMEN

BACKGROUND: Our pilot study suggested that the angiotensin-converting enzyme inhibitor perindopril might reduce some subjective effects produced by i.v. methamphetamine. We characterized the impact of a wider range of perindopril doses on methamphetamine-induced effects in a larger group of non-treatment-seeking, methamphetamine-using volunteers. METHODS: Before treatment, participants received 30mg methamphetamine. After 5 to 7 days of perindopril treatment (0, 4, 8, or 16mg/d), participants received 15 and 30mg of methamphetamine on alternate days. Before and after treatment, participants rated subjective effects and cardiovascular measures were collected. RESULTS: Prior to treatment with perindopril, there were no significant differences between treatment groups on maximum or peak subjective ratings or on peak cardiovascular effects. Following perindopril treatment, there were significant main effects of treatment on peak subjective ratings of "anxious" and "stimulated"; compared to placebo treatment, treatment with 8mg perindopril significantly reduced peak ratings of both anxious (P=.0009) and stimulated (P=.0070). There were no significant posttreatment differences between groups on peak cardiovascular effects. CONCLUSIONS: Moderate doses of perindopril (8mg) significantly reduced peak subjective ratings of anxious and stimulated as well as attenuated many other subjective effects produced by methamphetamine, likely by inhibiting angiotensin II synthesis. Angiotensin II is known to facilitate the effects of norepinephrine, which contributes to methamphetamine's subjective effects. The lack of a classic dose-response function likely results from either nonspecific effects of perindopril or from between-group differences that were not accounted for in the current study (i.e., genetic variations and/or caffeine use). The current findings suggest that while angiotensin-converting enzyme inhibitors can reduce some effects produced by methamphetamine, more consistent treatment effects might be achieved by targeting components of the renin-angiotensin system that are downstream of angiotensin-converting enzyme.


Asunto(s)
Ansiedad/tratamiento farmacológico , Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Metanfetamina/administración & dosificación , Metanfetamina/farmacología , Perindopril/farmacología , Administración Intravenosa , Adolescente , Adulto , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Ansiedad/inducido químicamente , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Consumidores de Drogas/psicología , Femenino , Voluntarios Sanos/psicología , Humanos , Masculino , Metanfetamina/antagonistas & inhibidores , Persona de Mediana Edad , Perindopril/antagonistas & inhibidores , Adulto Joven
6.
J Appl Toxicol ; 35(8): 927-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25523949

RESUMEN

Recently, we have demonstrated that ginsenoside Re protects methamphetamine (MA)-induced dopaminergic toxicity in mice via genetic inhibition of PKCδ and attenuation of mitochondrial stress. In addition, we have reported that induction of mitochondrial glutathione peroxidase (GPx) is also important for neuroprotection mediated by ginsenoside Re. To extend our knowledge, we examined the effects of ginsenoside Re against MA toxicity in vitro condition using SH-SY5Y neuroblastoma cells. Treatment with ginsenoside Re resulted in significant attenuations against a decrease in the activity of GPx and an increase in the activity of superoxide dismutase (SOD) in the cytosolic and mitochondrial fraction. The changes in glutathione (GSH) paralleled those in GPx in the same experimental condition. Consistently, ginsenoside Re treatment exhibited significant protections against cytosolic and mitochondrial oxidative damage (i.e. lipid peroxidation and protein oxidation), mitochondrial translocation of PKCδ, mitochondrial dysfunction (mitochondrial transmembrane potential and intra-mitochondrial Ca(2+)), apoptotic events [i.e., cytochrome c release from mitochondria, cleavage of caspase-3 and poly(ADP-ribose)polymerase-1, nuclear condensation, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive apoptotic cells], and a reduction in the tyrosine hydroxylase (TH) expression and TH activity induced by MA in SH-SY5Y neuroblastoma cells. These protective effects of ginsenoside Re were comparable to those of PKCδ antisense oligonucleotide (ASO). However, ginsenoside Re did not significantly provide additional protective effects mediated by genetic inhibition of PKCδ. Our results suggest that PKCδ is a specific target for ginsenoside Re-mediated protective activity against MA toxicity in SH-SY5Y neuroblastoma cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Ginsenósidos/farmacología , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteína Quinasa C-delta/genética , Antioxidantes/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Citosol/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
7.
Int J Neuropsychopharmacol ; 15(9): 1275-85, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21939588

RESUMEN

Recent studies indicate that bipolar disorder is associated with mitochondrial dysfunction and oxidative stress. Previous studies in our laboratory have shown that the mood stabilizer lithium inhibits oxidative stress. The α,ß-unsaturated aldehyde 4-hydroxy-2-nonenal (4-HNE), a major product of lipid peroxidation, is able to exert cytotoxicity and disturb cellular function by forming protein adducts. The purpose of this study is to determine whether chronic lithium treatment prevents 4-HNE-protein adduction in an amphetamine-induced hyperactive mania-like model. We found that repeated amphetamine stimulation significantly induced hyperactive behaviour, decreased activities of mitochondrial complexes I and III, and increased 4-HNE-protein adducts in rat frontal cortex, and that chronic lithium treatment inhibited both amphetamine-induced hyperactivity and 4-HNE-protein adduction. Monoamine neurotransmitters are involved in the aetiology and pathology of bipolar disorder and other psychiatric diseases, and also contribute significantly to amphetamine-induced behavioural effects. Vesicular monoamine transporter 2 (VMAT2) is critical in packaging monoamine neurotransmitters. We found that 4-HNE can form protein adducts with VMAT2. Repeated amphetamine stimulation significantly increased 4-HNE-VMAT2 adducts, while chronic lithium treatment reduced amphetamine-increased 4-HNE-VMAT2 adducts in rat frontal cortex. Our findings suggest that chronic lithium treatment may inhibit amphetamine-induced hyperactive mania-like behaviour by preventing 4-HNE-VMAT2 adduction. This finding also indicates that prevention of 4-HNE-VMAT2 adduction may contribute in part to the pharmacological action of lithium for the treatment of bipolar disorder.


Asunto(s)
Aldehídos/metabolismo , Anfetamina/antagonistas & inhibidores , Anfetamina/farmacología , Antimaníacos/farmacología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Litio/farmacología , Proteínas del Tejido Nervioso/metabolismo , Corteza Prefrontal/metabolismo , Animales , Trastorno Bipolar/inducido químicamente , Trastorno Bipolar/metabolismo , Western Blotting , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Masculino , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
8.
Metab Brain Dis ; 27(3): 341-50, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22447521

RESUMEN

Methamphetamine is a highly addictive stimulant drug whose illicit use and resultant addiction has become an alarming global phenomenon. The mesolimbic dopaminergic pathway has been shown to be fundamental to the establishment of addictive behaviour. This pathway, as part of the reward system of the brain, has also been shown to be important in classical conditioning, which is a learnt response. Within the modulation of learning and memory, the neurohypophyseal hormones vasopressin and oxytocin have been reported to play a vital role, with vasopressin exerting a long- term facilitatory effect and oxytocin exerting an inhibitory effect. Therefore we adopted a conditioned place preference model to investigate whether vasopressin V1b receptor antagonist SSR 149415 or oxytocin treatment would cause a decrease in the seeking behaviour in a reinstatement paradigm. Behavioural findings indicated that methamphetamine induced a change in the place preference in the majority of our animals. This change in place preference was not seen when vasopressin was administered during the extinction phase. On the other hand the methamphetamine-induced change in place preference was enhanced during the reinstatement phase in the animals that were treated with oxytocin. Striatal dopamine levels were determined, as methamphetamine is known to increase dopamine transmission in this area. Significant changes in dopamine levels were observed in some of our animals. Rats that received both methamphetamine and oxytocin had significantly higher striatal dopamine than those that received oxytocin alone. Western blot analysis for hippocampal cyclic AMP response element binding protein (CREB) was also conducted as a possible indicator of glutamatergic NMDA receptor activity, a pathway that is important for learning and memory. The Western blot analysis showed no changes in hippocampal pCREB expression. Overall our data led us to conclude that methamphetamine treatment can change place preference behaviour in rats and that this change may be partially restored by vasopressin antagonism, but exaggerated by oxytocin.


Asunto(s)
Trastornos Relacionados con Anfetaminas/tratamiento farmacológico , Metanfetamina/antagonistas & inhibidores , Oxitocina/farmacología , Vasopresinas/farmacología , Trastornos Relacionados con Anfetaminas/metabolismo , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Modelos Animales de Enfermedad , Masculino , Metanfetamina/toxicidad , Oxitocina/fisiología , Ratas , Ratas Sprague-Dawley , Vasopresinas/antagonistas & inhibidores , Vasopresinas/fisiología
9.
J Neurosci ; 30(50): 17051-62, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159975

RESUMEN

L-type Ca(2+) channel (LTCC)-activated signaling cascades contribute significantly to psychostimulant-induced locomotor sensitization; however, the precise contribution of the two brain-specific subunits Ca(v)1.2 and Ca(v)1.3 remains mostly unknown. In this study, by using amphetamine and cocaine locomotor sensitization in mutant mice expressing dihydropyridine (DHP)-insensitive Ca(v)1.2 LTCCs (Ca(v)1.2DHP(-/-)), we find that, as opposed to a previously identified role of the Ca(v)1.3 subunit of LTCCs in development of sensitization, the Ca(v)1.2 subunit mediates expression of amphetamine and cocaine sensitization when examined after a 14 d drug-free period. Molecular studies to further elucidate the role of Ca(v)1.2 versus Ca(v)1.3 LTCCs in activating signaling pathways in the nucleus accumbens (NAc) of drug-naive versus drug-preexposed mice examined 14 d later revealed that an acute amphetamine and cocaine challenge in drug-naive mice increases Ser133 cAMP response element-binding protein (CREB) phosphorylation in the NAc via Ca(v)1.3 channels and via a dopamine D(1)-dependent mechanism, independent of the extracellular signal-regulated kinase (ERK) pathway, an important mediator of psychostimulant-induced plasticity. In contrast, in amphetamine- and cocaine-preexposed mice, an amphetamine or cocaine challenge no longer activates CREB unless Ca(v)1.2 LTCCs are blocked. This Ca(v)1.2-dependent blunting of CREB activation that underlies expression of locomotor sensitization occurs only after extended drug-free periods and involves recruitment of D(1) receptors and the ERK pathway. Thus, our results demonstrate that specific LTCC subunits are required for the development (Ca(v)1.3) versus expression (Ca(v)1.2) of psychostimulant sensitization and that subunit-specific signaling pathways recruited by psychostimulants underlies long-term drug-induced behavioral responses.


Asunto(s)
Anfetamina/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Actividad Motora/efectos de los fármacos , Nifedipino/farmacología , Núcleo Accumbens/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Aminoacetonitrilo/análogos & derivados , Aminoacetonitrilo/farmacología , Anfetamina/antagonistas & inhibidores , Animales , Benzazepinas/farmacología , Canales de Calcio Tipo L/genética , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Cocaína/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Interacciones Farmacológicas , Masculino , Ratones , Ratones Noqueados , Actividad Motora/genética , Núcleo Accumbens/metabolismo
10.
Int J Neuropsychopharmacol ; 13(4): 541-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20047715

RESUMEN

The atypical antipsychotic aripiprazole has been demonstrated to reduce symptoms of bipolar mania. To further profile the antimanic-like properties of aripiprazole in relevant preclinical models, we examined its efficacy in d-amphetamine-based behavioural models of acute mania in rats. The effects of acute and repeated administration of aripiprazole were assessed in the facilitation of intracranial self-stimulation (ICSS) and hyperlocomotion after acute d-amphetamine, and in the sensitized facilitation of ICSS function and hyperlocomotion after repeated d-amphetamine. Acutely, aripiprazole (0.75, 1.5 and 2.5 mg/kg i.p.) increased ICSS thresholds, attenuated the reward-facilitating effects of d-amphetamine (0.5 mg/kg i.p.), decreased motor activity and prevented d-amphetamine-induced hyperlocomotion. Co-administration of aripiprazole and d-amphetamine for 7 d resulted in aripiprazole counteracting the d-amphetamine-induced sensitization in facilitation of brain reward function and hyperlocomotion. These results indicate the efficacy of aripiprazole in d-amphetamine-based preclinical models of acute mania that are characterized by increased motivational drive and/or hyperfunction of brain reward.


Asunto(s)
Trastorno Bipolar/inducido químicamente , Trastorno Bipolar/tratamiento farmacológico , Dextroanfetamina/farmacología , Modelos Animales de Enfermedad , Piperazinas/farmacología , Quinolonas/farmacología , Autoestimulación/efectos de los fármacos , Animales , Antipsicóticos/administración & dosificación , Antipsicóticos/farmacología , Aripiprazol , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Dextroanfetamina/administración & dosificación , Dextroanfetamina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Locomoción/efectos de los fármacos , Masculino , Piperazinas/administración & dosificación , Quinolonas/administración & dosificación , Ratas , Ratas Sprague-Dawley
11.
Horm Behav ; 58(3): 533-43, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20399212

RESUMEN

Stimulant abuse continues to be a problem, particularly for women. There is increasing preclinical and clinical evidence showing that the hormone progesterone attenuates the behavioral effects of cocaine, and this effect is primarily observed in females. The purpose of the present study was to determine if progesterone would also alter the behavioral effects of another stimulant, oral d-amphetamine (AMPH) in women. Eighteen normal non-drug abusing women completed eight outpatient sessions over two menstrual cycles. During the follicular phase of each cycle, women were administered AMPH (0, 10, 20 mg); in one cycle they were pretreated with oral micronized progesterone (200 mg) and in another cycle they were pretreated with placebo progesterone. Each session, participants completed a range of tasks including subjective measures of abuse liability, cognitive performance tasks, and behavioral measures of impulsivity and risk-taking. AMPH produced dose-related increases in positive subjective effects and these effects were enhanced by progesterone pretreatment. AMPH alone, or in combination with progesterone, had little effect on performance or behavioral measures of impulsivity. These results are in contrast with previous studies showing that progesterone attenuates the subjective response to cocaine and nicotine. Additional studies are needed to explore the modulatory role of progesterone on the effects of AMPH to determine whether progesterone has any clinical utility for AMPH abuse.


Asunto(s)
Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Cognición/efectos de los fármacos , Dextroanfetamina/antagonistas & inhibidores , Progesterona/administración & dosificación , Administración Oral , Adulto , Estimulantes del Sistema Nervioso Central/administración & dosificación , Dextroanfetamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Conducta Impulsiva , Ciclo Menstrual , Placebos , Asunción de Riesgos
12.
Addict Biol ; 14(4): 408-18, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19489752

RESUMEN

Opioid receptor antagonism has been shown to attenuate behavioral and neurochemical effects of amphetamine in humans and rodents. The effects of acute (oral or subcutaneous) or extended-release naltrexone (XR-NTX) were tested on the reward-enhancing effects of amphetamine using the intracranial self-stimulation (ICSS) paradigm. Acute exposure to drugs of abuse reduces the locus of rise (LOR) in the ICSS procedure, reflecting enhanced brain stimulation reward (BSR). Rats were treated once a day with naltrexone orally (PO; 5.0 mg/kg) or subcutaneously (SC; 0.5 mg/kg) for four consecutive days and tested with D-amphetamine (0.5 mg/kg, intraperitoneal) in the ICSS paradigm 30 minutes later on days 1 and 4. Separate groups of rats received XR-NTX (50 mg/kg, SC) or placebo microspheres (similar mass to XR-NTX, SC) on day 0 and tested with D-amphetamine in the ICSS paradigm on days 4, 14, 21, 28 and 41 after administration. Naltrexone plasma concentrations were determined for each amphetamine testing session using liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). In rats pretreated with naltrexone acutely, amphetamine-potentiated BSR did not differ from vehicle-pretreated rats on either day 1 or day 4 (25-30% decrease in LOR). In XR-NTX-pretreated rats, amphetamine-potentiated BSR was reduced by 64 and 70% on days 4 and 14, respectively, compared to placebo microsphere-treated controls. This effect dissipated by day 21. Naltrexone plasma concentrations were comparable across all treatment groups (14-30 ng/ml) on days 1, 4 and 14. In summary, an extended-release formulation of naltrexone results in significant attenuation of psychostimulant-enhanced BSR that is not observed with acute naltrexone.


Asunto(s)
Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Dextroanfetamina/antagonistas & inhibidores , Dextroanfetamina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Administración Oral , Animales , Estimulantes del Sistema Nervioso Central/administración & dosificación , Dextroanfetamina/administración & dosificación , Vías de Administración de Medicamentos , Esquema de Medicación , Cromatografía de Gases y Espectrometría de Masas , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Masculino , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
13.
Am J Drug Alcohol Abuse ; 35(6): 412-6, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20014909

RESUMEN

BACKGROUND: Although preclinical studies support the contribution of the noradrenergic system activation in mediating the acute effects of amphetamines, these findings have not been followed up in clinical studies. OBJECTIVES: To examine the effects of atomoxetine, a norepinephrine transporter inhibitor, on subjective, physiological, and plasma cortisol responses to dextroamphetamine in 10 healthy volunteers. METHODS: Subjects were randomly assigned to a sequence of atomoxetine (40 mg/day) or placebo treatments each lasting for 4 days. On Day 4 of each treatment period, responses to a single 20 mg/70 kg dose of dextroamphetamine were assessed. RESULTS: Atomoxetine treatment attenuated dextroamphetamine-induced increases in systolic and diastolic blood pressure and plasma cortisol as well as the self-report ratings of "stimulated," "high," and "good drug effects." CONCLUSIONS: These findings are consistent with previous preclinical studies supporting the role of the noradrenergic system in mediating acute amphetamine responses. SCIENTIFIC SIGNIFICANCE: Atomoxetine's capacity to attenuate some of the physiological and subjective responses to dextroamphetamine supports its potential use for stimulant addiction.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Dextroanfetamina/antagonistas & inhibidores , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Propilaminas/farmacología , Adulto , Afecto/efectos de los fármacos , Clorhidrato de Atomoxetina , Presión Sanguínea/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Dextroanfetamina/farmacología , Interacciones Farmacológicas , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hidrocortisona/sangre , Masculino
14.
Psychopharmacology (Berl) ; 236(11): 3147-3158, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31139878

RESUMEN

RATIONALE: Previous research indicates that the selective sigma-1 receptor ligand PD144418 and the selective sigma-2 ligands YUN-252 can inhibit cocaine-induced hyperactivity. The effects of these ligands on other stimulants, such as methamphetamine, have not been reported. OBJECTIVES: The present study examined the effects of PD144418 and YUN-252 pretreatment on methamphetamine-induced hyperactivity after acute treatment. METHODS: Mice (n = 8-14/group) were injected with PD144418 (3.16, 10, or 31.6 µmol/kg), YUN-252 (0.316, 3.16, 31.6 µmol/kg), or saline. After 15 min, mice injected with 2.69 µmol/kg methamphetamine or saline vehicle, where distance traveled during a 60-min period was recorded. Additionally, the effect of PD144418 on the initiation and expression of methamphetamine sensitization was determined by treating mice (n = 8-14/group) with PD144418, methamphetamine or saline repeatedly over a 5-day period, and testing said mice with a challenge dose after a 7-day withdrawal period. RESULTS: Results indicate that both PD144418 and YUN-252, in a dose-dependent manner, attenuated hyperactivity induced by an acute methamphetamine injection. Specifically, 10 µmol/kg or 31.6 µmol/kg of PD144418 and 31 µmol/kg of YUN-252 suppressed methamphetamine-induced hyperactivity. In regard to methamphetamine sensitization, while 10 µmol/kg PD144418 prevented the initiation of methamphetamine sensitization, it did not have an effect on the expression. CONCLUSIONS: Overall, the current results suggest an intriguing potential for this novel sigma receptor ligand as a treatment for the addictive properties of methamphetamine. Future analysis of this novel sigma receptor ligand in assays directly measuring reinforcement properties will be critical.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Isoxazoles/metabolismo , Locomoción/efectos de los fármacos , Metanfetamina/farmacología , Piridinas/metabolismo , Receptores sigma/metabolismo , Animales , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Isoxazoles/farmacología , Ligandos , Locomoción/fisiología , Masculino , Ratones , Piridinas/farmacología , Receptores sigma/antagonistas & inhibidores , Refuerzo en Psicología , Receptor Sigma-1
15.
Curr Clin Pharmacol ; 14(2): 125-131, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30417792

RESUMEN

BACKGROUND: Psychostimulants can induce behavioral sensitization by their chronic use. The main target for the action of these drugs is dopamine, neither epinephrine nor serotonin transporters. Serotonin is synthesized by the precursor L-tryptophan. Tryptophan and methylphenidate being 5-HT agonists, both increase the level of serotonin thereby causing desensitization of 5-HT1a receptors. The present study investigated whether behavioral sensitization induced by Methylphenidate is decreased in tryptophan administrated animals. METHODS: The Experiment was divided into 2 phases (1). Behavioral effects of repeated administration of TRP 100 mg/kg and MPD for 14 days in three groups; (i) water (ii) MPD 1.0 mg/kg (iii) TRP. To explore the locomotor effects of treatment, the activity was monitored in a familiar and novel environment. (2) Behavioral consequences of repeatedly administrated MPD (1.0 mg/kg) on pretreated TRP (100 mg/kg) and MPD (1.0 mg/kg) animals following Co-MPD and TRP for 14 days, rats were divided in three groups (i) water, (ii) MPD and (iii) TRP as mentioned in Experiment no 1. After two weeks six subgroups were assigned i.e. (i) water-saline, (ii) water- MPD, (iii) TRP-saline (iv) TRP-MPD (v) MPD-saline and (vi) MPD-MPD+TRP and treated for further 14 days. Locomotor behavior was monitored in familiar environment on the next day and in novel environment on alternate days of each administration. RESULTS: The Results from phase 1 showed increased activity in both (TRP and MPD) treatments. However, the results of phase 2 showed significant decrease in methylphenidate-induced behavioral sensitization by both pretreatment and co-administration with TRP. CONCLUSION: The present study suggests the potential of tryptophan to decrease the risk of behavioral sensitization induced by methylphenidate.


Asunto(s)
Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Dendritas/efectos de los fármacos , Metilfenidato/antagonistas & inhibidores , Metilfenidato/farmacología , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Triptófano/farmacología , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Wistar
16.
Neurotoxicology ; 74: 91-99, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31163210

RESUMEN

There are no Food and Drug Administration approved pharmacotherapies for methamphetamine (METH) overdose, thus identifying novel drug targets to prevent this devastating adverse event is a public-health imperative. Previous research suggests that serotonin and sigma receptors may contribute to the adverse effects of METH. The present study assessed whether pretreatment with the 5-HT2A receptor antagonist M100907 or the sigma 1 (σ1) receptor antagonist BD 1047 attenuated METH-induced lethality, hyperthermia, convulsions, and seizures. Male, Swiss-Webster mice received intraperitoneal injections of M100907 (1 and 10 mg/kg), BD 1047 (10 mg/kg), or a combination of M100907 (1 mg/kg) and BD 1047 (10 mg/kg) prior to treatment with METH (78 mg/kg). Convulsions and lethality were assessed by observation, core body temperature was assessed by surgically implanted telemetric probes, and seizures were assessed by electroencephalography. M100907 reduced METH-elicited lethality from 67% to 33%, BD1047 reduced METH-elicited lethality from 67% to 50%, and combined administration of both agents eliminated lethality in all mice tested. Similarly, both agents and their combination reduced METH-elicited seizures and convulsions. None of the treatments decreased METH-induced hyperthermia. This research suggests that reducing METH-induced seizures is an important factor in reducing lethality associated with METH overdose. However, future studies should examine whether M100907 and BD 1047 modulate METH-induced hypertension and other adverse effects that may also contribute to METH overdose. Our data support the continued investigation of compounds that target 5-HT2A and σ1 receptors in METH-induced overdose, including their potential to yield emergency reversal agents.


Asunto(s)
Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Etilenodiaminas/farmacología , Fluorobencenos/farmacología , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Piperidinas/farmacología , Receptores sigma/antagonistas & inhibidores , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Fiebre/inducido químicamente , Fiebre/prevención & control , Dosificación Letal Mediana , Masculino , Ratones , Convulsiones/inducido químicamente , Convulsiones/prevención & control , Receptor Sigma-1
17.
Neuropsychopharmacology ; 33(8): 1856-63, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17957221

RESUMEN

Amphetamine abuse and dependence is a global health concern with a collateral increase in medical and social problems. Although some of the neurobiological mechanisms underlying amphetamine dependence and its devastating effects in humans are known, the development of rational and evidence-based treatment is lagging. There is evidence from preclinical studies suggesting that the endogenous opioid system plays a role in mediating some of the behavioral and neurochemical effects of amphetamine in a variety of controlled settings. In the present study we assessed the effects of naltrexone, an opioid antagonist (50 mg) on the subjective physiological and biochemical response to dexamphetamine (30 mg) in 20 amphetamine-dependent patients. Patients received naltrexone/amphetamine followed by placebo/amphetamine, 1 week apart in a randomized double-blind placebo-controlled design. The primary objective of the study was to evaluate the effect of pretreatment with naltrexone on the subjective response to amphetamine, using a Visual Analog Scale. The secondary objective was to investigate the effects of naltrexone on physiological and biochemical responses to amphetamine, as measured by changes in blood pressure, heart rate, skin conductance, and cortisol. Naltrexone significantly attenuated the subjective effects produced by dexamphetamine in dependent patients (p<0.001). Pretreatment with naltrexone also significantly blocked the craving for dexamphetamine (p<0.001). There was no difference between the groups on the physiological measures. The results suggest that the subjective effects of amphetamine could be modulated via the endogenous opioid system. The potential of naltrexone as an adjunct pharmaceutical for amphetamine dependence is promising.


Asunto(s)
Trastornos Relacionados con Anfetaminas/psicología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Dextroanfetamina/antagonistas & inhibidores , Dextroanfetamina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Adulto , Envejecimiento/psicología , Estimulantes del Sistema Nervioso Central/farmacocinética , Estudios Cruzados , Dextroanfetamina/farmacocinética , Método Doble Ciego , Endorfinas/fisiología , Semivida , Humanos , Hidrocortisona/sangre , Masculino , Naltrexona/farmacocinética , Antagonistas de Narcóticos/farmacocinética , Escalas de Valoración Psiquiátrica , Pruebas Psicológicas , Síndrome de Abstinencia a Sustancias/psicología
18.
Neuroscience ; 151(1): 92-103, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18082966

RESUMEN

Methamphetamine (MA) is a drug of abuse as well as a dopaminergic neurotoxin. We have previously demonstrated that pretreatment with bone morphogenetic protein 7 (BMP7) reduced 6-hydroxydopamine-mediated neurodegeneration in a rodent model of Parkinson's disease. In this study, we examined the neuroprotective effects of BMP7 against MA-mediated toxicity in dopaminergic neurons. Primary dopaminergic neurons, prepared from rat embryonic ventral mesencephalic tissue, were treated with MA. High doses of MA decreased tyrosine hydroxylase immunoreactivity (THir) while increasing terminal deoxynucleotidyl transferase-mediated dNTP nick end labeling. These toxicities were significantly antagonized by BMP7. Interaction of BMP7 and MA in vivo was first examined in CD1 mice. High doses of MA (10 mg/kgx4 s.c.) significantly reduced locomotor activity and THir in striatum. I.c.v. administration of BMP7 antagonized these changes. In BMP7 +/- mice, MA suppressed locomotor activity and reduced TH immunoreactivity in nigra reticulata to a greater degree than in wild type BMP7 +/+ mice, suggesting that deficiency in BMP7 expression increases vulnerability to MA insults. Since BMP7 +/- mice also carry a LacZ-expressing reporter allele at the BMP7 locus, the expression of BMP7 was indirectly measured through the enzymatic activity of beta-galactosidase (beta-gal) in BMP7 +/- mice. High doses of MA significantly suppressed beta-gal activity in striatum, suggesting that MA may inhibit BMP7 expression at the terminals of the nigrostriatal pathway. A similar effect was also found in CD1 mice in that high doses of MA suppressed BMP7 mRNA expression in nigra. In conclusion, our data indicate that MA can cause lesioning in the nigrostriatal dopaminergic terminals and that BMP7 is protective against MA-mediated neurotoxicity in central dopaminergic neurons.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Fármacos Neuroprotectores , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , Recuento de Células , Células Cultivadas , Femenino , Factor 2 de Diferenciación de Crecimiento , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Mesencéfalo/citología , Mesencéfalo/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Embarazo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirosina 3-Monooxigenasa/metabolismo , beta-Galactosidasa/metabolismo
19.
Pharmacol Biochem Behav ; 91(2): 217-22, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18703079

RESUMEN

Modafinil is approved for use in the treatment of excessive daytime sleepiness. The precise mechanism of modafinil action has not been elucidated, although both dopamine (DA) and norepinephrine (NE) systems have been implicated. To explore the roles of DA and NE in the mechanism of modafinil-induced arousal, dopamine beta-hydroxylase knockout (Dbh -/-) mice were examined in behavioral paradigms of arousal (photobeam breaks and behavioral scoring of sleep latency). Dbh -/- mice completely lack NE but have hypersensitive DA signaling. It was hypothesized that Dbh -/- mice would be unresponsive to modafinil if the compound acts primarily via NE, but would be hypersensitive to modafinil if it acts primarily via DA. Dbh -/- mice had increased sensitivity to the locomotor-activating and wake-promoting effects of modafinil. Paradoxically, the alpha1-adrenergic receptor antagonist, prazosin, attenuated the effects of modafinil in control mice, but not in Dbh -/- mice. Blockade of DA receptors with flupenthixol decreased modafinil-induced locomotion and wake in both control and Dbh -/- mice. These results suggest that both NE and DA are involved in the behavioral effects of modafinil in control mice, but the requirement for NE can be bypassed by hypersensitive DA signaling.


Asunto(s)
Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina beta-Hidroxilasa/genética , Dopamina beta-Hidroxilasa/fisiología , Dopamina/fisiología , Norepinefrina/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1 , Antagonistas Adrenérgicos alfa/farmacología , Animales , Compuestos de Bencidrilo/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Antagonistas de Dopamina/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Flupentixol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modafinilo , Actividad Motora/efectos de los fármacos , Norepinefrina/antagonistas & inhibidores , Prazosina/farmacología , Receptores Adrenérgicos/efectos de los fármacos , Receptores Dopaminérgicos/efectos de los fármacos , Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos
20.
Drug Alcohol Depend ; 186: 75-79, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29550625

RESUMEN

BACKGROUND AND PURPOSE: Little is known about how chemokine systems influence the behavioral effects of designer cathinones and psychostimulants. The chemokine CXCL12 and its principal receptor target, CXCR4, are of particular interest because CXCR4 activation enhances mesolimbic dopamine output that facilitates psychostimulant reward, reinforcement, and locomotor activation. Repeated cocaine enhances CXCL12 gene expression in the midbrain and produces conditioned place preference (CPP) that is inhibited by a CXCR4 antagonist. Yet, interactions between chemokines and synthetic cathinones remain elusive. METHODS: We tested the hypothesis that an FDA-approved CXCR4 antagonist (AMD3100) inhibits MDPV-induced reward, locomotor activation and positive affective state in rats using a triad of behavioral assays (CPP, open field, and 50-kHz ultrasonic vocalizations [USVs]). KEY RESULTS: AMD3100 (1, 2.5, 5, 10 mg/kg, ip) significantly reduced MDPV (2 mg/kg, ip)-evoked hyper-locomotion in a dose-related manner. AMD3100 (1, 5, 10 mg/kg) administered during CPP conditioning caused a significant, dose-dependent reduction of MDPV (2 mg/kg x 4 days) place preference. MDPV injection elicited significantly greater 50 kHz USVs in vehicle-pretreated rats but not in AMD3100-pretreated rats. CONCLUSION AND IMPLICATION: A CXCR4 antagonist reduced the rewarding and locomotor-activating effects of MDPV. Our results identify the existence of chemokine/cathinone interactions and suggest the rewarding and stimulant effects of MDPV, similar to cocaine, require an active CXCL12/CXCR4 system.


Asunto(s)
Alcaloides/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Quimiocinas/farmacología , Drogas de Diseño/farmacología , Actividad Motora/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Recompensa , Alcaloides/farmacología , Animales , Bencilaminas , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Ciclamas , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Vocalización Animal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA