Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Pharmacol Exp Ther ; 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38262742

RESUMEN

MJN110 inhibits the enzyme monoacylglycerol lipase (MAGL) to increase levels of the endocannabinoid (eCB) 2-arachidonoylglycerol (2-AG), an endogenous high-efficacy agonist of cannabinoid 1 and 2 receptors (CB1/2R). MAGL inhibitors are under consideration as candidate analgesics, and we reported previously that acute MJN110 produced partial antinociception in an assay of pain-related behavioral depression in mice. Given the need for repeated analgesic administration in many pain patients and the potential for analgesic tolerance during repeated treatment, this study examined antinociceptive effects of repeated MJN110 on pain-related behavioral depression and CB1R-mediated G-protein function. Male and female ICR mice were treated daily for 7 days in a 2x2 design with (a) 1.0 mg/kg/day MJN110or its vehicle followed by (b) intraperitoneal injection of dilute lactic acid (IP acid) or its vehicle as a visceral noxious stimulus to depress nesting behavior. After behavioral testing, G-protein activity was assessed in lumbar spinal cord andfive brain regions using an assay of CP55,940-stimulated [35S]GTPÉ£S activation. As reported previously, acute MJN110 produced partial but significant relief of IP acid-induced nesting depression on Day 1. After 7 days, MJN110 continued to produce significant but partial antinociception in males, while antinociceptive tolerance developed in females. Repeated MJN110 also produced modest decreases in maximum levels of CP55,940-induced [35S]GTPÉ£S binding in spinal cord and most brain regions. These results indicate that repeated treatment with a relatively low antinociceptive MJN110 dose produces only partial and sex-dependent transient antinociception associated with the emergence of CB1R desensitization in this model of IP acid-induced nesting depression. Significance Statement The drug MJN110 inhibits monoacylglycerol lipase (MAGL) to increase levels of the endogenous cannabinoid 2-arachidonoylglycerol and produce potentially useful therapeutic effects including analgesia. This study used an assay of pain-related behavioral depression in mice to show that repeated MJN110 treatment produced (1) weak but sustained antinociception in male mice, (2) antinociceptive tolerance in females, and (3) modest cannabinoid-receptor desensitization that varied by region and sex. Antinociceptive tolerance may limit the utility of MJN110 for treatment of pain.

2.
J Pharmacol Exp Ther ; 366(3): 509-518, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945931

RESUMEN

The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [35S]GTPγS autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35S]GTPγS binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Péptidos Opioides/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Temperatura , Animales , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/uso terapéutico , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/fisiopatología , Receptores de Lisoesfingolípidos/agonistas , Nociceptina
3.
Neurobiol Dis ; 92(Pt B): 124-36, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26845176

RESUMEN

Co-exposure to opiates and HIV/HIV proteins results in enhanced CNS morphological and behavioral deficits in HIV(+) individuals and in animal models. Opiates with abuse liability, such as heroin and morphine, bind preferentially to and have pharmacological actions through µ-opioid-receptors (MORs). The mechanisms underlying opiate-HIV interactions are not understood. Exposure to the HIV-1 transactivator of transcription (Tat) protein causes neurodegenerative outcomes that parallel many aspects of the human disease. We have also observed that in vivo exposure to Tat results in apparent changes in morphine efficacy, and thus have hypothesized that HIV proteins might alter MOR activation. To test our hypothesis, MOR-mediated G-protein activation was determined in neuroAIDS-relevant forebrain regions of transgenic mice with inducible CNS expression of HIV-1 Tat. G-protein activation was assessed by MOR agonist-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate ([(35)S]GTPγS) autoradiography in brain sections, and in concentration-effect curves of MOR agonist-stimulated [(35)S]GTPγS binding in membranes isolated from specific brain regions. Comparative studies were done using the MOR-selective agonist DAMGO ([D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin) and a more clinically relevant agonist, morphine. Tat exposure reduced MOR-mediated G-protein activation in an agonist, time, and regionally dependent manner. Levels of the GPCR regulatory protein ß-arrestin-2, which is involved in MOR desensitization, were found to be elevated in only one affected brain region, the amygdala; amygdalar ß-arrestin-2 also showed a significantly increased association with MOR by co-immunoprecipitation, suggesting decreased availability of MOR. Interestingly, this correlated with changes in anxiety and fear-conditioned extinction, behaviors that have substantial amygdalar input. We propose that HIV-1 Tat alters the intrinsic capacity of MOR to signal in response to agonist binding, possibly via a mechanism involving altered expression and/or function of ß-arrestin-2.


Asunto(s)
Ansiedad/metabolismo , Miedo/fisiología , Prosencéfalo/metabolismo , Receptores Opioides mu/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/metabolismo , Analgésicos Opioides/farmacología , Animales , Ansiedad/virología , Condicionamiento Psicológico/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Proteínas de Unión al GTP/metabolismo , VIH-1 , Masculino , Ratones , Ratones Transgénicos , Morfina/farmacología , Prosencéfalo/efectos de los fármacos , Receptores Opioides mu/agonistas , Arrestina beta 2/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
4.
J Neurosci ; 34(15): 5152-63, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24719095

RESUMEN

For many G-protein-coupled receptors (GPCRs), including cannabinoid receptor 1 (CB1R), desensitization has been proposed as a principal mechanism driving initial tolerance to agonists. GPCR desensitization typically requires phosphorylation by a G-protein-coupled receptor kinase (GRK) and interaction of the phosphorylated receptor with an arrestin. In simple model systems, CB1R is desensitized by GRK phosphorylation at two serine residues (S426 and S430). However, the role of these serine residues in tolerance and dependence for cannabinoids in vivo was unclear. Therefore, we generated mice where S426 and S430 were mutated to nonphosphorylatable alanines (S426A/S430A). S426A/S430A mutant mice were more sensitive to acutely administered delta-9-tetrahydrocannabinol (Δ(9)-THC), have delayed tolerance to Δ(9)-THC, and showed increased dependence for Δ(9)-THC. S426A/S430A mutants also showed increased responses to elevated levels of endogenous cannabinoids. CB1R desensitization in the periaqueductal gray and spinal cord following 7 d of treatment with Δ(9)-THC was absent in S426A/S430A mutants. Δ(9)-THC-induced downregulation of CB1R in the spinal cord was also absent in S426A/S430A mutants. Cultured autaptic hippocampal neurons from S426A/S430A mice showed enhanced endocannabinoid-mediated depolarization-induced suppression of excitation (DSE) and reduced agonist-mediated desensitization of DSE. These results indicate that S426 and S430 play major roles in the acute response to, tolerance to, and dependence on cannabinoids. Additionally, S426A/S430A mice are a novel model for studying pathophysiological processes thought to involve excessive endocannabinoid signaling such as drug addiction and metabolic disease. These mice also validate the approach of mutating GRK phosphorylation sites involved in desensitization as a general means to confer exaggerated signaling to GPCRs in vivo.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Tolerancia a Medicamentos , Mutación Missense , Receptor Cannabinoide CB1/metabolismo , Secuencias de Aminoácidos , Animales , Sensibilización del Sistema Nervioso Central , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiología , Potenciales de la Membrana , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Sustancia Gris Periacueductal/fisiología , Fosforilación , Unión Proteica , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/química , Receptor Cannabinoide CB1/genética , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiología
5.
J Pharmacol Exp Ther ; 354(3): 316-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26099530

RESUMEN

Δ(9)-Tetrahydrocannabinol (THC), the main psychoactive component of marijuana, produces motor and motivational effects via interactions with the dopaminergic system in the caudate-putamen and nucleus accumbens. However, the molecular events that underlie these interactions after THC treatment are not well understood. Our study shows that pretreatment with dopamine D1 receptor (D1R) antagonists before repeated administration of THC attenuated induction of Δ FBJ murine osteosarcoma viral oncogene homolog B (ΔFosB) in the nucleus accumbens, caudate-putamen, amygdala, and prefrontal cortex. Anatomical studies showed that repeated THC administration induced ΔFosB in D1R-containing striatal neurons. Dopamine signaling in the striatum involves phosphorylation-specific effects of the dopamine- and cAMP-regulated phosphoprotein Mr 32 kDa (DARPP-32), which regulates protein kinase A signaling. Genetic deletion of DARPP-32 attenuated ΔFosB expression measured after acute, but not repeated, THC administration in both the caudate-putamen and nucleus accumbens. THC was then acutely or repeatedly administered to wild-type (WT) and DARPP-32 knockout (KO) mice, and in vivo responses were measured. DARPP-32 KO mice exhibited enhanced acute THC-mediated hypolocomotion and developed greater tolerance to this response relative to the WT mice. Agonist-stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPγS) binding showed that cannabinoid-stimulated G-protein activity did not differ between DARPP-32 KO and WT mice treated with vehicle or repeated THC. These results indicate that D1Rs play a major role in THC-mediated ΔFosB induction in the forebrain, whereas the role of DARPP-32 in THC-mediated ΔFosB induction and modulation of motor activity appears to be more complex.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Dronabinol/farmacología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Dopamina D1/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Dopamina/metabolismo , Antagonistas de Dopamina/farmacología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Putamen/efectos de los fármacos , Putamen/metabolismo
6.
J Pharmacol Exp Ther ; 352(2): 195-207, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25398241

RESUMEN

A growing body of evidence implicates endogenous cannabinoids as modulators of the mesolimbic dopamine system and motivated behavior. Paradoxically, the reinforcing effects of Δ(9)-tetrahydrocannabinol (THC), the primary psychoactive constituent of cannabis, have been difficult to detect in preclinical rodent models. In this study, we investigated the impact of THC and inhibitors of the endocannabinoid hydrolytic enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) on operant responding for electrical stimulation of the medial forebrain bundle [intracranial self-stimulation (ICSS)], which is known to activate the mesolimbic dopamine system. These drugs were also tested in assays of operant responding for food reinforcement and spontaneous locomotor activity. THC and the MAGL inhibitor JZL184 (4-[bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester) attenuated operant responding for ICSS and food, and also reduced spontaneous locomotor activity. In contrast, the FAAH inhibitor PF-3845 (N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide) was largely without effect in these assays. Consistent with previous studies showing that combined inhibition of FAAH and MAGL produces a substantially greater cannabimimetic profile than single enzyme inhibition, the dual FAAH-MAGL inhibitor SA-57 (4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester) produced a similar magnitude of ICSS depression as that produced by THC. ICSS attenuation by JZL184 was associated with increased brain levels of 2-arachidonoylglycerol (2-AG), whereas peak effects of SA-57 were associated with increased levels of both N-arachidonoylethanolamine (anandamide) and 2-AG. The cannabinoid receptor type 1 receptor antagonist rimonabant, but not the cannabinoid receptor type 2 receptor antagonist SR144528, blocked the attenuating effects of THC, JZL184, and SA-57 on ICSS. Thus, THC, MAGL inhibition, and dual FAAH-MAGL inhibition not only reduce ICSS, but also decrease other reinforced and nonreinforced behaviors.


Asunto(s)
Dronabinol/farmacología , Endocannabinoides/metabolismo , Inhibidores Enzimáticos/farmacología , Haz Prosencefálico Medial/efectos de los fármacos , Refuerzo en Psicología , Autoestimulación , Amidohidrolasas/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Benzodioxoles/farmacología , Compuestos de Bifenilo/farmacología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Masculino , Haz Prosencefálico Medial/enzimología , Haz Prosencefálico Medial/metabolismo , Ratones Endogámicos C57BL , Monoacilglicerol Lipasas/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Piperidinas/farmacología , Compuestos de Piridinio/farmacología
7.
J Pharmacol Exp Ther ; 354(2): 111-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25998048

RESUMEN

Inhibition of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), the primary hydrolytic enzymes for the respective endocannabinoids N-arachidonoylethanolamine (AEA) and 2-arachidonylglycerol (2-AG), produces antinociception but with minimal cannabimimetic side effects. Although selective inhibitors of either enzyme often show partial efficacy in various nociceptive models, their combined blockade elicits augmented antinociceptive effects, but side effects emerge. Moreover, complete and prolonged MAGL blockade leads to cannabinoid receptor type 1 (CB1) receptor functional tolerance, which represents another challenge in this potential therapeutic strategy. Therefore, the present study tested whether full FAAH inhibition combined with partial MAGL inhibition would produce sustained antinociceptive effects with minimal cannabimimetic side effects. Accordingly, we tested a high dose of the FAAH inhibitor PF-3845 (N-​3-​pyridinyl-​4-​[[3-​[[5-​(trifluoromethyl)-​2-​pyridinyl]oxy]phenyl]methyl]-​1-​piperidinecarboxamide; 10 mg/kg) given in combination with a low dose of the MAGL inhibitor JZL184 [4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate] (4 mg/kg) in mouse models of inflammatory and neuropathic pain. This combination of inhibitors elicited profound increases in brain AEA levels (>10-fold) but only 2- to 3-fold increases in brain 2-AG levels. This combination produced significantly greater antinociceptive effects than single enzyme inhibition and did not elicit common cannabimimetic effects (e.g., catalepsy, hypomotility, hypothermia, and substitution for Δ(9)-tetrahydrocannabinol in the drug-discrimination assay), although these side effects emerged with high-dose JZL184 (i.e., 100 mg/kg). Finally, repeated administration of this combination did not lead to tolerance to its antiallodynic actions in the carrageenan assay or CB1 receptor functional tolerance. Thus, full FAAH inhibition combined with partial MAGL inhibition reduces neuropathic and inflammatory pain states with minimal cannabimimetic effects.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Analgésicos/administración & dosificación , Agonistas de Receptores de Cannabinoides/administración & dosificación , Antagonistas de Receptores de Cannabinoides/administración & dosificación , Monoacilglicerol Lipasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Animales , Benzodioxoles/administración & dosificación , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Quimioterapia Combinada , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Monoacilglicerol Lipasas/metabolismo , Piperidinas/administración & dosificación , Piridinas/administración & dosificación , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Factores de Tiempo , Resultado del Tratamiento
8.
Epilepsia ; 53(5): 897-907, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22509801

RESUMEN

PURPOSE: The endocannabinoid system is known to modulate seizure activity in several in vivo and in vitro models, and CB(1) -receptor activation is anticonvulsant in the rat pilocarpine model of acquired epilepsy (AE). In these epileptic rats, a unique redistribution of the CB(1) receptor occurs within the hippocampus; however, an anatomically inclusive analysis of the effect of status epilepticus (SE)-induced AE on CB(1) receptors has not been thoroughly evaluated. Therefore, statistical parametric mapping (SPM), a whole-brain unbiased approach, was used to study the long-term effect of pilocarpine-induced SE on CB(1) -receptor binding and G-protein activation in rats with AE. METHODS: Serial coronal sections from control and epileptic rats were cut at equal intervals throughout the neuraxis and processed for [(3) H]WIN55,212-2 (WIN) autoradiography, WIN-stimulated [(35) S]GTPγS autoradiography, and CB(1) -receptor immunohistochemistry (IHC). The autoradiographic techniques were evaluated with both region of interest (ROI) and SPM analyses. KEY FINDINGS: In rats with AE, regionally specific increases in CB(1) -receptor binding and activity were detected in cortex, discrete thalamic nuclei, and other regions including caudate-putamen and septum, and confirmed by IHC. However, CB(1) receptors were unaltered in several brain regions, including substantia nigra and cerebellum, and did not exhibit regional decreases in rats with AE. SIGNIFICANCE: This study provides the first comprehensive evaluation of the regional distribution of changes in CB(1) -receptor expression, binding, and G-protein activation in the rat pilocarpine model of AE. These regions may ultimately serve as targets for cannabinomimetic compounds or manipulation of the endocannabinoid system in epileptic brain.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Epilepsia/patología , Proteínas de Unión al GTP/metabolismo , Imagenología Tridimensional , Receptor Cannabinoide CB1/metabolismo , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Benzoxazinas/farmacocinética , Encéfalo/efectos de los fármacos , Mapeo Encefálico , Diazepam/farmacología , Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Masculino , Morfolinas/farmacocinética , Naftalenos/farmacocinética , Pilocarpina/toxicidad , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Isótopos de Azufre/farmacocinética , Factores de Tiempo , Tritio/farmacocinética
9.
Proc Natl Acad Sci U S A ; 106(48): 20270-5, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19918051

RESUMEN

Delta(9)-tetrahydrocannabinol (THC), the psychoactive component of marijuana, and other direct cannabinoid receptor (CB1) agonists produce a number of neurobehavioral effects in mammals that range from the beneficial (analgesia) to the untoward (abuse potential). Why, however, this full spectrum of activities is not observed upon pharmacological inhibition or genetic deletion of either fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), enzymes that regulate the two major endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG), respectively, has remained unclear. Here, we describe a selective and efficacious dual FAAH/MAGL inhibitor, JZL195, and show that this agent exhibits broad activity in the tetrad test for CB1 agonism, causing analgesia, hypomotilty, and catalepsy. Comparison of JZL195 to specific FAAH and MAGL inhibitors identified behavioral processes that were regulated by a single endocannabinoid pathway (e.g., hypomotility by the 2-AG/MAGL pathway) and, interestingly, those where disruption of both FAAH and MAGL produced additive effects that were reversed by a CB1 antagonist. Falling into this latter category was drug discrimination behavior, where dual FAAH/MAGL blockade, but not disruption of either FAAH or MAGL alone, produced THC-like responses that were reversed by a CB1 antagonist. These data indicate that AEA and 2-AG signaling pathways interact to regulate specific behavioral processes in vivo, including those relevant to drug abuse, thus providing a potential mechanistic basis for the distinct pharmacological profiles of direct CB1 agonists and inhibitors of individual endocannabinoid degradative enzymes.


Asunto(s)
Amidohidrolasas/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Carbamatos/farmacología , Endocannabinoides , Monoacilglicerol Lipasas/metabolismo , Piperazinas/farmacología , Piperidinas/farmacología , Amidohidrolasas/antagonistas & inhibidores , Animales , Ácidos Araquidónicos/metabolismo , Carbamatos/síntesis química , Hidrolasas de Éster Carboxílico/metabolismo , Glicéridos/metabolismo , Ratones , Estructura Molecular , Monoacilglicerol Lipasas/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Dimensión del Dolor , Piperazinas/síntesis química , Piperidinas/síntesis química , Alcamidas Poliinsaturadas/metabolismo
10.
J Pharmacol Exp Ther ; 339(1): 173-85, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21719468

RESUMEN

Δ(9)-Tetrahydrocannbinol (THC), the primary active constituent of Cannabis sativa, has long been known to reduce opioid withdrawal symptoms. Although THC produces most of its pharmacological actions through the activation of CB(1) and CB(2) cannabinoid receptors, the role these receptors play in reducing the variety of opioid withdrawal symptoms remains unknown. The endogenous cannabinoids, N-arachidonoylethanolamine (anandamide; AEA) and 2-arachidonylglycerol (2-AG), activate both cannabinoid receptors but are rapidly metabolized by fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively. The objective of this study was to test whether increasing AEA or 2-AG, via inhibition of their respective hydrolytic enzymes, reduces naloxone-precipitated morphine withdrawal symptoms in in vivo and in vitro models of opioid dependence. Morphine-dependent mice challenged with naloxone reliably displayed a profound withdrawal syndrome, consisting of jumping, paw tremors, diarrhea, and weight loss. THC and the MAGL inhibitor 4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate (JZL184) dose dependently reduced the intensity of most measures through the activation of CB(1) receptors. JZL184 also attenuated spontaneous withdrawal signs in morphine-dependent mice. The FAAH inhibitor N-(pyridin-3-yl)-4-(3-(5-(trifluoromethyl)pyridin-2-yloxy)benzyl)-piperdine-1-carboxamide (PF-3845) reduced the intensity of naloxone-precipitated jumps and paw flutters through the activation of CB(1) receptors but did not ameliorate incidence of diarrhea or weight loss. In the final series of experiments, we investigated whether JZL184 or PF-3845 would attenuate naloxone-precipitated contractions in morphine-dependent ilea. Both enzyme inhibitors attenuated the intensity of naloxone-induced contractions, although this model does not account mechanistically for the autonomic withdrawal responses (i.e., diarrhea) observed in vivo. These results indicate that endocannabinoid catabolic enzymes are promising targets to treat opioid dependence.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Monoacilglicerol Lipasas/antagonistas & inhibidores , Dependencia de Morfina/complicaciones , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Amidohidrolasas/genética , Animales , Ácido Araquidónico/metabolismo , Conducta Animal/efectos de los fármacos , Benzodioxoles/farmacología , Química Encefálica/efectos de los fármacos , Diarrea/prevención & control , Dronabinol/farmacología , Estimulación Eléctrica , Hidrólisis , Íleon/efectos de los fármacos , Íleon/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Monoacilglicerol Lipasas/genética , Contracción Muscular/efectos de los fármacos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Piperidinas/farmacología , Prostaglandinas/metabolismo , Piridinas/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB2/antagonistas & inhibidores , Síndrome de Abstinencia a Sustancias/psicología , Pérdida de Peso/efectos de los fármacos
11.
Nat Neurosci ; 9(2): 205-11, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16415864

RESUMEN

The transcription factor DeltaFosB is induced in the nucleus accumbens (NAc) and dorsal striatum by the repeated administration of drugs of abuse. Here, we investigated the role of DeltaFosB in the NAc in behavioral responses to opiates. We achieved overexpression of DeltaFosB by using a bitransgenic mouse line that inducibly expresses the protein in the NAc and dorsal striatum and by using viral-mediated gene transfer to specifically express the protein in the NAc. DeltaFosB overexpression in the NAc increased the sensitivity of the mice to the rewarding effects of morphine and led to exacerbated physical dependence, but also reduced their sensitivity to the analgesic effects of morphine and led to faster development of analgesic tolerance. The opioid peptide dynorphin seemed to be one target through which DeltaFosB produced this behavioral phenotype. Together, these experiments demonstrated that DeltaFosB in the NAc, partly through the repression of dynorphin expression, mediates several major features of opiate addiction.


Asunto(s)
Morfina/farmacología , Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Trastornos Relacionados con Opioides/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Western Blotting , Dinorfinas/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Núcleo Accumbens/fisiología , Recompensa
12.
Neuropharmacology ; 176: 108170, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32479813

RESUMEN

Nicotine, the primary psychoactive component in tobacco, plays a major role in the initiation and maintenance of tobacco dependence and addiction, a leading cause of preventable death worldwide. An essential need thus exists for more effective pharmacotherapies for nicotine-use cessation. Previous reports suggest that pharmacological and genetic blockade of CB1 receptors attenuate nicotine reinforcement and reward; while exogenous agonists enhanced these abuse-related behaviors. In this study, we utilized complementary genetic and pharmacologic approaches to test the hypothesis that increasing the levels of the endocannabinoid 2-arachindonoylglycerol (2-AG), will enhance nicotine reward by stimulating neuronal CB1 receptors. Contrary to our hypothesis, we found that inhibition of monoacylglycerol lipase (MAGL), the primary catabolic enzyme of 2-AG, attenuates nicotine conditioned place preference (CPP) in mice, through a non-CB1 receptor-mediated mechanism. MAGL inhibition did not alter palatable food reward or Lithium Chloride (LiCl) aversion. In support of our findings, repeated MAGL inhibition did not induce a reduction in CB1 brain receptor levels or hinder function. To explore the potential mechanism of action, we investigated if MAGL inhibition affected other fatty acid levels in our CPP paradigm. Indeed, MAGL inhibition caused a concomitant decrease in arachidonic acid (AA) levels in various brain regions of interest, suggesting an AA cascade-dependent mechanism. This idea is supported by dose-dependent attenuation of nicotine preference by the selective COX-2 inhibitors valdecoxib and LM-4131. Collectively, these findings, along with our reported studies on nicotine withdrawal, suggest that inhibition of MAGL represents a promising new target for the development of pharmacotherapies to treat nicotine dependence.


Asunto(s)
Condicionamiento Clásico/efectos de los fármacos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo , Nicotina/administración & dosificación , Recompensa , Tabaquismo/metabolismo , Animales , Ansiolíticos/farmacología , Ácidos Araquidónicos/farmacología , Benzodioxoles/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Condicionamiento Clásico/fisiología , Endocannabinoides/farmacología , Inhibidores Enzimáticos/farmacología , Glicéridos/farmacología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Piperidinas/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Tabaquismo/tratamiento farmacológico , Tabaquismo/psicología
13.
Neuropharmacology ; 166: 107935, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31917153

RESUMEN

Neuropathy is major source of chronic pain that can be caused by mechanically or chemically induced nerve injury. Intraplantar formalin injection produces local necrosis over a two-week period and has been used to model neuropathy in rats. To determine whether neuropathy alters dopamine (DA) receptor responsiveness in mesolimbic brain regions, we examined dopamine D1-like and D2-like receptor (D1/2R) signaling and expression in male rats 14 days after bilateral intraplantar formalin injections into both rear paws. D2R-mediated G-protein activation and expression of the D2R long, but not short, isoform were reduced in nucleus accumbens (NAc) core, but not in NAc shell, caudate-putamen or ventral tegmental area of formalin- compared to saline-treated rats. In addition, D1R-stimulated adenylyl cyclase activity was also reduced in NAc core, but not in NAc shell or prefrontal cortex, of formalin-treated rats, whereas D1R expression was unaffected. Other proteins involved in dopamine neurotransmission, including dopamine uptake transporter and tyrosine hydroxylase, were unaffected by formalin treatment. In behavioral tests, the potency of a D2R agonist to suppress intracranial self-stimulation (ICSS) was decreased in formalin-treated rats, whereas D1R agonist effects were not altered. The combination of reduced D2R expression and signaling in NAc core with reduced suppression of ICSS responding by a D2R agonist suggest a reduction in D2 autoreceptor function. Altogether, these results indicate that intraplantar formalin produces attenuation of highly specific DA receptor signaling processes in NAc core of male rats and suggest the development of a neuropathy-induced allostatic state in both pre- and post-synaptic DA receptor function.


Asunto(s)
Formaldehído/toxicidad , Neuralgia/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Transducción de Señal/fisiología , Animales , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Neuralgia/inducido químicamente , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D2/agonistas , Transducción de Señal/efectos de los fármacos
14.
J Neurochem ; 110(4): 1191-202, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19493165

RESUMEN

Sphingosine-1-phosphate (S1P) is a ubiquitous, lipophilic cellular mediator that acts in part by activation of G-protein-coupled receptor. Modulation of S1P signaling is an emerging pharmacotherapeutic target for immunomodulatory drugs. Although multiple S1P receptor types exist in the CNS, little is known about their function. Here, we report that S1P stimulated G-protein activity in the CNS, and results from [(35)S]GTPgammaS autoradiography using the S1P(1)-selective agonist SEW2871 and the S1P(1/3)-selective antagonist VPC44116 show that in several regions a majority of this activity is mediated by S1P(1) receptors. S1P receptor activation inhibited glutamatergic neurotransmission as determined by electrophysiological recordings in cortical neurons in vitro, and this effect was mimicked by SEW2871 and inhibited by VPC44116. Moreover, central administration of S1P produced in vivo effects resembling the actions of cannabinoids, including thermal antinociception, hypothermia, catalepsy and hypolocomotion, but these actions were independent of CB(1) receptors. At least one of the central effects of S1P, thermal antinociception, is also at least partly S1P(1) receptor mediated because it was produced by SEW2871 and attenuated by VPC44116. These results indicate that CNS S1P receptors are part of a physiologically relevant and widespread neuromodulatory system, and that the S1P(1) receptor contributes to S1P-mediated antinociception.


Asunto(s)
Corteza Cerebral/metabolismo , Lisofosfolípidos/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Neurotransmisores/metabolismo , Esfingosina/análogos & derivados , Animales , Animales Recién Nacidos , Unión Competitiva/fisiología , Cannabinoides/farmacología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Técnicas de Cocultivo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Neuronas/citología , Neuronas/efectos de los fármacos , Oxadiazoles/farmacología , Técnicas de Placa-Clamp , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Neurotransmisores/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/metabolismo , Radioisótopos de Azufre/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tiofenos/farmacología
15.
Int Rev Psychiatry ; 21(2): 113-21, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19367505

RESUMEN

Humans have used Cannabis sativa (marijuana) for at least 12,000 years, but researchers have only recently described an endogenous cannabinoid system. The endocannabinoid system modulates an array of physiological and psychological functions. Endocannabinoids are widely distributed throughout the body, including the central nervous system (CNS). This article gives a basic overview of endocannabinoid neuroanatomy and function. Several endocannabinoids have been discovered to date, and their roles are being elucidated. Two G-protein coupled cannabinoid receptors, CB1R and CB2R, have been identified, although other candidate receptors exist, including ion channel and nuclear receptors that might be components of the endocannabinoid system. It appears that cannabinoids are dysregulated in a number of psychiatric disorders and might be involved in their pathogenesis. There is now evidence that manipulation of the endocannabinoid system could be a therapeutic target for a variety of conditions.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/patología , Agonistas de Receptores de Cannabinoides , Moduladores de Receptores de Cannabinoides/farmacología , Cannabinoides/farmacología , Animales , Encéfalo/metabolismo , Humanos , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB2/agonistas
16.
Neuron ; 38(6): 941-52, 2003 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12818179

RESUMEN

Regulators of G protein signaling (RGS) modulate heterotrimeric G proteins in part by serving as GTPase-activating proteins for Galpha subunits. We examined a role for RGS9-2, an RGS subtype highly enriched in striatum, in modulating dopamine D2 receptor function. Viral-mediated overexpression of RGS9-2 in rat nucleus accumbens (ventral striatum) reduced locomotor responses to cocaine (an indirect dopamine agonist) and to D2 but not to D1 receptor agonists. Conversely, RGS9 knockout mice showed heightened locomotor and rewarding responses to cocaine and related psychostimulants. In vitro expression of RGS9-2 in Xenopus oocytes accelerated the off-kinetics of D2 receptor-induced GIRK currents, consistent with the in vivo data. Finally, chronic cocaine exposure increased RGS9-2 levels in nucleus accumbens. Together, these data demonstrate a functional interaction between RGS9-2 and D2 receptor signaling and the behavioral actions of psychostimulants and suggest that psychostimulant induction of RGS9-2 represents a compensatory adaptation that diminishes drug responsiveness.


Asunto(s)
Dopamina/metabolismo , Canales de Potasio de Rectificación Interna , Proteínas RGS/fisiología , Transducción de Señal , Animales , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Cuerpo Estriado/fisiología , Conductividad Eléctrica , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/química , Oocitos/metabolismo , Canales de Potasio/fisiología , Proteínas RGS/análisis , Proteínas RGS/deficiencia , Proteínas RGS/genética , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D2/fisiología , Transfección , Xenopus
17.
J Pharmacol Exp Ther ; 327(2): 546-53, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18682568

RESUMEN

Inhibition of the metabolism of the endocannabinoids, anandamide (AEA) and 2-arachidonyl glycerol (2-AG), by their primary metabolic enzymes, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively, has the potential to increase understanding of the physiological functions of the endocannabinoid system. To date, selective inhibitors of FAAH, but not MAGL, have been developed. The purpose of this study was to determine the selectivity and efficacy of N-arachidonyl maleimide (NAM), a putative MAGL inhibitor, for modulation of the effects of 2-AG. Our results showed that NAM unmasked 2-AG activity in a tetrad of in vivo tests sensitive to the effects of cannabinoids in mice. The efficacy of 2-AG (and AEA) to produce hypothermia was reduced compared with Delta(9)-tetrahydrocannabinol; however, 2-AG differed from AEA by its lower efficacy for catalepsy. All tetrad effects were partially CB(1) receptor-mediated because they were attenuated (but not eliminated) by SR141716A [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-H-pyrazole-3-carboxamide HCl] and in CB(1)(-/-) mice. In vitro, NAM increased endogenous levels of 2-AG in the brain. Furthermore, NAM raised the potency of 2-AG, but not AEA, in agonist-stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate binding assay, a measure of G-protein activation. These results suggest that NAM is an MAGL inhibitor with in vivo and in vitro efficacy. NAM and other MAGL inhibitors are valuable tools to elucidate the biological functions of 2-AG and to examine the consequences of dysregulation of this endocannabinoid. In addition, NAM's unmasking of 2-AG effects that are only partially reversed by SR141716A offers support for the existence of non-CB(1), non-CB(2) cannabinoid receptors.


Asunto(s)
Ácidos Araquidónicos/farmacología , Glicéridos/farmacología , Maleimidas/farmacología , Animales , Sinergismo Farmacológico , Endocannabinoides , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ratones , Ratones Endogámicos ICR , Monoacilglicerol Lipasas/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/fisiología , Rimonabant
18.
J Pharmacol Exp Ther ; 324(2): 664-73, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17967938

RESUMEN

Chronic treatment with Delta(9)-tetrahydrocannabinol (THC) produces tolerance to cannabinoid-mediated behaviors and region-specific adaptation of brain cannabinoid receptors. However, the relationship between receptor adaptation and tolerance is not well understood, and the dose-response relationship of THC-induced cannabinoid receptor adaptation is unknown. This study assessed cannabinoid receptor function in the brain and cannabinoid-mediated behaviors after chronic treatment with different dosing regimens of THC. Mice were treated twice per day for 6.5 days with the following: vehicle, 10 mg/kg THC, or escalating doses of 10 to 20 to 30 or 10 to 30 to 60 mg/kg THC. Tolerance to cannabinoid-mediated locomotor inhibition, ring immobility, antinociception, and hypothermia was produced by both ramping THC-dose paradigms. Administration of 10 mg/kg THC produced less tolerance development, the magnitude of which depended upon the particular behavior. Decreases in cannabinoid-mediated G-protein activation, which varied with treatment dose and region, were observed in autoradiographic and membrane guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS)-binding assays in brains from THC-treated mice. Agonist-stimulated [(35)S]GTPgammaS binding was reduced in the hippocampus, cingulate cortex, periaqueductal gray, and cerebellum after all treatments. Decreased agonist-stimulated [(35)S]GTPgammaS binding in the caudate-putamen, nucleus accumbens, and preoptic area occurred only after administration of 10 to 30 to 60 mg/kg THC, and no change was found in the globus pallidus or entopeduncular nucleus after any treatment. Changes in the CB(1) receptor B(max) values also varied by region, with hippocampus and cerebellum showing reductions after all treatments and striatum/globus pallidus showing effects only at higher dosing regimens. These results reveal that tolerance and CB(1) receptor adaptation exhibit similar dose-dependent development, and they are consistent with previous studies demonstrating less cannabinoid receptor adaptation in striatal circuits.


Asunto(s)
Adaptación Biológica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Dronabinol/farmacología , Tolerancia a Medicamentos , Receptores de Cannabinoides/metabolismo , Adaptación Biológica/fisiología , Animales , Encéfalo/metabolismo , Agonistas de Receptores de Cannabinoides , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos/fisiología , Masculino , Ratones , Ratones Endogámicos ICR , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología
19.
Neuropharmacology ; 55(7): 1183-90, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18708079

RESUMEN

Dysregulation of the endocannabinoid and dopamine systems has been implicated in schizophrenia. The purpose of this study was to examine the effects of sub-chronic treatment with two antipsychotics on CB1 receptor-mediated in vitro and in vivo effects. Adult and adolescent male and female rats were injected twice daily with haloperidol (0.3 mg/kg), clozapine (10 mg/kg), or saline for 10 days. Subsequently, CB1 receptor number and function were assessed by [3H]SR141716 and WIN55,212-2-stimulated [35S]GTPgammaS binding, respectively. The effects of sub-chronic antipsychotic treatment on the in vivo actions of Delta9-tetrahydrocannabinol (Delta9-THC) were also evaluated. In adult female rats, antipsychotic treatment attenuated maximal stimulation of CB1 receptor-mediated G-protein activity in the striatum (clozapine) and prefrontal cortex (both antipsychotics), but not in the ventral midbrain. Associated changes in CB1 receptor number were not observed, suggesting that this attenuation was not due to downregulation. In vivo, sub-chronic treatment with clozapine, but not haloperidol, attenuated Delta9-THC-induced suppression of activity in adult females, whereas neither drug altered hypothermia or catalepsy. In contrast, antipsychotic treatment did not change CB1 receptor-mediated G-protein activation in any brain region in adult male rats and in adolescents of either sex. In vivo, haloperidol, but not clozapine, enhanced Delta9-THC-mediated suppression of activity and hypothermia in adult male rats whereas neither antipsychotic affected Delta9-THC-induced in vivo effects in adolescent rats. These findings suggest that modulation of the endocannabinoid system might contribute in a sex- and age-selective manner to differences in motor side effects of clozapine versus haloperidol.


Asunto(s)
Antipsicóticos/farmacología , Proteínas de Unión al GTP/fisiología , Receptor Cannabinoide CB1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Envejecimiento/fisiología , Envejecimiento/psicología , Animales , Células CHO , Células Cultivadas , Clozapina/farmacología , Cricetinae , Cricetulus , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Haloperidol/farmacología , Masculino , Piperidinas/metabolismo , Pirazoles/metabolismo , Ratas , Ratas Long-Evans , Rimonabant , Caracteres Sexuales
20.
Pharmacol Biochem Behav ; 173: 58-64, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30125591

RESUMEN

RATIONALE: There is extensive literature regarding nicotine-opioid functional interactions. The possibility that use of nicotine products during adolescence might increase the risk of substance abuse such as morphine later in adulthood is particularly relevant to the current opioid crisis. OBJECTIVES: To investigate the effects of nicotine exposure for seven days during adolescence in mice on morphine reward as well as morphine physical dependence later in adulthood. METHODS: Mice were exposed to nicotine in either early or late adolescence then evaluated for morphine reward and withdrawal symptoms in adulthood. A separate group of mice was exposed to nicotine during adolescent and tissue was evaluated for changes in MOR-mediated G-protein activity using [35S]GTPγS binding assays. RESULTS: We report that a 7-day exposure to a low dose of nicotine during early adolescence significantly enhanced morphine preference in the CPP test in adult mice. In contrast, the same treatment with nicotine had no effect on expression of somatic withdrawal signs in morphine-dependent adult mice. MOR-mediated G-protein activity in hippocampus, but not thalamus and striatum of adult mice, was significantly altered by adolescent nicotine treatment. CONCLUSIONS: Adolescence is a unique developmental stage during which nicotine has long-term effects on future drug-taking behavior. Further studies are needed to identify the neurotransmitters and mechanisms involved in increased vulnerability to drug abuse.


Asunto(s)
Hipocampo/efectos de los fármacos , Morfina/farmacología , Nicotina/administración & dosificación , Receptores Opioides mu/metabolismo , Recompensa , Animales , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Nicotina/farmacología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA