Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
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.
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
3.
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
4.
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
5.
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
6.
Cannabis Cannabinoid Res ; 2(1): 224-234, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29082320

RESUMEN

Introduction: Repeated administration of abused drugs, including Δ9-tetrahydrocannabinol (THC), induces the stable transcription factor ΔFosB in dopaminergic terminal field regions of the mesolimbic system. These studies investigated the effect of prior repeated THC treatment on THC-induced ΔFosB expression and regulation of downstream targets in the forebrain. Methods: Mice received THC (10 mg/kg) or vehicle twice daily for 13 days, and then half of each group received a single injection of THC or vehicle 45 min before brain collection. ΔFosB messenger RNA (mRNA) and protein were measured by polymerase chain reaction and immunoblotting, respectively. Potential downstream targets of ΔFosB induction were measured by immunoblot. Results: THC injection in mice with a history of repeated THC treatment enhanced ΔFosB expression as compared with vehicle in the prefrontal cortex (PFC), nucleus accumbens (NAc), and amygdala. This change occurred concomitantly with an increase in ΔFosB mRNA in the PFC and NAc. THC injection in mice with a history of repeated THC treatment increased expression of cyclin-dependent kinase 5 (Cdk5) and its regulatory protein p35 only in the PFC. This increase in Cdk5 and p35 expression in PFC was also found in mice that had only received repeated THC administration, suggesting that this effect might be due to induction of ΔFosB. Extracellular signal-regulated kinase (ERK) phosphorylation was increased in PFC after THC injection in repeated THC-treated mice. Phosphorylation of glycogen synthase kinase-3ß (GSK3ß), a Cdk5 target, was reduced in PFC after repeated THC treatment regardless of THC history, and phosphorylation of dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) at the Cdk5-regulated threonine 75 site was unchanged. Conclusion: These results suggest that a history of repeated THC administration primes THC-mediated induction of ΔFosB in the NAc and PFC, and that expression of both downstream targets of ΔFosB (e.g., Cdk5 and p35) and upstream activators (e.g., pERK) in the PFC is dependent on THC history, which might have functional implications in addiction and neuropsychiatric disease.

7.
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
8.
Neuropharmacology ; 105: 308-317, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26808314

RESUMEN

Adolescence represents a unique developmental period associated with increased risk-taking behavior and experimentation with drugs of abuse, in particular nicotine. We hypothesized that exposure to nicotine during early adolescence might increase the risk for drug reward in adulthood. To test this hypothesis, male ICR mice were treated with a subchronic regimen of nicotine or saline during adolescence, and their preference for cocaine, morphine and amphetamine was examined using the conditioned place preference (CPP) test in adulthood. Long-term behavioral changes induced by nicotine suggested a possible role of altered gene transcription. Thus, immunoblot for ΔFosB, a member of the Fos family of transcription factors, was conducted in the nucleus accumbens of these mice. Mice treated with nicotine during early but not late adolescence showed an increase in CPP for cocaine, morphine and amphetamine later in adulthood. This effect was not seen in mice pretreated with a subchronic regimen of nicotine as adults, suggesting that exposure to nicotine specifically during early adolescence increases the rewarding effects of other drugs in adulthood. However, adolescent nicotine exposure did not alter highly palatable food conditioning in mice. The enhancement of cocaine CPP by nicotine was strain-dependent and was blocked by pretreatment with nicotinic antagonists. In addition, nicotine exposure during early adolescence induced ΔFosB expression to a greater extent than identical nicotine exposure in adulthood, and enhanced cocaine-induced locomotor sensitization later in adulthood. These results suggest that nicotine exposure during early adolescence increases drug-induced reward in adulthood through mechanisms that may involve the induction of ΔFosB.


Asunto(s)
Trastornos Relacionados con Cocaína/psicología , Cocaína/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Recompensa , Envejecimiento/psicología , Anfetamina/farmacología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante , Ingestión de Alimentos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética
9.
Cell Rep ; 12(5): 798-808, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212325

RESUMEN

The endocannabinoid 2-arachidonoylglycerol (2-AG) is a retrograde lipid messenger that modulates synaptic function, neurophysiology, and behavior. 2-AG signaling is terminated by enzymatic hydrolysis-a reaction that is principally performed by monoacylglycerol lipase (MAGL). MAGL is broadly expressed throughout the nervous system, and the contributions of different brain cell types to the regulation of 2-AG activity in vivo remain poorly understood. Here, we genetically dissect the cellular anatomy of MAGL-mediated 2-AG metabolism in the brain and show that neurons and astrocytes coordinately regulate 2-AG content and endocannabinoid-dependent forms of synaptic plasticity and behavior. We also find that astrocytic MAGL is mainly responsible for converting 2-AG to neuroinflammatory prostaglandins via a mechanism that may involve transcellular shuttling of lipid substrates. Astrocytic-neuronal interplay thus provides distributed oversight of 2-AG metabolism and function and, through doing so, protects the nervous system from excessive CB1 receptor activation and promotes endocannabinoid crosstalk with other lipid transmitter systems.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Astrocitos/metabolismo , Comunicación Celular/fisiología , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Neuronas/metabolismo , Animales , Ácidos Araquidónicos/genética , Astrocitos/citología , Endocannabinoides/genética , Glicéridos/genética , Ratones , Ratones Noqueados , Monoacilglicerol Lipasas/genética , Monoacilglicerol Lipasas/metabolismo , Neuronas/citología , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo
10.
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
11.
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
12.
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
13.
Biochem Pharmacol ; 91(3): 380-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25093286

RESUMEN

Repeated Δ(9)-tetrahydrocannabinol (THC) administration produces cannabinoid type 1 receptor (CB1R) desensitization and downregulation, as well as tolerance to its in vivo pharmacological effects. However, the magnitude of CB1R desensitization varies by brain region, with CB1Rs in the striatum and its output nuclei undergoing less desensitization than other regions. A growing body of data indicates that regional differences in CB1R desensitization are produced, in part, by THC-mediated induction of the stable transcription factor, ΔFosB, and subsequent regulation of CB1Rs. The purpose of the present study was to determine whether THC-mediated induction of ΔFosB in the striatum inhibits CB1R desensitization in the striatum and output nuclei. This hypothesis was tested using bitransgenic mice with inducible expression of ΔFosB or ΔcJun, a dominant negative inhibitor of AP-1-mediated transcription, in specific forebrain regions. Mice were treated repeatedly with escalating doses of THC or vehicle for 6.5 days, and CB1R-mediated G-protein activation was assessed using CP55,940-stimulated [(35)S]GTPγS autoradiography. Overexpression of ΔFosB in striatal dopamine type 1 receptor-containing (D1R) medium spiny neurons (MSNs) attenuated CB1R desensitization in the substantia nigra, ventral tegmental area (VTA) and amygdala. Expression of ΔcJun in striatal D1R- and dopamine type 2 receptor (D2R)-containing MSNs enhanced CB1R desensitization in the caudate-putamen and attenuated desensitization in the hippocampus and VTA. THC-mediated in vivo pharmacological effects were then assessed in ΔcJun-expressing mice. Tolerance to THC-mediated hypomotility was enhanced in ΔcJun-expressing mice. These data reveal that ΔFosB and possibly other AP-1 binding proteins regulate CB1R signaling and adaptation in the striatum and limbic system.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Cuerpo Estriado/efectos de los fármacos , Dronabinol/farmacología , Sistema Límbico/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cannabinoide CB1/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Cuerpo Estriado/metabolismo , Tolerancia a Medicamentos , Proteínas de Unión al GTP/metabolismo , Genes jun , Sistema Límbico/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-fos/genética , Receptor Cannabinoide CB1/genética , Factor de Transcripción AP-1/genética
14.
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
15.
Neuropharmacology ; 77: 224-33, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24090766

RESUMEN

Repeated Δ(9)-tetrahydrocannabinol (THC) administration produces desensitization and downregulation of cannabinoid type 1 receptors (CB1Rs) in the brain, but the magnitude of these adaptations varies among regions. CB1Rs in the striatum and its output regions exhibit the least magnitude and slowest development of desensitization and downregulation. The molecular mechanisms that confer these region-dependent differences are not known. The stable transcription factor, ΔFosB, is induced in the striatum following repeated THC administration and could regulate CB1Rs. To directly compare the regional profile of ΔFosB induction and CB1R desensitization and downregulation, mice were treated with THC (10 mg/kg) or vehicle for 13.5 days. CP55,940-stimulated [(35)S]GTPγS autoradiography and immunohistochemistry were performed to measure CB1R desensitization and downregulation, respectively, and ΔFosB expression was measured by immunoblot. Significant CB1R desensitization and downregulation occurred in the prefrontal cortex, lateral amygdala and hippocampus; desensitization was found in the basomedial amygdala and no changes were seen in remaining regions. ΔFosB was induced in the prefrontal cortex, caudate-putamen, nucleus accumbens and lateral amygdala. An inverse regional relationship between ΔFosB expression and CB1R desensitization was found, such that regions with the greatest ΔFosB induction did not exhibit CB1R desensitization and areas without ΔFosB induction had the greatest desensitization, with remaining regions exhibiting intermediate levels of both. Dual immunohistochemistry in the striatum showed both CB1R co-localization with ΔFosB in cells and CB1R puncta surrounding ΔFosB-positive cells. THC-induced expression of ΔFosB was absent in the striatum of CB1R knockout mice. These data suggest that transcriptional targets of ΔFosB might inhibit CB1R desensitization and/or that ΔFosB induction could be limited by CB1R desensitization.


Asunto(s)
Encéfalo/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Dronabinol/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cannabinoide CB1/metabolismo , Animales , Encéfalo/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/genética , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Receptor Cannabinoide CB1/genética
16.
Life Sci ; 93(5-6): 187-93, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23782998

RESUMEN

A significant number of patients experience chronic pain and the intractable side effects of currently prescribed pain medications. Recent evidence indicates important pain-modulatory roles for two classes of G-protein-coupled receptors that are activated by endogenous lipid ligands, the endocannabinoid (eCB) and sphingosine-1-phosphate (S1P) receptors, which are widely expressed in both the immune and nervous systems. In the central nervous system (CNS), CB1 cannabinoid and S1P1 receptors are most abundantly expressed and exhibit overlapping anatomical distributions and similar signaling mechanisms. The eCB system has emerged as a potential target for treatment of chronic pain, but comparatively little is known about the roles of S1P in pain regulation. Both eCB and S1P systems modulate pain perception via the central and peripheral nervous systems. In most paradigms studied, the eCB system mainly inhibits pain perception. In contrast, S1P acting peripherally at S1P1 and S1P3 receptors can enhance sensitivity to various pain stimuli or elicit spontaneous pain. However, S1P acting at S1P1 receptors and possibly other targets in the CNS can attenuate sensitivity to various pain stimuli. Interestingly, other endogenous sphingolipid derivatives might play a role in central pain sensitization. Moreover, these sphingolipids can also act as CB1 cannabinoid receptor antagonists, but the physiological relevance of this interaction is unknown. Overall, both eCB and sphingolipid systems offer promising targets for the treatment of chronic pain. This review compares and contrasts the eCB and S1P systems with a focus on their roles in pain modulation, and considers possible points of interaction between these systems.


Asunto(s)
Antagonistas de Receptores de Cannabinoides/líquido cefalorraquídeo , Cannabinoides/antagonistas & inhibidores , Cannabinoides/líquido cefalorraquídeo , Sistema Nervioso Central/fisiología , Endocannabinoides/líquido cefalorraquídeo , Lisofosfolípidos/líquido cefalorraquídeo , Dolor/líquido cefalorraquídeo , Esfingosina/líquido cefalorraquídeo , Animales , Antagonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/uso terapéutico , Sistema Nervioso Central/efectos de los fármacos , Endocannabinoides/farmacología , Endocannabinoides/uso terapéutico , Humanos , Lisofosfolípidos/farmacología , Lisofosfolípidos/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Esfingolípidos/líquido cefalorraquídeo , Esfingolípidos/farmacología , Esfingolípidos/uso terapéutico , Esfingosina/farmacología , Esfingosina/uso terapéutico
17.
Biochem Pharmacol ; 84(12): 1551-62, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22971335

RESUMEN

Lysolipids are important mediators of cellular communication in multiple physiological processes. Sphingosine-1-phosphate (S1P) is a major lysolipid in many organs, including the central nervous system (CNS). This commentary discusses recent findings on the role of S1P in regulating pain perception, and highlights advances and challenges in the field. S1P interacts with multiple cellular targets, including G-protein-coupled receptors. Known S1P receptors include five types, four of which are expressed in the CNS (S1P(1,2,3,5)) where they are localized on neurons and glia. S1P receptor-mediated G-protein activation has been demonstrated throughout the CNS, including regions that regulate nociception. S1P receptors couple to multiple G-proteins to produce various intracellular responses, and can mediate both excitatory and inhibitory neuromodulation, depending on the receptor type and cellular context. Both antinociceptive and pro-nociceptive effects of S1P have been reported, and both actions can involve S1P(1) receptors. Current evidence suggests that antinociception is mediated by CNS neurons, whereas pro-nociception is mediated by primary afferent neurons or immune cells in the periphery, or CNS glia. Nonetheless, peripheral administration of the S1P(1,3,4,5) agonist pro-drug, FTY720, produces antinociception. FTY720 is approved to treat multiple sclerosis, and produces potent anti-inflammatory effects, which suggests potential utility for painful autoimmune diseases. Furthermore, evidence suggests that the S1P system interacts with other pain-modulatory systems, such as endogenous cannabinoid and opioid systems, and putative novel sphingolipid targets in the CNS. These findings suggest that drugs targeting the S1P system could be developed as novel analgesics, either as monotherapy or potential adjuncts to established analgesics.


Asunto(s)
Manejo del Dolor , Receptores de Lisoesfingolípidos/efectos de los fármacos , Animales , Humanos , Ratones , Ratones Noqueados , Conformación Proteica , Receptores de Lisoesfingolípidos/química , Receptores de Lisoesfingolípidos/genética
18.
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
19.
Biol Psychiatry ; 71(8): 714-24, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22264443

RESUMEN

BACKGROUND: Cannabinoid CB(1) receptors (CB(1)Rs) mediate the effects of ▵(9)-tetrahydrocannabinol (THC), the psychoactive component in marijuana. Repeated THC administration produces tolerance and dependence, which limit therapeutic development. Moreover, THC produces motor and psychoactive side effects. ß-arrestin2 mediates receptor desensitization, internalization, and signaling, but its role in these CB(1)R effects and receptor regulation is unclear. METHODS: CB(1)R signaling and behaviors (antinociception, hypothermia, catalepsy) were assessed in ß-arrestin2-knockout (ßarr2-KO) and wild-type mice after THC administration. Cannabinoid-stimulated [(35)S]GTPγS and [(3)H]ligand autoradiography were assessed by statistical parametric mapping and region-of-interest analysis. RESULTS: ß-arrestin2 deletion increased CB(1)R-mediated G-protein activity in subregions of the cortex but did not affect CB(1)R binding, in vehicle-treated mice. ßarr2-KO mice exhibited enhanced acute THC-mediated antinociception and hypothermia, with no difference in catalepsy. After repeated THC administration, ßarr2-KO mice showed reduced CB(1)R desensitization and/or downregulation in cerebellum, caudal periaqueductal gray, and spinal cord and attenuated tolerance to THC-mediated antinociception. In contrast, greater desensitization was found in hypothalamus, cortex, globus pallidus, and substantia nigra of ßarr2-KO compared with wild-type mice. Enhanced tolerance to THC-induced catalepsy was observed in ßarr2-KO mice. CONCLUSIONS: ß-arrestin2 regulation of CB(1)R signaling following acute and repeated THC administration was region-specific, and results suggest that multiple, overlapping mechanisms regulate CB(1)Rs. The observations that ßarr2-KO mice display enhanced antinociceptive responses to acute THC and decreased tolerance to the antinociceptive effects of the drug, yet enhanced tolerance to catalepsy, suggest that development of cannabinoid drugs that minimize CB(1)R interactions with ß-arrestin2 might produce improved cannabinoid analgesics with reduced motor suppression.


Asunto(s)
Arrestinas/metabolismo , Dronabinol/metabolismo , Tolerancia a Medicamentos/fisiología , Receptor Cannabinoide CB1/metabolismo , Animales , Autorradiografía , Catalepsia/metabolismo , Sistema Nervioso Central/metabolismo , Regulación hacia Abajo , Dronabinol/farmacología , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipotermia/metabolismo , Ratones , Ratones Noqueados , Nocicepción/efectos de los fármacos , Transducción de Señal , Radioisótopos de Azufre , beta-Arrestinas
20.
Neuropharmacology ; 61(8): 1470-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21907220

RESUMEN

The stable transcription factor ΔFosB is induced in the nucleus accumbens (NAc) by chronic exposure to several drugs of abuse, and transgenic expression of ΔFosB in the striatum enhances the rewarding properties of morphine and cocaine. However, the mechanistic basis for these observations is incompletely understood. We used a bitransgenic mouse model with inducible expression of ΔFosB in dopamine D(1) receptor/dynorphin-containing striatal neurons to determine the effect of ΔFosB expression on opioid and cannabinoid receptor signaling in the NAc. Results showed that mu opioid-mediated G-protein activity and inhibition of adenylyl cyclase were enhanced in the NAc of mice that expressed ΔFosB. Similarly, kappa opioid inhibition of adenylyl cyclase was enhanced in the ΔFosB expressing mice. In contrast, cannabinoid receptor-mediated signaling did not differ between mice overexpressing ΔFosB and control mice. These findings suggest that opioid and cannabinoid receptor signaling are differentially modulated by expression of ΔFosB, and indicate that ΔFosB expression might produce some of its effects via enhanced mu and kappa opioid receptor signaling in the NAc.


Asunto(s)
Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cannabinoide CB1/metabolismo , Transducción de Señal/fisiología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Nucleótidos de Adenina/farmacología , Adenosina Trifosfato/farmacocinética , Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , Animales , Benzoxazinas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Masculino , Ratones , Ratones Transgénicos , Morfolinas/farmacología , Naftalenos/farmacología , Núcleo Accumbens/efectos de los fármacos , Fosfopiruvato Hidratasa/genética , Isótopos de Fósforo/farmacocinética , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/genética , Transducción de Señal/genética , Isótopos de Azufre/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...