Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Neuropsychopharmacol ; 26(7): 513-521, 2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37343217

RESUMEN

BACKGROUND: Evidence has accumulated demonstrating the existence of opioid receptor heteromers, and recent data suggest that targeting these heteromers could reduce opioid side effects while retaining therapeutic effects. Indeed, CYM51010 characterized as a MOR (mu opioid receptor)/DOR (delta opioid receptor) heteromer-preferring agonist promoted antinociception comparable with morphine but with less tolerance. In the perspective of developing these new classes of pharmacological agents, data on their putative side effects are mandatory. METHODS: Therefore, in this study, we investigated the effects of CYM51010 in different models related to drug addiction in mice, including behavioral sensitization, conditioned place preference and withdrawal. RESULTS: We found that, like morphine, CYM51010 promoted acute locomotor activity as well as psychomotor sensitization and rewarding effect. However, it induced less physical dependence than morphine. We also investigated the ability of CYM51010 to modulate some morphine-induced behavior. Whereas CYM51010 was unable to block morphine-induced physical dependence, it blocked reinstatement of an extinguished morphine induced-conditioned place preference. CONCLUSIONS: Altogether, our results reveal that targeting MOR-DOR heteromers could represent a promising strategy to block morphine reward.


Asunto(s)
Morfina , Receptores Opioides delta , Ratones , Animales , Morfina/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu , Analgésicos Opioides/farmacología , Recompensa
2.
Int J Neuropsychopharmacol ; 22(6): 394-401, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-30915438

RESUMEN

BACKGROUND: Chronic morphine treatments produce important morphological changes in multiple brain areas including the nucleus accumbens. METHODS: In this study, we have investigated the effect of chronic morphine treatment at a relatively low dose on the morphology of medium spiny neurons in the core and shell of the nucleus accumbens in rats 1 day after the last injection of a chronic morphine treatment (5 mg/kg once per day for 14 days). Medium spiny neurons were labeled with 1,1' dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate crystal and analyzed by confocal laser-scanning microscope. RESULTS: Our results show an increase of thin spines and a decrease of stubby spines specifically in the shell of morphine-treated rats compared with control. Since morphine-treated rats also presented an elevation of corticosterone level in plasma, we explored whether spine alterations induced by morphine treatment in the nucleus accumbens could be affected by the depletion of the hormone. Thus, bilaterally adrenalectomized rats were treated with morphine in the same conditions. No more alteration in stubby spines in the shell was detected in morphine-treated rats with a depletion of corticosterone, while a significant increase was observed in mushroom spines in the shell and stubby spines in the core. Regarding the thin spines, the increase observed with morphine compared with saline was lower in adrenalectomized rats than in nonadrenalectomized animals. CONCLUSION: These results indicate that dendritic spine remodeling in nucleus accumbens following chronic morphine treatment at relatively low doses is dependent on corticosterone levels.


Asunto(s)
Corticosterona/fisiología , Espinas Dendríticas/fisiología , Morfina/farmacología , Plasticidad Neuronal/fisiología , Núcleo Accumbens/citología , Adrenalectomía , Animales , Peso Corporal/efectos de los fármacos , Corticosterona/sangre , Espinas Dendríticas/efectos de los fármacos , Masculino , Microscopía Confocal , Actividad Motora/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Ratas
3.
Addict Biol ; 23(1): 448-460, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28074533

RESUMEN

Netrin G1 is a presynaptic ligand involved in axonal projection. Although molecular mechanisms underlying cocaine addiction are still poorly understood, Netrin G1 might have a role as a regulator of anxiety, fear and spatial memory, behavioural traits impaired in the context of cocaine exposure. In this study, the Netrin G1 (Ntng1) expression was investigated in the nucleus accumbens of mice primarily conditioned to cocaine using a place preference paradigm. A genetic association study was then conducted on 146 multiplex families of the Collaborative study on Genetics of Alcoholism, in which seven single nucleotide polymorphisms located in the NTNG1 gene were genotyped. NTNG1 expression levels were also quantified in BA10, BA46 and the cerebellum of healthy controls (with no Axis 1 psychopathology). Decreased Ntng1 expression was initially observed in the nucleus accumbens of mice conditioned to cocaine. Significant genetic family-based associations were detected between NTNG1 polymorphisms and cocaine dependence. NTNG1 expression in BA10, BA46 and the cerebellum, however, were not significantly associated with any allele or haplotype of this gene. These results confirm that Ntng1 expression is disturbed in the nucleus accumbens of mice, after cocaine conditioning. A haplotype of NTNG1 was found to constitute a vulnerability factor for cocaine use disorder in patients, although none of its single nucleotide polymorphisms were associated with a differential expression pattern in healthy controls. The data suggest that change in the Ntng1 expression is a consequence of cocaine exposure, and that some of its genetic markers are associated with a greater risk for cocaine use disorder.


Asunto(s)
Trastornos Relacionados con Cocaína/genética , Cocaína , Condicionamiento Psicológico , Inhibidores de Captación de Dopamina , Netrinas/genética , Núcleo Accumbens/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Cerebelo/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Ratones , Netrinas/metabolismo , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-26390873

RESUMEN

BACKGROUND: Opiates such as morphine are the most powerful analgesics, but their protracted use is restrained by the development of tolerance to analgesic effects. Recent works suggest that tolerance to morphine might be due to its inability to promote mu opioid receptor endocytosis, and the co-injection of morphine with a mu opioid receptor internalizing agonist like [D-Ala(2),N-Me-Phe(4),Gly-ol(5)]enkephalin reduces tolerance to morphine. So far, no studies have been conducted to evaluate the ability of methadone to reduce morphine tolerance in morphine-pretreated animals, a treatment sequence that could be encountered in opiate rotation protocol. We investigated the ability of methadone (a mu opioid receptor internalizing agonist used in therapy) to reverse morphine tolerance and the associated cellular mechanisms in the periaqueductal gray matter, a region involved in pain control. METHODS: We measured analgesic response following a challenge dose of morphine in the hot plate test and investigated regulation of mu opioid receptor (coupling and endocytosis) and some cellular mechanisms involved in tolerance such as adenylate cyclase superactivation and changes in N-methyl-d-aspartate receptor subunits expression and phosphorylation state. RESULTS: A chronic treatment with morphine promoted tolerance to its analgesic effects and was associated with a lack of mu opioid receptor endocytosis, adenylate cyclase overshoot, NR2A and NR2B downregulation, and phosphorylation of NR1. We reported that a methadone treatment in morphine-treated mice reversed morphine tolerance to analgesia by promoting mu opioid receptor endocytosis and blocking cellular mechanisms of tolerance. CONCLUSIONS: Our data might lead to rational strategies to tackle opiate tolerance in the frame of opiate rotation.


Asunto(s)
Tolerancia a Medicamentos , Metadona/farmacología , Morfina/antagonistas & inhibidores , Morfina/farmacología , Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Endocitosis , Masculino , Ratones , Dimensión del Dolor/efectos de los fármacos , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores Opioides mu/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 307(5): H649-57, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25015969

RESUMEN

High-protein-low-carbohydrate (HP-LC) diets have become widespread. Yet their deleterious consequences, especially on glucose metabolism and arteries, have already been underlined. Our previous study (2) has already shown glucose intolerance with major arterial dysfunction in very old mice subjected to an HP-LC diet. The hypothesis of this work was that this diet had an age-dependent deleterious metabolic and cardiovascular outcome. Two groups of mice, young and adult (3 and 6 mo old), were subjected for 12 wk to a standard or to an HP-LC diet. Glucose and lipid metabolism was studied. The cardiovascular system was explored from the functional stage with Doppler-echography to the molecular stage (arterial reactivity, mRNA, immunohistochemistry). Young mice did not exhibit any significant metabolic modification, whereas adult mice presented marked glucose intolerance associated with an increase in resistin and triglyceride levels. These metabolic disturbances were responsible for cardiovascular damages only in adult mice, with decreased aortic distensibility and left ventricle dysfunction. These seemed to be the consequence of arterial dysfunctions. Mesenteric arteries were the worst affected with a major oxidative stress, whereas aorta function seemed to be maintained with an appreciable role of cyclooxygenase-2 to preserve endothelial function. This study highlights for the first time the age-dependent deleterious effects of an HP-LC diet on metabolism, with glucose intolerance and lipid disorders and vascular (especially microvessels) and cardiac functions. This work shows that HP-LC lead to equivalent cardiovascular alterations, as observed in very old age, and underlines the danger of such diet.


Asunto(s)
Aorta/metabolismo , Dieta Baja en Carbohidratos/efectos adversos , Proteínas en la Dieta/administración & dosificación , Intolerancia a la Glucosa/etiología , Miocardio/metabolismo , Disfunción Ventricular Izquierda/etiología , Factores de Edad , Animales , Aorta/patología , Glucemia/metabolismo , Proteínas en la Dieta/efectos adversos , Ecocardiografía , Intolerancia a la Glucosa/metabolismo , Metabolismo de los Lípidos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Resistina/sangre , Triglicéridos/sangre , Disfunción Ventricular Izquierda/metabolismo
6.
Int J Neuropsychopharmacol ; 17(9): 1367-73, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24606726

RESUMEN

Buprenorphine is used as a sublingual medication in the treatment of opioid dependence. However, its misuse by i.v. injection may limit its acceptability and dissemination. A buprenorphine/naloxone (ratio 4:1) combination has been developed to reduce diversion and abuse. So far, the relevance of this combination has not been investigated in the animal models traditionally used to study the reinforcing effects of drugs of abuse. The aim of this study was to compare the rewarding effects, assessed by conditioned place preference (CPP), of buprenorphine and buprenorphine/naloxone combination following i.v. administration in mice. Animals were treated with different doses of buprenorphine or buprenorphine/naloxone combination (ratio 4:1), and CPP conditioning trial duration was 5 or 30 min. At the longest trial duration, a bell-shaped dose-response curve was obtained with buprenorphine, which was shifted significantly to the right with naloxone combination. At the shortest trial duration, an aversive effect was observed with the buprenorphine/naloxone combination in animals, involving opioid receptor-like 1 (ORL1). These findings may explain the discrepancies reported in the literature as some authors have shown a reduced buprenorphine/naloxone misuse compared to buprenorphine in opioid abusers, while others have not.


Asunto(s)
Analgésicos Opioides/farmacología , Reacción de Prevención/efectos de los fármacos , Buprenorfina/farmacología , Condicionamiento Operante/efectos de los fármacos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Recompensa , Análisis de Varianza , Animales , Combinación Buprenorfina y Naloxona , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Masculino , Ratones
7.
Int J Neuropsychopharmacol ; 18(5)2014 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-25522421

RESUMEN

BACKGROUND: Brain-derived neurotrophic factor (BDNF) is a neurotrophin that has long been studied in the field of addiction and its importance in regulating drug addiction-related behavior has been widely demonstrated. The aim of our study was to analyze the consequences of a repeated exposure to drugs of abuse or natural reward on plasma BDNF levels during withdrawal. METHODS: Rats were chronically injected with morphine (subcutaneously, 5mg/kg) or cocaine (intraperitoneally, 20mg/kg) or fed with a butter biscuit (per os, 4g) once per day for 14 days. Blood collection was performed on the 1st (withdrawal day 1 or WD1) or on (WD14), either at the same time point rats had been exposed to drugs or natural reward or at a different time point (used to quantify basal brain-derived neurotrophic factor levels). RESULTS: Cocaine treatment led to a rapid (WD1) and persistent (WD14) decrease of basal BDNF levels compared with saline-treated animals, whereas morphine induced an increase on WD14 without any alteration on WD1. On the contrary, the natural reward induced a significant increase of basal brain-derived neurotrophic factor levels only on WD1. The analysis of BDNF levels at the usual time point at which animals had been exposed showed that both drugs, but not the natural reward, increased BDNF levels compared with basal levels. CONCLUSION: Our data highlight that only drugs of abuse are able to persistently alter BDNF levels and to induce specific variations of this neurotrophic factor at the usual hour of injection.


Asunto(s)
Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/sangre , Cocaína/farmacología , Morfina/farmacología , Recompensa , Analgésicos Opioides/farmacología , Animales , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/farmacología , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Masculino , Morfina/administración & dosificación , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias
8.
J Neurosci ; 32(21): 7301-10, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22623675

RESUMEN

G-protein-coupled receptors (GPCRs) mediate numerous physiological functions and represent prime therapeutic targets. Receptor trafficking upon agonist stimulation is critical for GPCR function, but examining this process in vivo remains a true challenge. Using knock-in mice expressing functional fluorescent delta opioid receptors under the control of the endogenous promoter, we visualized in vivo internalization of this native GPCR upon physiological stimulation. We developed a paradigm in which animals were made dependent on morphine in a drug-paired context. When re-exposed to this context in a drug-free state, mice showed context-dependent withdrawal signs and activation of the hippocampus. Receptor internalization was transiently detected in a subset of CA1 neurons, uncovering regionally restricted opioid peptide release. Importantly, a pool of surface receptors always remained, which contrasts with the in vivo profile previously established for exogenous drug-induced internalization. Therefore, a distinct response is observed at the receptor level upon a physiological or pharmacological stimulation. Altogether, direct in vivo GPCR visualization enables mapping receptor stimulation promoted by a behavioral challenge and represents a powerful approach to study endogenous GPCR physiology.


Asunto(s)
Hipocampo/metabolismo , Transporte de Proteínas , Receptores Opioides delta/metabolismo , Animales , Encefalina Metionina/metabolismo , Femenino , Técnicas de Sustitución del Gen , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Imagen Molecular , Morfina/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Síndrome de Abstinencia a Sustancias/metabolismo
9.
Front Psychiatry ; 14: 1229439, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38152360

RESUMEN

The over-prescription of opioid analgesics is a growing problem in the field of addiction, which has reached epidemic-like proportions in North America. Over the past decade, oxycodone has gained attention as the leading opioid responsible for the North America opioid crisis. Oxycodone is the most incriminated drug in the early years of the epidemic of opioid use disorder in USA (roughly 1999-2016). The number of preclinical articles on oxycodone is rapidly increasing. Several publications have already compared oxycodone with other opioids, focusing mainly on their analgesic properties. The aim of this review is to focus on the genomic and epigenetic regulatory features of oxycodone compared with other opioid agonists. Our aim is to initiate a discussion of perceptible differences in the pharmacological response observed with these various opioids, particularly after repeated administration in preclinical models commonly used to study drug dependence potential.

10.
Artículo en Inglés | MEDLINE | ID: mdl-37479108

RESUMEN

Repeated exposure to substances of abuse results in an increase in some behavioral responses. This phenomenon, called behavioral sensitization (BS), is well described in preclinical models. However, its existence in humans is still a matter of debate. After a review of preclinical evidence of BS and its mechanisms in animal models, we reviewed the evidence supporting the existence of BS in humans, despite the limited research available in this regard. We focused our review on opioids and psychostimulants, since they share the ability to promote addictive behaviors. Further, they induce BS despite their distinct sedative and stimulant properties. Moreover, we proposed future research perspectives in this review to address the remaining unsolved questions, especially regarding BS in humans using a harm reduction approach.


Asunto(s)
Conducta Adictiva , Estimulantes del Sistema Nervioso Central , Animales , Humanos , Analgésicos Opioides , Roedores , Estimulantes del Sistema Nervioso Central/farmacología , Aprendizaje , Conducta Animal
11.
Addict Biol ; 17(3): 528-38, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21309946

RESUMEN

A great number of studies have shown the presence of physiological interactions between brain neurotransmitter systems in behavioural responses. This is the case for opioid, cholecystokinin (CCK) and dopamine systems. However, so far the role that the CCK system may play in vulnerability to consumption of drugs of abuse is not clear. This was investigated in this study using Lewis rats that are more sensitive to the reinforcing properties of drugs of abuse than Fischer rats. The extraneuronal CCK(8) levels and brain CCK(2) receptors were found higher in Fischer than in Lewis rats in the nucleus accumbens, one of the most important structures involved in drug consumption. Moreover, pharmacological modulation of the CCK system by administration of a selective CCK(2) agonist blocked, in the conditioned place preference, the reinforcing effects of morphine in Lewis rats, whereas a selective CCK(2) antagonist revealed reinforcing effects of the alkaloid in Fischer rats. These results obtained following systemic administrations of the CCK ligands were confirmed following microinjection into the nucleus accumbens. Thus, a low level of CCK efflux in the nucleus accumbens could be one of the many factors involved in drug reinforcing effects, whereas a high level of CCK efflux could attenuate it.


Asunto(s)
Morfina/farmacología , Narcóticos/farmacología , Receptores de Colecistoquinina/efectos de los fármacos , Refuerzo en Psicología , Análisis de Varianza , Animales , Colecistoquinina/análogos & derivados , Colecistoquinina/metabolismo , Colecistoquinina/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Encefalinas/metabolismo , Antagonistas de Hormonas/farmacología , Indoles/farmacología , Masculino , Meglumina/análogos & derivados , Meglumina/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Receptores de Colecistoquinina/agonistas , Receptores de Colecistoquinina/antagonistas & inhibidores
12.
Front Pharmacol ; 13: 856672, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35571111

RESUMEN

Substance use disorders (SUD) may emerge from an individual's attempt to limit negative affective states and symptoms linked to stress. Indeed, SUD is highly comorbid with chronic stress, traumatic stress, or post-traumatic stress disorder (PTSD), and treatments approved for each pathology individually often failed to have a therapeutic efficiency in such comorbid patients. The kappa-opioid receptor (KOR) and its endogenous ligand dynorphin (DYN), seem to play a key role in the occurrence of this comorbidity. The DYN/KOR function is increased either in traumatic stress or during drug use, dependence acquisition and DYN is released during stress. The behavioural effects of stress related to the DYN/KOR system include anxiety, dissociative and depressive symptoms, as well as increased conditioned fear response. Furthermore, the DYN/KOR system is implicated in negative reinforcement after the euphoric effects of a drug of abuse ends. During chronic drug consumption DYN/KOR functions increase and facilitate tolerance and dependence. The drug-seeking behaviour induced by KOR activation can be retrieved either during the development of an addictive behaviour, or during relapse after withdrawal. DYN is known to be one of the most powerful negative modulators of dopamine signalling, notably in brain structures implicated in both reward and fear circuitries. KOR are also acting as inhibitory heteroreceptors on serotonin neurons. Moreover, the DYN/KOR system cross-regulate with corticotropin-releasing factor in the brain. The sexual dimorphism of the DYN/KOR system could be the cause of the gender differences observed in patients with SUD or/and traumatic stress-related pathologies. This review underlies experimental and clinical results emphasizing the DYN/KOR system as common mechanisms shared by SUD or/and traumatic stress-related pathologies, and suggests KOR antagonist as a new pharmacological strategy to treat this comorbidity.

13.
Front Pain Res (Lausanne) ; 2: 722820, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35295469

RESUMEN

Opioid are the most powerful analgesics ever but their use is still limited by deleterious side effects such as tolerance, dependence, and respiratory depression that could eventually lead to a fatal overdose. The opioid crisis, mainly occurring in north America, stimulates research on finding new opioid ligands with reduced side effects. Among them, biased ligands are likely the most promising compounds. We will review some of the latest discovered biased opioid ligands and see if they were able to fulfill these expectations.

14.
ACS Med Chem Lett ; 12(11): 1739-1744, 2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34795862

RESUMEN

Kappa opioid receptor (KOR) PET imaging remains attractive to understand the role of KOR in health and diseases and to help the development of drugs especially for psychiatric disorders such as depression, anxiety, and addiction. The potent and selective KOR antagonist RTI-97 labeled with carbon-11 was previously demonstrated to display specific KOR binding in mouse brain by ex vivo autoradiography studies. Herein, we evaluated [11C]RTI-97 in rat by in vitro autoradiography and by in vivo PET imaging. The radiosynthesis of [11C]RTI-97 was optimized to obtain high molar activities. Despite a low cerebral uptake, the overall results showed a heterogeneous repartition and specific KOR binding of [11C]RTI-97 in brain and a high and specific accumulation of [11C]RTI-97 in pituitary in accordance with KOR expression.

15.
J Endocrinol ; 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34582357

RESUMEN

The Growth Hormone Secretagogue Receptor (GHSR) mediates key properties of the gut hormone ghrelin on metabolism and behavior. Nevertheless, most recent observations also support that the GHSR is a constitutively active G protein-coupled receptor endowed of a sophisticated tuning involving a balance of endogenous ligands. Demonstrating the feasibility of shifting GHSR canonical signaling in vivo, we previously reported that a model with enhanced sensitivity to ghrelin (GhsrQ343X mutant rats) developed fat accumulation and glucose intolerance. Herein, we investigated the contribution of energy homeostasis to the onset of this phenotype, as well as behavioral responses to feeding or pharmacological challenges, by comparing GhsrM/M rats to wild-type littermate rats 1) as freely behaving animals and 2) in feeding and locomotor paradigms. Herein, GhsrM/M rats showed enhanced locomotor response to a GHSR agonist while locomotor or anorexigenic responses to amphetamine or cabergoline (dopamine receptor 2 agonist), respectively, were preserved. Ad libitum fed GhsrM/M rats consumed and conditioned for sucrose similarly to littermate control rats. In calorie-restricted conditions, GhsrM/M rats retained food anticipatory activity and maintained better their body weight and glycemia. Importantly, prior to fat accumulation, male GhsrM/M rats preferentially used carbohydrates as fuel substrate without alterations of energy intake, energy expenditure or physical activity and showed alterations of the GHSR system (i.e. enhanced ratio of GHSR hormones LEAP2:acyl-ghrelin and increased Ghsr expression in the hypothalamus). Overall, the present study provides proof of concept that shifted GHSR signaling can specifically alter nutrient partitioning resulting in modified balance of carbohydrate/lipid utilization.

16.
Nat Neurosci ; 9(9): 1134-41, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16906152

RESUMEN

Depression is a devastating illness with a lifetime prevalence of up to 20%. The neurotransmitter serotonin or 5-hydroxytryptamine (5-HT) is involved in the pathophysiology of depression and in the effects of antidepressant treatments. However, molecular alterations that underlie the pathology or treatment of depression are still poorly understood. The TREK-1 protein is a background K+ channel regulated by various neurotransmitters including 5-HT. In mice, the deletion of its gene (Kcnk2, also called TREK-1) led to animals with an increased efficacy of 5-HT neurotransmission and a resistance to depression in five different models and a substantially reduced elevation of corticosterone levels under stress. TREK-1-deficient (Kcnk2-/-) mice showed behavior similar to that of naive animals treated with classical antidepressants such as fluoxetine. Our results indicate that alterations in the functioning, regulation or both of the TREK-1 channel may alter mood, and that this particular K+ channel may be a potential target for new antidepressants.


Asunto(s)
Trastorno Depresivo/fisiopatología , Eliminación de Gen , Canales de Potasio de Dominio Poro en Tándem/fisiología , Análisis de Varianza , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Trastorno Depresivo/genética , Trastorno Depresivo/prevención & control , Resistencia a Medicamentos/genética , Fluoxetina/farmacología , Genotipo , Ratones , Ratones Noqueados , Fenotipo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Serotonina/metabolismo , Transmisión Sináptica/efectos de los fármacos
17.
Front Pharmacol ; 11: 623, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477119

RESUMEN

Comorbidity between drug abuse and post-traumatic stress disorder (PTSD), a stress-related dysregulation of fear responses, is very high. While some drugs are known to increase fear and anxiety, there are only few data regarding interactions between voluntary drug consumption and fear memory. The spontaneous chronic consumption of either alcohol or cocaine under a 3-week free-choice progressive paradigm of alcohol (3/6/10%) or cocaine (0.2/0.4/0.6 mg/ml), was assessed in VGV transgenic mice, having full 5-HT2C receptor editing and displaying PTSD-like behaviors. The consequences of these drug consumptions on the potentiated contextual and cued fear conditioning responses of VGV mice were assessed. The effects of drugs on hippocampal brain-derived neurotrophic factor (Bdnf) mRNA were measured as its expression was previously found to be decreased in VGV mice. Chronic alcohol consumption was similar in WT and VGV mice. In the alcohol condition, fear acquisition was not different at the end of the learning session and cue-fear extinction was facilitated. Regarding cocaine, in contrast to WT mice, VGV mice did not increase their drug consumption along with increasing doses, an effect that might be related with enhanced drug stimuli discrimination via increased 5-HT2C receptors. Cocaine-intake VGV mice did not display the contextual fear generalization usually observed in control VGV mice. In addition, Bdnf expression was upregulated after either chronic alcohol or cocaine intake. Altogether, these results suggest that both chronic alcohol and cocaine voluntary oral consumptions can exert some therapeutic-like effects in a mutant model of PTSD predisposition.

18.
Br J Pharmacol ; 177(20): 4782-4795, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32851651

RESUMEN

BACKGROUND AND PURPOSE: Opioids are effective painkillers. However, their risk-benefit ratio is dampened by numerous adverse effects and opioid misuse has led to a public health crisis. Safer alternatives are required, but isolating the antinociceptive effect of opioids from their adverse effects is a pharmacological challenge because activation of the µ opioid receptor triggers both the antinociceptive and adverse effects of opioids. EXPERIMENTAL APPROACH: The TREK1 potassium channel is activated downstream of µ receptor and involved in the antinociceptive activity of morphine but not in its adverse effects. Bypassing the µ opioid receptor to directly activate TREK1 could therefore be a safer analgesic strategy. KEY RESULTS: We developed a selective TREK1 activator, RNE28, with antinociceptive activity in naive rodents and in models of inflammatory and neuropathic pain. This activity was lost in TREK1 knockout mice or wild-type mice treated with the TREK1 blocker spadin, showing that TREK1 is required for the antinociceptive activity of RNE28. RNE28 did not induce respiratory depression, constipation, rewarding effects, or sedation at the analgesic doses tested. CONCLUSION AND IMPLICATIONS: This proof-of-concept study shows that TREK1 activators could constitute a novel class of painkillers, inspired by the mechanism of action of opioids but devoid of their adverse effects.


Asunto(s)
Analgésicos Opioides , Neuralgia , Analgésicos , Analgésicos Opioides/efectos adversos , Animales , Ratones , Ratones Noqueados , Morfina , Receptores Opioides mu
19.
Toxicol Appl Pharmacol ; 241(3): 339-47, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19781562

RESUMEN

The use of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) has increased in recent years; it can lead to life-threatening hyperthermia and serotonin syndrome. Human and rodent males appear to be more sensitive to acute toxicity than are females. MDMA is metabolized to five main metabolites by the enzymes CYP1A2, CYP2D and COMT. Little is presently known about sex-dependent differences in the pharmacokinetics of MDMA and its metabolites. We therefore analyzed MDMA disposition in male and female rats by measuring the plasma and urine concentrations of MDMA and its metabolites using a validated LC-MS method. MDA AUC(last) and C(max) were 1.6- to 1.7-fold higher in males than in females given MDMA (5 mg/kg sc), while HMMA C(max) and AUC(last) were 3.2- and 3.5-fold higher, respectively. MDMA renal clearance was 1.26-fold higher in males, and that of MDA was 2.2-fold higher. MDMA AUC(last) and t(1/2) were 50% higher in females given MDMA (1 mg/kg iv). MDA C(max) and AUC(last) were 75-82% higher in males, with a 2.8-fold higher metabolic index. Finally, the AUC(last) of MDA was 0.73-fold lower in males given 1 mg/kg iv MDA. The volumes of distribution of MDMA and MDA at steady-state were similar in the two sexes. These data strongly suggest that differences in the N-demethylation of MDMA to MDA are major influences on the MDMA and MDA pharmacokinetics in male and female rats. Hence, males are exposed to significantly more toxic MDA, which could explain previously reported sexual dysmorphism in the acute effects and toxicity of MDMA in rats.


Asunto(s)
Alucinógenos/farmacocinética , N-Metil-3,4-metilenodioxianfetamina/farmacocinética , 3,4-Metilenodioxianfetamina/farmacocinética , Animales , Área Bajo la Curva , Biotransformación , Calibración , Cromatografía Liquida , Femenino , Inyecciones Intravenosas , Masculino , Espectrometría de Masas , Control de Calidad , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales
20.
Addict Biol ; 14(3): 245-52, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19523041

RESUMEN

The delta opioid system is involved in the behavioral effects of various drugs of abuse. However, only a few studies have focused on the possible interactions between the opioid system and the effects of 3,4-methylenedioxymethamphetamine (MDMA). In order to examine the possible role of the delta opioid system in MDMA-induced behaviors in mice, locomotor activity and conditioned place preference (CPP) were investigated in the presence of naltrindole (NTI), a selective delta opioid antagonist. Moreover, the consequences of acute and chronic MDMA administration on pro-enkephalin (Penk) and pro-opiomelanocortin (Pomc) gene expression were assessed by real-time quantitative polymerase chain reaction (QPCR). The results showed that, after acute MDMA administration (9 mg/kg; i.p.), NTI (5 mg/kg, s.c.) was able to totally block MDMA-induced hyperlocomotion. Penk gene expression was not modulated by acute MDMA, but a decrease of Pomc gene expression was observed, which was not antagonized by NTI. Administration of the antagonist prevented the acquisition of MDMA-induced CPP, suggesting an implication of the delta opioid receptors in this behavior. Following chronic MDMA treatment, only the level of Pomc was modulated. The observed increase was totally blocked by NTI pre-treatment. All these results confirm the interactions between the delta opioid system (receptors and peptides) and the effects of MDMA.


Asunto(s)
Trastornos Relacionados con Anfetaminas/genética , Conducta Animal/efectos de los fármacos , Encefalinas/genética , Alucinógenos/toxicidad , N-Metil-3,4-metilenodioxianfetamina/toxicidad , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Proopiomelanocortina/genética , Precursores de Proteínas/genética , Receptores Opioides delta/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Ratones , Actividad Motora/efectos de los fármacos , Naltrexona/farmacología , ARN Mensajero/genética , Medio Social
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA