Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
CNS Neurosci Ther ; 27(7): 831-841, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34018697

RESUMEN

AIMS: Naltrexone is a mu opioid receptor (MOR) antagonist used to treat drug dependence in patients. Previous reports indicated that MOR antagonists reduced neurodegeneration and inflammation after brain injury. The purpose of this study was to evaluate the neuroprotective effect of naltrexone in cell culture and a mouse model of traumatic brain injury (TBI). METHODS: The neuroprotective effect of naltrexone was examined in primary cortical neurons co-cultured with BV2 microglia. Controlled cortical impact (CCI) was delivered to the left cerebral cortex of adult male MOR wild-type (WT) and knockout (KO) mice. Naltrexone was given daily for 4 days, starting from day 2 after lesioning. Locomotor activity was evaluated on day 5 after the CCI. Brain tissues were collected for immunostaining, Western, and qPCR analysis. RESULTS: Glutamate reduced MAP2 immunoreactivity (-ir), while increased IBA1-ir in neuron/BV2 co-culture; both responses were antagonized by naltrexone. TBI significantly reduced locomotor activity and increased the expression of IBA1, iNOS, and CD4 in the lesioned cortex. Naltrexone significantly and equally antagonized the motor deficits and expression of IBA1 and iNOS in WT and KO mice. TBI-mediated CD4 protein production was attenuated by naltrexone in WT mice, but not in KO mice. CONCLUSION: Naltrexone reduced TBI-mediated neurodegeneration and inflammation in MOR WT and KO mice. The protective effect of naltrexone involves non-MOR and MOR mechanisms.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Receptores Opioides mu/deficiencia , Animales , Técnicas de Cocultivo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Fármacos Neuroprotectores/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/genética
2.
Cell Mol Neurobiol ; 41(5): 1031-1038, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32989585

RESUMEN

Opioids, like morphine and naloxone, regulate the proliferation and neuronal differentiation of neural stem cells (NSCs) in a receptor-independent and ten-eleven translocation methylcytosine dioxygenase (TET1)-dependent manner in vitro. Whether naloxone regulates hippocampal NSCs and contextual learning in vivo in a similar manner was determined. Naloxone infusion increased the Ki67 and Doublecortin positive cells in subgranular zone of wild type mice, which suggested the increased proliferation and differentiation of hippocampal NSCs in vivo and was consistent with the in vitro functions of naloxone. In addition, naloxone infusion also facilitated the contextual learning and memory of wild type mice. To determine the contribution of µ-opioid receptor (OPRM1) and TET1 to these functions of naloxone, several types of knockout mice were used. Since Tet1-/- mice have high deficiency in contextual learning and memory, Tet1+/- mice were used instead. The abilities of naloxone to regulate NSCs and to facilitate contextual learning were significantly impaired in Tet1+/- mice. In addition, these abilities of naloxone were not affected in Oprm1-/- mice. Therefore, naloxone facilitates contextual learning and memory in a receptor-independent and Tet1-dependent manner, which provides new understanding on the receptor-independent functions of opioids.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Proteínas Proto-Oncogénicas/deficiencia , Receptores Opioides mu/deficiencia , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Noqueados , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Proteínas Proto-Oncogénicas/genética , Receptores Opioides mu/genética
3.
Drug Alcohol Depend ; 216: 108310, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33017752

RESUMEN

BACKGROUND: Made as a tea, the Thai traditional drug "kratom" reportedly possesses pharmacological actions that include both a coca-like stimulant effect and opium-like depressant effect. Kratom has been used as a substitute for opium in physically-dependent subjects. The objective of this study was to evaluate the antinociception, somatic and physical dependence produced by kratom tea, and then assess if the tea ameliorated withdrawal in opioid physically-dependent subjects. METHODS: Lyophilized kratom tea (LKT) was evaluated in C57BL/6J and opioid receptor knockout mice after oral administration. Antinociceptive activity was measured in the 55 °C warm-water tail-withdrawal assay. Potential locomotor impairment, respiratory depression and locomotor hyperlocomotion, and place preference induced by oral LKT were assessed in the rotarod, Comprehensive Lab Animal Monitoring System, and conditioned place preference assays, respectively. Naloxone-precipitated withdrawal was used to determine potential physical dependence in mice repeatedly treated with saline or escalating doses of morphine or LKT, and LKT amelioration of morphine withdrawal. Data were analyzed using one- and two-way ANOVA. RESULTS: Oral administration of LKT resulted in dose-dependent antinociception (≥1 g/kg, p.o.) absent in mice lacking the mu-opioid receptor (MOR) and reduced in mice lacking the kappa-opioid receptor. These doses of LKT did not alter coordinated locomotion or induce conditioned place preference, and only briefly reduced respiration. Repeated administration of LKT did not produce physical dependence, but significantly decreased naloxone-precipitated withdrawal in morphine dependent mice. CONCLUSIONS: The present study confirms the MOR agonist activity and therapeutic effect of LKT for the treatment of pain and opioid physical dependence.


Asunto(s)
Mitragyna , Dependencia de Morfina/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Receptores Opioides mu/agonistas , , Analgésicos Opioides/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Liofilización/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/administración & dosificación , Dependencia de Morfina/fisiopatología , Dependencia de Morfina/psicología , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Dimensión del Dolor/métodos , Extractos Vegetales/aislamiento & purificación , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/deficiencia , Receptores Opioides mu/deficiencia
4.
Neuropharmacology ; 180: 108307, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941853

RESUMEN

Opioid signaling controls the activity of the brain's reward system. It is involved in signaling the hedonic effects of rewards and has essential roles in reinforcement and motivational processes. Here, we focused on opioid signaling through mu and delta receptors on dopaminoceptive neurons and evaluated the role these receptors play in reward-driven behaviors. We generated a genetically modified mouse with selective double knockdown of mu and delta opioid receptors in neurons expressing dopamine receptor D1. Selective expression of the transgene was confirmed using immunostaining. Knockdown was validated by measuring the effects of selective opioid receptor agonists on neuronal membrane currents using whole-cell patch clamp recordings. We found that in the nucleus accumbens of control mice, the majority of dopamine receptor D1-expressing neurons were sensitive to a mu or delta opioid agonist. In mutant mice, the response to the delta receptor agonist was blocked, while the effects of the mu agonist were strongly attenuated. Behaviorally, the mice had no obvious impairments. The mutation did not affect the sensitivity to the rewarding effects of morphine injections or social contact and had no effect on preference for sweet taste. Knockdown had a moderate effect on motor activity in some of the tests performed, but this effect did not reach statistical significance. Thus, we found that knocking down mu and delta receptors on dopamine receptor D1-expressing cells does not appreciably affect some of the reward-driven behaviors previously attributed to opioid signaling.


Asunto(s)
Neuronas/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores Opioides delta/deficiencia , Receptores Opioides mu/deficiencia , Recompensa , Analgésicos Opioides/farmacología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfina/farmacología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Receptores de Dopamina D1/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
5.
J Neurosci Res ; 98(8): 1532-1548, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32424971

RESUMEN

Restless legs syndrome (RLS) is characterized by an irresistible need to move the legs while sitting or lying at night with insomnia as a frequent consequence. Human RLS has been associated with abnormalities in the endogenous opioid system, the dopaminergic system, the iron regulatory system, anemia, and inflammatory and auto-immune disorders. Our previous work indicates that mice lacking all three subtypes of opioid receptors have a phenotype similar to that of human RLS. To study the roles of each opioid receptor subtype in RLS, we first used mu opioid receptor knockout (MOR KO) mice based on our earlier studies using postmortem brain and cell culture. The KO mice showed decreased hemoglobin, hematocrit, and red blood cells (RBCs), with an appearance of microcytic RBCs indicating anemia. Together with decreased serum iron and transferrin, but increased ferritin levels, the anemia is similar to that seen with chronic inflammation in humans. A decreased serum iron level was also observed in the wildtype mice treated with an MOR antagonist. Iron was increased in the liver and spleen of the KO mice. Normal circadian variations in the dopaminergic and serotoninergic systems were absent in the KO mice. The KO mice showed hyperactivity and increased thermal sensitivity in wakefulness primarily during what would normally be the sleep phase similar to that seen in human RLS. Deficits in endogenous opioid system transmission could predispose to anemia of inflammation and loss of circadian variations in dopaminergic or serotonergic systems, thereby contributing to an RLS-like phenotype.


Asunto(s)
Receptores Opioides mu/deficiencia , Síndrome de las Piernas Inquietas/sangre , Síndrome de las Piernas Inquietas/genética , Anemia , Animales , Monoaminas Biogénicas/sangre , Ritmo Circadiano , Cuerpo Estriado , Dopamina/metabolismo , Eritrocitos , Hierro/sangre , Ratones , Ratones Noqueados , Actividad Motora , Dolor , Agitación Psicomotora
6.
Neuropsychopharmacology ; 45(2): 247-255, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31005059

RESUMEN

The medial habenula (MHb) is considered a brain center regulating aversive states. The mu opioid receptor (MOR) has been traditionally studied at the level of nociceptive and mesolimbic circuits, for key roles in pain relief and reward processing. MOR is also densely expressed in MHb, however, MOR function at this brain site is virtually unknown. Here we tested the hypothesis that MOR in the MHb (MHb-MOR) also regulates aversion processing. We used chnrb4-Cre driver mice to delete the Oprm1 gene in chnrb4-neurons, predominantly expressed in the MHb. Conditional mutant (B4MOR) mice showed habenula-specific reduction of MOR expression, restricted to chnrb4-neurons (50% MHb-MORs). We tested B4MOR mice in behavioral assays to evaluate effects of MOR activation by morphine, and MOR blockade by naloxone. Locomotor, analgesic, rewarding, and motivational effects of morphine were preserved in conditional mutants. In contrast, conditioned place aversion (CPA) elicited by naloxone was reduced in both naïve (high dose) and morphine-dependent (low dose) B4MOR mice. Further, physical signs of withdrawal precipitated by either MOR (naloxone) or nicotinic receptor (mecamylamine) blockade were attenuated. These data suggest that MORs expressed in MHb B4-neurons contribute to aversive effects of naloxone, including negative effect and aversive effects of opioid withdrawal. MORs are inhibitory receptors, therefore we propose that endogenous MOR signaling normally inhibits chnrb4-neurons of the MHb and moderates their known aversive activity, which is unmasked upon receptor blockade. Thus, in addition to facilitating reward at several brain sites, tonic MOR activity may also limit aversion within the MHb circuitry.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Habénula/efectos de los fármacos , Habénula/metabolismo , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/deficiencia , Animales , Reacción de Prevención/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Opioides mu/genética
7.
Mol Pain ; 14: 1744806917754142, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29353538

RESUMEN

Background Lithium is widely used to treat bipolar disorders and displays mood stabilizing properties. In addition, lithium relieves painful cluster headaches and has a strong analgesic effect in neuropathic pain rat models. Objectives To investigate the analgesic effect of lithium on the cuff model of neuropathic pain. Methods We used behavioral and pharmacological approaches to study the analgesic effect of a single injection of lithium in wild-type and mu opioid receptor (MOR) null cuffed neuropathic mice. Mass spectrometry and enzyme-linked immunosorbent assay allowed to measure the levels of endogenous MOR agonist beta-endorphin as well as monoamines in brain and plasma samples 4 h after lithium administration. Results A single injection of lithium chloride (100 mg/kg, ip) alleviated mechanical allodynia for 24 h, and this effect was absent in MOR null neuropathic mice. Biochemical analyses highlight a significant increase in beta-endorphin levels by 30% in the brain of lithium-treated mice compared to controls. No variation of beta-endorphin was detected in the blood. Conclusions Together, our results provide evidence that lithium induces a long-lasting analgesia in neuropathic mice presumably through elevated brain levels of beta-endorphin and the activation of MORs.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Litio/uso terapéutico , Receptores Opioides mu/metabolismo , Analgesia , Animales , Monoaminas Biogénicas/sangre , Catecolaminas/sangre , Modelos Animales de Enfermedad , Hiperalgesia/sangre , Límite de Detección , Litio/farmacología , Masculino , Ratones Endogámicos C57BL , Neuralgia/sangre , Neuralgia/tratamiento farmacológico , Neuralgia/patología , Nocicepción/efectos de los fármacos , Receptores Opioides mu/deficiencia
9.
EMBO Mol Med ; 9(11): 1521-1536, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28818835

RESUMEN

Opiate withdrawal/negative reinforcement has been implicated as one of the mechanisms for the progression from impulsive to compulsive drug use. Increase in the intracellular cAMP level and protein kinase A (PKA) activities within the neurocircuitry of addiction has been a leading hypothesis for opiate addiction. This increase requires the phosphorylation of µ-opioid receptor (MOR) at Tyr336 by Src after prolonged opiate treatment in vitro Here, we report that the Src-mediated MOR phosphorylation at Tyr336 is a prerequisite for opiate withdrawal in mice. We observed the recruitment of Src in the vicinity of MOR and an increase in phosphorylated Tyr336 (pY336) levels during naloxone-precipitated withdrawal. The intracerebroventricular or stereotaxic injection of a Src inhibitor (AZD0530), or Src shRNA viruses attenuated pY336 levels, and several somatic withdrawal signs. This was also observed in Fyn-/- mice. The stereotaxic injection of wild-type MOR, but not mutant (Y336F) MOR, lentiviruses into the locus coeruleus of MOR-/- mice restored somatic withdrawal jumping. Regulating pY336 levels during withdrawal might be a future target for drug development to prevent opiate addictive behaviors.


Asunto(s)
Receptores Opioides mu/metabolismo , Tirosina/metabolismo , Familia-src Quinasas/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Benzodioxoles/farmacología , Peso Corporal/efectos de los fármacos , Células HEK293 , Humanos , Locomoción/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fyn/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fyn/deficiencia , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Tirosina/química , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
10.
Psychopharmacology (Berl) ; 234(12): 1891-1900, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28343361

RESUMEN

RATIONALE: Morphine is the prototypic mu opioid, producing its analgesic actions through traditional 7 transmembrane domain (7TM) G-protein-coupled receptors generated by the mu opioid receptor gene (Oprm1). However, the Oprm1 gene undergoes extensive alternative splicing to yield three structurally distinct sets of splice variants. In addition to the full-length 7TM receptors, it produces a set of truncated variants comprised of only 6 transmembrane domains (6TM). OBJECTIVES: This study explored the relative contributions of 7TM and 6TM variants in a range of morphine actions. METHODS: Groups of male and mixed-gender wild-type and exon 11 Oprm1 knockout mice were examined in a series of behavioral assays measuring analgesia, hyperalgesia, respiration, and reward in conditioned place preference assays. RESULTS: Loss of the 6TM variants in an exon 11 knockout (E11 KO) mouse did not affect morphine analgesia, reward, or respiratory depression. However, E11 KO mice lacking 6TM variants failed to show morphine-induced hyperalgesia, developed tolerance more slowly than wild-type mice, and did not display hyperlocomotion. CONCLUSIONS: Together, our findings confirm the established role of 7TM mu receptor variants in morphine analgesia, reward, and respiratory depression, but reveal an unexpected obligatory role for 6TM variants in morphine-induced hyperalgesia and a modulatory role in morphine tolerance and dependence.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Morfina/uso terapéutico , Variantes Farmacogenómicas/genética , Receptores Opioides mu/genética , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Analgesia/métodos , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos/genética , Femenino , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Variantes Farmacogenómicas/efectos de los fármacos , Receptores Opioides mu/deficiencia
11.
Int J Neuropsychopharmacol ; 20(5): 403-409, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031268

RESUMEN

Background: Opioid and dopamine systems play crucial roles in reward. Similarities and differences in the neural mechanisms of reward that are mediated by these 2 systems have remained largely unknown. Thus, in the present study, we investigated the differences in reward function in both µ-opioid receptor knockout mice and dopamine transporter knockout mice, important molecules in the opioid and dopamine systems. Methods: Mice were implanted with electrodes into the right lateral hypothalamus (l hour). Mice were then trained to put their muzzle into the hole in the head-dipping chamber for intracranial electrical stimulation, and the influences of gene knockout were assessed. Results: Significant differences are observed between opioid and dopamine systems in reward function. µ-Opioid receptor knockout mice exhibited enhanced intracranial electrical stimulation, which induced dopamine release. They also exhibited greater motility under conditions of "despair" in both the tail suspension test and water wheel test. In contrast, dopamine transporter knockout mice maintained intracranial electrical stimulation responding even when more active efforts were required to obtain the reward. Conclusions: The absence of µ-opioid receptor or dopamine transporter did not lead to the absence of intracranial electrical stimulation responsiveness but rather differentially altered it. The present results in µ-opioid receptor knockout mice are consistent with the suppressive involvement of µ-opioid receptors in both positive incentive motivation associated with intracranial electrical stimulation and negative incentive motivation associated with depressive states. In contrast, the results in dopamine transporter knockout mice are consistent with the involvement of dopamine transporters in positive incentive motivation, especially its persistence. Differences in intracranial electrical stimulation in µ-opioid receptor and dopamine transporter knockout mice underscore the multidimensional nature of reward.


Asunto(s)
Analgésicos Opioides/metabolismo , Dopamina/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Receptores Opioides mu/deficiencia , Animales , Biofisica , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/deficiencia , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Estimulación Eléctrica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Motivación , Actividad Motora/efectos de los fármacos , Receptores Opioides mu/genética , Recompensa , Autoadministración , Factores de Tiempo
12.
Nature ; 537(7619): 185-190, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27533032

RESUMEN

Morphine is an alkaloid from the opium poppy used to treat pain. The potentially lethal side effects of morphine and related opioids-which include fatal respiratory depression-are thought to be mediated by µ-opioid-receptor (µOR) signalling through the ß-arrestin pathway or by actions at other receptors. Conversely, G-protein µOR signalling is thought to confer analgesia. Here we computationally dock over 3 million molecules against the µOR structure and identify new scaffolds unrelated to known opioids. Structure-based optimization yields PZM21-a potent Gi activator with exceptional selectivity for µOR and minimal ß-arrestin-2 recruitment. Unlike morphine, PZM21 is more efficacious for the affective component of analgesia versus the reflexive component and is devoid of both respiratory depression and morphine-like reinforcing activity in mice at equi-analgesic doses. PZM21 thus serves as both a probe to disentangle µOR signalling and a therapeutic lead that is devoid of many of the side effects of current opioids.


Asunto(s)
Analgésicos Opioides/efectos adversos , Analgésicos Opioides/química , Descubrimiento de Drogas , Receptores Opioides mu/agonistas , Tiofenos/química , Tiofenos/farmacología , Urea/análogos & derivados , Analgesia/métodos , Analgésicos Opioides/farmacología , Animales , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación del Acoplamiento Molecular , Dolor/tratamiento farmacológico , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Compuestos de Espiro/farmacología , Relación Estructura-Actividad , Tiofenos/efectos adversos , Urea/efectos adversos , Urea/química , Urea/farmacología
13.
Neural Plast ; 2016: 5346161, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27274875

RESUMEN

Environmental enrichment has been proven to have positive effects on both behavioral and physiological phenotypes in rodent models of mental and neurodevelopmental disorders. In this study, we used mice lacking the µ-opioid receptor gene (Oprm1 (-/-)), which has been shown to have deficits in social competence and communication, to assess the hypothesis that early enrichment can ameliorate sociability during development and adulthood. Due to the immaturity of sensory-motor capabilities of young pups, we chose as environmental stimulation a second lactating female, who provided extra maternal care and stimulation from birth. The results show that double mothering normalized the abnormal response to maternal separation in Oprm1 (-/-) pups and increased social motivation in juveniles and adult knockout mice. Additionally, we observed that Oprm1 (-/-) mice act as less attractive social partners than wild types, which suggests that social motivation can be modulated by the stimulus employed. This experiment supports previous findings suggesting that early social environmental stimulation has profound and long-term beneficial effects, encouraging the use of nonpharmacological interventions for the treatment of social defects in neurodevelopmental diseases.


Asunto(s)
Trastorno Autístico/metabolismo , Modelos Animales de Enfermedad , Ambiente , Motivación/fisiología , Receptores Opioides mu/deficiencia , Conducta Social , Animales , Trastorno Autístico/genética , Reacción de Prevención/fisiología , Femenino , Masculino , Privación Materna , Ratones , Ratones Noqueados , Receptores Opioides mu/genética
14.
Proc Natl Acad Sci U S A ; 113(13): 3663-8, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976581

RESUMEN

The clinical management of severe pain depends heavily on opioids acting through mu opioid receptors encoded by the Oprm1 gene, which undergoes extensive alternative splicing. In addition to generating a series of prototypic seven transmembrane domain (7TM) G protein-coupled receptors (GPCRs), Oprm1 also produces a set of truncated splice variants containing only six transmembrane domains (6TM) through which selected opioids such as IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) mediate a potent analgesia without many undesirable effects. Although morphine analgesia is independent of these 6TM mu receptor isoforms, we now show that the selective loss of the 6TM variants in a knockout model eliminates the analgesic actions of delta and kappa opioids and of α2-adrenergic compounds, but not cannabinoid, neurotensin, or muscarinic drugs. These observations were confirmed by using antisense paradigms. Despite their role in analgesia, loss of the 6TM variants were not involved with delta opioid-induced seizure activity, aversion to the kappa drug U50, 488H, or α2-mediated hypolocomotion. These observations support the existence of parallel opioid and nonopioid pain modulatory systems and highlight the ability to dissociate unwanted delta, kappa1, and α2 actions from analgesia.


Asunto(s)
Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Empalme Alternativo , Analgesia , Analgésicos Opioides/farmacología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Manejo del Dolor , Dimensión del Dolor , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Receptores Opioides mu/deficiencia
15.
Sci Rep ; 5: 11384, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26072707

RESUMEN

Opportunistic lung infection and inflammation is a hallmark of chronic recreational/clinical use of morphine. We show that early induction of IL17 from the bronchial epithelium, following pathogenic encounter is a protective response, which contributes to pathogenic clearance and currently attributed to TLR2 activation in immune cells. Concurrent activation of TLR2 and IL17R in bronchial epithelium results in the sequestration of MyD88 (TLR2 adapter) by Act1/CIKS (IL17R adapter), thereby turning off TLR2 signaling to restore homeostasis. Morphine inhibits the early IL17 release and interaction between Act1 and MyD88, leading to decreased pathogenic clearance and sustained inflammation. Hence, we propose that therapeutically targeting either TLR2 or IL17 in bronchial epithelia, in the context of morphine, can restore inflammatory homeostasis.


Asunto(s)
Interleucina-17/inmunología , Pulmón/inmunología , Morfina/efectos adversos , Infecciones Neumocócicas/inmunología , Receptores de Interleucina-17/inmunología , Receptor Toll-Like 2/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Regulación de la Expresión Génica , Homeostasis , Humanos , Inflamación , Interleucina-17/genética , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Infecciones Neumocócicas/etiología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/inmunología , Receptores de Interleucina-17/genética , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Receptores Opioides mu/inmunología , Transducción de Señal , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
16.
J Clin Invest ; 125(7): 2626-30, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26011641

RESUMEN

The generation of potent opioid analgesics that lack the side effects of traditional opioids may be possible by targeting truncated splice variants of the µ-opioid receptor. µ-Opioids act through GPCRs that are generated from the Oprm1 gene, which undergoes extensive alternative splicing. The most abundant set of Oprm1 variants encode classical full-length 7 transmembrane domain (7TM) µ-opioid receptors that mediate the actions of the traditional µ-opioid drugs morphine and methadone. In contrast, 3-iodobenzoyl-6ß-naltrexamide (IBNtxA) is a potent analgesic against thermal, inflammatory, and neuropathic pain that acts independently of 7TM µ-opioid receptors but has no activity in mice lacking a set of 6TM truncated µ-opioid receptor splice variants. Unlike traditional opioids, IBNtxA does not depress respiration or result in physical dependence or reward behavior, suggesting it acts through an alternative µ-opioid receptor target. Here we demonstrated that a truncated 6TM splice variant, mMOR-1G, can rescue IBNtxA analgesia in a µ-opioid receptor-deficient mouse that lacks all Oprm1 splice variants, ablating µ-opioid activity in these animals. Intrathecal administration of lentivirus containing the 6TM variant mMOR-1G restored IBNtxA, but not morphine, analgesia in Oprm1-deficient animals. Together, these results confirm that a truncated 6TM GPCR is both necessary and sufficient for IBNtxA analgesia.


Asunto(s)
Analgésicos Opioides/farmacología , Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Empalme Alternativo , Animales , Exones , Marcación de Gen , Metadona/farmacología , Ratones , Ratones Noqueados , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/fisiología , Estructura Terciaria de Proteína , Receptores Opioides mu/deficiencia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Neuropharmacology ; 89: 325-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25446676

RESUMEN

Social defeat stress causes social avoidance and long-lasting cross-sensitization to psychostimulants, both of which are associated with increased brain-derived neurotrophic factor (BDNF) expression in the ventral tegmental area (VTA). Moreover, social stress upregulates VTA mu-opioid receptor (MOR) mRNA. In the VTA, MOR activation inhibits GABA neurons to disinhibit VTA dopamine neurons, thus providing a role for VTA MORs in the regulation of psychostimulant sensitization. The present study determined the effect of lentivirus-mediated MOR knockdown in the VTA on the consequences of intermittent social defeat stress, a salient and profound stressor in humans and rodents. Social stress exposure induced social avoidance and attenuated weight gain in animals with non-manipulated VTA MORs, but both these effects were prevented by VTA MOR knockdown. Rats with non-manipulated VTA MOR expression exhibited cross-sensitization to amphetamine challenge (1.0 mg/kg, i.p.), evidenced by a significant augmentation of locomotion. By contrast, knockdown of VTA MORs prevented stress-induced cross-sensitization without blunting the locomotor-activating effects of amphetamine. At the time point corresponding to amphetamine challenge, immunohistochemical analysis was performed to examine the effect of stress on VTA BDNF expression. Prior stress exposure increased VTA BDNF expression in rats with non-manipulated VTA MOR expression, while VTA MOR knockdown prevented stress-induced expression of VTA BDNF. Taken together, these results suggest that upregulation of VTA MOR is necessary for the behavioral and biochemical changes induced by social defeat stress. Elucidating VTA MOR regulation of stress effects on the mesolimbic system may provide new therapeutic targets for treating stress-induced vulnerability to substance abuse.


Asunto(s)
Anfetamina/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Receptores Opioides mu/deficiencia , Estrés Psicológico , Área Tegmental Ventral/metabolismo , Analgésicos Opioides/farmacocinética , Animales , Peso Corporal/efectos de los fármacos , Recuento de Células , Modelos Animales de Enfermedad , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/fisiología , Manejo Psicológico , Masculino , Actividad Motora/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptores Opioides mu/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/prevención & control , Transducción Genética
18.
Psychopharmacology (Berl) ; 232(8): 1471-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25363463

RESUMEN

RATIONALE: Evidence based on clinical and experimental animal studies indicates that adolescent social deprivation influences alcohol consumption in a sex-dependent manner, perhaps by influencing stress responses. However, the mechanisms underlying the interaction between these phenomena remain to be elucidated. Since the µ-opioid receptor (MOP) has been reported to have key roles in social stress responses as well as the reinforcing/addictive effects of ethanol, MOP is a candidate molecule that may link adolescent social deprivation and subsequent alterations in alcohol consumption. OBJECTIVES: To evaluate the involvement of MOP and social isolation-induced changes in alcohol consumption, as well as the effect of sex differences on responses to social isolation, alcohol consumption was assessed using a two-bottle home-cage consumption procedure (8 % ethanol vs. water) in MOP knockout (MOP-KO) and wild type (WT) mice of both sexes exposed to adolescent social deprivation or reared socially. RESULTS: Isolation rearing had no effects upon alcohol consumption of WT mice, whereas it significantly altered alcohol consumption in both male and female MOP-KO mice. Interestingly, social isolation affected ethanol consumption differently in male and female mice. Ethanol consumption was increased in male MOP-KO mice, but decreased in female MOP-KO mice, by isolation rearing. CONCLUSION: These results indicate that disturbances of MOP function influence the effects of isolation rearing on ethanol consumption in a sex-dependent manner. Consequently, this suggests the possibility that genetic variation that influences MOP function may have differential roles in alcoholism in men and women, and alcoholism treatments that target MOP function may be differentially effective in males and females.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Caracteres Sexuales , Aislamiento Social , Factores de Edad , Consumo de Bebidas Alcohólicas/psicología , Animales , Etanol/administración & dosificación , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Aislamiento Social/psicología , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología
19.
Neuropsychopharmacology ; 39(13): 2974-88, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24943644

RESUMEN

The repeated cycles of cessation of consumption and relapse remain the major clinical concern in treating drug addiction. The endogenous opioid system is a crucial component of the reward circuit that participates in the adaptive changes leading to relapse in the addictive processes. We have used genetically modified mice to evaluate the involvement of µ-opioid receptor (MOR) and δ-opioid receptor (DOR) and their main endogenous ligands, the enkephalins derived from proenkephalin (PENK) and prodynorphin (PDYN), in the reinstatement of cocaine-seeking behavior. Constitutive knockout mice of MOR, DOR, PENK, and PDYN, and their wild-type littermates were trained to self-administer cocaine or to seek for palatable food, followed by a period of extinction and finally tested on a cue-induced reinstatement of seeking behavior. The four lines of knockout mice acquired operant cocaine self-administration behavior, although DOR and PENK knockout mice showed less motivation for cocaine than wild-type littermates. Moreover, cue-induced relapse was significantly decreased in MOR and DOR knockout mice. In contrast, PDYN knockout mice showed a slower extinction and increased relapse than wild-type littermates. C-Fos expression analysis revealed differential activation in brain areas related with memory and reward in these knockout mice. No differences were found in any of the four genotypes in operant responding to obtain palatable food, indicating that the changes revealed in knockout mice were not due to unspecific deficit in operant performance. Our results indicate that MOR, DOR, and PDYN have a differential role in cue-induced reinstatement of cocaine-seeking behavior.


Asunto(s)
Condicionamiento Operante/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Encefalinas/deficiencia , Precursores de Proteínas/deficiencia , Receptores Opioides mu/deficiencia , Refuerzo en Psicología , Análisis de Varianza , Anestésicos Locales/administración & dosificación , Animales , Cocaína/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Encefalinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ratones , Ratones Noqueados , Precursores de Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Opioides delta/deficiencia , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Esquema de Refuerzo , Autoadministración
20.
PLoS One ; 9(3): e91577, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24662916

RESUMEN

Recent epidemiologic studies implying differences in cancer recurrence based on anesthetic regimens raise the possibility that the mu opioid receptor (MOR) can influence cancer progression. Based on our previous observations that overexpression of MOR in human non-small cell lung cancer (NSCLC) cells increased tumor growth and metastasis, this study examined whether MOR regulates growth factor receptor signaling and epithelial mesenchymal transition (EMT) in human NSCLC cells. We utilized specific siRNA, shRNA, chemical inhibitors and overexpression vectors in human H358 NSCLC cells that were either untreated or treated with various concentrations of DAMGO, morphine, fentanyl, EGF or IGF. Cell function assays, immunoblot and immunoprecipitation assays were then performed. Our results indicate MOR regulates opioid and growth factor-induced EGF receptor signaling (Src, Gab-1, PI3K, Akt and STAT3 activation) which is crucial for consequent human NSCLC cell proliferation and migration. In addition, human NSCLC cells treated with opioids, growth factors or MOR overexpression exhibited an increase in snail, slug and vimentin and decrease ZO-1 and claudin-1 protein levels, results consistent with an EMT phenotype. Further, these effects were reversed with silencing (shRNA) or chemical inhibition of MOR, Src, Gab-1, PI3K, Akt and STAT3 (p<0.05). Our data suggest a possible direct effect of MOR on opioid and growth factor-signaling and consequent proliferation, migration and EMT transition during lung cancer progression. Such an effect provides a plausible explanation for the epidemiologic findings.


Asunto(s)
Analgésicos Opioides/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Pulmonares/patología , Receptores Opioides mu/metabolismo , Anestésicos/efectos adversos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Silenciador del Gen , Humanos , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...