Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Chem Neuroanat ; 132: 102324, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37557929

RESUMEN

Advances have been made in recent years in using opioid receptor antagonists as an adjunct therapy to psychotropic medication to reduce debilitating weight gain and metabolic adverse effects associated with in particular second generation antipsychotics. However, it is unknown whether second generation antipsychotics produce a change in opioid receptor expression in the brain. The present study investigated early changes in opioid receptor expression in the female rat hypothalamus, a master controller of hunger and metabolic regulation, after acute treatment with olanzapine, a commonly used second generation antipsychotic. Using quantitative spatial in situ hybridization and receptor autoradiography, expression levels of the three opioid receptors; kappa, mu and delta, were determined at mRNA and protein level, respectively, in the five hypothalamic areas: paraventricular nucleus, arcuate nucleus, ventromedial nucleus, dorsomedial nucleus and lateral hypothalamus. After 48 h of olanzapine treatment at clinically relevant plasma concentration weight gain and food intake changes, and increased plasma glucose were observed in female rats. Olanzapine treatment also led to a significant increase in mu opioid receptor availability in the arcuate nucleus, which contains both satiety and hunger controlling neurons. No other areas showed any opioid receptor expressional changes with olanzapine treatment on neither at mRNA nor protein level. Technical difficulties made it impossible to analyze mRNA levels in the lateral hypothalamus and overall binding of delta opioid receptors. Thus, the present study provided insights in to how olanzapine at clinically relevant plasma levels already at an early stage modulated the opioid system in the hypothalamus.


Asunto(s)
Antipsicóticos , Receptores Opioides mu , Ratas , Femenino , Animales , Olanzapina/farmacología , Olanzapina/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Antipsicóticos/farmacología , Hipotálamo/metabolismo , Receptores Opioides/metabolismo , Conducta Alimentaria , Aumento de Peso , ARN Mensajero
2.
Nat Commun ; 12(1): 3858, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158473

RESUMEN

Mitragynine (MG) is the most abundant alkaloid component of the psychoactive plant material "kratom", which according to numerous anecdotal reports shows efficacy in self-medication for pain syndromes, depression, anxiety, and substance use disorders. We have developed a synthetic method for selective functionalization of the unexplored C11 position of the MG scaffold (C6 position in indole numbering) via the use of an indole-ethylene glycol adduct and subsequent iridium-catalyzed borylation. Through this work we discover that C11 represents a key locant for fine-tuning opioid receptor signaling efficacy. 7-Hydroxymitragynine (7OH), the parent compound with low efficacy on par with buprenorphine, is transformed to an even lower efficacy agonist by introducing a fluorine substituent in this position (11-F-7OH), as demonstrated in vitro at both mouse and human mu opioid receptors (mMOR/hMOR) and in vivo in mouse analgesia tests. Low efficacy opioid agonists are of high interest as candidates for generating safer opioid medications with mitigated adverse effects.


Asunto(s)
Mitragyna/química , Extractos Vegetales/farmacología , Receptores Opioides mu/agonistas , Alcaloides de Triptamina Secologanina/farmacología , Analgésicos/química , Analgésicos/farmacología , Animales , Glicol de Etileno/química , Humanos , Ratones Noqueados , Modelos Químicos , Estructura Molecular , Extractos Vegetales/química , Unión Proteica , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina Secologanina/química
3.
Alcohol ; 95: 1-6, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34022405

RESUMEN

Clinical studies have postulated that ß-endorphin deficiency generates excessive alcohol consumption, and it has been shown that the reduction of ß-endorphin neurons increases alcohol intake in animal models. The ß-endorphin produce their rewarding effect when they act mainly on the µ-opioid receptors (MOR) located in mesolimbic structures. Thus, it is possible that individual differences in these components of the endogenous opioid system are related to different levels of alcohol consumption. The present study thus examines the relation between two levels of alcohol consumption and intrinsic characteristics of the components of the opioid system in outbred Wistar rats that were not genetically selected. We analyzed the number of ß-endorphin-positive neurons in the arcuate nucleus (ArN) and the expression of µ-opioid receptors (MOR) in regions of the reward system, such as the nucleus accumbens (NAc), amygdala (Amy), and ventral tegmental area (VTA) in outbred rats with low (LC) or high (HC) voluntary alcohol consumption. Findings showed that the HC rats had a lower number of ß-endorphin-positive neurons in the hypothalamic ArN and a higher expression of MOR in the NAc and VTA, compared to the LC rats. No changes in the expression of MOR in the Amy were observed between the two groups. Results suggest that intrinsic variability in the number of ß-endorphin neurons and in the expression of MOR in the LC and HC rats could explain their different patterns for alcohol intake.


Asunto(s)
Endorfinas , Consumo de Bebidas Alcohólicas , Animales , Hipotálamo/metabolismo , Neuronas/metabolismo , Ratas , Ratas Wistar , Receptores Opioides mu/genética
4.
Physiol Rep ; 9(5): e14788, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33661571

RESUMEN

Anorexia nervosa (AN) has a lifetime prevalence of up to 4% and a high mortality rate (~5-10%), yet little is known regarding the etiology of this disease. In an attempt to fill the gaps in knowledge, activity-based anorexia (ABA) in rodents has been a widely used model as it mimics several key features of AN including severely restricted food intake and excessive exercise. Using this model, a role for the hypothalamic proopiomelanocortin (POMC) system has been implicated in the development of ABA as Pomc mRNA is elevated in female rats undergoing the ABA paradigm. Since the Pomc gene product α-MSH potently inhibits food intake, it could be that elevated α-MSH might promote ABA. However, the α-MSH receptor antagonist SHU9119 does not protect against the development of ABA. Interestingly, it has also been shown that female mice lacking the mu opioid receptor (MOR), the primary receptor activated by the Pomc-gene-derived opioid ß-endorphin, display blunted food anticipatory behavior (FAA), a key feature of ABA. Thus, we hypothesized that the elevation in Pomc mRNA observed during ABA may lead to increased ß-endorphin concentrations and MOR activation to promote ABA. Further, given the known sex differences in AN and ABA, we hypothesized that MORs may contribute differentially in male and female mice. Using wild-type and MOR knockout mice of both sexes, a MOR antagonist and careful analysis of food anticipatory behavior and ß-endorphin levels, we found 1) increased Pomc mRNA levels in both female and male mice that underwent ABA, 2) increased ß-endorphin in female mice that underwent ABA, and 3) blunted FAA in both sexes in response to MOR genetic deletion yet blunted FAA only in males in response to MOR antagonism. The results presented provide support for both hypotheses and suggest that it may be the ß-endorphin resulting from increased Pomc transcription that supports the development of some features of ABA.


Asunto(s)
Anorexia/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , betaendorfina/farmacología , Animales , Anorexia/metabolismo , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Proopiomelanocortina/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
5.
Alcohol Clin Exp Res ; 44(6): 1224-1233, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32406553

RESUMEN

BACKGROUND: Human laboratory paradigms are a pillar in medication development for alcohol use disorders (AUD). Neuroimaging paradigms, in which individuals are exposed to cues that elicit neural correlates of alcohol craving (e.g., mesocorticolimbic activation), are increasingly utilized to test the effects of AUD medications. Elucidation of the translational effects of these neuroimaging paradigms on human laboratory paradigms, such as self-administration, is warranted. The current study is a secondary analysis examining whether alcohol cue-induced activation in the ventral striatum is predictive of subsequent alcohol self-administration in the laboratory. METHODS: Non-treatment-seeking heavy drinkers of East Asian descent (n = 41) completed a randomized, placebo-controlled, double-blind, crossover experiment on the effects of naltrexone on neuroimaging and human laboratory paradigms. Participants completed 5 days of study medication (or placebo); on day 4, they completed a neuroimaging alcohol taste cue-reactivity task. On the following day (day 5), participants completed a 60-minute alcohol self-administration paradigm. RESULTS: Multilevel Cox regressions indicated a significant effect of taste cue-elicited ventral striatum activation on latency to first drink, Wald χ2  = 2.88, p = 0.05, such that those with higher ventral striatum activation exhibited shorter latencies to consume their first drink. Similarly, ventral striatum activation was positively associated with total number of drinks consumed, F(1, 38) = 5.90, p = 0.02. These effects were significant after controlling for alcohol use severity, OPRM1 genotype, and medication. Other potential regions of interest (anterior cingulate, thalamus) were not predictive of self-administration outcomes. CONCLUSIONS: Neuroimaging alcohol taste cue paradigms may be predictive of laboratory paradigms such as self-administration. Elucidation of the relationships among different paradigms will inform how these paradigms may be used synergistically in experimental medicine and medication development.


Asunto(s)
Trastornos Relacionados con Alcohol/diagnóstico por imagen , Depresores del Sistema Nervioso Central/administración & dosificación , Señales (Psicología) , Etanol/administración & dosificación , Estriado Ventral/diagnóstico por imagen , Adulto , Disuasivos de Alcohol/farmacología , Trastornos Relacionados con Alcohol/fisiopatología , Familia de Aldehído Deshidrogenasa 1/genética , Aldehído Deshidrogenasa Mitocondrial/genética , Femenino , Neuroimagen Funcional , Genotipo , Giro del Cíngulo/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Análisis Multinivel , Naltrexona/farmacología , Modelos de Riesgos Proporcionales , Distribución Aleatoria , Receptores Opioides mu/genética , Autoadministración , Tálamo/diagnóstico por imagen , Estriado Ventral/efectos de los fármacos , Estriado Ventral/fisiopatología , Adulto Joven
6.
Eur J Pharmacol ; 872: 172948, 2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-31991139

RESUMEN

Compounds with high affinity at kappa and mu opioid receptors may have clinical utility in treating major depressive disorder. Nalmefene (NMF) is a partial kappa opioid receptor agonist and potent mu opioid receptor antagonist, but there has been no preclinical evaluation of NMF in rodent tests relevant to depression and anxiety. To address this, the effects of NMF on neurochemical and behavioral endpoints in C57BL/6J mice were examined and contrasted with a structurally related analog, naltrexone (NTX). NMF exhibited kappa opioid receptor agonist activity, measured as a reduction in extracellular dopamine release in the nucleus accumbens using in vivo microdialysis following acute but not chronic administration. In the mouse forced swim test, female mice were more responsive to higher doses of NMF and NTX compared to male mice. The behavioral effects of NMF in the forced swim test were blocked in Oprk1-/- and Oprm1-/- mice. Conversely, the effects of NTX were blocked only in Oprm1-/- mice. These results indicate that both kappa and mu opioid receptors mediate the behavioral effects of NMF, but the effects of NTX in this test were modified only by mu opioid receptor engagement. Unlike NTX, NMF did not produce conditioned place aversion in either sex. Finally, NMF's activity in the marble burying test and forced swim test were retained following chronic administration. The sustained effects exerted by NMF on tests that are sensitive to antidepressant and anxiolytic compounds support further investigation of NMF as a potential therapeutic for depression.


Asunto(s)
Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Reposicionamiento de Medicamentos , Naltrexona/análogos & derivados , Receptores Opioides kappa/agonistas , Animales , Técnicas de Observación Conductual , Depresión/diagnóstico , Modelos Animales de Enfermedad , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Naltrexona/farmacología , Naltrexona/uso terapéutico , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Factores Sexuales
7.
Sci Rep ; 9(1): 17837, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780740

RESUMEN

Brain µ-opioid receptors (MOR) mediate reward and help coping with pain, social rejection, anxiety and depression. The dorsal midline thalamus (dMT) integrates visceral/emotional signals and biases behavior towards aversive or defensive states through projections to the amygdala. While a dense MOR expression in the dMT has been described, the exact cellular and synaptic mechanisms of µ-opioidergic modulation in the dMT-amygdala circuitry remain unresolved. Here, we hypothesized that MORs are important negative modulators of dMT-amygdala excitatory networks. Using retrograde tracers and targeted channelrhodopsin expression in combination with patch-clamp electrophysiology, we found that projections of dMT neurons onto both basal amygdala principal neurons (BA PN) and central amygdala (CeL) neurons are attenuated by stimulation of somatic or synaptic MORs. Importantly, dMT efferents to the amygdala drive feedforward excitation of centromedial amygdala neurons (CeM), which is dampened by MOR activation. This downregulation of excitatory activity in dMT-amygdala networks puts the µ-opioid system in a position to ameliorate aversive or defensive behavioral states associated with stress, withdrawal, physical pain or social rejection.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Neuronas/metabolismo , Receptores Opioides mu/metabolismo , Tálamo/metabolismo , Potenciales de Acción , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Regulación hacia Abajo , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Neuronas/fisiología , Receptores Opioides mu/genética , Tálamo/citología , Tálamo/fisiología
8.
Genes Brain Behav ; 18(8): e12600, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31339663

RESUMEN

Persistent alterations of proopiomelanocortin (Pomc) and mu-opioid receptor (Oprm1) activity and stress responses after alcohol are critically involved in vulnerability to alcohol dependency. Gene transcriptional regulation altered by alcohol may play important roles. Mice with genome-wide deletion of neuronal Pomc enhancer1 (nPE1-/- ), had hypothalamic-specific partial reductions of beta-endorphin and displayed lower alcohol consumption, compared to wildtype littermates (nPE1+/+ ). We used RNA-Seq to measure steady-state nuclear mRNA transcripts of opioid and stress genes in hypothalamus of nPE1+/+ and nPE1-/- mice after 1-day acute withdrawal from chronic excessive alcohol drinking or after water. nPE1-/- had lower basal Pomc and Pdyn (prodynorphin) levels compared to nPE1+/+ , coupled with increased basal Oprm1 and Oprk1 (kappa-opioid receptor) levels, and low alcohol drinking increased Pomc and Pdyn to the basal levels of nPE1+/+ in the water group, without significant effects on Oprm1 and Oprk1. In nPE1+/+ , excessive alcohol intake increased Pomc and Oprm1, with no effect on Pdyn or Oprk1. For stress genes, nPE1-/- had lowered basal Oxt (oxytocin) and Avp (arginine vasopressin) that were restored by low alcohol intake to basal levels of nPE1+/+ . In nPE1+/+ , excessive alcohol intake decreased Oxt and Avpi1 (AVP-induced protein1). Functionally examining the effect of pharmacological blockade of mu-opioid receptor, we found that naltrexone reduced excessive alcohol intake in nPE1+/+ , but not nPE1-/- . Our results provide evidence relevant to the transcriptional profiling of the critical genes in mouse hypothalamus: enhanced opioid and reduced stress gene transcripts after acute withdrawal from excessive alcohol may contribute to altered reward and stress responses.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/genética , Elementos de Facilitación Genéticos/genética , Hipotálamo/metabolismo , Proopiomelanocortina/genética , Animales , Arginina Vasopresina/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Etanol/farmacología , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxitocina/metabolismo , Proopiomelanocortina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transcriptoma
9.
Naunyn Schmiedebergs Arch Pharmacol ; 392(9): 1141-1149, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31079200

RESUMEN

Berberine, a chemical found in plants, is used as a supplement for diabetes. This study aimed to investigate the effects and the underlying molecular regulations of berberine in diabetic neuropathic pain in a rat model of diabetes. Rats were injected with streptozotocin (STZ) to induce diabetes and then were treated with berberine. Blood glucose levels and body weight were measured. Thermal and mechanical nociception were assessed by paw pressure test and hot tail immersion test. Oxidative stress was assessed by lipid peroxidation, production of reactive oxygen species (ROS) and catalase activity. Neuroinflammation was assessed by tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) protein levels. Finally, µ-opioid receptor (MOR) protein and mRNA levels were measured. We found that berberine treatment partially suppressed blood glucose levels and restored body weight in diabetic rats. Berberine also suppressed STZ-induced oversensitivity of mechanical and thermal nociception. Additionally, berberine partially suppressed oxidative stress and inflammation of diabetic rats. Finally, berberine significantly enhanced protein and mRNA expression levels of µ-opioid receptor (MOR). Our findings suggest that berberine is a potential therapeutic alleviating diabetes and diabetic neuropathic pain, probably through suppression of oxidative stress and neuroinflammation that may be mediated by MOR.


Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Berberina/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Dolor/tratamiento farmacológico , Analgésicos/farmacología , Animales , Antiinflamatorios/farmacología , Berberina/farmacología , Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Interleucina-6/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Dolor/metabolismo , Ratas Wistar , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Sci Rep ; 9(1): 3602, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837649

RESUMEN

Aldehyde dehydrogenase 1A1 (ALDH1A1), a retinoic acid (RA) synthase, is selectively expressed by the nigrostriatal dopaminergic (nDA) neurons that preferentially degenerate in Parkinson's disease (PD). ALDH1A1-positive axons mainly project to the dorsal striatum. However, whether ALDH1A1 and its products regulate the activity of postsynaptic striatal neurons is unclear. Here we show that µ-type opioid receptor (MOR1) levels were severely decreased in the dorsal striatum of postnatal and adult Aldh1a1 knockout mice, whereas dietary supplement of RA restores its expression. Furthermore, RA treatment also upregulates striatal MOR1 levels and signaling and alleviates L-DOPA-induced dyskinetic movements in pituitary homeobox 3 (Pitx3)-deficient mice that lack of ALDH1A1-expressing nDA neurons. Therefore, our findings demonstrate that ALDH1A1-synthesized RA is required for postsynaptic MOR1 expression in the postnatal and adult dorsal striatum, supporting potential therapeutic benefits of RA supplementation in moderating L-DOPA-induced dyskinesia.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/fisiología , Cuerpo Estriado/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Discinesias/prevención & control , Proteínas de Homeodominio/fisiología , Receptores Opioides mu/metabolismo , Retinal-Deshidrogenasa/fisiología , Factores de Transcripción/fisiología , Tretinoina/farmacología , Animales , Cuerpo Estriado/patología , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Discinesias/etiología , Discinesias/metabolismo , Discinesias/patología , Femenino , Masculino , Ratones , Ratones Noqueados , Receptores Opioides mu/genética
11.
Nat Commun ; 9(1): 1500, 2018 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-29662095

RESUMEN

The endogenous µ-opioid receptor (MOR) system regulates motivational and hedonic processing. We tested directly whether individual differences in MOR are associated with neural reward responses to food pictures in humans. We scanned 33 non-obese individuals with positron emission tomography (PET) using the MOR-specific radioligand [11C]carfentanil. During a functional magnetic resonance imaging (fMRI) scan, the subjects viewed pictures of appetizing versus bland foods to elicit reward responses. MOR availability was measured in key components of the reward and emotion circuits and used to predict BOLD-fMRI responses to foods. Viewing palatable versus bland foods activates regions involved in homeostatic and reward processing, such as amygdala, ventral striatum, and hypothalamus. MOR availability in the reward and emotion circuit is negatively associated with the fMRI reward responses. Variation in MOR availability may explain why some people feel an urge to eat when encountering food cues, increasing risk for weight gain and obesity.


Asunto(s)
Amígdala del Cerebelo/fisiología , Hipotálamo/fisiología , Reconocimiento Visual de Modelos/fisiología , Receptores Opioides mu/metabolismo , Estriado Ventral/fisiología , Adulto , Amígdala del Cerebelo/anatomía & histología , Amígdala del Cerebelo/diagnóstico por imagen , Analgésicos Opioides/farmacocinética , Mapeo Encefálico , Fentanilo/análogos & derivados , Fentanilo/farmacocinética , Alimentos , Expresión Génica , Humanos , Hipotálamo/anatomía & histología , Hipotálamo/diagnóstico por imagen , Ligandos , Imagen por Resonancia Magnética , Masculino , Fotograbar , Tomografía de Emisión de Positrones , Receptores Opioides mu/genética , Estriado Ventral/anatomía & histología , Estriado Ventral/diagnóstico por imagen
12.
Alcohol Clin Exp Res ; 42(6): 1034-1043, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29667742

RESUMEN

BACKGROUND: The gene encoding phosphatidylinositol-4-phosphate 5-kinase (PIP5K1C) has been recently implicated in pain regulation. Interestingly, a recent cross-tissue and cross-phenotypic epigenetic analysis identified the same gene in alcohol use disorder (AUD). Given the high comorbidity between AUD and chronic pain, we hypothesized that genetic variation in PIP5K1C might contribute to susceptibility to AUD. METHODS: We conducted a case-control association study of genetic variants in PIP5K1C. Association analyses of 16 common PIP5K1C single nucleotide polymorphisms (SNPs) were conducted in cases and controls of African (427 cases and 137 controls) and European ancestry (488 cases and 324 controls) using standard methods. In addition, given the prominent role of the opioid system in pain signaling, we investigated the effects of acute alcohol exposure on PIP5K1C expression in humanized transgenic mice for the µ-opioid receptor that included the OPRM1 A118G polymorphism, a widely used mouse model to study analgesic response to opioids in pain. PIP5K1C expression was measured in the thalamus and basolateral amygdala (BLA) in mice after short-term administration (single 2 g/kg dose) of alcohol or saline using immunohistochemistry and analyzed by 2-way analysis of variance. RESULTS: In the case-control association study using an NIAAA discovery sample, 8 SNPs in PIP5K1C were significantly associated with AUD in the African ancestry (AA) group (p < 0.05 after correction; rs4807493, rs10405681, rs2074957, rs10432303, rs8109485, rs1476592, rs10419980, and rs4432372). However, a replication analysis using an independent sample (N = 3,801) found no significant associations after correction for multiple testing. In the humanized transgenic mouse model with the OPRM1 polymorphism, PIP5K1C expression was significantly different between alcohol and saline-treated mice, regardless of genotype, in both the thalamus (p < 0.05) and BLA (p < 0.01). CONCLUSIONS: Our discovery sample shows that genetic variants in PIP5K1C are associated with AUD in the AA group, and acute alcohol exposure leads to up-regulation of PIP5K1C, potentially explaining a mechanism underlying the increased risk for chronic pain conditions in individuals with AUD.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Etanol/farmacología , Predisposición Genética a la Enfermedad/genética , Dolor/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Transducción de Señal/genética , Negro o Afroamericano/genética , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Estudios de Casos y Controles , Estudios de Asociación Genética , Humanos , Ratones , Ratones Transgénicos , Polimorfismo de Nucleótido Simple , Receptores Opioides mu/genética , Transducción de Señal/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/metabolismo , Población Blanca/genética
13.
Int J Clin Exp Hypn ; 66(1): 106-118, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29319460

RESUMEN

The possible cooperation between hypnotizability-related and placebo mechanisms in pain modulation has not been consistently assessed. Here, we investigate possible genetic bases for such cooperation. The OPRM1 gene, which encodes the µ1 opioid receptor-the primary site of action for endogenous and exogenous opioids-is polymorphic in the general population for the missense mutation Asn40Asp (A118G, rs1799971). The minor allele 118G results in decreased levels of OPRM1 mRNA and protein. As a consequence, G carriers are less responsive to opioids. The aim of the study was to investigate whether hypnotizability is associated with the presence of the OPRM1 polymorphism. Forty-three high and 60 low hypnotizable individuals, as well as 162 controls, were genotyped for the A118G polymorphism of OPRM1. The frequency of the G allele was significantly higher in highs compared to both lows and controls. Findings suggest that an inefficient opioid system may be a distinctive characteristic of highs and that hypnotic assessment may predict lower responsiveness to opioids.


Asunto(s)
Hipnosis , Receptores Opioides mu/genética , Estudios de Casos y Controles , Frecuencia de los Genes/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Polimorfismo de Nucleótido Simple/fisiología , Receptores Opioides mu/fisiología
14.
J Ethnopharmacol ; 205: 178-185, 2017 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-28501425

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Mitragyna speciosa and its extracts are called kratom (dried leaves, extract). They contain several alkaloids with an affinity for different opioid receptors. They are used in traditional medicine for the treatment of different diseases, as a substitute by opiate addicts, and to mitigate opioid withdrawal symptoms. Apart from their medical properties, they are used to enhance physical endurance and as a means of overcoming stress. PURPOSE: The aim of this study was to determine the mechanisms underlying the effects of kratom on restraint-stress-induced analgesia which occurs during or following exposure to a stressful or fearful stimulus. METHODS: To gain further insights into the action of kratom on stress, we conducted experiments using restraint stress as a test system and stress-induced analgesia as a test parameter. Using transgenic mu opioid-receptor (MOR) deficient mice, we studied the involvement of this receptor type. We used nor-binaltorphimine (BNT), an antagonist at kappa opioid receptors (KOR), to study functions of this type of receptor. Membrane potential assay was also employed to measure the intrinsic activity of kratom in comparison to U50,488, a highly selective kappa agonist. RESULTS: Treatment with kratom diminished stress-induced analgesia in wildtype and MOR knockout animals. Pretreatment of MOR deficient mice with BNT resulted in similar effects. In comparison to U50,488, kratom exhibited negligible intrinsic activity at KOR alone. CONCLUSIONS: The results suggest that the use of kratom as a pharmacological tool to mitigate withdrawal symptoms is related to its action on KOR.


Asunto(s)
Mitragyna/química , Extractos Vegetales/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgesia , Animales , Masculino , Ratones , Ratones Transgénicos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Extractos Vegetales/química , Hojas de la Planta/química , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Restricción Física , Estrés Fisiológico
15.
Behav Brain Res ; 320: 391-399, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27825896

RESUMEN

Conditioned taste aversion (CTA) causes a shift in the hedonic evaluation of a conditioned stimulus (CS) from positive to negative, and reduces the CS intake. Mu-opioid receptors (MORs) in the ventral pallidum (VP) are known to be involved in the hedonic evaluation of positive rewarding stimuli; however, their involvement in evaluation of a negative aversive stimulus is still unclear. To explore the neural mechanisms of the negative hedonic evaluation of the CS in CTA, we examined the effects of the activation of VP MORs on the behavioral responses of rats to a CS. Rats implanted with guide cannulae into the bilateral VP received a pairing of 5mM saccharin solution as a CS with an intraperitoneal injection of 0.15M lithium chloride as an unconditioned stimulus. On the test day, after microinjections of MOR agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) into the VP, we observed the behavioral responses to the intraorally infused CS solution. The DAMGO injections caused a larger number of ingestive taste reactivity responses to the CS solution. We also measured the consumption of the CS solution in a separate group of rats, using a single-bottle test. The DAMGO injected rats drank a higher volume of the CS solution than the saline injected rats. These results indicate that the activation of MORs in the VP results in the attenuation of aversion to the CS solution, thereby inducing the larger CS intake. Therefore, it is likely that VP MORs are involved in not only positive but also negative hedonic evaluation.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Globo Pálido/metabolismo , Receptores Opioides mu/metabolismo , Percepción del Gusto/efectos de los fármacos , Gusto/fisiología , Adyuvantes Inmunológicos/farmacología , Analgésicos Opioides/farmacología , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Condicionamiento Psicológico/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Globo Pálido/efectos de los fármacos , Cloruro de Litio/farmacología , Masculino , Microinyecciones , Ratas , Ratas Wistar , Receptores Opioides mu/genética , Sacarina/administración & dosificación , Gusto/efectos de los fármacos , Percepción del Gusto/fisiología
16.
Electromagn Biol Med ; 35(3): 206-13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26151161

RESUMEN

During the cell communication process, endogenous and exogenous signaling affect normal as well as pathological developmental conditions. Exogenous influences such as extra-low-frequency electromagnetic field (EMF) have been shown to effect pain and inflammation by modulating G-protein receptors, down-regulating cyclooxygenase-2 activity, and affecting the calcium/calmodulin/nitric oxide pathway. Investigators have reported changes in opioid receptors and second messengers, such as cyclic adenosine monophosphate (cAMP), in opiate tolerance and dependence by showing how repeated exposure to morphine decreases adenylate cyclase activity causing cAMP to return to control levels in the tolerant state, and increase above control levels during withdrawal. Resonance responses to biological systems using exogenous EMF signals suggest that frequency response characteristics of the target can determine the EMF biological response. In our past research we found significant down regulation of inflammatory markers tumor necrosis factor alpha (TNF-α) and nuclear factor kappa B (NFκB) using 5 Hz EMF frequency. In this study cAMP was stimulated in Chinese Hamster Ovary (CHO) cells transfected with human mu-opioid receptors, then exposed to 5 Hz EMF, and outcomes were compared with morphine treatment. Results showed a 23% greater inhibition of cAMP-treating cells with EMF than with morphine. In order to test our results for frequency specific effects, we ran identical experiments using 13 Hz EMF, which produced results similar to controls. This study suggests the use of EMF as a complementary or alternative treatment to morphine that could both reduce pain and enhance patient quality of life without the side-effects of opiates.


Asunto(s)
AMP Cíclico/metabolismo , Campos Electromagnéticos , Receptores Opioides mu/metabolismo , Animales , Células CHO , Colforsina/metabolismo , Cricetinae , Cricetulus , Espacio Extracelular/metabolismo , Espacio Extracelular/efectos de la radiación , Humanos , Espacio Intracelular/metabolismo , Espacio Intracelular/efectos de la radiación , Receptores Opioides mu/genética , Transducción de Señal/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
17.
ACS Chem Neurosci ; 6(11): 1813-24, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26325040

RESUMEN

3-Iodobenzoyl naltrexamine (IBNtxA) is a potent analgesic belonging to the pharmacologically diverse 6ß-amidoepoxymorphinan group of opioids. We present the synthesis and pharmacological evaluation of five analogs of IBNtxA. The scaffold of IBNtxA was modified by removing the 14-hydroxy group, incorporating a 7,8 double bond and various N-17 alkyl substituents. The structural modifications resulted in analogs with picomolar affinities for opioid receptors. The lead compound (MP1104) was found to exhibit approximately 15-fold greater antinociceptive potency (ED50 = 0.33 mg/kg) compared with morphine, mediated through the activation of kappa- and delta-opioid receptors. Despite its kappa agonism, this lead derivative did not cause place aversion or preference in mice in a place-conditioning assay, even at doses 3 times the analgesic ED50. However, pretreatment with the lead compound prevented the reward behavior associated with cocaine in a conditioned place preference assay. Together, these results suggest the promise of dual acting kappa- and delta-opioid receptor agonists as analgesics and treatments for cocaine addiction.


Asunto(s)
Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacología , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Morfinanos/síntesis química , Morfinanos/farmacología , Analgésicos Opioides/química , Animales , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Trastornos Relacionados con Cocaína/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Evaluación Preclínica de Medicamentos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Morfinanos/química , Naltrexona/análogos & derivados , Naltrexona/química , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/farmacología , Distribución Aleatoria , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Recompensa , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
18.
Brain Res Bull ; 117: 32-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26235542

RESUMEN

The therapeutic use of opioids is limited by the development of tolerance to the analgesic effect and the cellular and molecular mechanisms underlying this phenomenon are still not completely understood. For this reason the search for new analgesic derivatives, endowed with lower tolerance, is always an active field. The newly synthesized 14-O-Methylmorphine-6-sulfate (14-O-MeM6SU) shows high efficacy in in vitro assays and a strong analgesic action in the rat tail flick test. The aim of present work was to investigate: the analgesic effect of 14-O-MeM6SU in mouse tail-flick test; the tolerance to analgesic effect of 14-O-MeM6SU compared to morphine in mice, the effects of test compounds on glutamatergic neurotransmission by measuring spontaneous excitatory postsynaptic currents (sEPSCs) of layer V pyramidal cells from rat prefrontal cortices; and the effect of acute and chronic 14-O-MeM6SU treatments on opioid receptor gene expression in SH-SY5Y neuroblastoma cells expressing µ-opioid (MOP) and nociceptin/opioid receptor-like 1 (NOP) receptors. 14-O-MeM6SU was 17 times more potent than morphine in analgesia and had long duration of action in analgesic dose equipotent to morphine. Mice were treated subcutaneously (s.c.) either with 200 µmol/kg morphine or with 14-O-MeM6SU (12 µmol/kg) twice daily for three days. The magnitude of tolerance or cross-tolerance indicated by the shift in antinociceptive ED50 measured was greater for morphine compared to 14-O-MeM6SU. Subsequent to behavioral testing, patch-clamp experiments in layer V pyramidal neurons of rat prefrontal cortical slices in the presence of bicuculline were performed. Both 14-O-MeM6SU (0.1 µM) and morphine (1 µM) decreased the frequency of sEPSCs, indicating reduction of glutamate release. The effect of the novel compound was reversed by the opioid receptor antagonist naloxone, indicating an opioid mediated action. In contrast, the amplitude was not affected. Finally, gene expression data showed a dose dependent down-regulation of MOP receptor after 24h and 48 h exposure to 14-O-MeM6SU. Interestingly, no changes were detected for NOP receptor gene expression. The specific lack of this effect could be related to the lower tolerance development to analgesic effect of 14-O-MeM6SU. Furthermore, 14-O-MeM6SU displayed high intrinsic efficacy possibly an important factor in the observed effects. Further, the observed inhibition of glutamatergic signaling might be attributed also to the reduction of opioid tolerance. Based on our results the development of a new clinically important, safe analgesic agent might be possible.


Asunto(s)
Analgésicos Opioides/farmacología , Codeína/análogos & derivados , Morfina/farmacología , Analgésicos Opioides/efectos adversos , Animales , Línea Celular Tumoral , Codeína/efectos adversos , Codeína/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Tolerancia a Medicamentos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Morfina/efectos adversos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas Wistar , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos , Receptor de Nociceptina
19.
J Pharmacol Sci ; 128(4): 208-11, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26318673

RESUMEN

Neuropathic pain is often insensitive to morphine. Our previous study has demonstrated that neuron-restrictive silencer factor represses mu opioid receptor (MOP) gene expression in the dorsal root ganglion (DRG) via histone hypoacetylation-mediated mechanisms after peripheral nerve injury, thereby causing loss of peripheral morphine analgesia. Here, we showed that histone deacetylase (HDAC) inhibitors, such as trichostatin A and valproic acid, restored peripheral and systemic morphine analgesia in neuropathic pain. Also, these agents blocked nerve injury-induced MOP down-regulation in the DRG. These results suggest that HDAC inhibitors could serve as adjuvant analgesics to morphine for the management of neuropathic pain.


Asunto(s)
Analgésicos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Morfina/farmacología , Morfina/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Traumatismos de los Nervios Periféricos/complicaciones , Receptores Opioides mu/genética , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Acetilación , Analgesia , Animales , Ganglios Espinales/metabolismo , Histona Desacetilasas/metabolismo , Histona Desacetilasas/fisiología , Histonas/metabolismo , Ácidos Hidroxámicos , Masculino , Ratones Endogámicos C57BL , Receptores Opioides mu/metabolismo
20.
Zhongguo Zhen Jiu ; 35(9): 917-21, 2015 Sep.
Artículo en Chino | MEDLINE | ID: mdl-26721148

RESUMEN

OBJECTIVE: To observe the analgesic effect of electroacupuncture (EA) on collagen-induced arthritis (CIA) rats and its regulating effect on inflammation reaction and the endogenous opioid system of synovial tissues. Methods A total of 30 healthy male Wistar rats were randomly divided into a control group, a model group and an EA group, 10 rats in each one. The chronic pain model of CIA rats was made by cattle type-II collagen in the model group and EA group. Rats in the EA group were treated with EA at "Zusanli" (ST 36) and "Kunlun" (BL 60) for 30 min from 16th day after model establishment, once a day for consecutive 10 days. Rats in the control group did not receive any treatment. Rats in the model group were treated with fixation as the EA group. Threshold of pain, arthritis index, paw swelling were measured before model establishment and 16 d, 20 d, 23 d and 25 d after model establishment. The levels of beta-endorphin (ß-END), met-enkephalin (met-ENK), dynorphin A (Dyn A) were measured by radioimmunoassay; the mRNA expressions of mu opioid receptor (MOR), kappa opioid receptor (KOR) and delta opioid receptor (DOR) in synovial tissues of CIA rats were detected by I quantitative polymerase chain reaction (qPCR). RESULTS: Compared with the control group, threshold of pain was reduced (all P<0. 01), arthritis index was increased (all P<0. 01) and paw swelling was increased (all P<0. 01) in the model group on the 16th day, 20th day, 23rd day, 25th day after model establishment. Compared with the model group, the threshold of pain was increased in the EA group (all P<0. 01), arthritis index and paw swelling were reduced (all P<0. 01) on the 23rd day and 25th day after model establishment. Compared with the control group, the level of Dyn A in synovial tissues of CIA rats was increased in the model group (P<0. 01); the mRNA expressions of MOR, KOR and DOR were down-regulated lower than 0. 5 fold of normal level. Compared with the model group, the level of ß-END in synovial tissues of the knee joint was increased in the EA group (P<0. 05), and the mRNA expressions of MOR, KOR and DOR in synovial tissues of CIA rats were up-regulated more than 2 folds of normal level. CONCLUSION: The intervention of EA on chronic pain of CIA rats is superior, which is likely to be related with effects of EA on anti-inflammation and up-regulation of synovial tissue ß-END and MOR, KOR, DOR.


Asunto(s)
Analgésicos Opioides/inmunología , Artritis Reumatoide/terapia , Dolor Crónico/terapia , Electroacupuntura , Líquido Sinovial/inmunología , Analgesia por Acupuntura , Puntos de Acupuntura , Animales , Artritis Reumatoide/inmunología , Bovinos , Dolor Crónico/inmunología , Dinorfinas/genética , Dinorfinas/inmunología , Encefalina Metionina/genética , Encefalina Metionina/inmunología , Humanos , Masculino , Ratas , Ratas Wistar , Receptores Opioides mu/genética , Receptores Opioides mu/inmunología , betaendorfina/genética , betaendorfina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA