Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Chem Commun (Camb) ; 57(81): 10560-10563, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34557886

RESUMEN

The mu-opioid receptor (MOR) regulates the neuronal pathways involved in pain, reward, and respiration. To increase our understanding of MOR's roles in these pathways, there is a need to detect opioids at cellular resolution. Here, we engineered an improved opioid-sensor, called M-SPOTIT2, which is 11x brighter than our previously engineered M-SPOTIT1.1. We engineered M-SPOTIT2 by adding the amino acids YNSH, located near the fluorophore of the enhanced green fluorescent protein, to the circular permuted green fluorescent protein in M-SPOTIT2. M-SPOTIT2 is 11x brighter than our previously engineered M-SPOTIT1.1 in HEK293T cell culture and 2.7x brighter in neuronal culture. M-SPOTIT2 will potentially be useful for the detection of opioids in cell culture for drug screening and the detection of opioids at cellular resolution in animal tissues. By using M-SPOTIT2, researchers can gain more understanding about the mechanisms of addiction, respiratory suppression, and pain-modulation involved in opioid signaling.


Asunto(s)
Fluorescencia , Proteínas Fluorescentes Verdes/genética , Receptores Opioides mu/análisis , Células Cultivadas , Células HEK293 , Humanos , Modelos Moleculares
2.
JCI Insight ; 6(10)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33848266

RESUMEN

Bariatric surgery is the most effective method for weight loss in morbid obesity. There is significant individual variability in the weight loss outcomes, yet factors leading to postoperative weight loss or weight regain remain elusive. Alterations in the µ-opioid receptor (MOR) and dopamine D2 receptor (D2R) systems are associated with obesity and appetite control, and the magnitude of initial brain receptor system perturbation may predict long-term surgical weight loss outcomes. We tested this hypothesis by studying 19 morbidly obese women (mean BMI 40) scheduled to undergo bariatric surgery. We measured their preoperative MOR and D2R availabilities using positron emission tomography with [11C]carfentanil and [11C]raclopride, respectively, and then assessed their weight development association with regional MOR and D2R availabilities at 24-month follow-up. MOR availability in the amygdala consistently predicted weight development throughout the follow-up period, but no associations were found for D2R. This is the first study to our knowledge to demonstrate that neuroreceptor markers prior to bariatric surgery are associated with postoperative weight development. Postoperative weight regain may derive from dysfunction in the opioid system, and weight loss outcomes after bariatric surgery may be partially predicted based on preoperative brain receptor availability, opening up new potential for treatment possibilities.


Asunto(s)
Encéfalo , Obesidad Mórbida , Receptores Opioides mu , Aumento de Peso/fisiología , Adulto , Cirugía Bariátrica , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Neuroimagen , Obesidad Mórbida/diagnóstico por imagen , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Tomografía de Emisión de Positrones , Periodo Preoperatorio , Receptores de Dopamina D2/análisis , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/análisis , Receptores Opioides mu/metabolismo , Pérdida de Peso/fisiología
3.
Cell Mol Life Sci ; 78(2): 415-426, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32671427

RESUMEN

µ-opioid receptor (MOR) is a class of opioid receptors that is critical for analgesia, reward, and euphoria. MOR is distributed in various brain regions, including the hippocampus, where traditionally, it is believed to be localized mainly at the presynaptic terminals of the GABAergic inhibitory interneurons to exert a strong disinhibitory effect on excitatory pyramidal neurons. However, recent intensive research has uncovered the existence of MOR in hippocampal astrocytes, shedding light on how astrocytic MOR participates in opioid signaling via glia-neuron interaction in the hippocampus. Activation of astrocytic MOR has shown to cause glutamate release from hippocampal astrocytes and increase the excitability of presynaptic axon fibers to enhance the release of glutamate at the Schaffer Collateral-CA1 synapses, thereby, intensifying the synaptic strength and plasticity. This novel mechanism involving astrocytic MOR has been shown to participate in hippocampus-dependent conditioned place preference. Furthermore, the signaling of hippocampal MOR, whose action is sexually dimorphic, is engaged in adult neurogenesis, seizure, and stress-induced memory impairment. In this review, we focus on the two profoundly different hippocampal opioid signaling pathways through either GABAergic interneuronal or astrocytic MOR. We further compare and contrast their molecular and cellular mechanisms and their possible roles in opioid-associated conditioned place preference and other hippocampus-dependent behaviors.


Asunto(s)
Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/fisiología , Interneuronas/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal , Animales , Astrocitos/citología , Ácido Glutámico/análisis , Hipocampo/citología , Humanos , Interneuronas/citología , Receptores Opioides mu/análisis , Transmisión Sináptica
4.
J Nat Prod ; 84(1): 71-80, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33326237

RESUMEN

The seeds of the akuamma tree (Picralima nitida) have been used as a traditional treatment for pain and fever. Previous studies have attributed these effects to a series of indole alkaloids found within the seed extracts; however, these pharmacological studies were significantly limited in scope. Herein, an isolation protocol employing pH-zone-refining countercurrent chromatography was developed to provide six of the akuamma alkaloids in high purity and quantities sufficient for more extensive biological evaluation. Five of these alkaloids, akuammine (1), pseudo-akuammigine (3), akuammicine (4), akuammiline (5), and picraline (6), were evaluated against a panel of >40 central nervous system receptors to identify that their primary targets are the opioid receptors. Detailed in vitro investigations revealed 4 to be a potent kappa opioid receptor agonist, and three alkaloids (1-3) were shown to have micromolar activity at the mu opioid receptor. The mu opioid receptor agonists were further evaluated for analgesic properties but demonstrated limited efficacy in assays of thermal nociception. These findings contradict previous reports of the antinociceptive properties of the P. nitida alkaloids and the traditional use of akuamma seeds as analgesics. Nevertheless, their opioid-preferring activity does suggest the akuamma alkaloids provide distinct scaffolds from which novel opioids with unique pharmacologic properties and therapeutic utility can be developed.


Asunto(s)
Alcaloides/farmacología , Analgésicos/uso terapéutico , Apocynaceae/química , Indoles/farmacología , Receptores Opioides mu/uso terapéutico , Terpenos/farmacología , Alcaloides/química , Alcaloides/aislamiento & purificación , Analgésicos/química , Animales , Indoles/química , Indoles/aislamiento & purificación , Receptores Opioides kappa , Receptores Opioides mu/agonistas , Receptores Opioides mu/análisis , Alcaloides de Triptamina Secologanina/química , Semillas/química , Terpenos/química , Terpenos/aislamiento & purificación
5.
Neurotoxicology ; 82: 119-129, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33248188

RESUMEN

Childhood lead (Pb2+) intoxication is a global public health problem best known for producing deficits in learning and poor school performance. Human and preclinical studies have suggested an association between childhood Pb2+ intoxication and proclivity to substance abuse and delinquent behavior. While environmental factors have been implicated in opioid addiction, less is known about the role of exposure to environmental pollutants on the brain opioid system. Opioid receptors are involved in the biological effects of opioids and other drugs of abuse. In this study, we examine the effect of chronic developmental Pb2+ exposure (1500 ppm in the diet) on µ-opioid receptor (MOR) levels in the rat brain using [3H]-d-Ala2-MePhe4-Gly-ol5 enkephalin ([3H]-DAMGO) quantitative receptor autoradiography at different developmental stages (juvenile, early-adolescent, late adolescent and adult) in male and female rats. Our results indicate that chronic developmental Pb2+ exposure increases the levels of [3H]-DAMGO specific binding to MOR in juvenile and early adolescent Pb2+-exposed male and female rat brain with no changes in late-adolescent (PN50) and minor changes in Pb2+-exposed adult male rats (PN120). Specifically, at PN14, Pb2+-exposed males had an increase in MOR binding in the lateral posthalamic nuclei (LPTN), and Pb2+-exposed females had increased MOR binding in LPTN, medial thalamus, and hypothalamus. At PN28, Pb2+-exposed males had increased MOR levels in the striatum, stria medullaris of the thalamus, LPTN, medial thalamus, and basolateral amygdala, while Pb2+-exposed females showed an increase in nucleus accumbens core, LPTN, and medial thalamus. No changes were detected in any brain region of male and female rats at PN50, and at PN120 there was a decrease in MOR binding of Pb2+-exposed males in the medial thalamus. Our findings demonstrate age and gender specific effects of MOR levels in the rat brain as a result of chronic developmental Pb2+ exposure. These results indicate that the major changes in brain MOR levels were during pre-adolescence and early adolescence, a developmental period in which there is higher engagement in reward and drug-seeking behaviors in humans. In summary, we show that chronic exposure to Pb2+, an ubiquitous and well-known environmental contaminant and neurotoxicant, alters MOR levels in brain regions associated with addiction circuits in the adolescent period, these findings have important implications for opioid drug use and abuse.


Asunto(s)
Química Encefálica/efectos de los fármacos , Intoxicación del Sistema Nervioso por Plomo/metabolismo , Receptores Opioides mu/análisis , Animales , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Femenino , Plomo/sangre , Intoxicación del Sistema Nervioso por Plomo/complicaciones , Masculino , Ratas/crecimiento & desarrollo , Ratas Long-Evans , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo
6.
JAMA Psychiatry ; 77(10): 1072-1080, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32492095

RESUMEN

Importance: With the current opioid crisis, it is important to improve understanding of the biological mechanisms of opioid use disorder (OUD). Objectives: To detect genetic risk variants for OUD and determine genetic correlations and causal association with OUD and other traits. Design, Setting, and Participants: A genome-wide association study of electronic health record-defined OUD in the Million Veteran Program sample was conducted, comprising 8529 affected European American individuals and 71 200 opioid-exposed European American controls (defined by electronic health record trajectory analysis) and 4032 affected African American individuals and 26 029 opioid-exposed African American controls. Participants were enrolled from January 10, 2011, to May 21, 2018, with electronic health record data for OUD diagnosis from October 1, 1999, to February 7, 2018. Million Veteran Program results and additional OUD case-control genome-wide association study results from the Yale-Penn and Study of Addiction: Genetics and Environment samples were meta-analyzed (total numbers: European American individuals, 10 544 OUD cases and 72 163 opioid-exposed controls; African American individuals, 5212 cases and 26 876 controls). Data on Yale-Penn participants were collected from February 14, 1999, to April 1, 2017, and data on Study of Addiction: Genetics and Environment participants were collected from 1990 to 2007. The key result was replicated in 2 independent cohorts: proxy-phenotype buprenorphine treatment in the UK Biobank and newly genotyped Yale-Penn participants. Genetic correlations between OUD and other traits were tested, and mendelian randomization analysis was conducted to identify potential causal associations. Main Outcomes and Measures: Main outcomes were International Classification of Diseases, Ninth Revision-diagnosed OUD or International Statistical Classification of Diseases and Related Health Problems, Tenth Revision-diagnosed OUD (Million Veteran Program), and DSM-IV-defined opioid dependence (Yale-Penn and Study of Addiction: Genetics and Environment). Results: A total of 114 759 individuals (101 016 men [88%]; mean [SD] age, 60.1 [12.8] years) were included. In 82 707 European American individuals, a functional coding variant (rs1799971, encoding Asn40Asp) in OPRM1 (µ-opioid receptor gene, the main biological target for opioid drugs; OMIM 600018) reached genome-wide significance (G allele: ß = -0.066 [SE = 0.012]; P = 1.51 × 10-8). The finding was replicated in 2 independent samples. Single-nucleotide polymorphism-based heritability of OUD was 11.3% (SE = 1.8%). Opioid use disorder was genetically correlated with 83 traits, including multiple substance use traits, psychiatric illnesses, cognitive performance, and others. Mendelian randomization analysis revealed the following associations with OUD: risk of tobacco smoking, depression, neuroticism, worry neuroticism subcluster, and cognitive performance. No genome-wide significant association was detected for African American individuals or in transpopulation meta-analysis. Conclusions and Relevance: This genome-wide meta-analysis identified a significant association of OUD with an OPRM1 variant, which was replicated in 2 independent samples. Post-genome-wide association study analysis revealed associated pleiotropic characteristics. Recruitment of additional individuals with OUD for future studies-especially those of non-European ancestry-is a crucial next step in identifying additional significant risk loci.


Asunto(s)
Trastornos Relacionados con Opioides/genética , Receptores Opioides mu/análisis , Anciano , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Persona de Mediana Edad , Trastornos Relacionados con Opioides/epidemiología , Receptores Opioides mu/sangre , Estados Unidos , United States Department of Veterans Affairs/organización & administración , United States Department of Veterans Affairs/estadística & datos numéricos
7.
Neurogastroenterol Motil ; 31(11): e13688, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31336406

RESUMEN

BACKGROUND AND AIMS: The gut immune, cannabinoid, and opioid systems constitute an integrated network contributing to visceral sensation and pain modulation. We aimed to assess the expression of the µ-opioid receptor (MOR), its ligand ß-endorphin (ß-END), and cannabinoid receptor-2 (CB2 ) in patients with irritable bowel syndrome (IBS) and asymptomatic controls (AC) and their correlation with sex and symptom perception. METHODS: Mucosal biopsies were obtained from the left colon of 31 IBS patients (45% women) with predominant constipation (IBS-C, 9) or diarrhea (IBS-D, 10) or with mixed bowel habits (IBS-M, 12) and 32 AC (44% women) and processed for qRT-PCR, Western blotting, and immunohistochemistry. KEY RESULTS: µ-opioid receptor and CB2 mRNA and protein expression and ß-END protein levels were increased in patients with IBS compared to AC (all Ps=0.021). A significant sex by IBS interaction was found in relation to CB2 mRNA expression (P = .003) with women showing a markedly higher expression to men (P = .035). In contrast, in AC, men had higher expression than women (P = .033). ß-END, MOR, and CB2 immunoreactivities (IR) were localized to CD4+T cells including EMR-1+ eosinophils and CD31+ T cells but not to mast cells. CONCLUSIONS: The increased expression of MOR, ß-END, and CB2 in the mucosa of IBS patients, where they are localized to immune cells, suggests that opioid and cannabinoid systems play an immune-related compensatory role in visceral pain in IBS patients. Further work is necessary to support this hypothesis.


Asunto(s)
Mucosa Intestinal/metabolismo , Síndrome del Colon Irritable/metabolismo , Receptor Cannabinoide CB2/biosíntesis , Receptores Opioides mu/biosíntesis , betaendorfina/biosíntesis , Femenino , Humanos , Masculino , Receptor Cannabinoide CB2/análisis , Receptores Opioides mu/análisis , Caracteres Sexuales , betaendorfina/análisis
8.
Elife ; 72018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29932421

RESUMEN

The intrinsic efficacy of orthosteric ligands acting at G-protein-coupled receptors (GPCRs) reflects their ability to stabilize active receptor states (R*) and is a major determinant of their physiological effects. Here, we present a direct way to quantify the efficacy of ligands by measuring the binding of a R*-specific biosensor to purified receptor employing interferometry. As an example, we use the mu-opioid receptor (µ-OR), a prototypic class A GPCR, and its active state sensor, nanobody-39 (Nb39). We demonstrate that ligands vary in their ability to recruit Nb39 to µ-OR and describe methadone, loperamide, and PZM21 as ligands that support unique R* conformation(s) of µ-OR. We further show that positive allosteric modulators of µ-OR promote formation of R* in addition to enhancing promotion by orthosteric agonists. Finally, we demonstrate that the technique can be utilized with heterotrimeric G protein. The method is cell-free, signal transduction-independent and is generally applicable to GPCRs.


Asunto(s)
Técnicas Biosensibles , Interferometría/métodos , Receptores Opioides mu/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación Alostérica , Sitio Alostérico , Biotina/química , Humanos , Ligandos , Loperamida/metabolismo , Loperamida/farmacología , Metadona/metabolismo , Metadona/farmacología , Unión Proteica , Receptores Opioides mu/metabolismo , Sensibilidad y Especificidad , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Soluciones , Estreptavidina/química , Tiofenos/metabolismo , Tiofenos/farmacología , Urea/análogos & derivados , Urea/metabolismo , Urea/farmacología
9.
Anesthesiology ; 128(5): 967-983, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29334500

RESUMEN

BACKGROUND: The current study used recombinant herpes simplex virus type I to increase expression of µ-opiate receptors and the opioid ligand preproenkephalin in peripheral nerve fibers in a mouse model of neuropathic pain. It was predicted that viral vector delivery of a combination of genes encoding the µ-opioid receptor and preproenkephalin would attenuate neuropathic pain and enhance opioid analgesia. The behavioral effects would be paralleled by changes in response properties of primary afferent neurons. METHODS: Recombinant herpes simplex virus type 1 containing cDNA sequences of the µ-opioid receptor, human preproenkephalin, a combination, or Escherichia coli lacZ gene marker (as a control) was used to investigate the role of peripheral opioids in neuropathic pain behaviors. RESULTS: Inoculation with the µ-opioid receptor viral vector (n = 13) reversed mechanical allodynia and thermal hyperalgesia and produced leftward shifts in loperamide (ED50 = 0.6 ± 0.2 mg/kg vs. ED50 = 0.9 ± 0.2 mg/kg for control group, n = 8, means ± SD) and morphine dose-response curves (ED50 = 0.3 ± 0.5 mg/kg vs. ED50 = 1.1 ± 0.1 mg/kg for control group). In µ-opioid receptor viral vector inoculated C-fibers, heat-evoked responses (n = 12) and ongoing spontaneous activity (n = 18) were decreased after morphine application. Inoculation with both µ-opioid receptor and preproenkephalin viral vectors did not alter mechanical and thermal responses. CONCLUSIONS: Increasing primary afferent expression of opioid receptors can decrease neuropathic pain-associated behaviors and increase systemic opioid analgesia through inhibition of peripheral afferent fiber activity.


Asunto(s)
Analgésicos Opioides/farmacología , Encefalinas/fisiología , Neuralgia/prevención & control , Neuronas Aferentes/fisiología , Receptores Opioides mu/fisiología , Analgesia , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Herpesvirus Humano 1/genética , Masculino , Ratones , Morfina/farmacología , Proteínas Proto-Oncogénicas c-fos/análisis , Receptores Opioides mu/análisis
10.
Braz. J. Pharm. Sci. (Online) ; 54(1): e17217, 2018. tab
Artículo en Inglés | LILACS | ID: biblio-951905

RESUMEN

ABSTRACT O sistema opioidérgico envolve a regulação do sono e da vigília. É possível, portanto, que os polimorfismos genéticos no OPRM1 influenciem na qualidade do sono. Este estudo investigou a associação de polimorfismos do OPRM1 com a qualidade subjetiva do sono entre indivíduos sem tratamento prévio com opióides. Este estudo observacional de corte transversal envolveu 161 homens que nunca haviam se tornado opióides (média de idade = 27,74 anos; variação: 18 a 63 anos). A qualidade subjetiva do sono foi avaliada com a versão traduzida e validada em malaio do Índice de Qualidade do Sono de Pittsburgh (PSQI). O DNA foi extraído do sangue total e submetido à reação em cadeia da polimerase (PCR) para dois polimorfismos OPRM1 (118A> G e IVS2 + 691G> C). Sujeitos combinados com 118A e IVS2 + 691Galelos (haplótipo AC) apresentaram escores significativamente mais baixos do PSQI [média (DP) = 4,29 (1,76)] em comparação àqueles sem o haplótipo [4,99 (2,50)] (p = 0,004). Por outro lado, os indivíduos com genótipo heterozigótico combinado (GC / AG diplotipo) apresentaram escores significativamente mais altos do PSQI em comparação àqueles sem o diplótipo [6,04 (2,48) vs 4,54 (2,22), p = 0,004]. Em indivíduos sem tratamento prévio com opiáceos, o haplótipo AC e o diplótipo GC / AG para os polimorfismos 118A> G e IVS2 + 691G> C do OPRM1 estão associados a uma melhor e pior qualidade do sono, respectivamente.


Asunto(s)
Humanos , Masculino , Adolescente , Adulto , Persona de Mediana Edad , Sueño/genética , Trastornos del Sueño-Vigilia , Receptores Opioides mu/análisis , Polimorfismo Genético/genética , Receptores Opioides/análisis
11.
J Surg Res ; 219: 214-221, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29078884

RESUMEN

BACKGROUND: The opioid epidemic is a growing concern, and emerging evidence suggests that morphine use may be associated with sepsis. Enteric glial cells (EGCs) are the most numerous cell type in the enteric nervous system and regulate gastrointestinal function through the production of trophic factors, including glial-derived neurotrophic factor (GDNF). We sought to determine the effect of morphine on enteric glia and hypothesized that morphine contributes to EGC dysfunction and increased gut permeability. MATERIALS AND METHODS: Rat intestinal epithelial cells (IECs) and EGC lines were purchased from ATCC. Immunocytochemistry was used to evaluate the impact of EGCs on IEC barrier proteins and detect the µ-opioid receptor. Co-culture assays were used to determine the effect of EGCs, GDNF, and morphine on barrier integrity. Quantitative polymerase chain reaction and western blotting were performed to determine the impact of morphine in GDNF production. Transepithelial resistance of IEC-6 cell monolayers was measured in the presence of EGC-conditioned media (EGC-CM) and morphine treated EGC-CM using electrical cell impedance sensing. RESULTS: EGC-CM enhanced tight junction organization in IECs. IEC barrier integrity was enhanced when co-cultured with unstimulated EGCs or with GDNF alone; this barrier protective effect was lost with morphine-treated EGCs. GDNF RNA and protein expression were decreased by morphine treatment. Transepithelial resistance was decreased in IEC confluent monolayers when exposed to morphine-treated EGC-CM compared with control. CONCLUSIONS: Morphine compromises intestinal epithelial cell barrier function through a mechanism which appears to involve GDNF. Further studies are warranted to delineate the role of enteric glial cell function in opioid signaling and sepsis.


Asunto(s)
Analgésicos Opioides/efectos adversos , Mucosa Intestinal/efectos de los fármacos , Morfina/efectos adversos , Neuroglía/efectos de los fármacos , Animales , Línea Celular , Factores Neurotróficos Derivados de la Línea Celular Glial/metabolismo , Neuroglía/química , Neuroglía/metabolismo , Ratas , Receptores Opioides mu/análisis
12.
Mol Neurobiol ; 54(1): 461-469, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26742523

RESUMEN

Opiates are commonly used analgesics that often cause clinical respiratory depression. However, their underlying mechanisms remain unclear. Endomorphin-2 (EM2) is a novel, endogenous tetrapeptide opioid with very high affinity and selectivity for the µ-opioid receptor (MOR). The pre-Bötzinger complex (pre-BötC) is considered the center of respiratory rhythm generation, and the synaptic connections in this region are essential for respiratory rhythm. The present study identified EM2-like immunoreactive (LI) axonal terminals in the pre-BötC of adult rats. Some EM2-LI axonal terminals made principally symmetric synapses with neurokinin 1 receptor (NK1R)-LI or MOR-LI neuronal dendritic processes in the pre-BötC. Unilateral microinjection of EM2 into the pre-BötC decreased breathing frequency and amplitude. A prior microinjection of the selective MOR antagonist ß-funaltrexamine (ß-FNA) into the pre-BötC prevented the effects of EM2. The present results suggest that EM2-LI axonal terminals modulate NK1R-expressing neurons in the pre-BötC and that EM2 plays a role in respiratory depression through MORs in the pre-BötC.


Asunto(s)
Tronco Encefálico/metabolismo , Oligopéptidos/metabolismo , Receptores Opioides mu/metabolismo , Mecánica Respiratoria/fisiología , Animales , Tronco Encefálico/química , Tronco Encefálico/efectos de los fármacos , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Oligopéptidos/análisis , Oligopéptidos/farmacología , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/análisis , Receptores Opioides mu/antagonistas & inhibidores , Mecánica Respiratoria/efectos de los fármacos
13.
Acta Derm Venereol ; 97(5): 564-570, 2017 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-27958613

RESUMEN

Psoriasis is an inflammatory immunogenetic skin disease, often accompanied by itch. Opioid receptors are known regulators of itch sensation in the central nervous system. In the brain, µ-opioid receptors may potentiate itch, while activation of κ-opioid receptors may reduce or even alleviate itch; however, the role of opioid receptors in itch perception in the skin is poorly understood. To further elucidate the role of opioid receptors in the neurobiology of psoriatic itch, punch biopsies of non-lesional and lesional skin of patients with psoriasis and healthy controls were studied. Real-time polymerase chain reaction and immunofluorescence microscopy were used to detect opioid receptor genes and protein expression, respectively. The OPRK1/κ-opioid receptor pathway was found to be downregulated in lesional skin of psoriasis, correlating positively with itch sensation. In contrast, the OPRM1/µ-opioid receptor system was uniformly expressed by epidermal keratinocytes in all analysed groups. These findings suggest that imbalance of epidermal opioid receptors may result in disordered neuroepidermal homeostasis in psoriasis, which could potentiate transmission of itch.


Asunto(s)
Epidermis/química , Prurito/metabolismo , Psoriasis/metabolismo , Receptores Opioides kappa/análisis , Receptores Opioides mu/análisis , Adulto , Anciano , Biopsia , Estudios de Casos y Controles , Epidermis/patología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Queratinocitos/química , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Prurito/genética , Prurito/patología , Psoriasis/genética , Psoriasis/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Umbral Sensorial , Transducción de Señal , Adulto Joven
14.
PLoS One ; 11(3): e0152162, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27031701

RESUMEN

The presence of endogenous opioid peptides in different testicular cell types has been extensively characterized and provides evidence for the participation of the opioid system in the regulation of testicular function. However, the exact role of the opioid system during the spermatogenesis has remained controversial since the presence of the mu-, delta- and kappa-opioid receptors in spermatogenic cells was yet to be demonstrated. Through a combination of quantitative real-time PCR, immunofluorescence, immunohistochemistry and flow cytometry approaches, we report for the first time the presence of active mu-, delta- and kappa-opioid receptors in mouse male germ cells. They show an exposition time-dependent response to opioid agonist, hence suggesting their active involvement in spermatogenesis. Our results contribute to understanding the role of the opioid receptors in the spermatogenesis and could help to develop new strategies to employ the opioid system as a biochemical tool for the diagnosis and treatment of male infertility.


Asunto(s)
Receptores Opioides delta/análisis , Receptores Opioides kappa/análisis , Receptores Opioides mu/análisis , Espermatogénesis , Espermatozoides/citología , Testículo/citología , Animales , Células Cultivadas , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo
15.
Reprod Biomed Online ; 32(3): 316-22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26803207

RESUMEN

The objectives of this study were to determine whether the main opioid receptor (OPRM1) is present on human granulosa cells and if exogenous opiates and their antagonists can influence granulosa cell vascular endothelial growth factor (VEGF) production via OPRM1. Granulosa cells were isolated from women undergoing oocyte retrieval for IVF. Complementary to the primary cells, experiments were conducted using COV434, a well-characterized human granulosa cell line. Identification and localization of opiate receptor subtypes was carried out using Western blot and flow cytometry. The effect of opiate antagonist on granulosa cell VEGF secretion was assessed by enzyme-linked immunosorbent assay. For the first time, the presence of OPRM1 on human granulosa cells is reported. Blocking of opiate signalling using naloxone, a specific OPRM1 antagonist, significantly reduced granulosa cell-derived VEGF levels in both COV434 and granulosa-luteal cells (P < 0.01). The presence of opiate receptors and opiate signalling in granulosa cells suggest a possible role in VEGF production. Targeting this signalling pathway could prove promising as a new clinical option in the prevention and treatment of ovarian hyperstimulation syndrome.


Asunto(s)
Células de la Granulosa/metabolismo , Alcaloides Opiáceos/farmacología , Receptores Opioides mu/metabolismo , Western Blotting , Línea Celular , Microambiente Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Naloxona/farmacología , Alcaloides Opiáceos/antagonistas & inhibidores , Receptores Opioides mu/análisis , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Mol Neurobiol ; 53(7): 4918-30, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26363797

RESUMEN

Imidazoline receptor antisera-selected (IRAS) protein, the mouse homologue named Nischarin, was found to target to early endosomes with properties of sorting nexins in vitro. Recently, we generated IRAS knockout mice and found IRAS deficiency exacerbated the analgesic tolerance and physical dependence caused by opioids, suggesting that IRAS plays a role in regulating µ opioid receptor (MOR) functions. In the present study, we found that IRAS interacts with MOR and regulates MOR trafficking in vitro. In the CHO or HEK293 cells co-expressing MOR and IRAS, IRAS, through its PX domain, interacted with MOR. The interaction facilitated the recycling of internalized MOR and prevented MOR downregulation induced by DAMGO, the MOR agonist. Functionally, IRAS accelerated MOR resensitization and attenuated DAMGO-induced MOR desensitization, which is believed as one of mechanisms mediating opioid tolerance and dependence. Taken together, we propose that IRAS is a new MOR interacting protein and regulates agonist-induced trafficking of MOR via sorting internalized MOR to the recycling pathway, which may be a molecular mechanism underlying IRAS modulating opioid tolerance and dependence.


Asunto(s)
Membrana Celular/metabolismo , Tolerancia a Medicamentos/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Animales , Células CHO , Membrana Celular/química , Cricetinae , Cricetulus , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/análisis , Unión Proteica/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Receptores Opioides mu/agonistas , Receptores Opioides mu/análisis
17.
Minerva Anestesiol ; 81(8): 894-900, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25300626

RESUMEN

Breast cancer is the most common type of cancer among women worldwide. Short-term postsurgical recovery is complicated by many factors, including imbalanced inflammatory and immune response, acute pain associated with functional impairment, and chronic postmastectomy pain (CPMP), developed by about 25-60% of patients. Opioids, most common drugs used for treatment of cancer pain, are immunosuppressive, and therefore, they might directly and/or indirectly influence long-term cancer recurrence. Moreover, they also produce endocrinopathy, which consists primarily of hypothalamic-pituitary-gonadal axis or hypothalamic-pituitary-adrenal axis dysfunction. The interindividual variability in both CPMP and opioid response is believed to be largely underlined by genetic variability in the gene locus for µ-opioid receptor (OPRM1) that modulates opioid pharmacodynamics. For this reason, OPRM1 genotype may play a key role both in short-term postmastectomy outcome and in long-term follow-up, becoming a new biomarker for breast cancer recurrence in patients suffering from chronic postmastectomy pain managed by opioid therapy. Hence OPRM1 might be used in near future to customize the opioid therapy, avoiding not only opioid side effects but also the disease progression. In this review we evaluate the literature state of the art on this topic and possible steps towards obtaining the safest individualized postmastectomy analgesic therapy. Therefore, a personalized pain treatment strategy might be useful to both manage pain and control cancer disease progression.


Asunto(s)
Biomarcadores/análisis , Neoplasias de la Mama/cirugía , Mastectomía/efectos adversos , Dolor Postoperatorio/diagnóstico , Receptores Opioides mu/análisis , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Recurrencia Local de Neoplasia , Valor Predictivo de las Pruebas , Receptores Opioides mu/metabolismo
18.
Brain Struct Funct ; 220(2): 677-702, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24623156

RESUMEN

Opioid receptors are G protein-coupled receptors (GPCRs) that modulate brain function at all levels of neural integration, including autonomic, sensory, emotional and cognitive processing. Mu (MOR) and delta (DOR) opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular levels remains unsolved. To challenge the hypothesis of MOR/DOR heteromerization in the brain, we generated redMOR/greenDOR double knock-in mice and report dual receptor mapping throughout the nervous system. Data are organized as an interactive database offering an opioid receptor atlas with concomitant MOR/DOR visualization at subcellular resolution, accessible online. We also provide co-immunoprecipitation-based evidence for receptor heteromerization in these mice. In the forebrain, MOR and DOR are mainly detected in separate neurons, suggesting system-level interactions in high-order processing. In contrast, neuronal co-localization is detected in subcortical networks essential for survival involved in eating and sexual behaviors or perception and response to aversive stimuli. In addition, potential MOR/DOR intracellular interactions within the nociceptive pathway offer novel therapeutic perspectives.


Asunto(s)
Encéfalo/metabolismo , Red Nerviosa/metabolismo , Neuronas/metabolismo , Receptores Opioides delta/análisis , Receptores Opioides mu/análisis , Animales , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL
19.
Ginekol Pol ; 85(10): 730-7, 2014 Oct.
Artículo en Polaco | MEDLINE | ID: mdl-25546922

RESUMEN

UNLABELLED: Fetal brain is considered to be the major body organ, critical for the future quality of human life. Offspring exposed to prenatal hypoxia has been evidenced to experience behavioral abnormalities as a result of the injury sustained by neuronal cells in the brain. The relatively early appearance of opioid receptors proved susceptible to endogenous and exogenous factors. Increased concentrations of neurotransmitters in the maternal circulation and amniotic fluid induced by hypoxic exposure imply their role in the regulation of cellular division and differentiation processes. Endogenous neuropeptides and specific opioid receptors are distributed in those brain structures that are associated with behavior and reproduction. Fetuses exposed to the adverse effects of increased opioid level incur structural brain tissue abnormalities. OBJECTIVES: The present study seeks to determine the effects of long-term hypoxic exposure during gestation on the expression of opioid receptors in specific brain regions in both sexes. MATERIAL AND METHODS: The study was conducted on pregnant Sprague-Dawley rats, (120 days old, body weight between 250 and 300 g). Experiments were carried out in order to determine the effect of long-term hypoxia on µ-opioid receptor density in selected structures of fetal central nervous system: caudate-putamen (CPu), zona germinata (ZG), nucleus accumbens (NA), olfactory tubercle (OT), Median Part Medial Preoptic Area (MMPoA) and Lateral Part Medial Preoptic Area (LMPoA). Pregnant female rats were assigned to two research groups: the control group (N=6) and the experimental group subject to prolonged hypoxia for 24 hours from the gestational day 15 to gestational day 20 (E-15-E20). At E-21 rats were sacrificed, their fetuses were removed and their brains were incubated with radioligands. The µ-opioid receptor incubation in selected brain structures was performed with a specific radioisotope [3H]DAMGO [tyrosyl-3,5,-3H(N)-D-Ala-Gly-N-methyl-Phe-Gly-enkephalin]. Optical density of µ-opioid receptors was determined at E-21 of gestation during long-term exposure to chronic hypoxia induced from E-15 to E-21 of gestation. Experimental model coupled with an innovative autoradiography allowed for a precise assessment of the lesions sustained by fetal brain tissues due to hypoxia and the adaptive mechanisms of the central nervous system in reaction to hypoxic exposure. RESULTS: Statistically significant chronic hypoxia (p<0.05) downregulated the values of µ-opioid receptors optical density in relation to control group in CPu and ZG. Chronic hypoxia in ZG substantially reduces the values of µ-opioid receptors optical density in males (p<0.05). The differences among remaining groups did not show to be statistically significant. CONCLUSIONS: The obtained results of µ-opioid receptor expression can be detected in specific fetal brain regions that mediate sexual behavior and may be attributable to behavioral changes of experimental animals due to hypoxic exposure during gestation.


Asunto(s)
Modelos Animales de Enfermedad , Hipoxia Fetal/metabolismo , Hipoxia Encefálica/metabolismo , Preñez/fisiología , Receptores Opioides mu/análisis , Animales , Enfermedad Crónica , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
20.
J Vis Exp ; (88)2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24962460

RESUMEN

A growing body of research, generated primarily from MRI-based studies, shows that migraine appears to occur, and possibly endure, due to the alteration of specific neural processes in the central nervous system. However, information is lacking on the molecular impact of these changes, especially on the endogenous opioid system during migraine headaches, and neuronavigation through these changes has never been done. This study aimed to investigate, using a novel 3D immersive and interactive neuronavigation (3D-IIN) approach, the endogenous µ-opioid transmission in the brain during a migraine headache attack in vivo. This is arguably one of the most central neuromechanisms associated with pain regulation, affecting multiple elements of the pain experience and analgesia. A 36 year-old female, who has been suffering with migraine for 10 years, was scanned in the typical headache (ictal) and nonheadache (interictal) migraine phases using Positron Emission Tomography (PET) with the selective radiotracer [(11)C]carfentanil, which allowed us to measure µ-opioid receptor availability in the brain (non-displaceable binding potential - µOR BPND). The short-life radiotracer was produced by a cyclotron and chemical synthesis apparatus on campus located in close proximity to the imaging facility. Both PET scans, interictal and ictal, were scheduled during separate mid-late follicular phases of the patient's menstrual cycle. During the ictal PET session her spontaneous headache attack reached severe intensity levels; progressing to nausea and vomiting at the end of the scan session. There were reductions in µOR BPND in the pain-modulatory regions of the endogenous µ-opioid system during the ictal phase, including the cingulate cortex, nucleus accumbens (NAcc), thalamus (Thal), and periaqueductal gray matter (PAG); indicating that µORs were already occupied by endogenous opioids released in response to the ongoing pain. To our knowledge, this is the first time that changes in µOR BPND during a migraine headache attack have been neuronavigated using a novel 3D approach. This method allows for interactive research and educational exploration of a migraine attack in an actual patient's neuroimaging dataset.


Asunto(s)
Encéfalo/metabolismo , Trastornos Migrañosos/metabolismo , Neuronavegación/métodos , Adulto , Encéfalo/patología , Femenino , Humanos , Trastornos Migrañosos/diagnóstico , Trastornos Migrañosos/patología , Receptores Opioides mu/análisis , Receptores Opioides mu/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA