Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Anal Biochem ; 451: 25-7, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24463015

RESUMEN

We have developed a modification of bioorthogonal click chemistry to assay the palmitoylation of cellular proteins. This assay uses 15-hexadecynoic acid (15-HDYA) as a chemical probe in combination with protein immunoprecipitation using magnetic beads in order to detect S-palmitoylation of proteins of interest. Here we demonstrate the utility of this approach for the mu-opioid receptor (MOR), a G-protein-coupled receptor (GPCR) responsible for mediating the analgesic and addictive properties of most clinically relevant opioid agonist drugs. This technique provides a rapid, non-isotopic, and efficient method to assay the palmitoylation status of a variety of cellular proteins, including most GPCRs.


Asunto(s)
Ácidos Grasos Insaturados/química , Immunoblotting , Receptores Opioides mu/análisis , Anticuerpos/inmunología , Química Clic , Células HEK293 , Humanos , Separación Inmunomagnética , Inmunoprecipitación , Lipoilación , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación
8.
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
9.
Cancer ; 119(23): 4103-10, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24104703

RESUMEN

BACKGROUND: Preclinical studies show that opioids stimulate angiogenesis and tumor progression through the mu opioid receptor (MOR). Although MOR is overexpressed in several human malignancies, the effect of chronic opioid requirement on cancer progression or survival has not been examined in humans. METHODS: We performed a retrospective analysis on 113 patients identified in the Minneapolis VA Tumor Registry (test cohort) and 480 patients from the national VA Central Cancer Registry (validation cohort) who had been diagnosed with stage IV prostate cancer between 1995 and 2010 to examine whether MOR expression or opioid requirement is associated with disease progression and survival. All opioids were converted to oral morphine equivalents for comparison. Laser scanning confocal microscopy was used to analyze MOR immunoreactivity in prostate cancer biopsies. The effects of variables on outcomes were analyzed in univariable and multivariable models. RESULTS: In patients with metastatic prostate cancer, MOR expression and opioid requirement were independently associated with inferior progression-free survival (hazard ratio [HR] 1.65, 95% confidence interval [CI] 1.33-2.07, P<.001 and HR 1.08, 95% CI 1.03-1.13, P<.001, respectively) and overall survival (HR 1.55, 95% CI 1.20-1.99, P<.001 and HR 1.05, 95% CI 1.00-1.10, P = .031, respectively). The validation cohort confirmed that increasing opioid requirement was associated with worse overall survival (HR 1.005, 95% CI 1.002-1.008, P = .001). CONCLUSION: Higher MOR expression and greater opioid requirement are associated with shorter progression-free survival and overall survival in patients with metastatic prostate cancer. Nevertheless, clinical practice should not be changed until prospective randomized trials show that opioid use is associated with inferior clinical outcomes, and that abrogation of the peripheral activities of opioids ameliorates this effect.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Neoplasias de la Próstata/mortalidad , Receptores Opioides mu/análisis , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Metástasis de la Neoplasia , Neoplasias de la Próstata/química , Neoplasias de la Próstata/patología , Estudios Retrospectivos , Resultado del Tratamiento
10.
Rheumatol Int ; 33(9): 2291-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23503893

RESUMEN

Activation of mitogen-activated protein kinase (MAPK) enzymes in nociceptive plasticity has been extensively studied. P38 MAPK enzyme, which can be activated by cytokines, acts as a crucial intracellular regulator of environmental changes. The aim of this study was to elucidate the cellular events during arthritis-induced hyperalgesia that are mediated by interleukin-6 and p38 MAPK, and their effects on the expression of spinal mu-opioid receptors (MORs), in different stages of arthritis in male Wistar rats. Complete Freund's adjuvant (CFA)-induced arthritis (AA) was caused by subcutaneous injection of CFA into the rats' hindpaw. Anti-IL-6 antibody and p38 MAPK phosphorylation inhibitor were administered during 21 days of study. Spinal MOR, p38, and phosphorylated-p38 (pp38) proteins expressions were detected by Western blotting. Daily treatment with anti-IL-6 antibody and p38 MAPK phosphorylation inhibitor, SB203580, significantly decreased paw edema in AA group. Daily anti-IL-6 and SB203580 administration caused a significant reduction in hyperalgesia in the first week of the study, but increased hyperalgesia in the next 2 weeks in experimental groups compared to the AA control group. Expression of pp38 MAPK protein significantly decreased on the 3, 7, 14, and 21 days in AA+SB203580 and AA+anti-IL6 groups compared to AA group. Additionally, daily treatment with anti-IL6 antibody and SB203580 in AA group caused significantly decrease in spinal MOR expression compared to AA control group. The results of our study can confirm that activated spinal p38 MAPK enzyme may play an important role in cellular IL-6 signaling pathways in hyperalgesia variation during different stages of AA inflammation. Also, it can be suggested that at least a part of p38 MAPK effects on hyperalgesia is mediated by spinal MOR expression variation.


Asunto(s)
Artritis Experimental/fisiopatología , Hiperalgesia/etiología , Interleucina-6/fisiología , Receptores Opioides mu/fisiología , Médula Espinal/química , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Imidazoles/farmacología , Interleucina-6/sangre , Masculino , Fosforilación , Piridinas/farmacología , Ratas , Ratas Wistar , Receptores Opioides mu/análisis
11.
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
12.
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
13.
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
14.
Mol Pain ; 6: 96, 2010 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-21172011

RESUMEN

BACKGROUND: Mice deficient for the stable tubule only peptide (STOP) display altered dopaminergic neurotransmission associated with severe behavioural defects including disorganized locomotor activity. Endogenous morphine, which is present in nervous tissues and synthesized from dopamine, may contribute to these behavioral alterations since it is thought to play a role in normal and pathological neurotransmission. RESULTS: In this study, we showed that STOP null brain structures, including cortex, hippocampus, cerebellum and spinal cord, contain high endogenous morphine amounts. The presence of elevated levels of morphine was associated with the presence of a higher density of mu opioid receptor with a higher affinity for morphine in STOP null brains. Interestingly, STOP null mice exhibited significantly lower nociceptive thresholds to thermal and mechanical stimulations. They also had abnormal behavioural responses to the administration of exogenous morphine and naloxone. Low dose of morphine (1 mg/kg, i.p.) produced a significant mechanical antinociception in STOP null mice whereas it has no effect on wild-type mice. High concentration of naloxone (1 mg/kg) was pronociceptive for both mice strain, a lower concentration (0.1 mg/kg) was found to increase the mean mechanical nociceptive threshold only in the case of STOP null mice. CONCLUSIONS: Together, our data show that STOP null mice displayed elevated levels of endogenous morphine, as well as an increase of morphine receptor affinity and density in brain. This was correlated with hypernociception and impaired pharmacological sensitivity to mu opioid receptor ligands.


Asunto(s)
Proteínas Asociadas a Microtúbulos/deficiencia , Morfina/farmacología , Dolor/fisiopatología , Analgésicos Opioides , Animales , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/fisiología , Morfina/análisis , Naloxona/administración & dosificación , Naloxona/farmacología , Antagonistas de Narcóticos , Proteínas del Tejido Nervioso , Alcaloides Opiáceos , Dolor/tratamiento farmacológico , Receptores Opioides mu/análisis , Receptores Opioides mu/metabolismo
15.
Connect Tissue Res ; 51(1): 67-70, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20067419

RESUMEN

Endogenous opioid peptides and opioid receptors form a neuromodulatory system, which plays an important part in the control of physiological pathways. In addition, some opioid peptides can function as endogenous messengers of the immune system and participate in the regulation of the immune response. The present studies indicated that mu, delta, and kappa opioid-like receptors were present in the gill and gonad of the scallop Chlamys farreri. Furthermore, the significance of opioid peptides involvement with the immune system is ascertained from the presence of mu, delta, and kappa opioid-like receptors on hemocytes of the scallop. Our report constitutes the first characterization of mu, delta, and kappa opioid-like receptors in the gill and gonad of the scallop Chlamys farreri.


Asunto(s)
Branquias/metabolismo , Gónadas/metabolismo , Hemocitos/metabolismo , Péptidos Opioides/metabolismo , Pectinidae/metabolismo , Receptores Opioides/metabolismo , Animales , Branquias/anatomía & histología , Gónadas/anatomía & histología , Hemocitos/citología , Hemocitos/inmunología , Sistema Inmunológico/fisiología , Inmunidad Celular/fisiología , Inmunohistoquímica/métodos , Pectinidae/anatomía & histología , Pectinidae/inmunología , Receptores Opioides/análisis , Receptores Opioides delta/análisis , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/análisis , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/análisis , Receptores Opioides mu/metabolismo , Reproducción/fisiología , Fenómenos Fisiológicos Respiratorios , Especificidad de la Especie
16.
Med Mycol ; 48(1): 73-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19225979

RESUMEN

Malassezia spp. may act as opportunistic skin pathogens in humans and animals. Malassezia pachydermatis proliferation and phospholipase production may play a pathogenic role in the occurrence of skin lesions in dogs. This study investigates the presence of mu-opioid receptor (MOR) in M. pachydermatis strains isolated from healthy dogs and dogs with skin lesions and its effects on phospholipase activity (p.a.). P.a. of 64 M. pachydermatis isolates was evaluated using different concentrations of naloxone (Nx), a MOR antagonist. Isolates were divided into Group A (i.e., 40 isolates from 26 dogs with dermatitis) and Group B (i.e., 24 isolates from 12 healthy dogs). The MOR expression was analyzed by Western blot and immunofluorescence. A statistically higher p.a. than that of the controls was found with isolates in Group A at a Nx concentration of 10(-6) M (P<0.05). No isolate in Group B displayed p.a. in either control samples or in the presence of any Nx concentration. Immunoblotting revealed two positive MOR immunoreactive bands of approximately 65 and 98 kDa. MOR expression and localization was also demonstrated by immunofluorescence in isolates from Groups A and B. This study provides the first evidence of MOR expression on M. pachydermatis cell membranes pointing to its possible role in modulating p.a. production in isolates from dogs with skin lesions.


Asunto(s)
Dermatomicosis/veterinaria , Enfermedades de los Perros/microbiología , Proteínas Fúngicas/análisis , Regulación Fúngica de la Expresión Génica , Malassezia/enzimología , Fosfolipasas/biosíntesis , Receptores Opioides mu/análisis , Animales , Western Blotting , Dermatomicosis/microbiología , Perros , Técnica del Anticuerpo Fluorescente , Proteínas Fúngicas/química , Proteínas Fúngicas/fisiología , Malassezia/química , Malassezia/aislamiento & purificación , Malassezia/fisiología , Peso Molecular , Naloxona/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/fisiología
17.
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
18.
Science ; 286(5444): 1566-8, 1999 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-10567264

RESUMEN

Neurokinin-1 receptor (NK1R) and mu-opioid receptor (muOR) agonists affected respiratory rhythm when injected directly into the preBötzinger Complex (preBötC), the hypothesized site for respiratory rhythmogenesis in mammals. These effects were mediated by actions on preBötC rhythmogenic neurons. The distribution of NK1R+ neurons anatomically defined the preBötC. Type 1 neurons in the preBötC, which have rhythmogenic properties, expressed both NK1Rs and muORs, whereas type 2 neurons expressed only NK1Rs. These findings suggest that the preBötC is a definable anatomic structure with unique physiological function and that a subpopulation of neurons expressing both NK1Rs and muORs generate respiratory rhythm and modulate respiratory frequency.


Asunto(s)
Bulbo Raquídeo/fisiología , Neuronas/fisiología , Receptores de Neuroquinina-1/fisiología , Receptores Opioides mu/fisiología , Mecánica Respiratoria/fisiología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Femenino , Técnicas In Vitro , Bulbo Raquídeo/citología , Bulbo Raquídeo/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Neuronas/química , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de GABA-B/análisis , Receptores de GABA-B/fisiología , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-1/análisis , Receptores Opioides mu/agonistas , Receptores Opioides mu/análisis , Mecánica Respiratoria/efectos de los fármacos , Sustancia P/farmacología , Transmisión Sináptica/efectos de los fármacos
19.
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
20.
J Neurochem ; 104(4): 937-45, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18005002

RESUMEN

The distribution of the mRNA of different C-terminal splice variants of the mu-opioid receptor in rat CNS was assessed by RT-PCR. The mRNA species for MOR1, MOR1A and MOR1B were readily detectable and distributed widely throughout the rat CNS, with levels of MOR1 and MOR1A mRNA being overall greater than for MOR1B. We did not find convincing evidence that significant levels of MOR1C, MOR1C1, MOR1C2 and MOR1D are present in rat CNS. To examine possible differences in the agonist-induced regulation of MOR1, MOR1A and MOR1B, we expressed these constructs in HEK293 cells along with G-protein-coupled inwardly rectifying K+ channel subunits and measured the rate and extent of desensitisation of (d-Ala2,N-Me-Phe4,glycinol5)-enkephalin (DAMGO)- and morphine-induced G-protein-coupled inwardly rectifying K+ currents. Morphine-induced desensitisation was rapid for all three splice variants (t1/2: 1.2-1.7 min) but DAMGO-induced desensitisation was significantly slower for MOR1B (t1/2 4.2 min). Inhibition of endocytosis by expression of a dynamin-dominant negative mutant increased the rate of DAMGO-induced desensitisation of MOR1B. These data show that some splice variants of mu-opioid receptor are widely expressed in rat CNS but question the existence of others that have been reported in the literature. In addition, whereas the rate of desensitisation of MOR1 and MOR1A is agonist-independent, that for MOR1B is agonist-dependent.


Asunto(s)
Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/fisiología , Isoformas de Proteínas/análisis , Isoformas de Proteínas/fisiología , Receptores Opioides mu/análisis , Receptores Opioides mu/fisiología , Secuencia de Aminoácidos , Analgésicos Opioides/farmacología , Animales , Línea Celular , Sistema Nervioso Central/química , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Humanos , Masculino , Datos de Secuencia Molecular , Fragmentos de Péptidos/agonistas , Isoformas de Proteínas/agonistas , Ratas , Ratas Wistar , Receptores Opioides mu/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA