Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pain ; 160(1): 117-135, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30169422

RESUMEN

The Federal Pain Research Strategy recommended development of nonopioid analgesics as a top priority in its strategic plan to address the significant public health crisis and individual burden of chronic pain faced by >100 million Americans. Motivated by this challenge, a natural product extracts library was screened and identified a plant extract that targets activity of voltage-gated calcium channels. This profile is of interest as a potential treatment for neuropathic pain. The active extract derived from the desert lavender plant native to southwestern United States, when subjected to bioassay-guided fractionation, afforded 3 compounds identified as pentacyclic triterpenoids, betulinic acid (BA), oleanolic acid, and ursolic acid. Betulinic acid inhibited depolarization-evoked calcium influx in dorsal root ganglion (DRG) neurons predominantly through targeting low-voltage-gated (Cav3 or T-type) and CaV2.2 (N-type) calcium channels. Voltage-clamp electrophysiology experiments revealed a reduction of Ca, but not Na, currents in sensory neurons after BA exposure. Betulinic acid inhibited spontaneous excitatory postsynaptic currents and depolarization-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices. Notably, BA did not engage human mu, delta, or kappa opioid receptors. Intrathecal administration of BA reversed mechanical allodynia in rat models of chemotherapy-induced peripheral neuropathy and HIV-associated peripheral sensory neuropathy as well as a mouse model of partial sciatic nerve ligation without effects on locomotion. The broad-spectrum biological and medicinal properties reported, including anti-HIV and anticancer activities of BA and its derivatives, position this plant-derived small molecule natural product as a potential nonopioid therapy for management of chronic pain.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo T/metabolismo , Infecciones por VIH/complicaciones , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Paclitaxel/toxicidad , Triterpenos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/toxicidad , Células CHO , Cricetulus , Diprenorfina/farmacocinética , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/citología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Triterpenos Pentacíclicos , Traumatismos de los Nervios Periféricos/inducido químicamente , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/etiología , Traumatismos de los Nervios Periféricos/virología , Ratas , Ratas Sprague-Dawley , Tritio/farmacocinética , Ácido Betulínico
2.
Neuropharmacology ; 118: 46-58, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28283391

RESUMEN

The development of G protein-biased agonists for the µ-opioid receptor (MOR) offers a clear drug discovery rationale for improved analgesia and reduced side-effects of opiate pharmacotherapy. However, our understanding of the molecular mechanisms governing ligand bias is limited, which hinders our ability to rationally design biased compounds. We have investigated the role of MOR binding site residues W320 and Y328 in controlling bias, by receptor mutagenesis. The pharmacology of a panel of ligands in a cAMP and a ß-arrestin2 assay were compared between the wildtype and mutated receptors, with bias factors calculated by operational analysis using ΔΔlog(τ/KA) values. [3H]diprenorphine competition binding was used to estimate affinity changes. Introducing the mutations W320A and Y328F caused changes in pathway bias, with different patterns of change between ligands. For example, DAMGO increased relative ß-arrestin2 activity at the W320A mutant, whilst its ß-arrestin2 response was completely lost at Y328F. In contrast, endomorphin-1 gained activity with Y328F but lost activity at W320A, in both pathways. For endomorphin-2 there was a directional shift from cAMP bias at the wildtype towards more ß-arrestin2 bias at W320A. We also observe clear uncoupling between mutation-driven changes in function and binding affinity. These findings suggest that the mutations influenced the balance of pathway activation in a ligand-specific manner, thus identifying residues in the MOR binding pocket that govern ligand bias. This increases our understanding of how ligand/receptor binding interactions can be translated into agonist-specific pathway activation.


Asunto(s)
Mutación/genética , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Triptófano/genética , Tirosina/genética , Analgésicos Opioides/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , AMP Cíclico/metabolismo , Diprenorfina/farmacocinética , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutagénesis , Antagonistas de Narcóticos/farmacocinética , Oligopéptidos/farmacología , Receptores Opioides mu/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección , Tritio/farmacocinética , Triptófano/metabolismo , Tirosina/metabolismo , beta-Arrestinas/metabolismo
3.
Neuroimage ; 146: 833-842, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27554530

RESUMEN

Deep brain stimulation (DBS) of the periaqueductal gray (PAG) is used in the treatment of severe refractory neuropathic pain. We tested the hypothesis that DBS releases endogenous opioids to exert its analgesic effect using [11C]diprenorphine (DPN) positron emission tomography (PET). Patients with de-afferentation pain (phantom limb pain or Anaesthesia Dolorosa (n=5)) who obtained long-lasting analgesic benefit from DBS were recruited. [11C]DPN and [15O]water PET scanning was performed in consecutive sessions; first without, and then with PAG stimulation. The regional cerebral tracer distribution and kinetics were quantified for the whole brain and brainstem. Analysis was performed on a voxel-wise basis using statistical parametric mapping (SPM) and also within brainstem regions of interest and correlated to the DBS-induced improvement in pain score and mood. Brain-wide analysis identified a single cluster of reduced [11C]DPN binding (15.5% reduction) in the caudal, dorsal PAG following DBS from effective electrodes located in rostral dorsal/lateral PAG. There was no evidence for an accompanying focal change in blood flow within the PAG. No correlation was found between the change in PAG [11C]DPN binding and the analgesic effect or the effect on mood (POMSSV) of DBS. The analgesic effect of DBS in these subjects was not altered by systemic administration of the opioid antagonist naloxone (400ug). These findings indicate that DBS of the PAG does indeed release endogenous opioid peptides focally within the midbrain of these neuropathic pain patients but we are unable to further resolve the question of whether this release is responsible for the observed analgesic benefit.


Asunto(s)
Estimulación Encefálica Profunda , Neuralgia/prevención & control , Péptidos Opioides/metabolismo , Sustancia Gris Periacueductal/metabolismo , Receptores Opioides/metabolismo , Adulto , Radioisótopos de Carbono , Diprenorfina/farmacocinética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuralgia/metabolismo , Dimensión del Dolor , Tomografía de Emisión de Positrones , Resultado del Tratamiento
4.
Pain ; 156(11): 2267-2275, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26176892

RESUMEN

The experience of pain in humans is modulated by endogenous opioids, but it is largely unknown how the opioid system adapts to chronic pain states. Animal models of chronic pain point to upregulation of opioid receptors (OpR) in the brain, with unknown functional significance. We sought evidence for a similar relationship between chronic pain and OpR availability in humans. Using positron emission tomography and the radiotracer (11)C-diprenorphine, patients with arthritis pain (n = 17) and healthy controls (n = 9) underwent whole-brain positron emission tomography scanning to calculate parametric maps of OpR availability. Consistent with the upregulation hypothesis, within the arthritis group, greater OpR availability was found in the striatum (including the caudate) of patients reporting higher levels of recent chronic pain, as well as regions of interest in the descending opioidergic pathway including the anterior cingulate cortex, thalamus, and periaqueductal gray. The functional significance of striatal changes were clarified with respect to acute pain thresholds: data across patients and controls revealed that striatal OpR availability was related to reduced pain perception. These findings are consistent with the view that chronic pain may upregulate OpR availability to dampen pain. Finally, patients with arthritis pain, compared with healthy controls, had overall less OpR availability within the striatum specifically, consistent with the greater endogenous opioid binding that would be expected in chronic pain states. Our observational evidence points to the need for further studies to establish the causal relationship between chronic pain states and OpR adaptation.


Asunto(s)
Artritis/complicaciones , Dolor Crónico/etiología , Dolor Crónico/patología , Cuerpo Estriado/metabolismo , Percepción del Dolor/fisiología , Receptores Opioides/metabolismo , Adulto , Anciano , Radioisótopos de Carbono/farmacocinética , Cuerpo Estriado/efectos de los fármacos , Diprenorfina/farmacocinética , Femenino , Humanos , Hiperalgesia/fisiopatología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Antagonistas de Narcóticos/farmacocinética , Radioisótopos de Oxígeno/farmacocinética , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Sustancia Gris Periacueductal/metabolismo , Tomografía de Emisión de Positrones , Encuestas y Cuestionarios
5.
J Med Chem ; 57(12): 5464-9, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24933507

RESUMEN

We report the synthesis and biological evaluation of a triplet of 6-O-(18)F-fluoroethylated derivatives of structurally related orvinols that span across the full range of intrinsic activities, the antagonist diprenorphine, the partial agonist buprenorphine, and the full agonist phenethyl-orvinol. [(18)F]fluoroethyl-diprenorphine, [(18)F]fluoroethyl-buprenorphine, and [(18)F]fluoroethyl-phenethyl-orvinol were prepared in high yields and quality from their 6-O-desmethyl-precursors. The results indicate suitable properties of the three 6-O-(18)F-fluoroethylated derivatives as functional analogues to the native carbon-11 labeled versions with similar pharmacological properties.


Asunto(s)
Buprenorfina/análogos & derivados , Diprenorfina/análogos & derivados , Morfinanos/síntesis química , Radiofármacos/síntesis química , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Buprenorfina/síntesis química , Buprenorfina/química , Buprenorfina/farmacocinética , Células CHO , Radioisótopos de Carbono , Cricetulus , Diprenorfina/síntesis química , Diprenorfina/química , Diprenorfina/farmacocinética , Radioisótopos de Flúor , Humanos , Morfinanos/química , Morfinanos/farmacocinética , Antagonistas de Narcóticos , Tomografía de Emisión de Positrones , Ensayo de Unión Radioligante , Radiofármacos/química , Radiofármacos/farmacocinética , Ratas , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Relación Estructura-Actividad
6.
Neuroimage ; 79: 72-80, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23597934

RESUMEN

Previous positron emission tomography (PET) studies in refractory temporal lobe epilepsy (TLE) using the non-selective opioid receptor antagonist [(11)C]diprenorphine (DPN) did not detect any changes in mesial temporal structures, despite known involvement of the hippocampus in seizure generation. Normal binding in smaller hippocampi is suggestive of increased receptor concentration in the remaining grey matter. Correction for partial-volume effect (PVE) has not been used in previous DPN PET studies. Here, we present PVE-corrected DPN-PET data quantifying post-ictal and interictal opioid receptor availability in humans with mTLE. Eight paired datasets of post-ictal and interictal DPN PET scans and eleven test/retest control datasets were available from a previously published study on opioid receptor changes in TLE following seizures (Hammers et al., 2007a). Five of the eight participants with TLE had documented hippocampal sclerosis. Data were re-analyzed using regions of interest and a novel PVE correction method (structural functional synergistic-resolution recovery (SFS-RR); (Shidahara et al., 2012)). Data were denoised, followed by application of SFS-RR, with anatomical information derived via precise anatomical segmentation of the participants' MRI (MAPER; (Heckemann et al., 2010)). [(11)C]diprenorphine volume-of-distribution (VT) was quantified in six regions of interest. Post-ictal increases were observed in the ipsilateral fusiform gyri and lateral temporal pole. A novel finding was a post-ictal increase in [(11)C]DPN VT relative to the interictal state in the ipsilateral parahippocampal gyrus, not observed in uncorrected datasets. As for voxel-based (SPM) analyses, correction for global VT values was essential in order to demonstrate focal post-ictal increases in [(11)C]DPN VT. This study provides further direct human in vivo evidence for changes in opioid receptor availability in TLE following seizures, including changes that were not evident without PVE correction. Denoising, resolution recovery and precise anatomical segmentation can extract valuable information from PET studies that would be missed with conventional post-processing procedures.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Diprenorfina/farmacocinética , Epilepsia/diagnóstico por imagen , Epilepsia/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores Opioides/metabolismo , Adulto , Radioisótopos de Carbono/farmacocinética , Femenino , Humanos , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagenología Tridimensional/métodos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
7.
J Neurosci ; 33(10): 4369-77, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23467353

RESUMEN

Regulator of G-protein signaling (RGS) proteins classically function as negative modulators of G-protein-coupled receptor signaling. In vitro, RGS proteins have been shown to inhibit signaling by agonists at the µ-opioid receptor, including morphine. The goal of the present study was to evaluate the contribution of endogenous RGS proteins to the antinociceptive effects of morphine and other opioid agonists. To do this, a knock-in mouse that expresses an RGS-insensitive (RGSi) mutant Gαo protein, Gαo(G184S) (Gαo RGSi), was evaluated for morphine or methadone antinociception in response to noxious thermal stimuli. Mice expressing Gαo RGSi subunits exhibited a naltrexone-sensitive enhancement of baseline latency in both the hot-plate and warm-water tail-withdrawal tests. In the hot-plate test, a measure of supraspinal nociception, morphine antinociception was increased, and this was associated with an increased ability of opioids to inhibit presynaptic GABA neurotransmission in the periaqueductal gray. In contrast, antinociception produced by either morphine or methadone was reduced in the tail-withdrawal test, a measure of spinal nociception. In whole-brain and spinal cord homogenates from mice expressing Gαo RGSi subunits, there was a small loss of Gαo expression and an accompanying decrease in basal G-protein activity. Our results strongly support a role for RGS proteins as negative regulators of opioid supraspinal antinociception and also reveal a potential novel function of RGS proteins as positive regulators of opioid spinal antinociceptive pathways.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Hiperalgesia/tratamiento farmacológico , Morfina/uso terapéutico , Proteínas RGS/metabolismo , Analgésicos Opioides/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Diprenorfina/farmacocinética , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacocinética , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Antagonistas del GABA/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Calor/efectos adversos , Humanos , Hiperalgesia/genética , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Isótopos/farmacocinética , Masculino , Metadona/farmacología , Metadona/uso terapéutico , Ratones , Ratones Transgénicos , Morfina/farmacología , Mutación , Naloxona/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oligopéptidos/farmacología , Dimensión del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Toxina del Pertussis/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteínas RGS/genética , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Ácido gamma-Aminobutírico/metabolismo
8.
J Neurochem ; 124(4): 466-77, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23173782

RESUMEN

Poly C binding protein 1 (PCBP1) is an expressional regulator of the mu-opioid receptor (MOR) gene. We hypothesized the existence of a PCBP1 co-regulator modifying human MOR gene expression by protein-protein interaction with PCBP1. A human brain cDNA library was screened using the two-hybrid system with PCBP1 as the bait. Receptor for activated protein kinase C (RACK1) protein, containing seven WD domains, was identified. PCBP1-RACK1 interaction was confirmed via in vivo validation using the two-hybrid system, and by co-immunoprecipitation with anti-PCBP1 antibody and human neuronal NMB cell lysate, endogenously expressing PCBP1 and RACK1. Further co-immunoprecipitation suggested that RACK1-PCBP1 interaction occurred in cytosol alone. Single and serial WD domain deletion analyses demonstrated that WD7 of RACK1 is the key domain interacting with PCBP1. RACK1 over-expression resulted in a dose-dependent decrease of MOR promoter activity using p357 plasmid containing human MOR promoter and luciferase reporter gene. Knock-down analysis showed that RACK1 siRNA decreased the endogenous RACK1 mRNA level in NMB, and elevated MOR mRNA level as indicated by RT-PCR. Likewise, a decrease of RACK1 resulted in an increase of MOR proteins, verified by (3) H-diprenorphine binding assay. Collectively, this study reports a novel role of RACK1, physically interacting with PCBP1 and participating in the regulation of human MOR gene expression in neuronal NMB cells.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Opioides mu/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN , Diprenorfina/farmacocinética , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Inmunoprecipitación , Antagonistas de Narcóticos/farmacocinética , Proteínas de Neoplasias/genética , Neuroblastoma/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Receptores Opioides mu/genética , Eliminación de Secuencia/genética , Transfección , Tritio/farmacocinética
9.
J Pharm Sci ; 98(12): 4928-40, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19370547

RESUMEN

Conclusions based on either in vitro or in vivo approach to evaluate the P-gp affinity status of opioids may be misleading. For example, in vitro studies indicated that fentanyl is a P-gp inhibitor while in vivo studies indicated that it is a P-gp substrate. Quite the opposite was evident for meperidine. The objective of this study was to evaluate the P-gp affinity status of methadone, buprenorphine and diprenorphine to predict P-gp-mediated drug-drug interactions and to determine a better candidate for management of opioid dependence. Two in vitro (P-gp ATPase and monolayer efflux) assays and two in vivo (tissue distribution and antinociceptive evaluation in mdr1a/b (-/-) mice) assays were used. Methadone stimulated the P-gp ATPase activity only at higher concentrations, while verapamil and GF120918 inhibited its efflux (p < 0.05). The brain distribution and antinociceptive activity of methadone were enhanced (p < 0.05) in P-gp knockout mice. Conversely, buprenorphine and diprenorphine were negative in all assays. P-gp can affect the PK/PD of methadone, but not buprenorphine or diprenorphine. Our report is in favor of buprenorphine over methadone for management of opioid dependence. Buprenorphine most likely is not a P-gp substrate and concerns regarding P-gp-mediated drug-drug interaction are not expected.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Analgésicos Opioides/farmacología , Analgésicos/farmacología , Buprenorfina/farmacología , Diprenorfina/farmacología , Absorción Intestinal/fisiología , Metadona/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Algoritmos , Analgésicos/farmacocinética , Analgésicos Opioides/farmacocinética , Animales , Buprenorfina/farmacocinética , Células CACO-2 , Proteínas Portadoras/genética , Diprenorfina/farmacocinética , Humanos , Manitol/análisis , Manitol/metabolismo , Metadona/farmacocinética , Ratones , Ratones Noqueados , Paclitaxel/análisis , Paclitaxel/metabolismo , Conformación Proteica , Tiempo de Reacción/efectos de los fármacos , Distribución Tisular
10.
Behav Brain Res ; 193(1): 63-8, 2008 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-18562019

RESUMEN

Functional neuroimaging studies suggest that a lateral network in the brain is associated with the sensory aspects of pain perception while a medial network is associated with affective aspects. The highest concentration of opioid receptors is in the medial network. There is significant evidence that endogenous opioids are central to the experience of pain and analgesia. We applied an integrative multimodal imaging approach during acupuncture. We found functional magnetic resonance imaging signal changes in the orbitofrontal cortex, insula, and pons and [11C]diprenorphine positron emission tomography signal changes in the orbitofrontal cortex, medial prefrontal cortex, insula, thalamus, and anterior cingulate cortex. These findings include brain regions within both the lateral and medial pain networks.


Asunto(s)
Analgesia por Acupuntura/métodos , Diprenorfina , Imagen por Resonancia Magnética/métodos , Tomografía de Emisión de Positrones/métodos , Analgesia/métodos , Radioisótopos de Carbono , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/metabolismo , Diprenorfina/farmacocinética , Femenino , Lóbulo Frontal/diagnóstico por imagen , Lóbulo Frontal/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Vías Nerviosas/diagnóstico por imagen , Vías Nerviosas/metabolismo , Dimensión del Dolor/métodos , Puente/diagnóstico por imagen , Puente/metabolismo , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo , Radiografía , Tálamo/diagnóstico por imagen , Tálamo/metabolismo
11.
J Neurochem ; 106(2): 781-92, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18419762

RESUMEN

Once internalized, some G protein-coupled receptors (GPCRs) can recycle back to the cell surface, while some of them are delivered to lysosomes for degradation. Because recycling and degradation represent two opposing receptor fates, understanding the mechanisms that determine post-endocytic fate of GPCRs is of great importance. Our recent work has verified that agonist-induced internalization of delta-opioid receptor (DOR) employs both phosphorylation-dependent and -independent mechanisms in HEK293 cells. To investigate whether these two internalization mechanisms work differently in receptor regulation, we monitored receptor post-endocytic fates using flow cytometry, surface receptor biotinylation and radioligand binding assays. Results showed that the internalized wild type DOR could either recycle to the cell surface or be degraded. Mutant DOR M4/5/6, which lacks all three G protein-coupled receptor kinase 2 (GRK2) phosphorylation sites, could also internalize upon agonist challenge although in a reduced level as compared with the wild type counterpart. However, the internalized mutant DOR could not recycle back to the cell surface and all mutant DOR was degraded after internalization. Inhibition of GRK2 expression by GRK2 RNAi also strongly attenuated recycling of DOR. Furthermore, overexpression of GRK2, which significantly increased receptor phosphorylation and internalization, also targeted more internalized receptors to the recycling pathway. These data suggest that GRK2-catalyzed receptor phosphorylation is critically involved in DOR internalization and recycling, and the phosphorylation-independent internalization leads to receptor degradation. Data obtained from beta-arrestin1 and beta-arrestin2 RNAi experiments indicated that both beta-arrestin1 and beta-arrestin2 participate in phosphorylation-dependent internalization and the subsequent recycling of DOR. However, phosphorylation-independent internalization and degradation of DOR were strongly blocked by beta-arrestin2 RNAi, but not beta-arrestin1 RNAi. Taken together, these data demonstrate for the first time that GRK2 phosphorylation-dependent internalization mediated by both beta-arrestin1 and beta-arrestin2 leads DOR to recycle, whereas GRK2-independent internalization mediated by beta-arrestin2 alone leads to receptor degradation. Thus, the post-endocytic fate of internalized DOR can be regulated by GRK2-catalyzed receptor phosphorylation as well as distinct beta-arrestin isoforms.


Asunto(s)
Arrestinas/metabolismo , Endocitosis/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Receptores Opioides delta/metabolismo , Analgésicos Opioides/farmacología , Arrestinas/genética , Biotina/metabolismo , Línea Celular Transformada , Diprenorfina/farmacocinética , Endocitosis/efectos de los fármacos , Encefalina D-Penicilamina (2,5)/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mutación/fisiología , Antagonistas de Narcóticos/farmacocinética , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/farmacología , Serina/metabolismo , Factores de Tiempo , Tritio/farmacocinética , beta-Arrestinas
12.
Neuroimage ; 38(1): 82-94, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17764977

RESUMEN

[(11)C]diprenorphine (DPN) is a non-subtype selective opioid receptor PET ligand with slow kinetics and no region devoid of specific binding. Parametric maps are desirable but have to overcome high noise at the voxel level. We obtained parameter values, parametric map image quality, test-retest reproducibility and reliability (using intraclass correlation coefficients (ICCs)) for conventional spectral analysis and a derived method (rank shaping), compared them with values obtained through sampling of volumes of interest (VOIs) on the dynamic data sets and tested whether smaller amounts of radioactivity injected maintained reliability. Ten subjects were injected twice with either approximately 185 MBq or approximately 135 MBq of [(11)C]DPN, followed by dynamic PET for 90 min. Data were movement corrected with a frame-to-frame co-registration method. Arterial plasma input functions corrected for radiolabelled metabolites were created. There was no overall effect of movement correction except for one subject with substantial movement whose test-retest differences decreased by approximately 50%. Actual parametric values depended heavily on the cutoff for slow frequencies (between 0.0008 s(-1) and 0.00063 s(-1)). Image quality was satisfactory for restricted base ranges when using conventional spectral analysis. The rank shaping method allowed maximising of this range but had similar bias. VOI-based methods had the widest dynamic range between regions. Average percentage test-retest differences were smallest for the parametric maps with restricted base ranges; similarly ICCs were highest for these (up to 0.86) but unacceptably low for VOI-derived VD estimates at the low doses of injected radioactivity (0.24/0.04). Our data can inform the choice of methodology for a given biological problem.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Diprenorfina/farmacocinética , Interpretación de Imagen Asistida por Computador/métodos , Tomografía de Emisión de Positrones/métodos , Receptores Opioides/metabolismo , Adulto , Anciano , Algoritmos , Artefactos , Radioisótopos de Carbono/farmacocinética , Femenino , Humanos , Aumento de la Imagen/métodos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
13.
Neurology ; 69(9): 827-34, 2007 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-17724284

RESUMEN

BACKGROUND: Motor cortex stimulation (MCS) for neuropathic pain control induces focal cerebral blood flow changes involving regions with high density of opioid receptors. We studied the possible contribution of the endogenous opioid system to MCS-related pain relief. METHODS: Changes in opioid receptor availability induced by MCS were studied with PET scan and [(11)C]diprenorphine in eight patients with refractory neuropathic pain. Each patient underwent two preoperative (test-retest) PET scans and one postoperative PET scan acquired after 7 months of chronic MCS. RESULTS: The two preoperative scans, performed at 2 weeks interval, did not show significant differences. Conversely, postoperative compared with preoperative PET scans revealed significant decreases of [(11)C]diprenorphine binding in the anterior middle cingulate cortex (aMCC), periaqueductal gray (PAG), prefrontal cortex, and cerebellum. Binding changes in aMCC and PAG were significantly correlated with pain relief. CONCLUSION: The decrease in binding of the exogenous ligand was most likely explained by receptor occupancy due to enhanced secretion of endogenous opioids. Motor cortex stimulation (MCS) may thus induce release of endogenous opioids in brain structures involved in the processing of acute and chronic pain. Correlation of this effect with pain relief in at least two of these structures supports the role of the endogenous opioid system in pain control induced by MCS.


Asunto(s)
Encéfalo/metabolismo , Terapia por Estimulación Eléctrica/métodos , Corteza Motora/fisiología , Péptidos Opioides/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/terapia , Adulto , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Radioisótopos de Carbono , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Diprenorfina/farmacocinética , Femenino , Giro del Cíngulo/diagnóstico por imagen , Giro del Cíngulo/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Sustancia Gris Periacueductal/diagnóstico por imagen , Sustancia Gris Periacueductal/metabolismo , Enfermedades del Sistema Nervioso Periférico/diagnóstico por imagen , Tomografía de Emisión de Positrones , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Resultado del Tratamiento
14.
J Pharmacol Exp Ther ; 322(2): 661-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17488881

RESUMEN

Previously, we reported minimal opioid receptor occupancy following a clinical dose of the micro-opioid agonist, methadone, measured in vivo using positron emission tomography (PET) with [(11)C]diprenorphine and subsequently used rats to obtain experimental data in support of a high receptor reserve hypothesis (Melichar et al., 2005). Here, we report on further preclinical studies investigating opioid receptor occupancy with oxycodone (micro- and kappa-receptor agonist), morphine (micro-receptor agonist), and buprenorphine (partial agonist at the micro-receptor and antagonist at the delta- and kappa-receptors), each given at antinociceptive doses. In vivo binding of [(11)C]diprenorphine was not significantly reduced after treatment with the full agonists but was reduced by approximately 90% by buprenorphine. In addition, given that [(11)C]diprenorphine is a non-subtype-specific PET tracer, there was no regional variation that might feasibly be interpreted as due to differences in opioid subtype distribution. The data support minimal competition between the high-efficacy agonists and the non-subtype-selective antagonist radioligand and highlight the limitations of [(11)C]diprenorphine PET to monitor in vivo occupancy. Alternative means may be needed to address clinical issues regarding opioid receptor occupancy that are required to optimize treatment strategies.


Asunto(s)
Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Diprenorfina/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores Opioides/metabolismo , Analgésicos Opioides/farmacocinética , Animales , Tronco Encefálico/metabolismo , Buprenorfina/metabolismo , Buprenorfina/farmacocinética , Radioisótopos de Carbono , Cerebelo/metabolismo , Propuestas de Licitación , Diprenorfina/sangre , Diprenorfina/farmacocinética , Sistema Límbico/metabolismo , Masculino , Morfina/metabolismo , Morfina/farmacocinética , Antagonistas de Narcóticos , Oxicodona/metabolismo , Oxicodona/farmacocinética , Prosencéfalo/metabolismo , Quinina/metabolismo , Quinina/farmacocinética , Ratas , Ratas Sprague-Dawley , Receptores Opioides/agonistas , Reproducibilidad de los Resultados , Distribución Tisular
15.
Pain ; 127(1-2): 183-94, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17137714

RESUMEN

This study used positron emission tomography (PET) and [11C]diprenorphine to compare the in vivo distribution abnormalities of brain opioid receptors (OR) in patients with peripheral (n=7) and central post-stroke pain (CPSP, n=8), matched for intensity and duration. Compared with age- and sex-matched controls, peripheral neuropathic pain (NP) patients showed bilateral and symmetrical OR binding decrease, while in CPSP binding decrease predominated in the hemisphere contralateral to pain. In CPSP patients, interhemispheric comparison demonstrated a significant decrease in opioid binding in posterior midbrain, medial thalamus and the insular, temporal and prefrontal cortices contralateral to the painful side. Peripheral NP patients did not show any lateralised decrease in opioid binding. Direct comparison between the central and peripheral groups confirmed a significant OR decrease in CPSP, contralateral to pain. While bilateral binding decrease in both NP groups may reflect endogenous opioid release secondary to chronic pain, the more important and lateralised decrease specific to CPSP suggests opioid receptor loss or inactivation in receptor-bearing neurons. Opioid binding decrease was much more extensive than brain anatomical lesions, and was not co-localised with them; metabolic depression (diaschisis) and/or degeneration of OR neurons-bearing secondary to central lesions appears therefore as a likely mechanism. Central and peripheral forms of NP may differ in distribution of brain opioid system changes and this in turn might underlie their different sensitivity to opiates.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Diprenorfina/farmacocinética , Neuralgia/metabolismo , Receptores Opioides/metabolismo , Adulto , Anciano , Disponibilidad Biológica , Radioisótopos de Carbono , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades del Sistema Nervioso Periférico/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Distribución Tisular
16.
Nuklearmedizin ; 45(5): 197-200, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17043729

RESUMEN

AIM: The morphinane-derivate 6-O-(2-[(18)F]fluoroethyl)-6-O-desmethyldiprenorphine ([(18)F]FDPN) is a nonselective opioid receptor ligand currently used in positron emission tomography (PET). Correction for plasma metabolites of the arterial input function is necessary for quantitative measurements of [(18)F]FDPN binding. A study was undertaken to investigate if there are gender dependent differences in the rate of metabolism of [(18)F]FDPN. METHODS: The rate of metabolism of [(18)F]FDPN was mathematically quantified by fitting a bi-exponential function to each individual's dynamic metabolite data. RESULTS: No statistically significant gender differences were found for age, weight, body mass index or dose. However, significant differences (p < 0.01) in two of the four kinetic parameters describing the rate of metabolism were found between the two groups, with women metabolizing [(18)F]FDPN faster than men. These differences were found in the contribution of the fast and slow kinetic components of the model describing the distribution of radioactive species in plasma, indicating a higher rate of enzyme-dependent degradation of [(18)F]FDPN in women than in men. CONCLUSION: The findings reinforce the need for individualized metabolite correction during [(18)F]FDPN-PET scans and also indicate that in certain cases, grouping according to gender could be performed in order to minimize methodological errors of the input function prior to kinetic analyses.


Asunto(s)
Diprenorfina/análogos & derivados , Tomografía de Emisión de Positrones/métodos , Adulto , Diprenorfina/sangre , Diprenorfina/farmacocinética , Femenino , Humanos , Cinética , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Caracteres Sexuales
17.
Neurology ; 66(7): 1108-10, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16606930

RESUMEN

Using PET with the opioidergic ligand [11C]diprenorphine, the authors demonstrate decreased tracer binding in the pineal gland of cluster headache patients vs healthy volunteers. Opioid receptor availability in the hypothalamus and cingulate cortex depended on the duration of the headache disorder. Therefore, the pathophysiology of cluster headache may relate to opioidergic dysfunction in circuitries generating the biologic clock.


Asunto(s)
Cefalalgia Histamínica/diagnóstico por imagen , Diprenorfina/farmacocinética , Hipotálamo/diagnóstico por imagen , Antagonistas de Narcóticos/farmacocinética , Glándula Pineal/diagnóstico por imagen , Adulto , Radioisótopos de Carbono , Cefalalgia Histamínica/patología , Lateralidad Funcional , Humanos , Hipotálamo/patología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Glándula Pineal/patología , Tomografía de Emisión de Positrones , Radiografía
18.
Pain ; 122(1-2): 63-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16527398

RESUMEN

Opioids modulate the affective component of pain and in vivo data indicate that opioids induce activation changes in the rostral ACC, insula and other brain areas. Hence, opioidergic release is to be expected in these brain regions following experimental pain stimulation. We examined healthy volunteers during heat pain and control subjects during rest using [18F]fluorodiprenorphine-PET. Pain stimulation led to significant reduction of diprenorphine binding in limbic and paralimbic brain areas including the rostral ACC and insula. The finding of altered opioidergic receptor availability in the rostral ACC after experimental nociceptive pain is novel and provides direct evidence for the involvement of this region in endogenous opioidergic inhibition of pain.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Diprenorfina/análogos & derivados , Calor/efectos adversos , Narcóticos/metabolismo , Transmisión Sináptica , Adulto , Mapeo Encefálico/métodos , Diprenorfina/farmacocinética , Femenino , Humanos , Hiperalgesia/etiología , Masculino , Persona de Mediana Edad , Neurotransmisores/metabolismo , Cintigrafía , Radiofármacos/farmacocinética , Receptores Opioides/metabolismo , Distribución Tisular
19.
J Pharmacol Exp Ther ; 314(2): 862-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15901806

RESUMEN

Zebrafish is considered a model organism, not only for the study of the biological functions of vertebrates but also as a tool to analyze the effects of some drugs or toxic agents. Five opioid precursor genes homologous to the mammalian opioid propeptide genes have recently been identified; one of these, the zebrafish proenkephalin, codes a novel heptapeptide, the Met-enkephalin-Gly-Tyr (MEGY). To analyze the pharmacological properties of this novel ligand, we have labeled it with tritium ([(3)H]MEGY). In addition, we have also synthesized two analogs: (d-Ala(2))-MEGY (Y-d-Ala-GFMGY) and (d-Ala(2), Val(5))-MEGY (Y-d-Ala-GFVGY). The binding profile of these three agents has been studied in zebrafish and rat brain membranes. [(3)H]MEGY presents one binding site in zebrafish, as well as in rat brain membranes, although it shows a slight higher affinity in zebrafish brain. The observed saturable binding is displaced by naloxone, thus confirming the opioid nature of the binding sites. Competition binding assays indicate that the methionine residue is essential for high-affinity binding of MEGY and probably of other peptidic agonists in zebrafish, whereas the change of a Gly for a d-Ala does not dramatically affect the ligand affinity. Our results show that the percentage of [(3)H]MEGY displaced by all the ligands studied is higher than 100%, thus inferring that naloxone (used to determine nonspecific binding) does not bind to all the sites labeled by [(3)H]MEGY. Therefore, we can deduct that some of the MEGY binding sites should not be considered classical opioid sites.


Asunto(s)
Encéfalo/metabolismo , Encefalina Metionina/análogos & derivados , Sustitución de Aminoácidos , Animales , Unión Competitiva/efectos de los fármacos , Diprenorfina/farmacocinética , Encefalina Metionina/síntesis química , Encefalina Metionina/farmacocinética , Femenino , Técnicas In Vitro , Masculino , Membranas/efectos de los fármacos , Membranas/metabolismo , Metionina/metabolismo , Naloxona/farmacocinética , Antagonistas de Narcóticos/farmacocinética , Ratas , Especificidad de la Especie , Pez Cebra
20.
Mov Disord ; 19(12): 1498-503, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15390064

RESUMEN

The opioid transmitters enkephalin and dynorphin are known to regulate pallidal output and consequently cortical excitability. Indeed, abnormal basal ganglia opioid transmission has been reported in several involuntary movement disorders, including levodopa-induced dyskinesias in Parkinson's disease (PD), tardive dyskinesias/dystonia, Huntington's disease, and Tourette's syndrome. Moreover, a previous 11C-diprenorphine PET study investigating levodopa-induced dyskinesias found reduced opioid receptor availability in PD with but not without dyskinesias. We wished to investigate if a similar alteration in basal ganglia opioid binding was present in DYT1 primary torsion dystonia (PTD). Regional cerebral 11C-diprenorphine binding was investigated in 7 manifesting carriers of the DYT1 gene and 15 age-matched normal controls using a region-of-interest (ROI) approach and statistical parametric mapping (SPM). No difference in regional mean 11C-diprenorphine binding was found between DYT1-PTD and controls, and no correlation between the severity of dystonia and opioid binding was seen. We conclude that aberrant opioid transmission is unlikely to be present in DYT1-PTD and altered opioid transmission is not a common mechanism underlying all disorders of involuntary movement.


Asunto(s)
Diprenorfina/farmacocinética , Distonía Muscular Deformante/genética , Distonía Muscular Deformante/metabolismo , Encefalinas/metabolismo , Chaperonas Moleculares/genética , Tomografía de Emisión de Positrones , Adulto , Anciano , Ganglios Basales/metabolismo , Sitios de Unión , Cuerpo Estriado/metabolismo , Diprenorfina/administración & dosificación , Femenino , Eliminación de Gen , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...