Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Pain ; 160(1): 117-135, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30169422

ABSTRACT

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.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/metabolism , HIV Infections/complications , Neuralgia/drug therapy , Neuralgia/etiology , Paclitaxel/toxicity , Triterpenes/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , CHO Cells , Cricetulus , Diprenorphine/pharmacokinetics , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Pentacyclic Triterpenes , Peripheral Nerve Injuries/chemically induced , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/etiology , Peripheral Nerve Injuries/virology , Rats , Rats, Sprague-Dawley , Tritium/pharmacokinetics , Betulinic Acid
2.
Neuropharmacology ; 118: 46-58, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28283391

ABSTRACT

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.


Subject(s)
Mutation/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Tryptophan/genetics , Tyrosine/genetics , Analgesics, Opioid/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , Cyclic AMP/metabolism , Diprenorphine/pharmacokinetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Models, Molecular , Mutagenesis , Narcotic Antagonists/pharmacokinetics , Oligopeptides/pharmacology , Receptors, Opioid, mu/chemistry , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Tritium/pharmacokinetics , Tryptophan/metabolism , Tyrosine/metabolism , beta-Arrestins/metabolism
3.
Neuroimage ; 146: 833-842, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27554530

ABSTRACT

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.


Subject(s)
Deep Brain Stimulation , Neuralgia/prevention & control , Opioid Peptides/metabolism , Periaqueductal Gray/metabolism , Receptors, Opioid/metabolism , Adult , Carbon Radioisotopes , Diprenorphine/pharmacokinetics , Female , Humans , Male , Middle Aged , Neuralgia/metabolism , Pain Measurement , Positron-Emission Tomography , Treatment Outcome
4.
Pain ; 156(11): 2267-2275, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26176892

ABSTRACT

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.


Subject(s)
Arthritis/complications , Chronic Pain/etiology , Chronic Pain/pathology , Corpus Striatum/metabolism , Pain Perception/physiology , Receptors, Opioid/metabolism , Adult , Aged , Carbon Radioisotopes/pharmacokinetics , Corpus Striatum/drug effects , Diprenorphine/pharmacokinetics , Female , Humans , Hyperalgesia/physiopathology , Magnetic Resonance Imaging , Male , Middle Aged , Narcotic Antagonists/pharmacokinetics , Oxygen Radioisotopes/pharmacokinetics , Pain Threshold/drug effects , Pain Threshold/physiology , Periaqueductal Gray/metabolism , Positron-Emission Tomography , Surveys and Questionnaires
5.
J Med Chem ; 57(12): 5464-9, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24933507

ABSTRACT

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.


Subject(s)
Buprenorphine/analogs & derivatives , Diprenorphine/analogs & derivatives , Morphinans/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Animals , Brain/diagnostic imaging , Brain/metabolism , Buprenorphine/chemical synthesis , Buprenorphine/chemistry , Buprenorphine/pharmacokinetics , CHO Cells , Carbon Radioisotopes , Cricetulus , Diprenorphine/chemical synthesis , Diprenorphine/chemistry , Diprenorphine/pharmacokinetics , Fluorine Radioisotopes , Humans , Morphinans/chemistry , Morphinans/pharmacokinetics , Narcotic Antagonists , Positron-Emission Tomography , Radioligand Assay , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Structure-Activity Relationship
6.
Neuroimage ; 79: 72-80, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23597934

ABSTRACT

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.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Diprenorphine/pharmacokinetics , Epilepsy/diagnostic imaging , Epilepsy/metabolism , Positron-Emission Tomography/methods , Receptors, Opioid/metabolism , Adult , Carbon Radioisotopes/pharmacokinetics , Female , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Male , Metabolic Clearance Rate , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
7.
J Neurosci ; 33(10): 4369-77, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23467353

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/therapeutic use , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hyperalgesia/drug therapy , Morphine/therapeutic use , RGS Proteins/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Diprenorphine/pharmacokinetics , Dose-Response Relationship, Drug , Electric Stimulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Hot Temperature/adverse effects , Humans , Hyperalgesia/genetics , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Isotopes/pharmacokinetics , Male , Methadone/pharmacology , Methadone/therapeutic use , Mice , Mice, Transgenic , Morphine/pharmacology , Mutation , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/physiology , Oligopeptides/pharmacology , Pain Measurement/drug effects , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , RGS Proteins/genetics , Reaction Time/drug effects , Reaction Time/genetics , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , gamma-Aminobutyric Acid/metabolism
8.
J Neurochem ; 124(4): 466-77, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23173782

ABSTRACT

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.


Subject(s)
GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Opioid, mu/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Diprenorphine/pharmacokinetics , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Immunoprecipitation , Narcotic Antagonists/pharmacokinetics , Neoplasm Proteins/genetics , Neuroblastoma/pathology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , RNA-Binding Proteins , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Receptors, Opioid, mu/genetics , Sequence Deletion/genetics , Transfection , Tritium/pharmacokinetics
9.
J Pharm Sci ; 98(12): 4928-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19370547

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Buprenorphine/pharmacology , Diprenorphine/pharmacology , Intestinal Absorption/physiology , Methadone/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Algorithms , Analgesics/pharmacokinetics , Analgesics, Opioid/pharmacokinetics , Animals , Buprenorphine/pharmacokinetics , Caco-2 Cells , Carrier Proteins/genetics , Diprenorphine/pharmacokinetics , Humans , Mannitol/analysis , Mannitol/metabolism , Methadone/pharmacokinetics , Mice , Mice, Knockout , Paclitaxel/analysis , Paclitaxel/metabolism , Protein Conformation , Reaction Time/drug effects , Tissue Distribution
10.
Behav Brain Res ; 193(1): 63-8, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18562019

ABSTRACT

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.


Subject(s)
Acupuncture Analgesia/methods , Diprenorphine , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Analgesia/methods , Carbon Radioisotopes , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Diprenorphine/pharmacokinetics , Female , Frontal Lobe/diagnostic imaging , Frontal Lobe/metabolism , Humans , Image Processing, Computer-Assisted , Male , Neural Pathways/diagnostic imaging , Neural Pathways/metabolism , Pain Measurement/methods , Pons/diagnostic imaging , Pons/metabolism , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Radiography , Thalamus/diagnostic imaging , Thalamus/metabolism
11.
J Neurochem ; 106(2): 781-92, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18419762

ABSTRACT

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.


Subject(s)
Arrestins/metabolism , Endocytosis/physiology , G-Protein-Coupled Receptor Kinase 2/metabolism , Receptors, Opioid, delta/metabolism , Analgesics, Opioid/pharmacology , Arrestins/genetics , Biotin/metabolism , Cell Line, Transformed , Diprenorphine/pharmacokinetics , Endocytosis/drug effects , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Green Fluorescent Proteins/metabolism , Humans , Mutation/physiology , Narcotic Antagonists/pharmacokinetics , Phosphorylation/drug effects , Protein Isoforms/metabolism , RNA, Small Interfering/pharmacology , Serine/metabolism , Time Factors , Tritium/pharmacokinetics , beta-Arrestins
12.
Neuroimage ; 38(1): 82-94, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17764977

ABSTRACT

[(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.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Diprenorphine/pharmacokinetics , Image Interpretation, Computer-Assisted/methods , Positron-Emission Tomography/methods , Receptors, Opioid/metabolism , Adult , Aged , Algorithms , Artifacts , Carbon Radioisotopes/pharmacokinetics , Female , Humans , Image Enhancement/methods , Male , Metabolic Clearance Rate , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
13.
Neurology ; 69(9): 827-34, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17724284

ABSTRACT

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.


Subject(s)
Brain/metabolism , Electric Stimulation Therapy/methods , Motor Cortex/physiology , Opioid Peptides/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/therapy , Adult , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/diagnostic imaging , Brain/physiopathology , Carbon Radioisotopes , Cerebellum/diagnostic imaging , Cerebellum/metabolism , Diprenorphine/pharmacokinetics , Female , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/metabolism , Humans , Male , Middle Aged , Periaqueductal Gray/diagnostic imaging , Periaqueductal Gray/metabolism , Peripheral Nervous System Diseases/diagnostic imaging , Positron-Emission Tomography , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Treatment Outcome
14.
J Pharmacol Exp Ther ; 322(2): 661-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17488881

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/metabolism , Brain/metabolism , Diprenorphine/metabolism , Positron-Emission Tomography/methods , Receptors, Opioid/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Brain Stem/metabolism , Buprenorphine/metabolism , Buprenorphine/pharmacokinetics , Carbon Radioisotopes , Cerebellum/metabolism , Competitive Bidding , Diprenorphine/blood , Diprenorphine/pharmacokinetics , Limbic System/metabolism , Male , Morphine/metabolism , Morphine/pharmacokinetics , Narcotic Antagonists , Oxycodone/metabolism , Oxycodone/pharmacokinetics , Prosencephalon/metabolism , Quinine/metabolism , Quinine/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Reproducibility of Results , Tissue Distribution
15.
Pain ; 127(1-2): 183-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17137714

ABSTRACT

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.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Central Nervous System Diseases/metabolism , Diprenorphine/pharmacokinetics , Neuralgia/metabolism , Receptors, Opioid/metabolism , Adult , Aged , Biological Availability , Carbon Radioisotopes , Female , Humans , Male , Middle Aged , Peripheral Nervous System Diseases/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
16.
Nuklearmedizin ; 45(5): 197-200, 2006.
Article in English | MEDLINE | ID: mdl-17043729

ABSTRACT

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.


Subject(s)
Diprenorphine/analogs & derivatives , Positron-Emission Tomography/methods , Adult , Diprenorphine/blood , Diprenorphine/pharmacokinetics , Female , Humans , Kinetics , Male , Metabolic Clearance Rate , Middle Aged , Sex Characteristics
17.
Neurology ; 66(7): 1108-10, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16606930

ABSTRACT

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.


Subject(s)
Cluster Headache/diagnostic imaging , Diprenorphine/pharmacokinetics , Hypothalamus/diagnostic imaging , Narcotic Antagonists/pharmacokinetics , Pineal Gland/diagnostic imaging , Adult , Carbon Radioisotopes , Cluster Headache/pathology , Functional Laterality , Humans , Hypothalamus/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Pineal Gland/pathology , Positron-Emission Tomography , Radiography
18.
Pain ; 122(1-2): 63-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16527398

ABSTRACT

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.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Diprenorphine/analogs & derivatives , Hot Temperature/adverse effects , Narcotics/metabolism , Synaptic Transmission , Adult , Brain Mapping/methods , Diprenorphine/pharmacokinetics , Female , Humans , Hyperalgesia/etiology , Male , Middle Aged , Neurotransmitter Agents/metabolism , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Receptors, Opioid/metabolism , Tissue Distribution
19.
J Pharmacol Exp Ther ; 314(2): 862-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15901806

ABSTRACT

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.


Subject(s)
Brain/metabolism , Enkephalin, Methionine/analogs & derivatives , Amino Acid Substitution , Animals , Binding, Competitive/drug effects , Diprenorphine/pharmacokinetics , Enkephalin, Methionine/chemical synthesis , Enkephalin, Methionine/pharmacokinetics , Female , In Vitro Techniques , Male , Membranes/drug effects , Membranes/metabolism , Methionine/metabolism , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Rats , Species Specificity , Zebrafish
20.
Mov Disord ; 19(12): 1498-503, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15390064

ABSTRACT

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.


Subject(s)
Diprenorphine/pharmacokinetics , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/metabolism , Enkephalins/metabolism , Molecular Chaperones/genetics , Positron-Emission Tomography , Adult , Aged , Basal Ganglia/metabolism , Binding Sites , Corpus Striatum/metabolism , Diprenorphine/administration & dosage , Female , Gene Deletion , Humans , Male , Middle Aged , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...