Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 174(22): 4173-4185, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28859225

ABSTRACT

BACKGROUND AND PURPOSE: The potential for therapeutic antibody treatment of neurological diseases is limited by poor penetration across the blood-brain barrier. I.c.v. delivery is a promising route to the brain; however, it is unclear how efficiently antibodies delivered i.c.v. penetrate the cerebrospinal spinal fluid (CSF)-brain barrier and distribute throughout the brain parenchyma. EXPERIMENTAL APPROACH: We evaluated the pharmacokinetics and pharmacodynamics of an inhibitory monoclonal antibody against ß-secretase 1 (anti-BACE1) following continuous infusion into the left lateral ventricle of healthy adult cynomolgus monkeys. KEY RESULTS: Animals infused with anti-BACE1 i.c.v. showed a robust and sustained reduction (~70%) of CSF amyloid-ß (Aß) peptides. Antibody distribution was near uniform across the brain parenchyma, ranging from 20 to 40 nM, resulting in a ~50% reduction of Aß in the cortical parenchyma. In contrast, animals administered anti-BACE1 i.v. showed no significant change in CSF or cortical Aß levels and had a low (~0.6 nM) antibody concentration in the brain. CONCLUSION AND IMPLICATIONS: I.c.v. administration of anti-BACE1 resulted in enhanced BACE1 target engagement and inhibition, with a corresponding dramatic reduction in CNS Aß concentrations, due to enhanced brain exposure to antibody.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Aspartic Acid Endopeptidases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/immunology , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/cerebrospinal fluid , Aspartic Acid Endopeptidases/immunology , Brain/metabolism , Female , Infusions, Intraventricular , Macaca fascicularis
2.
J Neurosci ; 35(17): 6931-6, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25926467

ABSTRACT

Multiple small-molecule inhibitors of the ß-secretase enzyme (BACE1) are under preclinical or clinical investigation for Alzheimer's disease (AD). Prior work has illustrated robust lowering of central amyloid ß (Aß) after acute administration of BACE1 inhibitors. However, very few studies have assessed the overall impact of chronically administered BACE1 inhibitors on brain amyloid burden, neuropathology, and behavioral function in aged preclinical models. We investigated the effects of a potent nonbrain-penetrant BACE1 inhibitor, delivered directly to the brain using intracerebroventricular infusion in an aged transgenic mouse model. Intracerebroventricular infusion of the BACE1 inhibitor (0.3-23.5 µg/d) for 8 weeks, initiated in 17-month-old Tg2576 mice, produced dose-dependent increases in brain inhibitor concentrations (0.2-13 µm). BACE1 inhibition significantly reversed the behavioral deficit in contextual fear conditioning, and reduced brain Aß levels, plaque burden, and associated pathology (e.g., dystrophic neurites), with maximal effects attained with ∼1 µg/d dose. Strikingly, the BACE1 inhibitor also reversed amyloid pathology below baseline levels (amyloid burden at the start of treatment), without adversely affecting cerebral amyloid angiopathy, microhemorrhages, myelination, or neuromuscular function. Inhibitor-mediated decline in brain amyloid pathology was associated with an increase in microglial ramification. This is the first demonstration of chronically administered BACE1 inhibitor to activate microglia, reverse brain amyloid pathology, and elicit functional improvement in an aged transgenic mouse model. Thus, engagement of novel glial-mediated clearance mechanisms may drive disease-modifying therapeutic benefit with BACE1 inhibition in AD.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/pathology , Cognition Disorders/drug therapy , Enzyme Inhibitors/therapeutic use , Microglia/drug effects , Age Factors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/drug effects , Brain/metabolism , Brain/physiology , Cognition Disorders/genetics , Cognition Disorders/pathology , Disease Models, Animal , Fear/drug effects , Humans , Infusions, Intraventricular , Male , Memory/drug effects , Mice , Mice, Transgenic , Microglia/pathology , Mutation/genetics , Neurons/drug effects , Neurons/pathology
3.
Neuropharmacology ; 72: 66-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603202

ABSTRACT

Our knowledge regarding the molecular pathophysiology underlying anxiety disorders remains incomplete. Increasing evidence points to a role of glutamate in anxiety. The group III metabotropic glutamate receptors (mGlu4, mGlu6, mGlu7 and mGlu8 receptors) remain the least investigated glutamate receptor subtypes partially due to a delay in the development of specific pharmacological tools. Early work using knockout animals and pharmacological tools aimed at investigating the role of mGlu7 receptor in the pathophysiology of anxiety disorders has yielded exciting yet not always consistent results. To further investigate the role this receptor plays in anxiety-like behaviour, we knocked down mGlu7 receptor mRNA levels in the adult mouse brain using siRNA delivered via an osmotic minipump. This reduced anxiety-like behaviour in the light-dark box coupled with an attenuation of stress-induced hyperthermia (SIH) and a reduction of the acoustic startle response (ASRs) in the fear-potentiated startle paradigm (FPS). These effects on anxiety-like behaviour were independent of any impairment of locomotor activity and surprisingly, no behavioural changes were observed in the forced swim test (FST), which is in contrast to mGlu7 receptor knockout animals. Furthermore, the previously reported epilepsy-prone phenotype seen in mGlu7 receptor knockout animals was not observed following siRNA-induced knockdown of the receptor. These data suggest targeting mGlu7 receptors with selective antagonist drugs may be an effective and safe strategy for the treatment of anxiety disorders.


Subject(s)
Anxiety/drug therapy , Anxiety/metabolism , RNA, Small Interfering/therapeutic use , Receptors, Metabotropic Glutamate/metabolism , Adaptation, Ocular/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Fear/drug effects , Hyperthermia, Induced/psychology , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Pentylenetetrazole/toxicity , Receptors, Metabotropic Glutamate/genetics , Reflex, Startle/drug effects , Seizures/chemically induced , Seizures/drug therapy , Stress, Physiological/physiology , Swimming/psychology
4.
Mol Pharmacol ; 83(5): 907-18, 2013 May.
Article in English | MEDLINE | ID: mdl-23395957

ABSTRACT

The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is the fourth and most recently discovered member of the opioid receptor superfamily that also includes µ, δ, and κ opioid receptor subtypes (MOR, DOR, and KOR, respectively). The widespread anatomic distribution of the NOP receptor enables the modulation of several physiologic processes by its endogenous agonist, N/OFQ. Accordingly, the NOP receptor has gained a lot of attention as a potential target for the development of ligands with therapeutic use in several pathophysiological states. NOP receptor activation frequently results in effects opposing classic opioid receptor action; therefore, regulation of the NOP receptor and conditions affecting its modulatory tone are important to understand. Mounting evidence reveals a heterologous interaction of the NOP receptor with other G protein-coupled receptors, including MOR, DOR, and KOR, which may subsequently influence their function. Our focus in this review is to summarize and discuss the findings that delineate the cellular mechanisms of NOP receptor signaling and regulation and the regulation of other receptors by N/OFQ and the NOP receptor.


Subject(s)
Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Animals , Humans , Ligands , Nociceptin Receptor , Nociceptin
5.
Brain ; 135(Pt 4): 1197-209, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22252996

ABSTRACT

Huntington's disease is caused by expression of a mutant form of Huntingtin protein containing an expanded polyglutamine repeat. One possible treatment for Huntington's disease may be to reduce expression of mutant Huntingtin in the brain via RNA interference. Unless the therapeutic molecule is designed to be allele-specific, both wild-type and mutant protein will be suppressed by an RNA interference treatment. A key question is whether suppression of wild-type as well as mutant Huntingtin in targeted brain regions can be tolerated and result in a net benefit to patients with Huntington's disease. Whether Huntingtin performs essential functions in the adult brain is unclear. Here, we tested the hypothesis that the adult primate brain can tolerate moderately reduced levels of wild-type Huntingtin protein for an extended period of time. A serotype 2 adeno-associated viral vector encoding for a short hairpin RNA targeting rhesus huntingtin messenger RNA (active vector) was bilaterally injected into the striatum of four adult rhesus monkeys. Four additional animals received a comparable vector encoding a scrambled control short hairpin RNA (control vector). General health and motor behaviour were monitored for 6 months. Upon termination, brain tissues were sampled and assessed blindly for (i) huntingtin messenger RNA knockdown; (ii) Huntingtin protein expression; and (iii) neuropathological changes. Reduction in wild-type huntingtin messenger RNA levels averaging ∼30% was measured in the striatum of active vector recipients 6 months post-injection. A widespread reduction in Huntingtin protein levels was also observed by immunohistochemistry in these animals, with an average protein reduction of ∼45% relative to controls measured by western blot analysis in the putamen of active vector recipients. As with control vector recipients, no adverse effects were observed behaviourally, and no neurodegeneration was found on histological examination of active vector recipients. Our results suggest that long-term partial suppression of wild-type Huntingtin may be safe, and thus if a comparable level of suppression of mutant Huntingtin is beneficial, then partial suppression of both wild-type and mutant Huntingtin may result in a net benefit in patients with heterozygous Huntington's disease.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , RNA Interference/physiology , Analysis of Variance , Animals , Arabidopsis Proteins/metabolism , Body Weight/genetics , Brain/metabolism , Brain/pathology , Cell Line, Transformed , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Eating/genetics , Female , Gene Expression Regulation/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , Glial Fibrillary Acidic Protein/metabolism , HLA-DR Antigens/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Intramolecular Transferases/metabolism , Macaca mulatta , Magnetic Resonance Imaging , Motor Activity/drug effects , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Psychomotor Performance/physiology , RNA, Small Interfering/administration & dosage , Transfection
6.
Proc Natl Acad Sci U S A ; 106(11): 4501-6, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19246392

ABSTRACT

Although immunization against amyloid-beta (Abeta) holds promise as a disease-modifying therapy for Alzheimer disease (AD), it is associated with an undesirable accumulation of amyloid in the cerebrovasculature [i.e., cerebral amyloid angiopathy (CAA)] and a heightened risk of micro-hemorrhages. The central and peripheral mechanisms postulated to modulate amyloid with anti-Abeta immunotherapy remain largely elusive. Here, we compared the effects of prolonged intracerebroventricular (i.c.v.) versus systemic delivery of anti-Abeta antibodies on the behavioral and pathological changes in an aged Tg2576 mouse model of AD. Prolonged i.c.v. infusions of anti-Abeta antibodies dose-dependently reduced the parenchymal plaque burden, astrogliosis, and dystrophic neurites at doses 10- to 50-fold lower than used with systemic delivery of the same antibody. Both i.c.v. and systemic anti-Abeta antibodies reversed the behavioral impairment in contextual fear conditioning. More importantly, unlike systemically delivered anti-Abeta antibodies that aggravated vascular pathology, i.c.v.-infused antibodies globally reduced CAA and associated micro-hemorrhages. We present data suggesting that the divergent effects of i.c.v.-delivered anti-Abeta antibodies result from gradually engaging the local (i.e., central) mechanisms for amyloid clearance, distinct from the mechanisms engaged by high doses of anti-Abeta antibodies that circulate in the vasculature following systemic delivery. With robust efficacy in reversing AD-related pathology and an unexpected benefit in reducing CAA and associated micro-hemorrhages, i.c.v.-targeted passive immunotherapy offers a promising therapeutic approach for the long-term management of AD.


Subject(s)
Amyloid beta-Peptides/immunology , Antibodies/administration & dosage , Cerebral Amyloid Angiopathy/prevention & control , Cerebral Hemorrhage/etiology , Immunization/methods , Age Factors , Alzheimer Disease , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Behavior, Animal/drug effects , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/therapy , Cerebral Hemorrhage/prevention & control , Fear/drug effects , Mice , Mice, Transgenic
7.
J Recept Signal Transduct Res ; 26(5-6): 527-47, 2006.
Article in English | MEDLINE | ID: mdl-17118797

ABSTRACT

RNA interference (RNAi) is a natural mechanism for regulating gene expression, which exists in plants, invertebrates, and mammals. We investigated whether non-viral infusion of short interfering RNA (siRNA) by the intracerebroventricular route would enable a sequence-specific gene knockdown in the mouse brain and whether the knockdown translates into disease-relevant behavioral changes. Initially, we targeted enhanced green fluorescent protein (EGFP) in mice overexpressing EGFP. A selective knockdown of both EGFP protein and mRNA was observed throughout the brain, with lesser down-regulation in regions distal to the infusion site. We then targeted endogenous genes, encoding the dopamine (DAT) and serotonin transporters (SERT). DAT-siRNA infusion in adult mice produced a significant down-regulation of DAT mRNA and protein and elicited hyperlocomotion similar, but delayed, to that produced on infusion of GBR-12909, a potent and selective DAT inhibitor. Similarly, SERT-siRNA infusion resulted in significant knockdown of SERT mRNA and protein and elicited reduced immobility in the forced swim test similar to that obtained on infusion of citalopram, a very selective and potent SSRI. Application of this non-viral RNAi approach may accelerate target validation for neuropsychiatric disorders that involve a complex interplay of gene(s) from various brain regions.


Subject(s)
Brain/drug effects , Gene Expression Regulation/drug effects , Plasma Membrane Neurotransmitter Transport Proteins/genetics , RNA Interference/drug effects , Receptors, G-Protein-Coupled/genetics , Animals , Brain/metabolism , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/genetics , Down-Regulation/genetics , Green Fluorescent Proteins/genetics , Locomotion/drug effects , Mental Disorders/drug therapy , Mice , Nervous System Diseases/drug therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics
8.
Pharmacol Ther ; 109(3): 413-38, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16183135

ABSTRACT

Psychiatric and neurological disorders are among the most complex, poorly understood, and debilitating diseases in medicine. The burgeoning advances in functional genomic technologies have led to the identification of a vast number of novel genes that are potentially implicated in the pathophysiology of such disorders. However, many of these candidate genes have not yet been functionalized and require validation in vivo. Traditionally, abrogating gene function is one of the primary means of examining the physiological significance of a given gene product. Several methods have been developed for gene ablation or knockdown, however, with limited levels of success. The recent discovery of RNA interference (RNAi), as a highly efficient method for gene knockdown, has been one of the major breakthroughs in molecular medicine. In vivo application of RNAi is further demonstrating the promise of this technology. Recent efforts have focused on applying RNAi-based knockdown to understand the genes implicated in neuropsychiatric disorders. However, the greatest challenge with this approach is translating the success of RNAi from mammalian cell cultures to the brain in animal models of disease and, subsequently, in patients. In this review, we describe the various methods that are being developed to deliver RNAi into the brain for down-regulating gene expression and subsequent phenotyping of genes in vivo. We illustrate the utility of various approaches with a few successful examples and also discuss the potential benefits and pitfalls associated with the use of each delivery approach. Appropriate tailoring of tools that deliver RNAi in the brain may not only aid our understanding of the complex pathophysiology of neuropsychiatric disorders, but may also serve as a valuable therapy for disorders, where there is an immense unmet medical need.


Subject(s)
RNA Interference , Animals , Brain/metabolism , Humans , Mental Disorders/physiopathology , Nervous System Diseases/physiopathology
9.
Mol Pharmacol ; 68(2): 447-56, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15890842

ABSTRACT

In this study, we investigate the molecular mechanisms by which acute orphanin FQ/nociceptin (OFQ/N), acting through the nociceptin opioid peptide (NOP) receptor, desensitizes the mu-opioid receptor. We described previously the involvement of protein kinase C and G-protein-coupled receptor kinases (GRK) 2 and 3 in OFQ/N-induced mu receptor desensitization. Because phosphorylation of the mu receptor triggers the successive regulatory mechanisms responsible for desensitization, such as receptor uncoupling, internalization, and down-regulation, we investigated the ability of OFQ/N to modulate [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO)-induced mu receptor phosphorylation in BE(2)-C human neuroblastoma cells transfected with epitope-tagged mu receptors. OFQ/N treatment (100 nM, 60 min) potentiated DAMGO-induced mu receptor phosphorylation; inhibition of GRK2 or protein kinase C concomitant with OFQ/N treatment blocked the OFQ/N-mediated increase in DAMGO-induced phosphorylation. Inclusion of the NOP antagonist peptide III-BTD during OFQ/N pretreatment blocked the potentiation of DAMGO-induced phosphorylation by OFQ/N, which is consistent with the potentiation being mediated via actions of the NOP receptor. In addition, in cells expressing mu receptors in which the GRK-mediated phosphorylation site Ser(375) was mutated to alanine, OFQ/N treatment failed to potentiate DAMGO-induced mu receptor phosphorylation and failed to desensitize the mu receptor. However, DAMGO-induced mu receptor phosphorylation and OFQ/N-induced mu receptor desensitization occurred in cells expressing mu receptors lacking non-GRK phosphorylation sites. These data suggest that OFQ/N binds to NOP receptors and activates protein kinase C, which then increases the ability of GRK2 to phosphorylate the agonist-occupied mu receptor, heterologously regulating homologous mu receptor desensitization.


Subject(s)
Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Opioid Peptides/pharmacology , Receptors, Opioid, mu/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Humans , Opioid Peptides/metabolism , Phosphorylation/drug effects , Nociceptin
10.
Proc Natl Acad Sci U S A ; 101(49): 17270-5, 2004 Dec 07.
Article in English | MEDLINE | ID: mdl-15569935

ABSTRACT

Gene expression analysis implicates an increasing number of novel genes in the brain as potential targets for the treatment of neurological and psychiatric disorders. Frequently, these genes are ubiquitously expressed in the brain and, thus, may contribute to a pathophysiological state through actions in several brain nuclei. Current strategies employing genetically modified animals for in vivo validation of such targets are time-consuming and often limited by developmental adaptations. Somatic gene manipulation using viral-mediated RNA interference (RNAi) has emerged recently, although restricting the target validation to specific brain nuclei. We investigated whether nonviral infusion of short interfering RNA (siRNA) into the ventricular system would enable a sequence-specific gene knockdown. The temporality and extent of siRNA-induced down-regulation were analyzed by targeting a transgene, EGFP, in mice overexpressing EGFP. Extensive knockdown of EGFP was observed, especially in regions adjacent or dorsoventrally and mediolaterally distant to the infusion site (dorsal third ventricle), with lesser knockdown in more distal regions. We challenged our RNAi approach to generate a specific knockdown of an endogenous gene, encoding the dopamine transporter (DAT) in regions (ventral midbrain) far distal to the infusion site. DAT-siRNA infusion in adult mice produced a significant down-regulation of DAT mRNA and protein in the brain and also elicited a temporal hyperlocomotor response similar to that (but delayed) obtained upon infusion of GBR-12909, a pharmacologically selective DAT inhibitor. Application of this nonviral RNAi approach may accelerate target validation for neuropsychiatric disorders that involve a complex interplay of gene(s) from various brain regions.


Subject(s)
Brain/drug effects , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Motor Activity/drug effects , Nerve Tissue Proteins/genetics , RNA Interference , Animals , Brain/metabolism , Cerebral Ventricles , Dopamine Plasma Membrane Transport Proteins , Down-Regulation/drug effects , Green Fluorescent Proteins/genetics , Male , Methods , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology
11.
J Pharmacol Exp Ther ; 306(3): 965-72, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12750434

ABSTRACT

Mu-Opioid receptors have been shown to contribute to orphanin FQ/nociceptin (OFQ/N)-mediated analgesia and hyperalgesia, indicating that both pro- and antinociceptive actions of OFQ/N are influenced by mu-opioid receptors. A 60-min activation of mu-or opioid receptor-like 1 (ORL1) opioid receptors natively expressed in BE(2)-C human neuroblastoma cells desensitized both mu- and ORL1 receptor-mediated inhibition of cAMP accumulation. The mechanism(s) of OFQ/N-mediated mu and ORL1 cross talk involves the conventional protein kinase C isozyme, PKC-alpha, and G protein-coupled receptor kinases (GRKs) 2 and 3. Unlike OFQ/N-mediated desensitization of ORL1 and mu-opioid receptors, [d-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO)-mediated ORL1 desensitization in BE(2)-C cells is PKC-independent. However, DAMGO (1 microM) pretreatment increased membrane levels of GRK2 and GRK3, indicating their translocation to the membrane upon activation. This suggests that DAMGO activation of mu-opioid receptors results in GRK2 and GRK3 inactivation of ORL1 upon challenge with OFQ/N. Antisense, but not sense, DNA selectively targeting GRK2 or GRK3 blocks DAMGO-mediated mu- and ORL1 desensitization, respectively. However, in SH-SY5Y neuroblastoma cells, DAMGO failed to desensitize ORL1 or alter membrane PKC-alpha or GRK levels. Instead, DAMGO stimulated PKC-epsilon translocation to the cell membrane and produced micro-receptor desensitization. These results indicate that acute exposure to mu-receptor agonists can regulate ORL1 function, but the ability to do so varies from cell type to cell type. These results also confirm the existence of multiple signaling mechanisms for mu-opioid receptors and the importance of these mechanisms for mu-receptor-mediated-heterologous effects.


Subject(s)
Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Receptors, Opioid, mu/metabolism , Receptors, Opioid/metabolism , Signal Transduction/physiology , Humans , Opioid Peptides/pharmacology , Receptor Cross-Talk/physiology , Tumor Cells, Cultured , Nociceptin Receptor
13.
Brain Res Mol Brain Res ; 105(1-2): 38-46, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12399106

ABSTRACT

The recently discovered endogenous peptide orphanin FQ/nociceptin (OFQ/N) activates the opioid receptor-like 1 (ORL1) receptor and produces diverse effects on pain perception. In addition to producing spinal analgesia, OFQ/N also exhibits an 'anti-opioid activity' against functional (supraspinal analgesia) and behavioral (conditioned place preference and withdrawal) properties of morphine. One manifestation of the behavioral changes resulting from chronic use of morphine is the upregulation of tyrosine hydroxylase (TH, the rate-limiting enzyme in catecholamine biosynthesis), which contributes to the dramatic increases in catecholamine release in the target regions of the locus coeruleus (LC) and the ventral tegmental area (VTA). The present study sought to determine the molecular mechanism(s) by which OFQ/N modulates the chronic actions of morphine by utilizing human neuroblastoma cell lines [BE(2)-C and SH-SY5Y] that endogenously express TH, and mu and ORL1 receptors. Activation of mu or ORL1 receptors in these cells in turn activates extracellular signal-regulated protein kinases (ERKs), ERK1 and ERK2. Chronic activation of mu, but not ORL1, receptors upregulated TH levels in these cells as previously reported in rat brain. Morphine-induced TH upregulation was blocked upon inclusion of a MEK-1 (mitogen-activated protein kinase kinase-1) inhibitor (PD98059), confirming the role for ERKs in this adaptive response to morphine. Inclusion of OFQ/N during chronic morphine exposure also blocked morphine-induced TH upregulation. Furthermore, chronic OFQ/N exposure increased levels of the TH gene repressor, Oct-2, irrespective of the presence or absence of morphine. This report suggests a potential role for Oct-2 in mediating the anti-opioid actions of OFQ/N against the behavioral manifestations resulting from chronic use of morphine.


Subject(s)
Brain/drug effects , Brain/enzymology , Morphine Dependence/enzymology , Morphine/antagonists & inhibitors , Opioid Peptides/pharmacology , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Up-Regulation/drug effects , Brain/physiopathology , Catecholamines/metabolism , Chronic Disease , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Morphine/metabolism , Morphine Dependence/physiopathology , Neurons/drug effects , Neurons/enzymology , Octamer Transcription Factor-2 , Opioid Peptides/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/physiology , Nociceptin Receptor , Nociceptin
14.
J Pharmacol Exp Ther ; 302(2): 502-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12130708

ABSTRACT

Morphine tolerance in vivo is reduced following blockade of the orphanin FQ/nociceptin (OFQ/N)/opioid receptor-like 1 (ORL1) receptor system, suggesting that OFQ/N contributes to the development of morphine tolerance. We previously reported that a 60-min activation of ORL1 receptors natively expressed in BE(2)-C cells desensitized both mu and ORL1 receptor-mediated inhibition of cAMP. Investigating the mechanism(s) of OFQ/N-mediated mu and ORL1 receptor cross-talk, we found that pretreatment with the protein kinase C inhibitor, chelerythrine chloride (1 microM), blocked OFQ/N-mediated homologous desensitization of ORL1 and heterologous desensitization of mu opioid receptors. Furthermore, depletion of PKC by 12-O-tetradecanoylphorbol-13-acetate exposure (48 h, 1 microM) also prevented OFQ/N-mediated mu and ORL1 desensitization. OFQ/N pretreatment resulted in translocation of PKC-alpha, G protein-coupled receptor kinase 2 (GRK2) and GRK3 from the cytosol to the membrane, and this translocation was also blocked by chelerythrine. Reduction of GRK2 and GRK3 levels by antisense, but not sense DNA treatment blocks ORL1 and mu receptor desensitization. This suggests that PKC-alpha is required for GRK2 and GRK3 translocation to the membrane, where GRK can inactivate ORL1 and mu opioid receptors upon rechallenge with the appropriate agonist. Our results demonstrate for the first time the involvement of conventional PKC isozymes in OFQ/N-induced mu-ORL1 cross-talk, and represent a possible mechanism for OFQ/N-induced anti-opioid actions.


Subject(s)
Opioid Peptides/pharmacology , Receptors, Opioid, mu/physiology , Receptors, Opioid/physiology , Alkaloids , Benzophenanthridines , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Inhibitors/pharmacology , G-Protein-Coupled Receptor Kinase 3 , Humans , Neuroblastoma , Oligodeoxyribonucleotides, Antisense/pharmacology , Phenanthridines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptor Cross-Talk/physiology , Receptors, Opioid/drug effects , Receptors, Opioid, mu/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , beta-Adrenergic Receptor Kinases , Nociceptin Receptor , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL
...