Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 81(2): 264-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22072818

ABSTRACT

The peripheral δ opioid receptor (DOR) is an attractive target for analgesic drug development. There is evidence that DOR can form heteromers with the κ-opioid receptor (KOR). As drug targets, heteromeric receptors offer an additional level of selectivity and, because of allosteric interactions between protomers, functionality. Here we report that selective KOR antagonists differentially altered the potency and/or efficacy of DOR agonists in primary cultures of adult rat peripheral sensory neurons and in a rat behavioral model of thermal allodynia. In vitro, the KOR antagonist nor-binaltorphimine (nor-BNI) enhanced the potency of [D-Pen(2,5)]-enkephalin (DPDPE), decreased the potency of [D-Ala(2),D-Leu(5)]-enkephalin (DADLE), and decreased the potency and efficacy of 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethylpiperazin-1-yl](3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC80) to inhibit prostaglandin E(2) (PGE(2))-stimulated adenylyl cyclase activity. In vivo, nor-BNI enhanced the effect of DPDPE and decreased the effect of SNC80 to inhibit PGE(2)-stimulated thermal allodynia. In contrast to nor-BNI, the KOR antagonist 5'-guanidinonaltrindole (5'-GNTI) reduced the response of DPDPE both in cultured neurons and in vivo. Evidence for DOR-KOR heteromers in peripheral sensory neurons included coimmunoprecipitation of DOR with KOR, a DOR-KOR heteromer selective antibody augmented the antinociceptive effect of DPDPE in vivo, and the DOR-KOR heteromer agonist 6'-GNTI inhibited adenylyl cyclase activity in vitro as well as PGE(2)-stimulated thermal allodynia in vivo. Taken together, these data suggest that DOR-KOR heteromers exist in rat primary sensory neurons and that KOR antagonists can act as modulators of DOR agonist responses most likely through allosteric interactions between the protomers of the DOR-KOR heteromer.


Subject(s)
Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Sensory Receptor Cells/chemistry , Allosteric Regulation , Animals , Cells, Cultured , Drug Design , Hyperalgesia/etiology , Ligands , Protein Multimerization , Rats , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists
2.
J Pharmacol Exp Ther ; 338(1): 92-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487072

ABSTRACT

There is considerable interest in understanding the regulation of peripheral opioid receptors to avoid central nervous system side effects associated with systemically administered opioid analgesics. Here, we investigated the regulation of the κ-opioid receptor (KOR) on rat primary sensory neurons in vitro and in a rat model of thermal allodynia. Under basal conditions, application of the KOR agonist trans-(1S,2S)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide hydrochloride hydrate (U50488) did not inhibit adenylyl cyclase (AC) activity nor release of calcitonin gene-related peptide (CGRP) in vitro and did not inhibit thermal allodynia in vivo. However, after 15-min pretreatment with bradykinin (BK), U50488 became capable of inhibiting AC activity, CGRP release, and thermal allodynia. Inhibition of AC by 5-hydroxytryptamine 1 or neuropeptide Y(1) receptor agonists and stimulation of extracellular signal-regulated kinase activity by U50488 did not require BK pretreatment. The effect of U50488 in BK-primed tissue was blocked by the KOR antagonist nor-binaltorphimine both in vitro and in vivo. The effect of BK in vitro was blocked by either indomethacin or bisindolylmaleimide, suggesting that an arachidonic acid (AA) metabolite and protein kinase C (PKC) activation mediate BK-induced regulation of the KOR system. Furthermore, the effect of U50488 in BK-treated tissue was blocked by a soluble integrin-blocking peptide (GRGDSP), but not the inactive reverse sequence peptide (GDGRSP), suggesting that, in addition to AA and PKC, RGD-binding integrins participate in the regulation of KOR signaling in response to U50488. Understanding the mechanisms by which peripheral KOR agonist efficacy is regulated may lead to improved pharmacotherapy for the treatment of pain with reduced adverse effects.


Subject(s)
Receptors, Opioid, kappa/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Trigeminal Ganglion/physiology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Male , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Trigeminal Ganglion/drug effects
3.
J Pharmacol Exp Ther ; 321(3): 839-47, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17347322

ABSTRACT

Management of pain by opioid analgesics is confounded by central adverse effects that limit clinical dosages. Consequently, there is considerable interest to understand peripheral analgesic effects of opioids. The actions of opioids on peripheral sensory neurons have been difficult to study because of a general lack of effect of opioid agonists on nociceptor function in culture despite documented presence of opioid receptors. In this study, the micro-opioid receptor agonist, [D-Ala(2),N-MePhe(4),Gly-ol(5)]-enkephalin (DAMGO), did not alter guanosine 5'-O-(3-[(35)S]thio)-triphosphate (GTPgamma[(35)S]) binding, adenylyl cyclase activity, or neuropeptide release in primary cultures of rat trigeminal ganglion (TG). However, after brief exposure to bradykinin (BK), DAMGO stimulated GTPgamma[(35)S] binding and inhibited both prostaglandin E(2) (PGE(2))-stimulated adenylyl cyclase activity and BK/PGE(2)-stimulated neuropeptide release. The effect of BK was blocked by the B(2) antagonist HOE 140 [D-Arg[Hyp(3),Thi(5),D-Tic(7),Oic(8)]-bradykinin], but not by the B(1) antagonist, Lys-[Leu8]des-Arg9-BK, and was mimicked by the protease-activated receptor-2 agonist, Ser-Leu-Ile-Gly-Arg-Leu-NH(2), and by activation of protein kinase C (PKC) or by administration of arachidonic acid (AA). The enhanced responsiveness of micro-opioid receptor signaling by BK priming was blocked by both cyclooxygenase and PKC inhibitors; however, the effect of AA was blocked only by a cyclooxygenase inhibitor. The results indicate that micro-opioid receptor signaling in primary sensory TG neurons is enhanced by activation of phospholipase C-coupled receptors via a cyclooxygenase-dependent AA metabolite that is downstream of PKC.


Subject(s)
Neurons, Afferent/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Arachidonic Acid/pharmacology , Bradykinin/pharmacology , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Inositol Phosphates/metabolism , Male , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/metabolism , Receptors, Opioid, mu/analysis , Receptors, Opioid, mu/antagonists & inhibitors , Signal Transduction/drug effects , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
4.
J Neurochem ; 100(3): 617-27, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17181558

ABSTRACT

p-Methoxyamphetamine (PMA) has been implicated in fatalities as a result of 'ecstasy' (MDMA) overdose worldwide. Like MDMA, acute effects are associated with marked changes in serotonergic neurotransmission, but the long-term effects of PMA are poorly understood. The aim of this study was to determine the effect of repeated PMA administration on in vitro measures of neurodegeneration: serotonin (5-HT) uptake, 5-HT transporter (SERT) density and 5-HT content in the hippocampus, and compare with effects on in vivo 5-HT clearance. Male rats received PMA, MDMA (4 or 15 mg/kg s.c., twice daily) or vehicle for 4 days and 2 weeks later indices of SERT function were measured. [(3)H]5-HT uptake into synaptosomes and [(3)H]cyanoimipramine binding to the SERT were significantly reduced by both PMA and MDMA treatments. 5-HT content was reduced in MDMA-, but not PMA-treatment. In contrast, clearance of locally applied 5-HT measured in vivo by chronoamperometry was only reduced in rats treated with 15 mg/kg PMA. The finding that 5-HT clearance in vivo was unaltered by MDMA treatment suggests that in vitro measures of 5-HT axonal degeneration do not necessarily predict potential compensatory mechanisms that maintain SERT function under basal conditions.


Subject(s)
Amphetamines/pharmacology , Hippocampus/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Serotonin/pharmacokinetics , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Synaptosomes/chemistry , Synaptosomes/metabolism , Wallerian Degeneration/chemically induced , Wallerian Degeneration/metabolism , Wallerian Degeneration/physiopathology
5.
J Pharmacol Exp Ther ; 319(1): 260-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16807362

ABSTRACT

5-Methyl-1-[[2-[(2-methyl-3-pyridyl)oxyl]-5-pyridyl]carbamoyl]-6-trifluoromethylindone (SB 243213) is a selective, high-affinity 5-hydroxytryptamine (serotonin)(2C) receptor ligand that has been previously characterized as a competitive 5-HT(2C) receptor antagonist that has a long duration of activity in vivo. It is active in two preclinical models of anxiety and has an improved anxiolytic profile compared with benzodiazepines. In this study, we further characterized the pharmacological properties of SB 243213 by measuring its effects on each of multiple responses coupled to the 5-HT(2C) receptor. In Chinese hamster ovary cells, SB 243213 was an inverse agonist for the phospholipase A(2) response, for guanosine 5'-O-(3-[(35)S]thio)triphosphate binding, for reduction of constitutive desensitization, and for enhancement of dopamine release in the rat nucleus accumbens, with relative efficacies of 0.6, 1, 1, and 0.6, respectively. However, for the phospholipase C (PLC) signaling cascade, SB 243213 behaved as an antagonist. Although SB 243213 was previously characterized as a competitive antagonist for the PLC response, the magnitude of the dextral shift of the 5-HT concentration-response curve was time-dependent, and the maximal PLC response to 5-HT was decreased, probably as a result of the slow dissociation rate of SB 243213 (initial dissociation rate was 3.2 times slower than SB206553, a prototypical 5-HT(2C) receptor inverse agonist). Taken together, these data show that the pharmacological characteristics of SB 243213 at the 5-HT(2C) receptor differ depending upon the response measured, and they support the hypothesis that different drugs, acting at the same receptor subtype, can differentially regulate multiple cellular signaling systems.


Subject(s)
Indoles/pharmacology , Pyridines/pharmacology , Receptor, Serotonin, 5-HT2C/drug effects , Animals , Arachidonic Acid/metabolism , CHO Cells , Cricetinae , Dopamine/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Nucleus Accumbens/metabolism , Phospholipases A/physiology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2C/physiology , Serotonin Antagonists/pharmacology , Signal Transduction , Type C Phospholipases/physiology
6.
Biol Psychiatry ; 55(3): 313-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14744474

ABSTRACT

BACKGROUND: Downregulation of serotonin transporter was observed previously after chronic treatment with selective serotonin reuptake inhibitors (SSRIs) but not selective norepinephrine reuptake inhibitors (NRIs). This study investigated if chronic treatment of rats with selective NRIs or SSRIs also affected the norepinephrine transporter (NET). METHODS: Rats were treated for 3 to 6 weeks by osmotic minipumps with either the selective NRIs, desipramine, or the SSRI paroxetine. RESULTS: [(3)H]nisoxetine binding sites as well as [(3)H]norepinephrine uptake were decreased in hippocampus and cortex after treatment with desipramine. By contrast, paroxetine-treated rats showed no alteration in either [(3)H]nisoxetine binding or [(3)H]norepinephrine uptake. NET messenger RNA levels in the locus coeruleus were unchanged by desipramine treatment. CONCLUSIONS: These results demonstrate that the marked decrease in NET density 1) is not a consequence of a decrease in gene expression; 2) was caused only by a selective NRI; and 3) was associated with a parallel decrease in norepinephrine uptake.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Fluoxetine/analogs & derivatives , Symporters/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Antidepressive Agents/administration & dosage , Autoradiography , Brain/metabolism , Cerebral Cortex/drug effects , Desipramine/pharmacology , Fluoxetine/pharmacology , Hippocampus/drug effects , In Situ Hybridization , Locus Coeruleus/drug effects , Male , Norepinephrine Plasma Membrane Transport Proteins , Paroxetine/pharmacology , Rats , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology , Symporters/metabolism
7.
Neuroendocrinology ; 77(2): 132-40, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12624535

ABSTRACT

The dopamine transporter (DAT) regulates extracellular dopamine DA levels and is an important site of action for amphetamine and cocaine. Amphetamine and cocaine increase extracellular levels of DA by acting on the DAT; thus, variations in DAT binding sites or activity might influence the action of some drugs of abuse. It was hypothesized that streptozotocin-induced diabetes decreases amphetamine self-administration and that this behavioral change is accompanied by changes in DAT function. Separate groups of male rats responded to receive either amphetamine (0.03 mg/kg/infusion), cocaine (0.25 mg/kg/infusion), or food before and for 7 days after receiving streptozotocin. Rats were sacrificed and [(3)H]DA uptake and [(3)H]WIN 35,428 binding were measured in the striatum. In a second study, rats could self-administer one of several different doses of amphetamine (0.01-0.178 mg/kg/infusion) before and after receiving streptozotocin. In streptozotocin-treated rats, a marked decrease in staining for insulin in pancreatic sections was paralleled by a more than doubling in blood glucose levels. Streptozotocin significantly decreased the number of amphetamine infusions without changing the number of cocaine infusions or food pellets received. Streptozotocin increased DA uptake (V(max)) 1.6- or 2.4-fold in rats that responded for food or amphetamine and increased 3-fold the K(m) for DA only in rats that responded for food; however, [(3)H]WIN 35,428 binding was not changed in any rat. In the second study, streptozotocin only decreased amphetamine self-administration thereby supporting the view that streptozotocin does not simply decrease the potency of amphetamine. These results demonstrate a selective decrease in amphetamine self-administration in diabetic rats that was associated with increased DAT function in the striatum. Collectively, these studies suggest that insulin pathways in the brain may play an important role in regulating DAT activity and amphetamine action.


Subject(s)
Amphetamine/administration & dosage , Cocaine/analogs & derivatives , Diabetes Mellitus, Experimental/physiopathology , Dopamine/pharmacokinetics , Membrane Glycoproteins , Membrane Transport Proteins/analysis , Neostriatum/metabolism , Nerve Tissue Proteins , Streptozocin/pharmacology , Animals , Blood Glucose/metabolism , Cocaine/administration & dosage , Cocaine/metabolism , Dopamine Plasma Membrane Transport Proteins , Dose-Response Relationship, Drug , Drug Interactions , Eating/physiology , Insulin/metabolism , Male , Pancrelipase/metabolism , Rats , Rats, Sprague-Dawley , Self Administration , Synaptosomes/drug effects , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...