Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Neuroscience ; 310: 73-90, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26386297

ABSTRACT

Tibia fracture in rodents induces substance P (SP)-dependent keratinocyte activation and inflammatory changes in the hindlimb, similar to those seen in complex regional pain syndrome (CRPS). In animal pain models spinal glial cell activation results in nociceptive sensitization. This study tested the hypothesis that limb fracture triggers afferent C-fiber SP release in the dorsal horn, resulting in chronic glial activation and central sensitization. At 4 weeks after tibia fracture and casting in rats, the cast was removed and hind paw allodynia, unweighting, warmth, and edema were measured, then the antinociceptive effects of microglia (minocycline) or astrocyte (L-2-aminoadipic acid (LAA)) inhibitors or an SP receptor antagonist (LY303870) were tested. Immunohistochemistry and PCR were used to evaluate microglial and astrocyte activation in the dorsal horn. Similar experiments were performed in intact rats after brief sciatic nerve electric stimulation at C-fiber intensity. Microglia and astrocytes were chronically activated at 4 weeks after fracture and contributed to the maintenance of hind paw allodynia and unweighting. Furthermore, LY303870 treatment initiated at 4 weeks after fracture partially reversed both spinal glial activation and nociceptive sensitization. Similarly, persistent spinal microglial activation and hind paw nociceptive sensitization were observed at 48 h after sciatic nerve C-fiber stimulation and this effect was inhibited by treatment with minocycline, LAA, or LY303870. These data support the hypothesis that C-fiber afferent SP signaling chronically supports spinal neuroglial activation after limb fracture and that glial activation contributes to the maintenance of central nociceptive sensitization in CRPS. Treatments inhibiting glial activation and spinal inflammation may be therapeutic for CRPS.


Subject(s)
Complex Regional Pain Syndromes/pathology , Neuroglia/metabolism , Nociception/physiology , Signal Transduction/physiology , Spinal Cord/pathology , Substance P/metabolism , 2-Aminoadipic Acid/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Complex Regional Pain Syndromes/etiology , Disease Models, Animal , Edema/drug therapy , Edema/etiology , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/physiopathology , Male , Minocycline/therapeutic use , Neuroglia/pathology , Pain Measurement , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tibial Fractures/complications , Time Factors
2.
Neuroscience ; 281: 99-109, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25267387

ABSTRACT

In certain forms of nerve injury and inflammation, noradrenaline augments pain via actions on up-regulated α1-adrenoceptors (α1-ARs). The aim of this study was to use immunohistochemistry to examine α1-AR expression on peripheral neurons, cutaneous blood vessels and keratinocytes after distal tibia fracture and cast immobilization, a model of complex regional pain syndrome type 1. We hypothesized that there would be increased α1-AR expression on neurons and keratinocytes in the injured limb in comparison to the contralateral unaffected limb after distal tibia fracture, in association with inflammatory changes and pain. α1-AR expression was increased on plantar keratinocytes, dermal blood vessels and peripheral nerve fibers at 16weeks after injury both in the fractured and contralateral uninjured limb. Similar changes were seen in controls whose limb had been immobilized in a cast for 4weeks but not fractured. Neurofilament 200 (NF200), a marker of myelinated neurons, and calcitonin gene-related peptide (CGRP), a neuropeptide involved in neuro-inflammatory signaling, decreased 4weeks after fracture and casting but then increased at the 16-week time point. As some of these changes were also detected in the contralateral hind limb, they probably were triggered by a systemic response to fracture and casting. Soon after the cast was removed, intraplantar injections of the α1-AR antagonist prazosin released local vasoconstrictor tone but had no effect on pain behaviors. However, systemic injection of prazosin inhibited behavioral signs of pain, suggesting that fracture and/or casting triggered an up-regulation of α1-ARs in central nociceptive pathways that augmented pain. Together, these findings indicate that α1-AR expression increases in the hind limbs after distal tibia fracture and cast immobilization. However, these peripheral increases do not contribute directly to residual pain.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Blood Vessels/metabolism , Calcitonin Gene-Related Peptide/metabolism , Chronic Pain/metabolism , Keratinocytes/metabolism , Neurofilament Proteins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Sciatic Nerve/metabolism , Skin/blood supply , Tibial Fractures/metabolism , Tibial Nerve/metabolism , Animals , Behavior, Animal , Casts, Surgical , Chronic Pain/drug therapy , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Time Factors
3.
Phytomedicine ; 18(7): 544-50, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21530209

ABSTRACT

Eight phenolic compounds, including (-)-epicatechin (1) and seven proanthocyanidins (2-8), were obtained from the butanol extract of Parabarium huaitingii (PHB). Their chemical structures were identified based on analyses of mass spectra (MS), NMR, CD spectra, and partial acid catalyzed thiolytic degradation. The observation made by laser scanning confocal microscope found a significant increase of the concentration of intracellular Ca²+ ([Ca²+](i)) in single myocytes when the PHB was added, while compounds 1 and 3 had the same physiological effect. Further investigations showed PHB had a dose-dependent positive inotropic effect on isolated right atria and papillary muscle of left ventricle of the rat, while having no significant influence on the spontaneous beating rate of the isolated right atria. The inotropic effect of PHB could be greatly abolished by pretreating the myocardium in Ca²+-free solution. These findings indicated that PHB could significantly increase [Ca²+](i) in myocytes, which was greatly dependent on the influx of extracellular Ca²+. Compounds 1 and 3 might be the effective ingredients of the inotropic effect of PHB. In addition, PHB could also significantly decrease the infarct size of the heart on acute myocardial infarction (AMI) model rats, which suggested its myocardial protective effect on ischemic myocardium. The positive inotropic effect of PHB, together with its myocardial protective effect on AMI, suggested that PHB had a promising potential for the prevention and treatment of heart failure, especially the one that was caused by AMI.


Subject(s)
Apocynaceae/chemistry , Cardiotonic Agents/pharmacology , Catechin/pharmacology , Myocardial Contraction/drug effects , Myocardial Infarction/prevention & control , Proanthocyanidins/pharmacology , Animals , Calcium/metabolism , Cardiotonic Agents/chemistry , Catechin/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Heart/drug effects , Medicine, Chinese Traditional , Plant Extracts/chemistry , Plant Stems/chemistry , Plants, Medicinal/chemistry , Proanthocyanidins/chemistry , Prohibitins , Random Allocation , Rats , Rats, Wistar , Stimulation, Chemical
4.
Nat Neurosci ; 5(10): 979-84, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12195434

ABSTRACT

We investigated the role of regionally discrete GABA (gamma-aminobutyric acid) receptors in the sedative response to pharmacological agents that act on GABA(A) receptors (muscimol, propofol and pentobarbital; 'GABAergic agents') and to ketamine, a general anesthetic that does not affect GABA(A) receptors. Behavioral studies in rats showed that the sedative response to centrally administered GABAergic agents was attenuated by the GABA(A) receptor antagonist gabazine (systemically administered). The sedative response to ketamine, by contrast, was unaffected by gabazine. Using c-Fos as a marker of neuronal activation, we identified a possible role for the tuberomammillary nucleus (TMN): when gabazine was microinjected directly into the TMN, it attenuated the sedative response to GABAergic agents. Furthermore, the GABA(A) receptor agonist muscimol produced a dose-dependent sedation when it was administered into the TMN. We conclude that the TMN is a discrete neural locus that has a key role in the sedative response to GABAergic anesthetics.


Subject(s)
Anesthesia , Hypnotics and Sedatives/pharmacology , Hypothalamic Area, Lateral/physiology , Receptors, GABA-A/physiology , Sleep/physiology , Anesthesia/methods , Anesthetics/pharmacology , Animals , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Hypothalamic Area, Lateral/drug effects , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Rats , Rats, Inbred F344 , Sleep/drug effects
5.
Anesthesiology ; 95(1): 184-91, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11465557

ABSTRACT

BACKGROUND: Studies show that the sedative and analgesic effects of alpha2 adrenergic agonists decrease over time, which is a form of synaptic plasticity referred to as tolerance. Because both the N-methyl-D-aspartate (NMDA) receptor complex and nitric oxide synthase are pivotal for some forms of synaptic plasticity, their role in tolerance to the hypnotic and analgesic effects of alpha2 agonists was investigated. METHODS: After institutional approval, rats were made tolerant to the hypnotic or analgesic action of an alpha2 agonist, dexmedetomidine. The hypnotic response to dexmedetomidine was assessed by the duration of loss of righting reflex, and the analgesic response to dexmedetomidine was assessed by the tail-flick assay. In separate cohorts, either the NMDA receptors or nitric oxide synthase was antagonized by coadministration of MK-801, ketamine, or NO2-arginine, respectively, during induction of tolerance. In a separate series of experiments, after tolerance was induced, the hypnotic and analgesic responses to dexmedetomidine were assessed in the presence of acutely administered MK-801 or NO2-arginine. RESULTS: Induction of tolerance to the hypnotic effect of dexmedetomidine is blocked by coadministration of MK-801, ketamine, and NO2-arginine. However, after tolerance developed, acute administration of MK-801, ketamine, or NO2-arginine did not prevent the expression of tolerance. Coadministration of MK-801 or NO2-arginine neither prevents the development nor reverses the expression of tolerance to the analgesic action of dexmedetomidine. CONCLUSION: The underlying processes responsible for the development of tolerance to the hypnotic and analgesic actions of systemically administered alpha2 agonists were different, with only the sedative tolerance involving the NMDA receptor and nitric oxide synthase system.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Analgesics/pharmacology , Enzyme Inhibitors/pharmacology , Hypnotics and Sedatives/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Dexmedetomidine/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Tolerance , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Male , Nitroarginine/pharmacology , Pain Measurement/drug effects , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects
6.
Anesthesiology ; 91(5): 1401-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10551592

ABSTRACT

BACKGROUND: The authors and others have demonstrated that supraspinal opiate receptors and spinal alpha2 adrenoceptors are involved in the analgesic mechanism for nitrous oxide (N2O). The authors hypothesize that activation of opiate receptors in the periaqueductal gray results in the activation of a descending noradrenergic pathway that releases norepinephrine onto alpha2 adrenoceptors in the dorsal horn of the spinal cord. METHODS: The spinal cord was transected at the level of T3-T4 in rats and the analgesic response to 70% N2O in oxygen was determined by the tail flick latency test. In a separate experiment in rats a dialysis fiber was positioned transversely in the dorsal horn of the spinal cord at the T12 level. The following day, the dialysis fiber was infused with artificial cerebrospinal fluid at a rate of 1.3 microl/min, and the effluent was sampled at 30-min intervals. After a 60-min equilibration period, the animals were exposed to 70% N2O in oxygen. The dialysis experiment was repeated in animals that were pretreated with naltrexone (10 mg/kg, intraperitoneally) before N2O. In a third series, spinal norepinephrine was depleted with n-(2-chloroethyl)-n-ethyl-2-bromobenzylamine (DSP-4), and the analgesic response to 70% N2O in oxygen was determined. RESULTS: The analgesic effect of N2O was prevented by spinal cord transection. After exposure to N2O, there was a fourfold increase in norepinephrine released in the first 30-min period, and norepinephrine was still significantly elevated after 1 h of exposure. The increased norepinephrine release was prevented by previous administration of naltrexone. Depletion of norepinephrine in the spinal cord blocked the analgesic response to N2O. CONCLUSIONS: A descending noradrenergic pathway in the spinal cord links N2O-induced activation of opiate receptors in the periaqueductal gray, with activation of alpha2 adrenoceptors in the spinal cord. N2O-induced release of norepinephrine in the dorsal horn of the spinal cord is blocked by naltrexone, as is the analgesic response. Spinal norepinephrine is necessary for the analgesic response to the N2O.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Nitrous Oxide/pharmacology , Norepinephrine/metabolism , Spinal Cord/metabolism , Analgesics, Non-Narcotic/antagonists & inhibitors , Animals , Benzylamines/administration & dosage , Benzylamines/pharmacology , Chromatography, Ion Exchange , Cordotomy , Injections, Spinal , Male , Microdialysis , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurotransmitter Uptake Inhibitors/administration & dosage , Neurotransmitter Uptake Inhibitors/pharmacology , Nitrous Oxide/antagonists & inhibitors , Pain Measurement , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Opioid/drug effects , Spinal Cord/drug effects
7.
Anesthesiology ; 90(2): 470-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9952154

ABSTRACT

BACKGROUND: Opiate receptors in the periaqueductal gray region and alpha2 adrenoceptors in the spinal cord of the rat mediate the antinociceptive properties of nitrous oxide (N2O). The availability of genetically altered mice facilitates the detection of the precise protein species involved in the transduction pathway. In this study, the authors establish the similarity between rats and mice in the antinociceptive action of N2O and investigate which alpha2 adrenoceptor subtypes mediate this response. METHODS: After obtaining institutional approval, antinociceptive dose-response and time-course to N2O was measured in wild-type and transgenic mice (D79N), with a nonfunctional alpha2A adrenoceptor using tail-flick latency. The antinociceptive effect of N2O was tested after pretreatment systemically with yohimbine (nonselective alpha2 antagonist), naloxone (opiate antagonist), L659,066 (peripheral alpha2-antagonist) and prazosin (alpha2B- and alpha2C-selective antagonist). The tail-flick latency to dexmedetomidine (D-med), a nonselective alpha2 agonist, was tested in wild-type and transgenic mice. RESULTS: N2O produced antinociception in both D79N transgenic and wild-type litter mates, although the response was less pronounced in the transgenic mice. Antinociception from N2O decreased over time with continuing exposure, and the decrement was more pronounced in the transgenic mice. The antinociceptive response could be dose dependently antagonized by opiate receptor and selective alpha2B-/alpha2C-receptor antagonists but not by a central nervous system-impermeant alpha2 antagonist (L659,066). Whereas dexmedetomidine exhibited no antinociceptive response in the D79N mice, the robust antinociceptive response in the wild-type litter mates could not be blocked by a selective alpha2B-/alpha2C-receptor antagonist. CONCLUSION: These data confirm that the antinociceptive response to an exogenous alpha2-agonist is mediated by an alpha2A adrenoceptor and that there appears to be a role for the alpha2B- or alpha2C-adrenoceptor subtypes, or both, in the analgesic response to N2O.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Anesthetics, Inhalation/pharmacology , Nitrous Oxide/pharmacology , Pain/drug therapy , Receptors, Adrenergic, alpha-2/drug effects , Animals , Male , Mice , Mice, Transgenic , Pain/physiopathology , Rats , Receptors, Adrenergic, alpha-2/physiology
8.
Anesth Analg ; 87(4): 941-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9768799

ABSTRACT

UNLABELLED: This study investigated the analgesic potency and site of action of systemic dexmedetomidine, a selective alpha2-adrenoceptor (alpha2AR) agonist, in normal and neuropathic rats. Ligation of the L5-6 spinal nerves produced a chronic mechanical and thermal neuropathic hyperalgesia in rats. von Frey fibers and a thermoelectric Peltier device were used to measure mechanical and heat withdrawal thresholds over the hindpaw. Systemic dexmedetomidine dose-dependently increased the mechanical and thermal thresholds in the control animals (50% effective dose [ED50] 144 and 180 microg/kg intraperitoneally [i.p.], respectively). Neuropathic animals responded to much smaller doses of dexmedetomidine with mechanical and thermal ED50 values of 52 and 29 microg/kg i.p., respectively. There was no difference between the control and neuropathic animals with respect to dexmedetomidine-evoked sedation, as determined by decreased grid crossings in an open-field activity chamber (ED50 12 and 9 microg/kg i.p., respectively). Atipamezole, a selective alpha2AR antagonist, blocked the analgesic and sedative actions of dexmedetomidine inboth the neuropathic and control animals. However, L-659,066, a peripherally restricted alpha2AR antagonist, could only block the analgesic actions of dexmedetomidine in the neuropathic rats, with no effect in control animals. In conclusion, nerve injury enhanced the analgesic but not the sedative potency of systemic dexmedetomidine and may have shifted the site of alpha2 analgesic action to outside the blood-brain barrier. IMPLICATIONS: We tested the analgesic efficacy of the alpha2 agonist dexmedetomidine in normal and nerve-injured rats. The analgesic potency of dexmedetomidine was enhanced after nerve injury with a site of action outside the central nervous system. Peripherally restricted alpha2 agonists may be useful in the management of neuropathic pain.


Subject(s)
Adrenergic alpha-Agonists/therapeutic use , Analgesics, Non-Narcotic/therapeutic use , Imidazoles/therapeutic use , Spinal Nerves/injuries , Adrenergic alpha-Antagonists/pharmacology , Analgesics, Non-Narcotic/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacology , Male , Medetomidine , Pain/drug therapy , Pain/etiology , Pain/physiopathology , Pain Threshold , Quinolizines/pharmacology , Rats , Rats, Sprague-Dawley
9.
Anesthesiology ; 88(6): 1634-42, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9637658

ABSTRACT

INTRODUCTION: The effects of long-term administration of the tricyclic antidepressant agent desipramine on the hypnotic, antinociceptive, anesthetic-sparing, and central norepinephrine turnover suppressant action of short-term dexmedetomidine, a highly selective alpha2-adrenergic agonist, were studied in rats. METHODS: Rats were given a 3- or 4-week course of twice daily administration of desipramine, 10 mg/kg, or saline. The effect of a hypnotic dose of dexmedetomidine, 250 microg/kg given intraperitoneally, on the duration of loss of righting reflex was determined. The tail flick latency response was determined before and after 50 microg/kg dexmedetomidine. The minimum anesthetic concentration of halothane and the central norepinephrine turnover rate were determined before and after administration of 30 microg/kg dexmedetomidine. Changes in the affinity and density of the alpha2-adrenergic receptor in locus coeruleus and spinal cord also were determined. RESULTS: Treatment with desipramine decreased dexmedetomidine-induced loss of righting reflex duration by 67% and eliminated the antinociceptive effect of dexmedetomidine. Dexmedetomidine produced a 55% decrease in minimum anesthetic concentration in the control group but no reduction in desipramine-treated rats. Desipramine did not change the receptor density or binding affinity of alpha2 receptors at the site for hypnotic (locus coeruleus) or antinociceptive (spinal cord) responses. No decrement in the central norepinephrine turnover rate was noted in the locus coeruleus of dexmedetomidine after 3 weeks of treatment with desipramine. The alpha1-adrenergic antagonist prazosin at 1 or 5 mg/kg completely (minimum anesthetic concentration reduction), almost completely (antinociceptive), or partially (hypnotic) restored responsiveness to normal. CONCLUSIONS: These data indicate that treatment with desipramine induces hyporesponsiveness to the hypnotic, analgesic, and minimum anesthetic concentration-reducing, but not to the suppression of central norepinephrine turnover, properties of dexmedetomidine. The hyporesponsiveness appears to involve an alpha1-adrenergic mechanism.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Analgesics, Non-Narcotic/pharmacology , Anesthetics, Inhalation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Desipramine/pharmacology , Imidazoles/pharmacology , Animals , Drug Synergism , Male , Medetomidine , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reflex/drug effects
11.
Proc Natl Acad Sci U S A ; 94(18): 9950-5, 1997 Sep 02.
Article in English | MEDLINE | ID: mdl-9275232

ABSTRACT

Norepinephrine contributes to antinociceptive, sedative, and sympatholytic responses in vivo, and alpha2 adrenergic receptor (alpha2AR) agonists are used clinically to mimic these effects. Lack of subtype-specific agonists has prevented elucidation of the role that each alpha2AR subtype (alpha2A, alpha2B, and alpha2C) plays in these central effects. Here we demonstrate that alpha2AR agonist-elicited sedative, anesthetic-sparing, and analgesic responses are lost in a mouse line expressing a subtly mutated alpha2AAR, D79N alpha2AAR, created by two-step homologous recombination. These functional changes are accompanied by failure of the D79N alpha2AAR to inhibit voltage-gated Ca2+ currents and spontaneous neuronal firing, a measure of K+ current activation. These results provide definitive evidence that the alpha2AAR subtype is the primary mediator of clinically important central actions of alpha2AR agonists and suggest that the D79N alpha2AAR mouse may serve as a model for exploring other possible alpha2AAR functions in vivo.


Subject(s)
Analgesics/pharmacology , Anesthetics/pharmacology , Behavior, Animal/physiology , Receptors, Adrenergic, alpha-2/physiology , Animals , Aspartic Acid/genetics , Mice , Mutation
12.
Pharmacol Biochem Behav ; 57(1-2): 23-9, 1997.
Article in English | MEDLINE | ID: mdl-9164550

ABSTRACT

alpha 2 adrenergic agonists are used clinically for their anesthetic, analgesic, and sympatholytic actions in surgical patients. All alpha 2 adrenergic receptors, when activated by alpha 2-adrenergic agonists, are able to inhibit adenylate cyclase. We have examined the alpha 2-adrenoceptor-mediated anesthetic actions of dexmedetomidine, a highly selective alpha 2-adrenergic agonist, after pretreatment of the animals with rolipram, a cyclic AMP (cAMP)-specific phosphodiesterase inhibitor, cAMP accumulation and monoamine turnover were measured in the locus coeruleus (LC) and hippocampus (HC) following administration of rolipram [275 mg/kg, intraperitoneally (IP)] and dexmedetomidine (100-500 mg/kg, IP). The hypnotic response to dexmedetomidine was also measured in these animals. In other experiments, rats were stereotactically cannulated in the LC with an indwelling catheter, and after the second day, the tail-flick analgesic response to dexmedetomidine (3.5 mg/0.2 ml LC), following rolipram (275 mg/kg, IP) pretreatment, was assessed. In the presence of elevated cAMP levels, the hypnotic, analgesic, and sympatholytic effects of dexmedetomidine persisted. These data suggest that adenylate cyclase activity does not mediate the cellular responses to alpha 2-adrenergic agonists but instead may act in concert with other alpha 2-adrenoceptor-coupled effector mechanisms to transduce the anesthetic actions of these agents.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Anesthetics/pharmacology , Cyclic AMP/metabolism , Imidazoles/pharmacology , Analgesics/pharmacology , Animals , Biogenic Monoamines/physiology , Hypnotics and Sedatives/pharmacology , Male , Medetomidine , Phosphodiesterase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram , Synaptic Transmission/drug effects
13.
Pharmacol Biochem Behav ; 57(1-2): 63-71, 1997.
Article in English | MEDLINE | ID: mdl-9164555

ABSTRACT

Tolerance to the hypnotic response was induced in rats by chronically infusing dexmedetomidine, a novel alpha 2-adrenergic agonist. The alpha 2-adrenoceptor affinity for dexmedtomidine and para-iodoclonidine was significantly reduced in tolerant rats, while Bmax was uncharged. The ability of pertussis toxin (PTX) to ribosylate guanine nucleotide regulatory proteins (G proteins) ex vivo was reduced in tolerant rats; the quantity of PTX-sensitive G proteins was unchanged. Forskolin-stimulated adenylyl cyclase was less sensitive to inhibition by dexmedetomidine in the tolerant rats; however, acute intraperitoneal injection of dexmedetomidine still reduced cyclic adenosine monophosphate levels in tolerant rats. Both the decrease in ribosylation and the lower alpha 2-adrenoceptor binding affinity may reflect a decrease in the ability of the G protein to couple to the alpha 2-adrenoceptors in the locus coeruleus of tolerant rats. In this state, the alpha 2 adrenoceptors are less capable of transducing the effector response (inhibition of adenylyl cyclase).


Subject(s)
Adenylate Cyclase Toxin , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Hypnotics and Sedatives/pharmacology , Imidazoles/pharmacology , Pertussis Toxin , Signal Transduction/drug effects , Virulence Factors, Bordetella/pharmacology , Adenylyl Cyclases/metabolism , Animals , Cyclic AMP/metabolism , Drug Evaluation, Preclinical , Drug Tolerance , GTP-Binding Proteins/metabolism , Infusion Pumps, Implantable , Medetomidine , Radioligand Assay , Rats , Receptors, Adrenergic, alpha-2/metabolism , Time Factors
14.
J Pharmacol Exp Ther ; 283(3): 993-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9399968

ABSTRACT

Rats were made tolerant to the hypnotic effects of the alpha-2 adrenergic agonist dexmedetomidine by a 7- or 14-day continuous systemic administration of the same, and the ability of nifedipine to reverse dexmedetomidine tolerance was assessed. Acute administration of nifedipine (10 mg/kg i.p.) restored the hypnotic response to dexmedetomidine in the alpha-2 tolerant rats. Concurrent administration of nifedipine during induction of tolerance, either partially (continuous administration 10 mg/kg/day delivered by minipumps) or completely (twice daily injections, 20 mg/kg s.c.) restored hypnotic responsiveness to control levels. Induction of tolerance reduced the affinity of [3H]PN200-110 for the L-type calcium channel. Chronically administered nifedipine treatment (20 mg/kg s.c. twice daily), at doses that partially restored the behavioral response to normal, did not change ligand binding affinity of [3H]PN200-110. An increase in Bmax for [3H]PN200-110 was noted in the dexmedetomidine tolerant state which did not change with chronic nifedipine. In naive rats, the phosphodiesterase inhibitor rolipram (275 microg/kg i.p.), mimicked the state of tolerance, as it resulted in a decreased hypnotic response to dexmedetomidine. Nifedipine (10 mg/kg i.p.) also reversed the rolipram-induced attenuation of the hypnotic response to dexmedetomidine. These data implicate a role for the L-type calcium channel in the mechanism of the hypnotic response in alpha-2 tolerant rats and suggest the involvement of the cAMP pathway.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Hypnotics and Sedatives/pharmacology , Imidazoles/pharmacology , Nifedipine/pharmacology , Animals , Calcium Channels/drug effects , Calcium Channels, L-Type , Cyclic AMP/physiology , Male , Medetomidine , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram , Tetrodotoxin/pharmacology
15.
Eur J Pharmacol ; 336(2-3): 137-41, 1997 Oct 08.
Article in English | MEDLINE | ID: mdl-9384225

ABSTRACT

The site of action and the pathways which are activated by nitrous oxide (N2O) to produce an analgesic effect are not well defined. Experiments were designed to determine whether N2O produces analgesia by activating opiate receptors or alpha2-adrenoceptors in periaqueductal gray. The analgesic effect of N2O was determined using the tail flick response to noxious radiant heat in lightly anesthetized rats. Different antagonists were bilaterally microinjected into ventrolateral periaqueductal gray to determine whether the analgesic effect produced by N2O was reversed. The increase in the tail flick latencies produced by N2O was reversed by bilateral microinjection into the ventrolateral part of periaqueductal gray with the opiate receptor antagonist naloxone 2.5 microg/0.5 microl, but not with the alpha2-adrenoceptors antagonist yohimbine 1.5 microg/0.5 microl. These results indicate that the N2O analgesic effect is mediated by activation of opiate receptors, but not alpha2-adrenoceptors, in the periaqueductal gray. Combined with the previous experiments that the N2O analgesic effect is reversed by intrathecal injection of an alpha2-adrenoceptor antagonist but not by an opiate receptor antagonist, it seems likely that N2O causes activation of the opiate receptors in the periaqueductal gray, which in turn activate the noradrenergic descending pathways to the spinal cord to produce the analgesic effect.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nitrous Oxide/pharmacology , Pain Threshold/drug effects , Periaqueductal Gray/drug effects , Receptors, Opioid/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Injections, Intraventricular , Male , Microinjections , Nitrous Oxide/antagonists & inhibitors , Pain Threshold/physiology , Periaqueductal Gray/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/physiology , Yohimbine/pharmacology
16.
Anesthesiology ; 85(4): 846-52, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8873555

ABSTRACT

BACKGROUND: Despite nearly 150 years of clinical use, the mechanism(s) of action of nitrous oxide (N2O) remains in doubt. In some but not all studies the analgesic properties of N2O can be attenuated by opiate receptor antagonists. The purported mechanism for the opiate antagonistic effect relates to the finding that N2O increases supraspinal levels of endogenous opiates, although this finding has been disputed. Based on the observations that (1) N2O promotes the release of catecholamines, including the endogenous alpha 2 adrenergic agonist norepinephrine, and (2) that descending noradrenergic inhibitory pathways are activated by opioid analgesics, this study sought to determine whether alpha 2 adrenergic receptors are involved in the antinociceptive action of nitrous oxide. METHODS: Institutional approval was obtained for the study. Rats breathed 70% N2O and 30% O2 in an enclosed chamber. After a 30-min exposure, significant antinociception was indicated by an increase in the latency response to a noxious stimulus (tail-flick latency). The tail-flick latency was tested in rats exposed to 70% N2O after either systemic or regional (intrathecal or intracerebroventricular) injections with either competitive (atipamezole; yohimbine) or noncompetitive (N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline) alpha 2 adrenoceptor antagonists, or the opiate receptor antagonist naloxone. RESULTS: When administered systemically, both the opiate (naloxone) and alpha 2 adrenoceptor antagonists (atipamezole, yohimbine, and N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline) blocked the enhanced tail-flick latency response to N2O-Naloxone administered intracerebroventricularly, but not intrathecally, blocked the enhanced tail-flick latency response to N2O. Conversely, atipamezole administered intrathecally, but not intracerebroventricularly, blocked the enhanced tail-flick latency response to N2O. CONCLUSIONS: These data suggest that both supraspinal opiate and spinal alpha 2 adrenoceptors play a mediating role in the antinociceptive response to N2O in rats. A possible mechanism may involve a descending inhibitory noradrenergic pathway that may be activated by opiate receptors in the periaqueductal gray region of the brain stem in the rat after exposure to N2O.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Analgesics/pharmacology , Nitrous Oxide/pharmacology , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Opioid/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Imidazoles/pharmacology , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociceptors/drug effects , Nociceptors/physiology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/physiology , Receptors, Opioid/physiology , Spinal Cord/drug effects , Spinal Cord/physiology , Yohimbine/pharmacology
17.
Anesthesiology ; 85(3): 565-73, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8853087

ABSTRACT

BACKGROUND: The development of tolerance to the sympatholytic and anesthetic-reducing effects of alpha(2) agonists after prolonged administration of dexmedetomidine and how the number of available alpha(2) adrenoceptors affects these dexmedetomidine-induced responses was studied. METHODS: The sympatholytic action of acute and chronic (3 and 10 micrograms.kg-1.h-1 for 7 days) dexmedetomidine, was assessed by the decrease in norepinephrine turnover in the locus coeruleus and hippocampus. The anesthetic-reducing effect of chronic (7 days) dexmedetomidine (5 and 10 micrograms.kg-1.h-1) was studied by determining the minimum alveolar concentration (MAC) for halothane that prevented rats from responding to a supramaximal noxious stimulus of dexmedetomidine (10 or 30 micrograms.kg-1), doses in the steep part of the dose-response curve. The receptor reserve for the norepinephrine turnover and anesthetic-sparing responses to dexmedetomidine was delineated with 0.3-1.0 mg.kg-1 N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline, an irreversible alkylating agent. RESULTS: After chronic administration of dexmedetomidine at both doses, acute dexmedetomidine significantly decreased norepinephrine turnover in the hippocampus and locus coeruleus. The baseline minimum anesthetic concentration (MAC) and the MAC-sparing effect to acutely administered dexmedetomidine were preserved after chronic dexmedetomidine treatment. In the N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline experiments, the dexmedetomidine-induced norepinephrine turnover effect required less than 20% and greater than 4% alpha(2) adrenoceptor availability in the locus coeruleus and the dexmedetomidine induced MAC-sparing effect required less than 40% and greater than 20% alpha(2) adrenoceptor availability in the locus coeruleus. CONCLUSION: Tolerance does not develop for either the sympatholytic or MAC-sparing actions of dexmedetomidine, although it is present for the hypnotic response. The durable quality of the sympatholytic and MAC-sparing responses to dexmedetomidine after chronic treatment is explained by a comparatively larger receptor reserve than is needed for the hypnotic and analgesic responses, which are blunted by the same drug treatment regimen.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Analgesics/pharmacology , Anesthetics/pharmacology , Hypnotics and Sedatives/pharmacology , Imidazoles/pharmacology , Sympatholytics/pharmacology , Animals , Drug Tolerance , Halothane/pharmacokinetics , Halothane/pharmacology , Imidazoles/pharmacokinetics , Male , Medetomidine , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley
18.
Anesth Analg ; 83(3): 606-10, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8780290

ABSTRACT

Combinations of alpha 2 agonists and opiates are used in the clinical management of pain to harness their potential synergistic interaction for analgesia while limiting their side-effects. To better predict the clinical consequences of this combination, we studied the behavioral effects of dexmedetomidine, a highly selective alpha 2 agonist with analgesic and hypnotic properties, during the development of, and recovery from, morphine tolerance. Rats were implanted with morphine pellets (or placebo), daily for 5 days. The analgesic response to morphine, dexmedetomidine, or the combination of the two drugs was assessed with the tail-flick latency response. The hypnotic response to dexmedetomidine, and to the combination of dexmedetomidine and morphine, was measured by the duration of the loss of righting reflex (sleep-time). One day after the last morphine pellet implantation, alpha 2 adrenoceptor binding was assessed in vitro in the locus coeruleus (LC) and the spinal cord (SC). Data were analyzed by analysis of variance (ANOVA), t-test, or Mann-Whitney test. The morphine tolerance was present after 1 day of morphine administration. At Days 1 and 3 of morphine administration, the hypnotic and analgesic responses to dexmedetomidine were significantly increased. After 5 days of morphine treatment, the analgesic response to dexmedetomidine was unaltered, while the hypnotic response to dexmedetomidine was now significantly decreased. The kd for the alpha 2 adrenoceptors was unaffected while the Bmax was significantly decreased only in the SC. Acutely administered morphine significantly enhanced the hypnotic and analgesic effects of dexmedetomidine in naive rats but not in morphine-tolerant rats. During morphine withdrawal, the hypnotic response to dexmedetomidine normalized; however, the analgesic response to dexmedetomidine was significantly decreased 5 days after withdrawal before returning to normal at Day 10 after withdrawal. We conclude that in the development of, and recovery from, the morphine-tolerant state, the hypnotic and analgesic responses to alpha 2 agonists are asynchronous.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Analgesics, Non-Narcotic/pharmacology , Hypnotics and Sedatives/pharmacology , Imidazoles/pharmacology , Morphine/pharmacology , Animals , Drug Synergism , Drug Tolerance , Locus Coeruleus/metabolism , Male , Medetomidine , Pain Measurement , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/metabolism , Spinal Cord/metabolism , Time Factors
19.
Eur J Pharmacol ; 306(1-3): 51-9, 1996 Jun 13.
Article in English | MEDLINE | ID: mdl-8813614

ABSTRACT

The role of serotonergic pathways in the hypnotic response to dexmedetomidine was examined in neurochemical and behavioral studies. Following acute administration of dexmedetomidine, loss of righting reflex and changes in serotonin (5-hydroxytryptamine, 5-HT) and norepinephrine turnover in different brain regions (locus coeruleus and hippocampus) were assessed. In separate experiments, the effect of dexmedetomidine on 5-HT turnover was measured in rats rendered tolerant to the hypnotic effects of dexmedetomidine. These neurochemical data were complemented by a study of dexmedetomidine-induced hypnotic response in the presence of a 5-HT2 receptor agonist and antagonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) and ritanserin, respectively. Dexmedetomidine (1-500 micrograms.kg-1) dose dependently reduced 5-HT and norepinephrine turnover in both the locus coeruleus and hippocampus. The decrease in 5-HT turnover more closely correlated with the dose-response curve for loss of righting reflex, a behavioral measure of hypnosis, than did the norepinephrine turnover. In previous studies with chronic administration of dexmedetomidine (3 micrograms.kg-1.h-1 for 7 days), the norepinephrine turnover effect of acute dexmedetomidine (30 micrograms.kg-1) persisted while the hypnotic effect was blunted. Following the same regimen, the drug's ability to diminish 5-HT turnover was also blunted. This biochemical evidence for the role of 5-HT in sleep was supported by the behavioral evidence that dexmedetomidine (100 micrograms.kg-1 i.p. or 7 micrograms.0.2 microliter-1 locus coeruleus)-induced hypnosis was dose dependently blocked by DOI (0.08-0.32 mg.kg-1 i.p.). The selectivity of this effect was demonstrated by the finding that ritanserin (0.16 mg.kg-1 i.p.) pretreatment blocked the effects of DOI (0.16 mg.kg-1 i.p.) on dexmedetomidine (100 micrograms.kg-1 i.p. or 7 micrograms.0.2 microliter-1 locus coeruleus)-induced loss of righting reflex. In conclusion, these findings suggest that the hypnotic effect of the alpha 2-adrenoceptor agonist, dexmedetomidine, is not mediated solely by changes in noradrenergic neurtransmission, but instead is strongly associated with a decrease in serotonergic neurotransmission and correspondingly diminished by stimulation of 5-HT2 receptors.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Hippocampus/drug effects , Imidazoles/pharmacology , Locus Coeruleus/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Serotonin/metabolism , Amphetamines/pharmacology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Hippocampus/metabolism , Imidazoles/agonists , Imidazoles/antagonists & inhibitors , Locus Coeruleus/metabolism , Male , Medetomidine , Neurotransmitter Agents/physiology , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Ritanserin/pharmacology , Sleep/drug effects
20.
Anesthesiology ; 84(4): 873-81, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8638842

ABSTRACT

BACKGROUND: Alpha(2)-Adrenergic agonists such as clonidine and dexmedetomidine are known to produce sedation and analgesia in humans. The sedative effect of these agents is thought to occur through supraspinal pathways, involving the locus ceruleus (LC) and its projections in rats. While the antinociceptive response to alpha(2) agonists, given intrathecally, is mediated predominantly in the spinal cord, other sites of action have not been systematically studied. The authors examined whether alpha(2)-adrenergic receptors in the LC mediate an antinociceptive effect. METHODS: For administration of different drugs into the LC, guide cannulas were placed with their tips in the LC in male Sprague-Dawley rats. Dexmedetomidine (3.5 micrograms/0.2 microliter) was microinjected into the LC through the cannula, or given systemically by intraperitoneal injecton (50 micrograms/kg). The antinociceptive effect of dexmedetomidine was measured using the tail-flick latency response. To determine the sites through which dexmedetomidine injection into the LC produces antinociception, the authors examined whether this response could be perturbed by the specific alpha(2)-adrenergic antagonists atipamezole and L659,066 and pertussis toxin administered either into the LC or intrathecally before injection of dexmedetomidine systemically or directly into the LC. To eliminate the possibility that drug administered in one site (LC or intrathecal) could reach the other site, the dispositional characteristics of radiolabeled dexmedetomidine (LC) or atipamezole (intrathecal) were studied. RESULTS: Dexmedetomidine placed into the LC produces a dose-dependent increase in the tail-flick latency. This antinociceptive effect was blocked by pertussis toxin and by the alpha(2) antagonists atipamezole and L659,066 placed in the LC. Intrathecal administration of atipamezole and pertussis toxin also blocked the antinociceptive effect of dexmedetomidine placed in the LC. (3)H-dexmedetomidine introduced into the LC did not reach the spinal cord in pharmacologically active concentrations; also, intrathecally administered (3)H-atipamezole did not reach the LC in appreciable amounts. The systemic administration of dexmedetomidine produced an increase in tail-flick latency, and this effect was attenuated by the injection of atipamezole and L659,066 into the LC. CONCLUSIONS: Part of the mechanism by which dexmedetomidine produces an antinociceptive effect is by an action directly on the LC, demonstrated by these studies in which antinociception produced by injection of this drug into the LC can be blocked by specific alpha(2) antagonists injected into the LC. Furthermore, the action of dexmedetomidine in the LC in turn may result in an increase in activation of alpha(2) adrenoceptors in the spinal cord, because the antinociceptive effect of LC dexmedetomidine injection also can be blocked by intrathecal injection of antipamezole and pertussis toxin.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Analgesics, Non-Narcotic/pharmacology , Imidazoles/pharmacology , Locus Coeruleus/drug effects , Animals , Imidazoles/administration & dosage , Male , Medetomidine , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...