Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
CNS Neurosci Ther ; 22(2): 146-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663888

ABSTRACT

AIMS: Acute postoperative pain remains a significant healthcare issue. Historically, the assessment of postoperative pain in rodents has relied on evoked withdrawal or reflexive measures. Using a recently developed, anatomically relevant rat model of acute postoperative pain (J Pain, 16, 2015, 421), the present experiments sought to investigate the affective component of acute postoperative pain associated with inguinal hernia repair. METHODS: Male Lister hooded rats underwent surgery to model Lichtenstein inguinal hernia repair (without hernia induction), or a sham procedure. Postsurgical characterization involved a modified place escape/avoidance paradigm (mPEAP), as well as home cage and open field locomotor activity monitoring. In pharmacological validation studies, rats received either morphine or carprofen prior to mPEAP testing. RESULTS: Surgery was associated with a significantly increased proportion of the trial duration in the light compartment of the mPEAP arena, in avoidance of the noxious stimulus, compared with sham animals. When retested in the mPEAP at day 7 postsurgery, there was no difference between sham and surgery animals for time spent in either compartment, but surgery animals displayed a persistent increase in the percentage response to noxious stimulation. Morphine and carprofen treatment in surgery animals reduced escape/avoidance behavior at discrete time points over the trial. Surgery-induced reductions in home cage and open field locomotor activity were also observed. CONCLUSION: The present studies report for the first time the characterization of the affective component of acute postoperative pain using the mPEAP in a rodent model, which may facilitate development of improved understanding and treatment of postoperative pain.


Subject(s)
Acute Pain/psychology , Disease Models, Animal , Hernia, Inguinal/psychology , Hernia, Inguinal/surgery , Pain Measurement/psychology , Pain, Postoperative/psychology , Acute Pain/drug therapy , Acute Pain/etiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Carbazoles/pharmacology , Carbazoles/therapeutic use , Hernia, Inguinal/drug therapy , Male , Pain Measurement/drug effects , Pain Measurement/methods , Pain, Postoperative/drug therapy , Pain, Postoperative/etiology , Rats
2.
J Pain ; 16(5): 421-35.e1-6, 2015 May.
Article in English | MEDLINE | ID: mdl-25640291

ABSTRACT

UNLABELLED: Acute postoperative pain remains a significant health care issue. Development of anatomically relevant animal models of postoperative pain, with improved predictive validity, would advance understanding of postoperative pain mechanisms and improve treatment outcomes. This study aimed to develop, characterize, and validate a rat model of acute postoperative pain associated with inguinal hernia repair based on the Lichtenstein inguinal hernia repair procedure (without hernia induction). We hypothesized that the surgery would result in reduced spontaneous locomotor activity, which would represent a pain-related phenotype. Postsurgical characterization involved extensive monitoring of home cage and open field locomotor activity, as well as mechanical hypersensitivity and assessment of c-Fos expression in the dorsal horn of the spinal cord. In pharmacologic validation studies, rats received morphine, carprofen, or paracetamol 1 hour before, and/or immediately after, surgery. Rats that underwent hernia repair surgery exhibited significantly lower horizontal and vertical activities in the home cage and open field in the early postsurgical period, compared with sham rats or rats that underwent skin incision only. Morphine, carprofen, and paracetamol attenuated the surgery-induced reductions in locomotor activity, to varying degrees. Surgery was associated with significantly increased c-Fos expression in the ipsilateral dorsal horn of the spinal cord, an effect attenuated by carprofen treatment. These results support the development and characterization of a novel, anatomically relevant animal model of acute postoperative pain that may facilitate development of improved treatment regimens. PERSPECTIVE: Acute pain following inguinal hernia repair can be difficult to treat. Here we report, for the first time, the development of a novel, anatomically relevant rat model to facilitate improved understanding and treatment of acute postoperative pain following inguinal hernia repair.


Subject(s)
Acute Pain/physiopathology , Analgesics/pharmacology , Behavior, Animal/physiology , Disease Models, Animal , Herniorrhaphy , Motor Activity/physiology , Pain, Postoperative/physiopathology , Spinal Cord Dorsal Horn/metabolism , Acetaminophen/administration & dosage , Acetaminophen/pharmacology , Acute Pain/drug therapy , Analgesics/administration & dosage , Animals , Behavior, Animal/drug effects , Carbazoles/administration & dosage , Carbazoles/pharmacology , Genes, fos/physiology , Hernia, Inguinal/surgery , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Morphine/administration & dosage , Morphine/pharmacology , Motor Activity/drug effects , Nociceptors/physiology , Pain, Postoperative/drug therapy , Phenotype , Rats
3.
Methods Mol Biol ; 617: 67-78, 2010.
Article in English | MEDLINE | ID: mdl-20336414

ABSTRACT

Creating a robust and unbiased assay for the study of current and novel analgesics has been a daunting task. Traditional rodent models of pain and inflammation typically rely on a negative reaction to various forms of evoked stimuli to elicit a pain response and are subject to rater interpretation. Recently, models such as weight bearing and gait analysis have been developed to address these drawbacks while detecting a drug's analgesic properties. We have recently developed the Reduction of Spontaneous Activity by Adjuvant (RSAA) model as a quick, unbiased method for the testing of potential analgesics. Rats, following prior administration of an activity-decreasing inflammatory insult, will positively increase spontaneous locomotor exploration when given single doses of known analgesics. The RSAA model capitalizes on a rat's spontaneous exploratory behavior in a novel environment with the aid of computer tracking software to quantify movement and eliminate rater bias.


Subject(s)
Exploratory Behavior/physiology , Inflammation/physiopathology , Motor Activity/physiology , Pain/physiopathology , Amphetamine/pharmacology , Amphetamine/therapeutic use , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Arthritis, Experimental/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/therapeutic use , Disease Models, Animal , Environment , Exploratory Behavior/drug effects , Male , Morphine/pharmacology , Morphine/therapeutic use , Motor Activity/drug effects , Pain/drug therapy , Pain Measurement/instrumentation , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
4.
Pain ; 142(1-2): 59-67, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135800

ABSTRACT

We have developed a model in which inflammation contiguous to and within a dorsal root ganglion (DRG) was generated by local application of complete Freund's adjuvant (CFA) to the L4 lumbar spinal nerve as it exits from the intervertebral foramen. The periganglionic inflammation (PGI) elicited a marked reduction in withdrawal threshold to mechanical stimuli and an increase in heat pain sensitivity in the ipsilateral hindpaw in the absence of any hindpaw inflammation. The pain sensitivity appeared within hours and lasted for a week. The PGI also induced a prominent increase in IL-1beta and TNF-alpha levels in the DRG and of cyclooxygenase-2 (COX-2) expression in neurons and satellite cells. A selective COX-2 inhibitor reduced the PGI-induced hyperalgesia. We also show that IL-1beta induces COX-2 expression and prostaglandin release in DRG neurons in vitro in a MAP kinase-dependent fashion. The COX-2 induction was prevented by ERK and p38 inhibitors. We conclude that periganglionic inflammation increases cytokine levels, including IL-1beta, leading to the transcription of COX-2 and prostaglandin production in the affected DRG, and thereby to the development of a dermatomally distributed pain hypersensitivity.


Subject(s)
Cyclooxygenase 2/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/etiology , Inflammation/complications , Pain Threshold/physiology , Analysis of Variance , Animals , Autonomic Fibers, Preganglionic/pathology , Behavior, Animal/drug effects , Biophysics , Cyclooxygenase 2/genetics , Cytokines/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Freund's Adjuvant/adverse effects , Ganglia, Spinal/drug effects , Gene Expression/drug effects , Gene Expression/physiology , In Vitro Techniques , Lactones/pharmacology , Male , Pain Measurement , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Sciatica/complications , Sulfones/pharmacology
5.
J Pharmacol Exp Ther ; 327(3): 620-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18772321

ABSTRACT

Recent evidence suggests that the P2X(7) receptor may play a role in the pathophysiology of preclinical models of pain and inflammation. Therefore, pharmacological agents that target this receptor may potentially have clinical utility as anti-inflammatory and analgesic therapy. We investigated and characterized the previously reported P2X(7) antagonist N-(adamantan-1-ylmethyl)-5-[(3R-amino-pyrrolidin-1-yl)methyl]-2-chloro-benzamide, hydrochloride salt (AACBA; GSK314181A). In vitro, AACBA was a relatively potent inhibitor of both human P2X(7)-mediated calcium flux and quinolinium,4-[(3-methyl-2(3H)-benzoxazolylidene)methyl]-1-[3-(triemethylammonio)propyl]-diiodide (YO-PRO-1) uptake assays, with IC(50) values of approximately 18 and 85 nM, respectively. Compared with the human receptor, AACBA was less potent at the rat P2X(7) receptor, with IC(50) values of 29 and 980 nM in the calcium flux and YO-PRO-1 assays, respectively. In acute in vivo models of pain and inflammation, AACBA dose-dependently reduced lipopolysaccharide-induced plasma interleukin-6 release and prevented or reversed carrageenan-induced paw edema and mechanical hypersensitivity. In chronic in vivo models of pain and inflammation, AACBA produced a prophylactic, but not therapeutic-like, prevention of the clinical signs and histopathological damage of collagen-induced arthritis. Finally, AACBA could not reverse L(5) spinal nerve ligation-induced tactile allodynia when given therapeutically. Consistent with previous literature, these results suggest that P2X(7) receptors do play a role in animal models of pain and inflammation. Further study of P2X(7) antagonists both in preclinical and clinical studies will help elucidate the role of the P2X(7) receptor in pain and inflammatory mechanisms and may help identify potential clinical benefits of such molecules.


Subject(s)
Adamantane/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzamides/pharmacology , Inflammation/drug therapy , Pain/drug therapy , Purinergic P2 Receptor Antagonists , Adamantane/pharmacology , Animals , Arthritis/chemically induced , Arthritis/drug therapy , Benzoxazoles , Calcium/metabolism , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Quinolinium Compounds , Rats , Receptors, Purinergic P2X7
6.
Expert Opin Drug Discov ; 3(9): 1099-108, 2008 Sep.
Article in English | MEDLINE | ID: mdl-23506182

ABSTRACT

BACKGROUND: Translating promising analgesic compounds into reliable pain therapeutics in humans is made particularly challenging by the difficulty in measuring the pain quantitatively. This problem is manifest not only in clinical settings in which patient pain assessments involve mostly subjective measures but also in preclinical settings wherein laboratory animals, most commonly rodents, are typically evaluated in stimulus-evoked response tests. OBJECTIVE: Given the limitations of traditional pain tests, we sought out new approaches to measure pain, and analgesia, in laboratory animals. METHODS: We reviewed the peer reviewed literature to identify pain tests that could be utilized in preclinical settings to understand the effects of new and established analgesics. RESULTS/CONCLUSIONS: The tests identified include weight bearing differential, suppression of feeding, reduction in locomotor activity, gait analysis, conditioning models and functional MRI. Although the pharmacology of known and new analgesics has not been broadly established in these models, they hold the promise of better predictive utility for the discovery of pain relievers.

7.
Biochim Biophys Acta ; 1772(8): 978-88, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17467247

ABSTRACT

Since the molecular identification of the capsaicin receptor, now known as TRPV1, transient receptor potential (TRP) channels have occupied an important place in the understanding of sensory nerve function in the context of pain. Several TRP channels exhibit sensitivity to substances previously known to cause pain or pain-like sensations; these include cinnamaldehyde, menthol, gingerol, and icillin. Many TRP channels also exhibit significant sensitivity to increases or decreases in temperature. Some TRP channels are sensitized in vitro by the activation of other receptors such that these channels may be activated by processes, such as inflammation that result in pain. TRP channels are suggested to be involved in processes as diverse as sensory neuron activation events, neurotransmitter release and action in the spinal cord, and release of inflammatory mediators. These functions strongly suggest that specific and selective inhibition of TRP channel activity will be of use in alleviating pain.


Subject(s)
Pain/physiopathology , Transient Receptor Potential Channels/physiology , Animals , Body Temperature Regulation/physiology , Humans , Inflammation/physiopathology , Models, Biological , Nociceptors/physiology , Pain/etiology , Phospholipids/metabolism , Spinal Cord/physiology , Stress, Mechanical
8.
J Pharmacol Exp Ther ; 320(1): 194-201, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050782

ABSTRACT

The majority of rodent models used to evaluate analgesic drug effects rely on evoked measures of nociceptive thresholds as primary outcomes. These approaches are often time-consuming, requiring extensive habituation sessions and repeated presentations of eliciting stimuli, and are prone to false-positive outcomes due to sedation or tester subjectivity. Here, we describe the reduction of spontaneous activity by adjuvant (RSAA) model as an objective and quantifiable behavioral model of inflammatory pain that can predict the analgesic activity of a variety of agents following single-dose administration. In the RSAA model, activity was measured in nonhabituated rats using standard, photocell-based monitors. Bilateral inflammation of the knee joints by complete Freund's adjuvant (CFA) reduced the normal level of activity (horizontal locomotion and vertical rearing) by approximately 60% in a novel environment. This reduction in activity was dose-dependently reversed by ibuprofen, rofecoxib, celecoxib, piroxicam, and dexamethasone, whereas gabapentin and amitriptyline were inactive. Morphine significantly reversed the activity-suppressing effects of CFA, at 1 mg/kg s.c., but at higher doses locomotor activity progressively declined, coincident with the induction of sedation. In contrast to morphine and anti-inflammatory therapies, amphetamine did not affect vertical rearing, even though it increased horizontal locomotion. Thus, unlike standard measures of analgesia such as alteration in thermal or mechanical sensitivity, the RSAA model operationally defines analgesia as a drug-induced increase in spontaneous behavior (vertical rearing in a novel environment). We conclude that the RSAA model is valuable as an objective measure of analgesic efficacy that is not dependent on an evoked stimulus response.


Subject(s)
Analgesics/pharmacology , Freund's Adjuvant/pharmacology , Inflammation/psychology , Motor Activity/drug effects , Analgesia , Animals , Carrageenan/pharmacology , Celecoxib , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Kaolin/pharmacology , Male , Models, Animal , Morphine/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology
9.
Neuron ; 49(3): 365-77, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16446141

ABSTRACT

In mammals, the perception of pain is initiated by the transduction of noxious stimuli through specialized ion channels and receptors expressed by nociceptive sensory neurons. The molecular mechanisms responsible for the specification of distinct sensory modality are, however, largely unknown. We show here that Runx1, a Runt domain transcription factor, is expressed in most nociceptors during embryonic development but in adult mice, becomes restricted to nociceptors marked by expression of the neurotrophin receptor Ret. In these neurons, Runx1 regulates the expression of many ion channels and receptors, including TRP class thermal receptors, Na+-gated, ATP-gated, and H+-gated channels, the opioid receptor MOR, and Mrgpr class G protein coupled receptors. Runx1 also controls the lamina-specific innervation pattern of nociceptive afferents in the spinal cord. Moreover, mice lacking Runx1 exhibit specific defects in thermal and neuropathic pain. Thus, Runx1 coordinates the phenotype of a large cohort of nociceptors, a finding with implications for pain therapy.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Neurons, Afferent/physiology , Nociceptors/physiology , Pain/physiopathology , Thermosensing/physiology , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal , Calcitonin Gene-Related Peptide/metabolism , Cell Count/methods , Core Binding Factor Alpha 2 Subunit/deficiency , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry/methods , In Situ Hybridization/methods , Ion Channels/classification , Ion Channels/metabolism , Lectins/metabolism , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Pain/genetics , Pain Measurement/methods , Pain Threshold/physiology , Physical Stimulation/adverse effects , Protein Kinase C/metabolism , Receptor, trkA/metabolism , Time Factors , Ubiquitin-Protein Ligases , Wnt1 Protein/genetics
10.
Mol Pain ; 1: 36, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16354295

ABSTRACT

BACKGROUND: Pain is elicited by cold, and a major feature of many neuropathic pain states is that normally innocuous cool stimuli begin to produce pain (cold allodynia). To expand our understanding of cold induced pain states we have studied cold pain behaviors over a range of temperatures in several animal models of chronic pain. RESULTS: We demonstrate that a Peltier-cooled cold plate with +/- 1 degrees C sensitivity enables quantitative measurement of a detection withdrawal response to cold stimuli in unrestrained rats. In naïve rats the threshold for eliciting cold pain behavior is 5 degrees C. The withdrawal threshold for cold allodynia is 15 degrees C in both the spared nerve injury and spinal nerve ligation models of neuropathic pain. Cold hyperalgesia is present in the spared nerve injury model animals, manifesting as a reduced latency of withdrawal response threshold at temperatures that elicit cold pain in naïve rats. We also show that following the peripheral inflammation produced by intraplantar injection of complete Freund's adjuvant, a hypersensitivity to cold occurs. CONCLUSION: The peltier-cooled provides an effective means of assaying cold sensitivity in unrestrained rats. Behavioral testing of cold allodynia, hyperalgesia and pain will greatly facilitate the study of the neurobiological mechanisms involved in cold/cool sensations and enable measurement of the efficacy of pharmacological treatments to reduce these symptoms.


Subject(s)
Cold Temperature/adverse effects , Hyperalgesia/diagnosis , Pain Measurement/methods , Pain/diagnosis , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Behavior, Animal/physiology , Denervation , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/complications , Inflammation/physiopathology , Ligation , Male , Nociceptors/drug effects , Nociceptors/physiology , Pain/chemically induced , Pain/physiopathology , Pain Measurement/standards , Pain Threshold/drug effects , Pain Threshold/physiology , Peripheral Nerve Injuries , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiopathology , Spinal Nerves/injuries , Spinal Nerves/physiopathology , Wakefulness/physiology
11.
Hum Mol Genet ; 14(22): 3507-21, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16210380

ABSTRACT

Mutations in enzymes involved in sphingolipid metabolism and trafficking cause a variety of neurological disorders, but details of the molecular pathophysiology remain obscure. SPTLC1 encodes one subunit of serine palmitoyltransferase (SPT), the rate-limiting enzyme in sphingolipid synthesis. Mutations in SPTLC1 cause hereditary sensory and autonomic neuropathy (type I) (HSAN1), an adult onset, autosomal dominant neuropathy. HSAN1 patients have reduced SPT activity. Expression of mutant SPTLC1 in yeast and mammalian cell cultures dominantly inhibits SPT activity. We created transgenic mouse lines that ubiquitously overexpress either wild-type (SPTLC1(WT)) or mutant SPTLC1 (SPTLC1(C133W)). We report here that SPTLC1(C133W) mice develop age-dependent weight loss and mild sensory and motor impairments. Aged SPTLC1(C133W) mice lose large myelinated axons in the ventral root of the spinal cord and demonstrate myelin thinning. There is also a loss of large myelinated axons in the dorsal roots, although the unmyelinated fibers are preserved. In the dorsal root ganglia, IB4 staining is diminished, whereas expression of the injury-induced transcription factor ATF3 is increased. These mice represent a novel mouse model of peripheral neuropathy and confirm the link between mutant SPT and neuronal dysfunction.


Subject(s)
Aging/genetics , Genes, Dominant , Hereditary Sensory and Autonomic Neuropathies/enzymology , Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation , Serine C-Palmitoyltransferase/genetics , Animals , Axons/pathology , Behavior, Animal/physiology , CHO Cells , Cricetinae , Cricetulus , Female , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Male , Mice , Mice, Transgenic , Pancreas, Exocrine/pathology , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/metabolism , Transfection
12.
J Neurosci ; 25(32): 7317-23, 2005 Aug 10.
Article in English | MEDLINE | ID: mdl-16093381

ABSTRACT

We show that transsynaptic apoptosis is induced in the superficial dorsal horn (laminas I-III) of the spinal cord by three distinct partial peripheral nerve lesions: spared nerve injury, chronic constriction, and spinal nerve ligation. Ongoing activity in primary afferents of the injured nerve and glutamatergic transmission cause a caspase-dependent degeneration of dorsal horn neurons that is slow in onset and persists for several weeks. Four weeks after spared nerve injury, the cumulative loss of dorsal horn neurons, determined by stereological analysis, is >20%. GABAergic inhibitory interneurons are among the neurons lost, and a marked decrease in inhibitory postsynaptic currents of lamina II neurons coincides with the induction of apoptosis. Blocking apoptosis with the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone (zVAD) prevents the loss of GABAergic interneurons and the reduction of inhibitory currents. Partial peripheral nerve injury results in pain-like behavioral changes characterized by hypersensitivity to tactile or cold stimuli. Treatment with zVAD, which has no intrinsic analgesic properties, attenuates this neuropathic pain-like syndrome. Preventing nerve injury-induced apoptosis of dorsal horn neurons by blocking caspase activity maintains inhibitory transmission in lamina II and reduces pain hypersensitivity.


Subject(s)
Apoptosis , Caspases/metabolism , Neural Inhibition , Neurons , Posterior Horn Cells , Sciatic Nerve/injuries , Synapses , Afferent Pathways/physiopathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Male , Neural Inhibition/drug effects , Neuralgia/psychology , Rats , Rats, Sprague-Dawley , Spinal Cord/physiopathology , Wounds and Injuries/physiopathology
13.
Psychopharmacology (Berl) ; 164(1): 42-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12373418

ABSTRACT

RATIONALE: Non-peptidic delta-opioid receptor agonists possess antidepressant-like activity in the forced swim assay in the rat. These compounds have also previously been shown to possess convulsant properties in mice. OBJECTIVE: The aim of the present study was to examine whether such convulsions occurred in rats and to investigate if delta-mediated convulsant activity was necessary for the mediation of delta-opioid agonist-induced antidepressant-like activity. METHODS: The peripheral administration of delta-opioid receptor agonists to male Sprague-Dawley rats was followed by a period of observation for convulsant activity. Following this period and 60 min after delta-opioid agonist administration, rats were tested in the forced swim assay. RESULTS: The non-peptidic delta-opioid receptor agonists (+)-4-[(R)-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC80) and (+)-4-[(R)-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-hydroxyphenyl)methyl]-N,N-diethylbenzamide dihydrochloride [(+)BW373U86] both produced dose-dependent convulsant activity in rats and decreased immobility in the forced swim assay. The delta-opioid receptor antagonist naltrindole prevented the convulsant activity of (+)BW373U86 and its effects in the forced swim assay. This suggested a delta-opioid mechanism for both effects. Midazolam prevented convulsions but did not prevent activity in the forced swim assay. Rats tolerant to the convulsive effects of (+)BW373U86 still displayed antidepressant-like effects. CONCLUSION: delta-Mediated convulsions do occur in rats and can be prevented without affecting the delta-mediated effects in the forced swim assay. Therefore the convulsant activity of (+)BW373U86 and possibly other non-peptidic delta-agonists is not required for activity in the forced swim assay.


Subject(s)
Antidepressive Agents/pharmacology , Benzamides/pharmacology , Convulsants/pharmacology , Motor Activity/drug effects , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , Animals , Dose-Response Relationship, Drug , Male , Motor Activity/physiology , Narcotics/pharmacology , Organic Chemicals , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/physiology
14.
J Pharmacol Exp Ther ; 303(2): 723-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388657

ABSTRACT

Delta-opioid receptor-selective agonists produce antinociception and convulsions in several species, including mice. This article examines two hypotheses in mice: 1) that antinociception and convulsive activity are mediated through the same type of delta-receptor and 2) that greater delta-agonist efficacy is required for antinociception than for convulsive activity. Delta-mediated antinociception was evaluated in the acetic acid-induced abdominal constriction assay, which involves a low-intensity noxious stimulus; convulsive activity was indicated as a mild tonic-clonic convulsive episode followed by a period of catalepsy. In delta-opioid receptor knockout mice [DOR-1(-/-)], the nonpeptidic delta-agonists (+/-)-4-[(R*)-[(2S*,5R*)-2,5-dimethyl-4-(2-propenyl)-1- piperazinyl]-(3-hydroxyphenyl)methyl]-N,N-diethylbenzamide hydrochloride (BW373U86) and (+)-4-[(R)-[(2S,5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N, N-diethylbenzamide (SNC80) failed to produce convulsive behavior demonstrating the absolute involvement of DOR-1 in this effect. In NIH Swiss mice expressing delta-opioid receptors, BW373U86 produced both antinociception and convulsive activity. These effects were antagonized by the putative delta(1)-receptor-selective antagonist 7-benzylidenenaltrexone and the putative delta(2)-receptor-selective antagonist naltriben. Tolerance developed to both the convulsive and antinociceptive effects of BW373U86. Tolerance to the convulsive, but not the antinociceptive, effects of BW373U86 was largely prevented when the antagonist naltrindole was given 20 min after each dose of the agonist in a 3-day treatment paradigm. The convulsive action of BW373U86 was also less sensitive than the antinociceptive action to treatment with the irreversible delta-antagonist naltrindole isothiocyanate. Collectively, these data suggest that the convulsive and antinociceptive activities of delta-agonists are mediated through the same receptor but that the receptor reserve for delta-mediated convulsive activity is greater than for delta-mediated antinociceptive activity.


Subject(s)
Analgesics/pharmacology , Convulsants/pharmacology , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Analgesics, Opioid/pharmacology , Animals , Benzamides/pharmacology , Benzylidene Compounds/pharmacology , Drug Tolerance , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Male , Mice , Mice, Knockout , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Piperazines/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/genetics
15.
Jpn J Pharmacol ; 90(1): 1-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12396021

ABSTRACT

The development of selective delta-opioid receptor agonists has revealed some very intriguing behavioral properties. delta-Opioid agonists have antinociceptive, seizuregenic and convulsive properties. A number of studies have identified a novel behavioral effect of delta-opioid-receptor agonists, implicating a role for the delta-opioid receptor in depression. Early clinical experiments demonstrated that exogenously administered opioid peptides had antidepressant activity in human patients. Also, enkephalinase inhibitors, which prevent the degradation of endogenous enkephalins, produced antidepressant-like effects mediated through the delta-opioid receptor in animal models of depression. More recently, the selective non-peptidic delta-opioid agonists SNC80 and (+)BW373U86 demonstrated antidepressant-like activity in the forced swim assay in rats. These studies propose that the delta-opioid receptor may provide a new therapeutic target for treating human depression.


Subject(s)
Antidepressive Agents/pharmacology , Motor Activity/drug effects , Receptors, Opioid, delta/agonists , Animals , Antidepressive Agents/chemistry , Benzamides/chemistry , Benzamides/pharmacology , Dose-Response Relationship, Drug , Humans , Motor Activity/physiology , Piperazines/chemistry , Piperazines/pharmacology , Receptors, Opioid, delta/physiology
16.
Neuropsychopharmacology ; 26(6): 744-55, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12007745

ABSTRACT

The present study examined the effect of opioid receptor agonists in the rat forced swim assay. The delta-opioid receptor agonists SNC80 ((+)-4-[(alpha R)-alpha-((2S,5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide) and (+)BW373U86 ((+)-[1(S*),2 alpha,5 beta]-4-[[2,5-dimethyl-4-(2-propenyl)-1-piperazinyl] (3-hydroxyphenyl)methyl]-N,N-diethyl-benzamide dihydrochloride) produced a decrease in immobility indicating an antidepressant-like effect. At antinociceptive doses, neither the kappa-opioid selective agonist CI977 (5R-(5 alpha,7 alpha,8 beta)-N-methyl-N-[7-(1-pyrrolidinyl-1-oxaspiro[4,5]dec-8-yl]-4-benzofuranacetamide) showed a change in immobility that was identifiable by dose, nor were changes in immobility seen with morphine. A delta-opioid mechanism of action in the forced swim assay was likely since naltrindole prevented the effects of both delta-agonists. When compared to desipramine and fluoxetine, SNC80 was more active with a single dose whereas both desipramine and fluoxetine produced greater effects with subchronic dosing (3 doses). All three compounds were active when administered before the initial swim exposure. SNC80 was, however, more effective following a single dose than by subchronic administration demonstrating both a fast onset of activity and potential tolerance. Thus, delta-agonists differ from typical antidepressants in the forced swim assay.


Subject(s)
Benzamides/pharmacology , Motor Activity/drug effects , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , Animals , Benzamides/therapeutic use , Depression/drug therapy , Depression/physiopathology , Dose-Response Relationship, Drug , Immobilization/physiology , Male , Motor Activity/physiology , Narcotics/pharmacology , Narcotics/therapeutic use , Organic Chemicals , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/physiology , Swimming/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...