Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Neuroscience ; 169(3): 1414-20, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20570607

ABSTRACT

Improgan, the prototype compound of a novel class of non-opioid analgesic drugs derived from histamine antagonists, attenuates thermal and mechanical nociception in rodents following intracerebroventricular (i.c.v.) administration. Improgan does not bind to known opioid, histamine or cannabinoid receptors, and its molecular target has not been identified. It is known however, that improgan acts directly in the periaqueductal gray and the rostral ventromedial medulla to produce its antinociceptive effects, and that inactivation of the rostral ventromedial medulla prevents the antinociceptive effect of improgan given i.c.v. Here we used in vivo single-cell recording in lightly anesthetized rats to show that improgan engages pain-modulating neurons in the medulla to produce antinociception. Following improgan administration, OFF-cells, which inhibit nociception, became continuously active and no longer paused during noxious stimulation. The increase in OFF-cell firing does not represent a non-specific neuroexcitant effect of this drug, since ON-cell discharge, associated with net nociceptive facilitation, was depressed. NEUTRAL-cell firing was unaffected by improgan. The net response of rostral ventromedial medulla (RVM) neurons to improgan is thus comparable to that evoked by mu-opioids and cannabinoids, well known RVM-active analgesic drugs. This common basis for improgan, opioid, and cannabinoid antinociception in the RVM supports the idea that improgan functions as a specific analgesic agent.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Pain/drug therapy , Action Potentials , Analgesics, Non-Narcotic/administration & dosage , Animals , Cimetidine/administration & dosage , Cimetidine/pharmacology , Injections, Intraventricular , Male , Medulla Oblongata/drug effects , Medulla Oblongata/physiopathology , Neurons/drug effects , Neurons/physiology , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Reaction Time
2.
Bioorg Med Chem Lett ; 17(20): 5715-9, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17766108

ABSTRACT

Furan-containing congeners of the histamine H(2) receptor antagonist ranitidine were synthesized and tested for improgan-like antinociceptive activity. The most potent ligand of the series, VUF5498, is the most potent improgan-like agent described to date (ED(50)=25 nmol, icv). This compound is approximately equal in potency with morphine. These non-imidazole, improgan-like pain relievers further define the structural requirements for analgesics of this class and are important tools for ongoing mechanism-based studies.


Subject(s)
Cimetidine/analogs & derivatives , Furans/chemistry , Nociceptors/metabolism , Ranitidine/chemistry , Ranitidine/pharmacology , Animals , Cimetidine/chemistry , Cimetidine/pharmacology , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
Pharmacol Biochem Behav ; 80(3): 505-10, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15740793

ABSTRACT

Improgan, an analgesic derived from histamine antagonists, acts in the brain stem to activate descending non-opioid, pain-relieving circuits, but the mechanism of action of this drug remains elusive. Because improgan has a moderate affinity for 5-HT(3) receptors, and, since cholinergic and serotonergic drugs can modulate descending analgesic circuits, roles for 5-HT(3), nicotinic and muscarinic receptors in improgan antinociception were presently investigated in rats. Improgan (80 microg, icv) induced nearly maximal inhibition of hot plate and tail flick nociceptive responses, and these actions we unaffected by antagonists of muscarinic (atropine, 5.9 mg/kg, i.p.) and nicotinic (mecamylamine, 2 mg/kg, i.p.) receptors. Control experiments verified that these antagonist treatments were maximally effective against muscarinic and nicotinic antinociception in both tests. In addition, improgan antinociception was unaffected by icv pretreatment with a 5-HT(3) antagonist (ondansetron, 20 microg). When given alone, icv treatment with neither this antagonist nor a 5-HT(3) agonist (m-chlorophenylbiguanide, 1000 nmol, icv) modified thermal nociceptive latencies. These results show no role for supraspinal cholinergic and 5-HT(3) receptors in improgan antinociception. The findings help to narrow the search for the relevant mediators of the action of this novel analgesic agent.


Subject(s)
Analgesics/pharmacology , Cimetidine/analogs & derivatives , Cimetidine/pharmacology , Pain Measurement/drug effects , Receptors, Cholinergic/physiology , Receptors, Serotonin, 5-HT3/physiology , Animals , Dose-Response Relationship, Drug , Male , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Serotonin 5-HT3 Receptor Antagonists , Serotonin Antagonists/pharmacology
4.
Brain Res ; 1021(2): 248-55, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15342273

ABSTRACT

Improgan is a compound developed from histamine antagonists which shows the pre-clinical profile of a highly effective, non-opioid analgesic when administered into the rodent CNS. Pharmacological studies suggest that improgan activates descending pain-relieving circuits, but the brain and spinal sites of action of this drug have not been previously studied. Presently, the effects of intracerebral and intrathecal microinjections of improgan were evaluated on thermal nociceptive responses in rats. Improgan produced large, dose- and time-related reductions in nociceptive responses following administration into the ventrolateral periaqueductal gray (PAG), the dorsal PAG, and the rostral ventromedial medulla (RVM). The drug had no measurable effects after injections into the caudate nucleus, basolateral amygdala, hippocampus, ventromedial hypothalamus, superior colliculi, ventrolateral medulla, or the spinal subarachnoid space. Inactivation of the RVM by muscimol microinjections completely attenuated antincociceptive responses produced by intraventricular improgan. These findings, taken with earlier results, show that, like opioids and cannabinoids, improgan acts in the PAG and RVM to activate descending analgesic systems. Unlike these other analgesics, improgan does not act in the spinal cord or in CNS areas outside of the brain stem.


Subject(s)
Analgesics, Non-Narcotic/administration & dosage , Brain Mapping , Brain Stem/drug effects , Cimetidine/analogs & derivatives , Cimetidine/administration & dosage , Animals , Dose-Response Relationship, Drug , Injections, Intraventricular , Injections, Spinal , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Time Factors
6.
J Pharmacol Exp Ther ; 303(1): 314-22, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12235266

ABSTRACT

Improgan, a nonopioid antinociceptive agent, activates descending, pain-relieving mechanisms in the brain stem, but the receptor for this compound has not been identified. Because cannabinoids also activate nonopioid analgesia by a brain stem action, experiments were performed to assess the significance of cannabinoid mechanisms in improgan antinociception. The cannabinoid CB(1) antagonist N-(piperidin-1-yl)-5-(4-chloro phenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (SR141716A) induced dose-dependent inhibition of improgan antinociception on the tail-flick test after i.c.v. administration in rats. The same treatments yielded comparable inhibition of cannabinoid [R-(+)-(2,3-dihydro-5-methyl-3-[(4-mor pholinyl)methyl]pyrol[1,2,3-de]-1,4-benzoxazin-6-yl)(1-naphthalenyl)methanone monomethanesulfonate, WIN 55,212-2] analgesia. Inhibition of improgan and WIN 55,212-2 antinociception by SR141716A was also observed in Swiss-Webster mice. Radioligand binding studies showed no appreciable affinity of improgan on rat brain, mouse brain, and human recombinant CB(1) receptors, ruling out a direct action at these sites. To test the hypothesis that CB(1) receptors indirectly participate in improgan signaling, the effects of improgan were assessed in mice with a null mutation of the CB(1) gene with and without SR141716A pretreatment. Surprisingly, improgan induced complete antinociception in both CB(1) (-/-) and wild-type control [CB(1) (+/+)] mice. Furthermore, SR141716A inhibited improgan antinociception in CB(1) (+/+) mice, but not in CB(1) (-/-) mice. Taken together, the results show that SR141716A reduces improgan antinociception, but neither cannabinoids nor CB(1) receptors seem to play an obligatory role in improgan signaling. Present and previous studies suggest that Delta(9)-tetrahydrocannabinol may act at both CB(1) and other receptors to relieve pain, but no evidence was found indicating that improgan uses either of these mechanisms. SR141716A will facilitate the study of improgan-like analgesics.


Subject(s)
Analgesics/pharmacology , Cannabinoids/antagonists & inhibitors , Cimetidine/analogs & derivatives , Cimetidine/pharmacology , Dronabinol/pharmacology , Pain/physiopathology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Drug/physiology , Analgesics/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Modulators , Cannabinoids/pharmacokinetics , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Cimetidine/administration & dosage , Cimetidine/antagonists & inhibitors , Endocannabinoids , Hot Temperature , Injections, Intraventricular , Male , Mice , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/drug effects , Rimonabant , Time Factors
7.
Brain Res ; 923(1-2): 12-9, 2001 Dec 27.
Article in English | MEDLINE | ID: mdl-11743967

ABSTRACT

Improgan is a derivative of cimetidine that induces non-opioid antinociception after intracerebroventricular (i.c.v.) administration, but the mechanism of action of this compound remains unknown. Since activation of either supraspinal or spinal alpha(2) adrenergic receptors can induce antinociception, and since improgan showed affinity for these receptors in vitro, the effects of the alpha(2) antagonist yohimbine on improgan antinociception were presently studied in rats on the hot plate and tail flick tests. Systemic yohimbine pretreatment (4 mg/kg, i.p.) completely blocked improgan antinociception (80 microg, i.c.v.), suggesting a mediator role for alpha(2) receptors. However, i.c.v. pretreatment with yohimbine (30 microg) had no effect on improgan antinociception. Since this treatment completely antagonized clonidine antinociception (40 microg, i.c.v.), supraspinal alpha(2) receptors seem to mediate the antinociceptive effects of clonidine, but not that produced by improgan. In contrast, intrathecal (i.t.) yohimbine pretreatment (30 microg) completely blocked the antinociception elicited by i.c.v. improgan and i.c.v. morphine. These results suggest that spinal (but not supraspinal) alpha(2) adrenergic receptors play a significant role in the pain-relieving actions of improgan. Furthermore, although improgan shows some affinity at alpha(2) receptors, this drug does not act directly at these receptors to induce antinociceptive responses. Like several other classes of analgesics, improgan-like drugs seem to activate non-opioid, descending pain-relieving circuits.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Cimetidine/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Spinal Cord/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Analgesics, Opioid/pharmacology , Animals , Clonidine/pharmacology , Injections, Intraventricular , Injections, Spinal , Male , Morphine/pharmacology , Nociceptors/drug effects , Nociceptors/metabolism , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Yohimbine/pharmacology
8.
Life Sci ; 68(24): 2751-7, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11400917

ABSTRACT

Improgan is the prototype drug from a new class of non-opioid analgesics chemically related to histamine and histamine antagonists, but the mechanism of action of these compounds has not been identified. Because several classes of analgesics act in the brain by reducing GABAergic inhibition of endogenous pain-relieving circuits, and because the activity of these substances is abolished by the GABA(A) agonist muscimol, the present study assessed the effects of muscimol on improgan antinociception in rats. Intracerebroventricular (icv) improgan (80 microg) and morphine (20 microg) both induced 80-100% of maximal analgesic responses on the tail flick test 10 to 30 min later. However, muscimol pretreatment (0.5 microg, icv) completely eliminated the antinociceptive activity of both compounds. Since improgan in vitro lacks activity at opioid and GABA(A) receptors, these findings: 1) confirm earlier literature showing that muscimol inhibits morphine analgesia, and 2) suggest that improgan activates a supraspinal, descending analgesic pathway, possibly via inhibition of GABAergic transmission. Since muscimol is the first compound discovered which inhibits improgan analgesia, muscimol will be a useful tool for the further characterization of this new class of pain-relieving substances.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Cimetidine/pharmacology , Receptors, GABA-A/drug effects , Animals , Male , Morphine/pharmacology , Muscimol/pharmacology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
9.
Brain Res ; 880(1-2): 102-8, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11032994

ABSTRACT

Improgan is an analog of the H(2) antagonist cimetidine that does not act on known histamine receptors, but induces highly effective analgesia in rodents following intracerebroventricular (icv) administration. Since the mechanism of action of this compound remains unknown, improgan analgesia was characterized presently with the tail immersion nociceptive test in mutant mice lacking either the mu (exon 1 of MOR-1), delta (exon 2 of DOR-1) or kappa (exon 3 of KOR-1) opioid receptor. Improgan (30 microg, icv) induced reversible, maximal analgesia in both sexes of all three genotypes (+/+, +/- and -/-) of MOR-1 mutant mice 10 and 20 min after administration, whereas morphine analgesia was reduced (+/-) or abolished (-/-) in these subjects. In DOR-1 mutant mice, improgan was equally effective in all three genotypes, despite the reduction (+/-) or complete loss (-/-) of delta opioid receptor (3H-[D-Pen(2), D-Pen(5)]enkephalin, DPDPE) binding. Similarly, improgan analgesia was equivalent in all three genotypes of KOR-1 mutant mice, whereas kappa-mediated analgesia (U50,488) and kappa opioid (3H-U69,593) binding were abolished in the homozygous (-/-) mice. These studies demonstrate that improgan analgesia does not require intact MOR-1, DOR-1, or KOR-1 genes, and support the hypothesis that improgan-like analgesics act in the CNS by non-opioid mechanisms.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cerebral Ventricles/physiology , Cimetidine/analogs & derivatives , Receptors, Opioid, mu/genetics , Receptors, Opioid/physiology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesia , Animals , Cerebral Ventricles/drug effects , Cimetidine/administration & dosage , Cimetidine/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Exons , Female , Gene Expression Regulation , Genotype , Injections, Intraventricular , Male , Mice , Mice, Knockout , Receptors, Opioid/deficiency , Receptors, Opioid/genetics , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/physiology
10.
Ann N Y Acad Sci ; 909: 25-40, 2000.
Article in English | MEDLINE | ID: mdl-10911922

ABSTRACT

Burimamide, a histamine (HA) derivative with both H2- and H3-blocking properties, induces antinociception when injected into the rodent CNS. Several related compounds share this property, and structure-activity studies have shown that this new class of analgesics is distinct from known HA antagonists. The prototype, named improgan, shows a preclinical profile of a highly effective analgesic, with activity against thermal, mechanical and inflammatory nociception after doses that do not alter motor balance or locomotor activity. Improgen analgesia is not blocked by opioid antagonists and is observed in opioid receptor knock-out mice. Unlike morphine, improgan does not induce tolerance after daily dosing. Extensive in vitro pharmacology studies have excluded known histaminergic, opioid, serotonergic, GABAergic and adrenergic receptor mechanisms, as well as 50 other sites of action. The improgan-like analgesic activity of some HA congeners suggests an analgesic action on a novel HA receptor, but further studies are required to substantiate this. Studies in progress are characterizing the sites and mechanisms of action of improgan, and developing brain-penetrating derivatives that could be useful for clinical pain.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Burimamide/pharmacology , Histamine H2 Antagonists/pharmacology , Animals , Humans , Mice , Receptors, Histamine/drug effects , Structure-Activity Relationship
11.
Brain Res ; 858(2): 227-36, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10708674

ABSTRACT

The current study examined the possible role of increased histamine release and granulocyte activity in the vascular changes that precede the onset of necrotic lesions with the thalamus of the pyrithiamine-induced thiamine deficiency (PTD) rat model of Wernicke's encephalopathy (WE). An increase in histamine release and the number of granulocytes was observed in lateral thalamus on day 9 and in medial thalamus on day 10 of PTD treatment, a duration of thiamine deficiency associated with perivascular edema in this brain region. Within the hippocampus, histamine release was significantly increased on day 9, declined to control levels on days 10-12, and was significantly elevated on days 12-14. No granulocytes were observed in hippocampus of either PTD or control rats. These observations suggest that the release of histamine from nerve terminals and histamine and other vasoactive substances from granulocytes may be responsible for thiamine deficiency-induced vascular breakdown and perivascular edema within thalamus.


Subject(s)
Basophils/metabolism , Histamine/metabolism , Thalamus/immunology , Wernicke Encephalopathy/immunology , Animals , Behavior, Animal , Cerebrovascular Circulation/immunology , Disease Models, Animal , Hippocampus/blood supply , Hippocampus/immunology , Hippocampus/pathology , Male , Mast Cells/metabolism , Microdialysis , Necrosis , Rats , Rats, Sprague-Dawley , Thalamic Nuclei , Thalamus/blood supply , Thalamus/pathology , Wernicke Encephalopathy/metabolism , Wernicke Encephalopathy/pathology
12.
Pharmacol Biochem Behav ; 65(1): 61-6, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10638637

ABSTRACT

Improgan, a compound related to H2 and H3 antagonists, induces antinociception in rodents after intraventricular administration. Characteristics of improgan and its congeners include: (a) morphine-like antinociception on thermal and mechanical tests in two species; (b) no impairment of motor coordination or locomotor activity; (c) evidence for a novel, nonopioid mechanism that is independent of known histamine receptors; (d) lack of tolerance with daily dosing; and (e) unique structure-activity relationships (SARs). Presently, the antinociceptive activity of several new derivatives of improgan was investigated in rats. Among compounds similar to burimamide, VUF4577 (possessing a two-carbon side chain) and VUF4582 (an N-phenyl derivative of VUF4577) induced complete, dose- and time-dependent antinociception on the hot-plate and tail-flick tests with no behavioral side effects. These compounds (with ED50 values of 71-117 nmol) were approximately twice as potent as burimamide itself (a four-carbon derivative). Two other derivatives in which the thiourea group (C=S, known to cause human toxicity) was replaced by either nitroethene (C=CH-NO2, VUF5405) or urea (C=O, VUF5407) also showed effective, potent antinociception on both assays. The latter compound is the most potent improgan-like drug discovered to date (ED50 = 71 nmol). Furthermore, positional isomers of antinociceptive compounds either lacked activity (VUF5394) or induced toxicity (VUF5393), revealing a high degree of pharmacological specificity. Although the mechanism of improgan antinociception remains unknown, the present results show promise for the further development of safe, effective, and potent pain-relieving compounds.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Burimamide/pharmacology , Cimetidine/analogs & derivatives , Histamine H2 Antagonists/pharmacology , Receptors, Histamine H3/drug effects , Animals , Cimetidine/pharmacology , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
Life Sci ; 64(5): PL79-86, 1999.
Article in English | MEDLINE | ID: mdl-10072195

ABSTRACT

The brain neuromodulator histamine induces antinociception when administered directly into the rodent CNS. However, several compounds derived from H2 and H3 antagonists also produce antinociception after central administration. Pharmacological studies have shown that a prototype of these agents, improgan, induces analgesia that is not mediated by actions on known histamine receptors. Presently, the antinociceptive properties of a compound that chemically resembles both improgan and histamine were investigated in rats. Intraventricular (i.v.t.) administration of impentamine (4-imidazolylpentylamine) induced reversible, near-maximal antinociception on the hot plate and tail flick tests (15 microg, 98 nmol). The dose-response function was extremely steep, however, since other doses showed either no effect or behavioral toxicity. On the tail flick test, impentamine antinociception was resistant to antagonism by blockers of H1, H2, or H3 receptors, similar to characteristics previously found for improgan. In contrast, histamine antinociception was highly attenuated by H1 and H2 antagonists. These findings suggest that: 1) the histamine congener impentamine may induce antinociception by a mechanism similar to that produced by improgan, and 2) additional histamine receptors may be discovered that are linked to pain-attenuating processes.


Subject(s)
Analgesics/pharmacology , Cerebral Ventricles/physiology , Histamine Antagonists/pharmacology , Imidazoles/pharmacology , Pain/physiopathology , Analgesics/administration & dosage , Animals , Cerebral Ventricles/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Histamine/pharmacology , Histamine Agonists/pharmacology , Histamine Antagonists/administration & dosage , Hot Temperature , Imidazoles/administration & dosage , In Vitro Techniques , Injections, Intraventricular , Jejunum/drug effects , Jejunum/physiology , Male , Microinjections , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Pain/prevention & control , Rats , Rats, Sprague-Dawley , Receptors, Histamine H1/drug effects , Receptors, Histamine H1/physiology
14.
Brain Res ; 814(1-2): 218-21, 1998 Dec 14.
Article in English | MEDLINE | ID: mdl-9838126

ABSTRACT

Improgan, an analog of the histamine receptor antagonist cimetidine, produces highly effective analgesia following intraventricular injection. The present study examined changes in the antinociceptive effects of improgan following once daily intraventricular injections. Improgan (100-150 microg) produced near maximal antinociception 10 and 30 min after daily administration on all 4 test days, whereas comparable morphine treatments (50 microg) induced considerable tolerance. Thus, improgan produced highly effective analgesia without the development of tolerance.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Animals , Cimetidine/pharmacology , Drug Tolerance , Injections, Intraventricular , Male , Pain Measurement , Rats , Rats, Sprague-Dawley
15.
Brain Res ; 748(1-2): 168-74, 1997 Feb 14.
Article in English | MEDLINE | ID: mdl-9067458

ABSTRACT

A recent study showed that SKF92374, a structural analog of the histamine H2 receptor antagonist cimetidine, induces antinociception after intraventricular (i.v.t.) administration in the rat. SKF92374 lacked significant activity on H1 or H2 receptors, but had weak activity on H3 receptors. To test the hypothesis that SKF92374-induced antinociception is mediated by an action on H3 receptors, the effects of the H3 agonist R-alpha-methylhistamine (RAMH) and the H3 antagonist thioperamide (both by i.v.t. administration) were investigated on SKF92374 antinociception. SKF92374-induced antinociception was slightly enhanced by thioperamide (30 microg), but unaffected by a range of doses of RAMH (up to 2 microg). Furthermore, SKF92374-induced antinociception was not reduced by large doses of systemically-administered antagonists of H1 (pyrilamine), H2 (zolantidine), H3 (GT-2016), or opioid (naltrexone) receptors. These findings show that the novel compound SKF92374 induces antinociception by a non-opioid mechanism that does not utilize brain H1, H2 or H3 receptors.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Histamine Antagonists/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Nociceptors/drug effects , Animals , Cimetidine/pharmacology , Histamine Agonists/pharmacology , Male , Methylhistamines/pharmacology , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism
16.
J Pharmacol Exp Ther ; 283(3): 1534-43, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9400031

ABSTRACT

Recent studies have shown that cimetidine, burimamide and improgan (also known as SKF92374, a cimetidine congener lacking H2 antagonist activity) induce antinociception after intracerebroventricular administration in rodents. Because these substances closely resemble the structure of histamine (a known mediator of some endogenous analgesic responses), yet no role for known histamine receptors has been found in the analgesic actions of these agents, the structure-activity relationships for the antinociceptive effects of 21 compounds chemically related to H2 and H3 antagonists were investigated in this study. Antinociceptive activity was assessed on the hot-plate and tail-flick tests after intracerebroventricular administration in rats. Eleven compounds induced time-dependent (10-min peak) and dose-dependent antinociceptive activity with no observable behavioral impairment. ED50 values, estimated by nonlinear regression, were highly correlated across nociceptive assays (r2 = 0.98, n = 11). Antinociceptive potencies varied more than 6-fold (80-464 nmol), but were not correlated with activity on H1, H2 or H3 receptors. Although highly potent H3 antagonists such as thioperamide lacked antinociceptive activity, homologs of burimamide and thioperamide containing N-aromatic substituents retained H3 antagonist activity and also showed potent, effective analgesia. A literature review of the pharmacology of these agents did not find a basis for their antinociceptive effects. Taken with previous findings, the present results suggest: 1) these compounds act on the brain to activate powerful analgesic responses that are independent of known histamine receptors, 2) the structure-activity profile of these agents is novel and 3) brain-penetrating derivatives of these compounds could be clinically useful analgesics.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Histamine Antagonists/pharmacology , Histamine H2 Antagonists/pharmacology , Receptors, Histamine H3/drug effects , Animals , Burimamide/pharmacology , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Male , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiourea/analogs & derivatives , Thiourea/pharmacology
17.
Brain Res ; 741(1-2): 258-62, 1996 Nov 25.
Article in English | MEDLINE | ID: mdl-9001731

ABSTRACT

The potential modulation of morphine antinociception by the putative anti-addictive agent ibogaine and its active metabolite (noribogaine) was investigated in rats with the radiant heat tail-flick test. Ibogaine pretreatment (40 mg/kg, i.p., 19 h) significantly decreased morphine (4 mg/kg, s.c.) antinociception, with no effects in the absence of morphine. However, co-administration of ibogaine (1-40 mg/kg, i.p.) and morphine (4 mg/kg, s.c.) exhibited a dose-dependent enhancement of morphine antinociception. Co-administration of noribogaine (40 mg/kg, i.p.) and morphine also resulted in an increase in morphine antinociception, while noribogaine pretreatment (19 h) had no effect on morphine antinociception. The results show that ibogaine acutely potentiates morphine antinociception and that noribogaine could be the active metabolite responsible for this effect. However, the inhibitory effects of a 19 h ibogaine pretreatment, which resemble ibogaine-induced inhibition of morphine's stimulant properties, cannot be accounted for by noribogaine.


Subject(s)
Analgesics, Opioid/pharmacology , Ibogaine/analogs & derivatives , Morphine/pharmacology , Psychotropic Drugs/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Interactions , Ibogaine/pharmacology , Male , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
18.
J Pharmacol Exp Ther ; 276(2): 500-8, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632315

ABSTRACT

The antinociceptive and pharmacological properties of the H2 receptor antagonist cimetidine and a novel cimetidine analog, SKF92374, were characterized. On both the hot-plate and tail-flick nociceptive tests, cimetidine and SKF92374 induced complete, dose-related analgesic responses when injected into the lateral ventricle of rats. SKF92374 showed strong similarities to cimetidine in analgesic efficacy, slope of dose-response curves and chemical structure, suggesting that these compounds share a common analgesic mechanism. In contrast, histamine induced submaximal antinociceptive effects, and the H3 antagonist thioperamide, a known HA-releasing drug, had little or no analgesic effects. Compared with cimetidine, SKF92374 showed very weak activity (400-fold lower affinity) on H2 receptors in vitro (isolated guinea pig atrium) and in vivo (rat gastric secretion). In addition, SKF92374 (100 microM) had neither agonist nor antagonist action on guinea pig ileum H1 receptors. SKF92374 was also a weak competitive antagonist of N alpha-methylhistamine-induced inhibition of electrically induced contractions of the guinea pig ileum (Kd = 5.2 microM), an H3 receptor-mediated response. Autoradiographic binding assays in guinea pig brain confirmed a weak antagonism of H3 receptors by SKF92374. The compound (up to 10 microM) also had no effect on unpurified rat brain histamine N-methyltransferase activity. These results support the hypothesis that cimetidine induces analgesia by a novel brain mechanism unrelated to H1, H2 or H3 receptors.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cimetidine/analogs & derivatives , Cimetidine/pharmacology , Histamine H2 Antagonists/pharmacology , Animals , Guinea Pigs , In Vitro Techniques , Male , Periaqueductal Gray/drug effects , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Histamine H1/drug effects , Receptors, Histamine H2/drug effects , Receptors, Histamine H3/drug effects
19.
J Pharmacol Exp Ther ; 275(2): 598-604, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7473144

ABSTRACT

GT-2016, a non-thiourea-containing imidazole, has been developed as a histamine H3 antagonist. In vitro and in vivo studies in rats were conducted to characterize receptor selectivity and autoreceptor functionality for GT-2016. GT-2016 demonstrated high affinity (43.8 +/- 3.0 nM) and selectivity for the histamine H3 receptor in vitro. In vivo, GT-2016 (3, 10 and 30 mg/kg i.p. and p.o.) was shown to cross the blood-brain barrier and dose-dependently bind to cortical histamine H3 receptors. GT-2016 induced dose-dependent increases in histamine turnover at concentrations that exhibited significant histamine H3 receptor occupancy. Also, in vivo microdialysis experiments were conducted in awake, freely moving rats treated with GT-2016. GT-2016 (10 and 30 mg/kg i.p.) increased histamine release by approximately 75% above baseline within 1 hr, and elevated histamine release was observed for up to 2.5 hr after the higher dose. In contrast, GT-2016 was devoid of activity on histamine methyltransferase in vitro at concentrations up to 3 microM. Taken together, the results show that GT-2016 crosses the blood-brain barrier, binds to H3 receptors and increases the release of histamine in the cerebral cortex, consistent with blockade of presynaptic H3 autoreceptors. In summary, these findings allowed us to identify and characterize the in vitro and in vivo biochemical properties of a novel H3 receptor antagonist, GT-2016.


Subject(s)
Histamine Antagonists/pharmacology , Imidazoles/pharmacology , Methylhistamines/metabolism , Receptors, Histamine H3/metabolism , Animals , Binding, Competitive , Cerebral Cortex/metabolism , Histamine N-Methyltransferase/metabolism , Histamine Release/drug effects , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...