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











Publication year range
1.
Life Sci ; 93(17): 637-45, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24012613

ABSTRACT

AIMS: The purpose of this study was to investigate the antinociceptive effect of epicatechin as well as the possible mechanisms of action in diabetic rats. MAIN METHODS: Rats were injected with streptozotocin to produce hyperglycemia. The formalin test was used to assess the nociceptive activity. KEY FINDINGS: Acute pre-treatment with epicatechin (0.03-30 mg/kg, i.p.) prevented formalin-induced nociception in diabetic rats. Furthermore, daily or every other day treatment for 2 weeks with epicatechin (0.03-30 mg/kg, i.p.) also prevented formalin-induced nociception in diabetic rats. Acute epicatechin-induced antinociception was prevented by l-NAME (N(ω)-nitro-l-arginine methyl ester hydrochloride, 1-10mg/kg, non-selective nitric oxide synthesis inhibitor), 7-nitroindazole (0.1-1mg/kg, selective neuronal nitric oxide synthesis inhibitor), ODQ (1H-(1,2,4)-oxadiazolo(4,2-a)quinoxalin-1-one, 0.2-2mg/kg, guanylyl cyclase inhibitor) or glibenclamide (1-10mg/kg, ATP-sensitive K(+) channel blocker). Moreover, epicatechin (3mg/kg)-induced antinociception was fully prevented by methiothepin (0.1-1mg/kg, serotonergic receptor antagonist), WAY-100635 (0.03-0.3mg/kg, selective 5-HT1A receptor antagonist) or SB-224289 (0.03-0.3mg/kg, selective 5-HT1B receptor antagonist). In contrast, BRL-15572 (0.03-0.3mg/kg, selective 5-HT1D receptor antagonist) only slightly prevented the antinociceptive effect of epicatechin. Naloxone (0.1-1mg/kg, opioid antagonist) did not modify epicatechin's effect. SIGNIFICANCE: Data suggest the involvement of the nitric oxide-cyclic GMP-K(+) channel pathway as well as activation of 5-HT1A and 5HT1B, and at a lesser extent, 5-HT1D, but not opioid, receptors in the antinociceptive effect of epicatechin in diabetic rats. Our data suggest that acute or chronic treatment with epicatechin may prove to be effective to treat nociceptive hypersensitivity in diabetic patients.


Subject(s)
Analgesics/pharmacology , Catechin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hyperalgesia/drug therapy , Analgesics/antagonists & inhibitors , Animals , Biphenyl Compounds/pharmacology , Catechin/antagonists & inhibitors , Diabetes Mellitus, Experimental/complications , Dose-Response Relationship, Drug , Female , Glyburide/pharmacology , Hyperalgesia/complications , Indazoles/pharmacology , Methiothepin/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Naloxone/pharmacology , Oxadiazoles/pharmacology , Pain Measurement/drug effects , Piperazines/pharmacology , Piperidones/pharmacology , Pyridines/pharmacology , Quinoxalines/pharmacology , Rats , Spiro Compounds/pharmacology
2.
Neuropharmacology ; 73: 261-73, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23791558

ABSTRACT

Pyrazole compounds are an intriguing class of compounds with potential analgesic activity; however, their mechanism of action remains unknown. Thus, the goal of this study was to explore the antinociceptive potential, safety and mechanism of action of novel 1-pyrazole methyl ester derivatives, which were designed by molecular simplification, using in vivo and in vitro methods in mice. First, tree 1-pyrazole methyl ester derivatives (DMPE, MPFE, and MPCIE) were tested in the capsaicin test and all presented antinociceptive effect; however the MPClE (methyl 5-trichloromethyl-3-methyl-1H-pyrazole-1-carboxylate) was the most effective. Thus, we selected this compound to assess the effects and mechanisms in subsequent pain models. MPCIE produced antinociception when administered by oral, intraperitoneal, intrathecal and intraplantar routes and was effective in the capsaicin and the acetic acid-induced nociception tests. Moreover, this compound reduced the hyperalgesia in diverse clinically-relevant pain models, including postoperative, inflammatory, and neuropathic nociception in mice. The antinociception produced by orally administered MPClE was mediated by κ-opioid receptors, since these effects were prevented by systemically pre-treatment with naloxone and the κ-opioid receptor antagonist nor-binaltorphimine. Moreover, MPCIE prevented binding of the κ-opioid ligand [(3)H]-CI-977 in vitro (IC50 of 0.68 (0.32-1.4) µM), but not the TRPV1 ([(3)H]-resiniferatoxin) or the α2-adrenoreceptor ([(3)H]-idazoxan) binding. Regarding the drug-induced side effects, oral administration of MPClE did not produce sedation, constipation or motor impairment at its active dose. In addition, MPCIE was readily absorbed after oral administration. Taken together, these results demonstrate that MPClE is a novel, potent, orally active and safe analgesic drug that targets κ-opioid receptors.


Subject(s)
Analgesics/pharmacology , Pyrazoles/pharmacology , Receptors, Opioid, kappa/agonists , Adrenergic alpha-2 Receptor Antagonists , Analgesics/administration & dosage , Analgesics/antagonists & inhibitors , Analgesics/chemistry , Animals , Benzofurans , Diterpenes , Drug Administration Routes , Drug Evaluation, Preclinical , Idazoxan , Male , Mice , Molecular Structure , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement , Pyrazoles/administration & dosage , Pyrazoles/antagonists & inhibitors , Pyrazoles/chemistry , Pyrrolidines , Radioligand Assay , Receptors, Opioid, kappa/antagonists & inhibitors , TRPV Cation Channels/drug effects , Tritium
3.
Neuropharmacology ; 63(5): 798-805, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22722030

ABSTRACT

In the current study, we investigated the effect of the activation of the alpha-7 nicotinic acetylcholine receptor (α7 nAchR) on dextran sulphate sodium (DSS)-induced colitis and referred mechanical hyperalgesia in mice. Colitis was induced in CD1 male mice through the intake of 4% DSS in tap water for 7 days. Control mice received unadulterated water. Referred mechanical hyperalgesia was evaluated for 7 days after the beginning of 4% DSS intake. Referred mechanical hyperalgesia started within 1 day after beginning DSS drinking, peaked at 3 days and persisted for 7 days. This time course profile perfectly matched with the appearance of signs of colitis. Both acute and chronic oral treatments with nicotine (0.1-1.0 mg/kg, p.o.) were effective in inhibiting the established referred mechanical hyperalgesia. The antinociceptive effect of nicotine was completely abrogated by cotreatment with the selective α7 nAchR antagonist methyllycaconitine (MLA) (1.0 mg/kg). Consistent with these results, i.p. treatment with the selective α7 nAchR agonist PNU 282987 (0.1-1.0 mg/kg) reduced referred mechanical hyperalgesia at all periods of evaluation. Despite their antinociceptive effects, nicotinic agonists did not affect DSS-induced colonic damage or inflammation. Taken together, the data generated in the present study show the potential relevance of using α7 nAchR agonists to treat referred pain and discomfort associated with inflammatory bowel diseases.


Subject(s)
Analgesics/therapeutic use , Colitis/physiopathology , Hyperalgesia/drug therapy , Molecular Targeted Therapy , Nicotinic Agonists/therapeutic use , Pain, Referred/drug therapy , Receptors, Nicotinic/metabolism , Aconitine/adverse effects , Aconitine/analogs & derivatives , Analgesics/administration & dosage , Analgesics/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Benzamides/administration & dosage , Benzamides/therapeutic use , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/therapeutic use , Colitis/chemically induced , Colitis/immunology , Dose-Response Relationship, Drug , Drug Antagonism , Hyperalgesia/etiology , Hyperalgesia/immunology , Male , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Nicotine/administration & dosage , Nicotine/antagonists & inhibitors , Nicotine/therapeutic use , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/chemistry , Nicotinic Antagonists/adverse effects , Pain Threshold/drug effects , Pain, Referred/etiology , Pain, Referred/immunology , Random Allocation , Receptors, Nicotinic/chemistry , alpha7 Nicotinic Acetylcholine Receptor
4.
Article in English | MEDLINE | ID: mdl-22627196

ABSTRACT

Previous studies have shown that uliginosin B inhibits dopamine reuptake in rat brain. This compound occurs in Hypericum polyanthemum and H. caprifoliatum for which was reported to have antinociceptive effect sensitive to naloxone. The aim of this study was to assess the antinociceptive effect of uliginosin B and to evaluate the involvement of opioid and dopaminergic receptors activation. Uliginosin B presented antinociceptive effect in hot-plate and abdominal writhing tests, in mice, at doses that did not impair the motor coordination (15 mg/kg, i.p.). Uliginosin B in high dose (90 mg/kg, i.p.) presented ataxic effect in the rotarod apparatus. These effects seem to be mediated by distinct receptors since the effect on the hot-plate was completely abolished by naloxone and sulpiride, but it was unaffected by SCH 23390. On the other hand, the motor impairment induced by uliginosin B was completely prevented by naloxone and partially prevented by sulpiride and SCH 23390. However, the receptors' activation appears to be indirect since uliginosin B did not bind to opioid and dopaminergic receptors. Thus, uliginosin B effects probably are due to its ability to inhibit monoamine reuptake with consequent activation of dopamine receptors and indirect stimulation of opioid system.


Subject(s)
Analgesics/pharmacology , Phloroglucinol/analogs & derivatives , Receptors, Dopamine/metabolism , Receptors, Opioid/metabolism , Analgesics/antagonists & inhibitors , Animals , Benzazepines/pharmacology , Brain/drug effects , Brain/metabolism , Dopamine Antagonists/pharmacology , Drug Interactions , Male , Mice , Mice, Inbred Strains , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Pain Measurement/statistics & numerical data , Phloroglucinol/antagonists & inhibitors , Phloroglucinol/pharmacology , Radioligand Assay/methods , Rats , Rats, Sprague-Dawley , Rotarod Performance Test/statistics & numerical data , Sulpiride/pharmacology
5.
Eur J Pharmacol ; 674(2-3): 255-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22134006

ABSTRACT

Studies have demonstrated that the L-arginine/NO/cGMP pathway and the potassium and calcium channels are involved in the mechanisms underlying opioid receptor activation. As additional pathways may participate in the observed antinociceptive effects following opioid exposure, the aim of our study was to determine whether Ca(2+)-activated Cl(-) channels (CaCCs) are involved in peripheral antinociception induced by µ-, δ- and κ-opioid receptor activation. Hyperalgesia was induced by intraplantar injection of prostaglandin E(2) (PGE(2), 2 µg). Nociceptive thresholds to pressure (grams) were measured using an algesimetric apparatus 3h following injection. The µ-opioid receptor agonist morphine (200 µg), δ-opioid receptor agonist (+)-4-[(alphaR)-alpha-((2S,5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80, 80 µg), κ-opioid receptor agonist bremazocine (50 µg), CaCCs blocker niflumic acid (8-64 µg), CaCCs blocker 5-Nitro-2-(3-phenylpropylamino) benzoic acid (NPPB, 32-128 µg), nitric oxide donor sodium nitroprusside (SNP, 500 µg) and cGMP exogenous analogs dibutyryl cGMP (db-cGMP, 100 µg) were also administered into the paw. The CaCCs blocker niflumic acid and NPPB partially reversed the peripheral antinociception induced by exposure to the SNC80 in a dose-dependent manner. In contrast, niflumic acid did not modify the antinociceptive effect observed following exposure to morphine or bremazocine. Additionally, the peripheral antinociception induced by the NO donor SNP or by db-cGMP was not inhibited by niflumic acid. These results provide evidence for the involvement of CaCCs in the peripheral antinociception induced by SNC80. CaCCs activation does not appear to be involved when µ- and κ-opioid receptors are activated. In addition, we did not observe a link between CaCCs and the L-arginine/NO/GMPc pathway.


Subject(s)
Analgesics/pharmacology , Chloride Channels/metabolism , Peripheral Nervous System/drug effects , Receptors, Opioid, delta/metabolism , Analgesics/antagonists & inhibitors , Animals , Arginine/metabolism , Male , Niflumic Acid/pharmacology , Nitric Oxide/metabolism , Nitrobenzoates/pharmacology , Nociception/drug effects , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Rats , Rats, Wistar , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects
6.
J Med Food ; 14(11): 1389-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21663477

ABSTRACT

The antioxidant, antinociceptive, and anti-inflammatory activities of the ethanolic extract from leaves of Combretum duarteanum (EEC) were assessed in rodents through in vitro tests. The antioxidant activity was investigated by using thiobarbituric acid reactive species (TBARS), hydroxyl radical-scavenging, and scavenging activity of nitric oxide assays. The antinociceptive activity was investigated by using acetic acid-induced writhing, formalin, and hot-plate tests in mice. The anti-inflammatory activity was assessed in rats by using the carrageenan-induced hind-paw edema test and arachidonic acid-induced paw edema test. EEC possesses a strong antioxidant potential according to the TBARS, nitric oxide, and hydroxyl radical-scavenging assays; it also presented scavenger activity in all in vitro tests. After intraperitoneal injection, EEC (100, 200, and 400 mg/kg) significantly reduced the number of writhes (38.1%, 90.6%, and 97.8%, respectively) in a writhing test and the number of paw licks during phase 1 (30.5% and 69.5%, higher doses) and phase 2 (38.1%, 90.6%, and 97.8%, all doses) of a formalin test when compared with the control group. Naloxone (1.5 mg/kg, intraperitoneally) antagonized the antinociceptive action of EEC (400 mg/kg), and this finding suggests participation of the opioid system. Administration of 200 and 400 mg/kg (intraperitoneally) of EEC exhibited an anti-inflammatory activity in the carrageenin test, which was based on interference with prostaglandin synthesis. This finding was confirmed by the arachidonic acid test. Together, these results indicate that properties of EEC might be further explored in the search for newer tools to treat painful inflammatory conditions, including those related to pro-oxidant states.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Combretum/chemistry , Plant Extracts/pharmacology , Analgesics/antagonists & inhibitors , Animals , Arachidonic Acid/toxicity , Carrageenan/toxicity , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Ethanol , Hydroxyl Radical/analysis , Male , Mice , Naloxone/metabolism , Nitric Oxide/analysis , Pain/drug therapy , Pain Measurement , Plant Leaves/chemistry , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Rodentia , Thiobarbituric Acid Reactive Substances/analysis
7.
J Biomed Biotechnol ; 2011: 678673, 2011.
Article in English | MEDLINE | ID: mdl-21660140

ABSTRACT

The antioxidant and antinociceptive activities of Citrus limon essential oil (EO) were assessed in mice or in vitro tests. EO possesses a strong antioxidant potential according to the scavenging assays. Moreover, it presented scavenger activity against all in vitro tests. Orally, EO (50, 100, and 150 mg/kg) significantly reduced the number of writhes, and, at highest doses, it reduced the number of paw licks. Whereas naloxone antagonized the antinociceptive action of EO (highest doses), this suggested, at least, the participation of the opioid system. Further studies currently in progress will enable us to understand the action mechanisms of EO.


Subject(s)
Analgesics/antagonists & inhibitors , Antioxidants/pharmacology , Citrus , Free Radical Scavengers/pharmacology , Oils, Volatile/administration & dosage , Pain/drug therapy , Plant Extracts/administration & dosage , Administration, Oral , Analgesics/analysis , Animals , Antioxidants/analysis , Citrus/chemistry , Disease Models, Animal , Free Radical Scavengers/analysis , Male , Mice , Naloxone , Oils, Volatile/isolation & purification , Pain Measurement , Plant Extracts/analysis , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Rutaceae
8.
Pain ; 143(3): 228-232, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19375225

ABSTRACT

The duration of noxious stimulus-induced antinociception (NSIA) has been shown to outlast the pain stimulus that elicited it, however, the mechanism that determines the duration of analgesia is unknown. We evaluated the role of spinal excitatory and inhibitory receptors (NMDA, mGluR(5), mu-opioid, GABA(A), and GABA(B)), previously implicated in NSIA initiation, in its maintenance. As in our previous studies, the supraspinal trigeminal jaw-opening reflex (JOR) in the rat was used for nociceptive testing because of its remoteness from the region of drug application, the lumbar spinal cord. NSIA was reversed by antagonists for two inhibitory receptors (GABA(B) and mu-opioid) but not by antagonists for either of the two excitatory receptors (NMDA and mGluR(5)), indicating that NSIA is maintained by ongoing activity at inhibitory synapses in the spinal cord. Furthermore, spinal administration of the GABA(B) agonist baclofen mimicked NSIA in that it could be blocked by prior injection of the mu-opioid receptor antagonist H-D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP) in nucleus accumbens. CTAP also blocked baclofen antinociception when administered in the spinal cord. We conclude that analgesia induced by noxious stimulation is maintained by activity in spinal inhibitory receptors.


Subject(s)
Neural Inhibition/physiology , Pain Threshold/physiology , Pain/physiopathology , Spinal Cord/physiology , Synaptic Transmission/physiology , Analgesics/antagonists & inhibitors , Animals , Baclofen/pharmacology , Capsaicin/antagonists & inhibitors , Drug Interactions/physiology , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA-B Receptor Agonists , GABA-B Receptor Antagonists , Male , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Pain/chemically induced , Pain Threshold/drug effects , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Sensory System Agents/antagonists & inhibitors , Spinal Cord/drug effects , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects
9.
Eur J Pharmacol ; 496(1-3): 93-7, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15288580

ABSTRACT

In this study, we investigated whether spinal noradrenergic and serotonergic systems are involved in the antinociception induced by the novel pyrazolines 3-methyl- and 3-phenyl-5-hydroxy-5-trichloromethyl-4,5-dihydro-1H-1-pyrazole-1-carboxyamide (MPCA and PPCA, respectively), and the pyrazolinone dipyrone in the acetic acid writhing (stretching) test in mice. Intrathecal (i.t.) administration of methysergide (3 and 10 microg) and yohimbine (3 microg), but not of prazosin (0.3 and 1 microg) prevented the antinociceptive action of MPCA and PPCA (500 micromol/kg, s.c.). Dipyrone-induced antinociception (500 micromol/kg, s.c.) was not affected by methysergide or adrenoceptor antagonists. These results suggest that spinal 5-HT receptors and alpha2-adrenoceptors are involved in the antinociception induced by MPCA and PPCA, but not in that elicited by dipyrone.


Subject(s)
Analgesics/antagonists & inhibitors , Analgesics/pharmacology , Dipyrone/pharmacology , Pyrazoles/pharmacology , Receptors, Adrenergic, alpha-2/physiology , Receptors, Serotonin/physiology , Animals , Dose-Response Relationship, Drug , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Pyrazoles/antagonists & inhibitors
10.
Eur J Pharmacol ; 494(2-3): 155-60, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15212969

ABSTRACT

The involvement of the nitric oxide (NO)/cyclic GMP pathway in the molecular mechanisms of antinociceptive drugs like morphine has been previously shown by our group. Additionally, it is known that the desensitisation of nociceptors by K(+) channel opening should be the final target for several analgesic drugs including nitric oxide donors and exogenous micro-opioid receptor agonists. In our previous study, we demonstrated that bremazocine, a kappa-opioid receptor agonist, induces peripheral antinociception by activating nitric oxide/cyclic GMP pathway. In the current study, we assessed whether bremazocine is capable to activate K(+) channels eliciting antinociception. Bremazocine (20, 40 and 50 microg) dose-dependently reversed the hyperalgesia induced in the rat paw by local injection of carrageenan (250 microg) or prostaglandin E(2) (2 microg), measured by the paw pressure test. Using the selective kappa-opioid receptor antagonist nor-binaltorphimine (Nor-BNI, 200 microg/paw), it was confirmed that bremazocine (50 microg/paw) acts specifically on the kappa-opioid receptors present at peripheral sites. Prior treatment with the ATP-sensitive K(+) channel blockers glibenclamide (40, 80 and 160 microg) and tolbutamide (40, 80 and 160 microg) did not antagonise the antinociceptive effect of bremazocine (50 microg). The same results were obtained when we used prostaglandin E(2) (2 microg) as the hyperalgesic stimulus. The supposed participation of other types of K(+) channels was tested using the Ca(2+)-activated K(+) channel blockers dequalinium (12.5, 25 and 50 microg) and charybdotoxin (0.5, 1 and 2 microg) and different types of the non-selective K(+) channel blockers tetraethylammonium (25, 50 and 100 microg) and 4-aminopyridine (10, 25 and 50 microg). None of the K(+) channel blockers reversed the antinociceptive effect of bremazocine. On the basis of these results, we suggest that K(+) channels are not involved in the peripheral antinociceptive effect of bremazocine, although this opioid receptor agonist induces nitric oxide/cGMP pathway activation.


Subject(s)
Analgesics/pharmacology , Benzomorphans/pharmacology , Naltrexone/analogs & derivatives , Peripheral Nervous System/drug effects , Potassium Channels/drug effects , Receptors, Opioid, kappa/agonists , 4-Aminopyridine/pharmacology , ATP-Binding Cassette Transporters , Analgesics/antagonists & inhibitors , Animals , Benzomorphans/antagonists & inhibitors , Charybdotoxin/pharmacology , Cyclic GMP/physiology , Dequalinium/pharmacology , Glyburide/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , KATP Channels , Male , Naltrexone/pharmacology , Nitric Oxide/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying , Rats , Rats, Wistar , Signal Transduction/drug effects , Tetraethylammonium/pharmacology , Tolbutamide/pharmacology
11.
Eur J Pharmacol ; 484(2-3): 201-8, 2004 Jan 26.
Article in English | MEDLINE | ID: mdl-14744604

ABSTRACT

The effect of K+ channel inhibitors on the antiallodynic activity induced by spinal gabapentin was assessed in rats. Ligation of L5 and L6 spinal nerves made the rats allodynic, whereas that intrathecal administration of gabapentin (25-200 microg) reduced tactile allodynia in a dose-dependent manner. Spinal pretreatment with glibenclamide (12.5-50 microg, ATP-sensitive K+ channel inhibitor), charybdotoxin (0.01-1 ng) or apamin (0.1-3 ng, large-and small-conductance Ca2+-activated K+ channel blockers, respectively), but not margatoxin (0.01-10 ng, voltage-dependent K+ channel inhibitor), significantly prevented gabapentin-induced antiallodynia. Pinacidil (1-30 microg, K+ channel opener) significantly reduced nerve ligation-induced allodynia. Intrathecal glibenclamide (50 microg), charybdotoxin (1 ng) and apamin (3 ng), but not margatoxin (10 ng), significantly reduced pinacidil-induced antiallodynia. K+ channel inhibitors alone did not modify allodynia produced by spinal nerve ligation. Results suggest that gabapentin and pinacidil may activate Ca2+-activated and ATP-sensitive K+ channels in order to produce part of its spinal antiallodynic effect in the Chung model.


Subject(s)
Acetates/pharmacology , Amines , Analgesics/pharmacology , Cyclohexanecarboxylic Acids , Pain Measurement/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , gamma-Aminobutyric Acid , Acetates/antagonists & inhibitors , Analgesics/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Female , Gabapentin , Pain Measurement/methods , Potassium Channels/physiology , Rats , Rats, Wistar
12.
La Plata; s.n; abr; 2001. 24 p. ilus.
Non-conventional in Spanish | BINACIS | ID: biblio-1201768

ABSTRACT

eN LA ACTUALIDAD, ATENDIENDO A LOS MODERNOS CONCEPTOS DE LA MEDICINA basada en la evidencia


Subject(s)
Analgesics/antagonists & inhibitors , Anti-Inflammatory Agents
13.
La Plata; s.n; abr; 2001. 24 p. ilus. (78483).
Non-conventional in Spanish | BINACIS | ID: bin-78483

ABSTRACT

eN LA ACTUALIDAD, ATENDIENDO A LOS MODERNOS CONCEPTOS DE LA MEDICINA basada en la evidencia


Subject(s)
INFORME DE CASO , Analgesics/antagonists & inhibitors , Anti-Inflammatory Agents
14.
J Ethnopharmacol ; 70(3): 323-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10837994

ABSTRACT

The essential oil (EO) from leaves of Cymbopogon citratus increased the reaction time to thermal stimuli both after oral (25 mg/kg) and intraperitoneal (25-100 mg/kg) administration. EO (50-200 mg/kg, p.o. or i.p.) strongly inhibited the acetic acid-induced writhings in mice. In the formalin test, EO (50 and 200 mg/kg, i.p.) inhibited preferentially the second phase of the response, causing inhibitions of 100 and 48% at 200 mg/kg, i.p. and 100 mg/kg, p.o., respectively. On the other hand, the opioid antagonist naloxone blocked the central antinociceptive effect of EO, suggesting that EO acts both at peripheral and central levels.


Subject(s)
Analgesics/pharmacology , Oils, Volatile/pharmacology , Plants, Medicinal/chemistry , Poaceae/chemistry , Acetates , Analgesics/antagonists & inhibitors , Analgesics/isolation & purification , Animals , Behavior, Animal/drug effects , Caribbean Region , Chromatography, Gas , Formaldehyde , Male , Mass Spectrometry , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oils, Volatile/isolation & purification , Pain Measurement/drug effects , Plant Leaves/chemistry , Reaction Time/drug effects
15.
Eur J Pharmacol ; 310(2-3): 123-7, 1996 Aug 29.
Article in English | MEDLINE | ID: mdl-8884207

ABSTRACT

The i.c.v. administration of bradykinin (4, 8 and 16 micrograms) induced antinociception in mice which was resistant to naloxone; furthermore, the induction of tolerance to morphine by a single s.c. injection (100 mg/kg, 24 h before test doses of the peptide) did not affect antinociception. Since bradykinin is known to increase nitric oxide (NO) in peripheral tissues, we studied the possibility that its antinociceptive action may be related to NO effects in the central nervous system. Bradykinin effects were antagonized by previous treatment with NG-nitro-L-arginine or concomitant i.c.v. administration of bradykinin and methylene blue. The immediate precursor of NO, L-arginine, which by itself produces analgesia, also reduced bradykinin effects; moreover, tolerance to L-arginine significantly decreased the response to the peptide. These results suggest that NO is involved in antinociception induced by i.c.v. administration of bradykinin.


Subject(s)
Analgesics/pharmacology , Bradykinin/pharmacology , Nitric Oxide/physiology , Analgesics/administration & dosage , Analgesics/antagonists & inhibitors , Animals , Arginine/pharmacology , Bradykinin/administration & dosage , Bradykinin/antagonists & inhibitors , Drug Tolerance , Injections, Intraventricular , Male , Methylene Blue/pharmacology , Mice , NG-Nitroarginine Methyl Ester/pharmacology
16.
Brain Res ; 647(2): 220-30, 1994 Jun 06.
Article in English | MEDLINE | ID: mdl-7922498

ABSTRACT

The changes in tail-flick latency (TFL) to noxious heating of the skin produced by the microinjection of carbachol (CCh) into the dorsal (dPAG), lateral (lPAG), and ventral (vPAG) portions of the mesencephalic periaqueductal gray matter (PAG) were studied in the rat. A significant increase in TFL was produced by CCh (0.2 microgram/0.5 microliter) microinjected into sites widely distributed within the PAG. The effect of CCh was stronger in the most caudal portion of the DPAG. Smaller effects were obtained after injection of CCh into the aqueduct, indicating that drug diffusion from the injection sites to the aqueduct lumen is unlikely to cause the antinociceptive effect of CCh. Dimethyl-phenyl-piperazinium (0.35 microgram/0.5 microliter), but not bethanechol (0.22 and 0.44 microgram/0.5 microliter), produced effects similar to CCh (0.2 microgram/0.5 microliter), when injected into the dPAG. The effects of CCh were inhibited by the previous administration of mecamylamine (1 microgram/0.5 microliter), but not atropine (1 microgram/0.5 microliter) or naloxone (1 microgram/0.5 microliter), into the dPAG. These results are indicative that antinociception produced by CCh from the dPAG depends on nicotinic, but not muscarinic or opioid mechanisms within the dPAG. The intraperitoneal administration of phenoxybenzamine (1 mg/kg) or mecamylamine (1 mg/kg), but not naloxone (1 mg/kg), methysergide (1 mg/kg), or atropine (1 mg/kg), inhibited the effects of CCh injected into the dPAG. In contrast, a higher dose of intraperitoneal phenoxybenzamine (5 mg/kg) was ineffective against the antinociception evoked by CCh when injected into the vPAG. Therefore, the effects of CCh from the dPAG may depend on the activation of centrifugal pathways involving both nicotinic and alpha-adrenergic mechanisms. In addition, the results indicate that different cholinergic substrates in the PAG may mediate both alpha-adrenergic and non-alpha-adrenergic descending pain mechanisms activated by the dPAG and vPAG, respectively.


Subject(s)
Analgesics/pharmacology , Carbachol/pharmacology , Mesencephalon/physiology , Periaqueductal Gray/physiology , Analgesics/administration & dosage , Analgesics/antagonists & inhibitors , Animals , Bethanechol/pharmacology , Brain Mapping , Carbachol/administration & dosage , Carbachol/antagonists & inhibitors , Dimethylphenylpiperazinium Iodide/pharmacology , Injections, Intraperitoneal , Male , Mesencephalon/anatomy & histology , Methysergide/pharmacology , Microinjections , Naloxone/pharmacology , Pain Measurement/drug effects , Periaqueductal Gray/anatomy & histology , Phenoxybenzamine/pharmacology , Rats , Rats, Wistar , Skin Temperature/drug effects
17.
Life Sci ; 50(12): 891-7, 1992.
Article in English | MEDLINE | ID: mdl-1545667

ABSTRACT

The involvement of opioid peptides in the mechanism of action of vouacapan, a new experimental compound extracted from seeds of Pterodon poligalaeflorus Benth, was investigated both in mice utilizing acetic acid writhing response and in rats utilizing inflammatory hyperalgesia induced by carrageenan and modified Randall-Selitto method. Vouacapan, in both models, caused a dose-dependent analgesia when injected p.o., s.c. and i.p. The analgesic effect was partially blocked by naloxone, nalorphine and n-methyl-nalorphine. Significant tolerance to analgesic effect was observed following repeated administration of vouacapan or morphine. On the last day of treatment, cross administration revealed symmetrical and asymmetrical cross-tolerance between vouacapan and morphine, in rats and mice, respectively. We conclude that a release of endorphins could be involved in the analgesic mechanism of vouacapan in both models tudied.


Subject(s)
Analgesics/administration & dosage , Diterpenes/administration & dosage , Endorphins/physiology , Pain Measurement/drug effects , Analgesics/antagonists & inhibitors , Animals , Diterpenes/antagonists & inhibitors , Drug Administration Routes , Drug Tolerance , Male , Mice , Morphine/administration & dosage , Nalorphine/analogs & derivatives , Nalorphine/pharmacology , Naloxone/pharmacology , Rats , Rats, Inbred Strains
18.
Rev. mex. anestesiol ; 9(4): 217-20, oct.-dic. 1986. tab
Article in Spanish | LILACS | ID: lil-65759

ABSTRACT

Se evaluó la eficacia e un antidepresivo tricíclico (amitriptilina) como potenciador de analgésicos en pacientes oncológico terminales, con dolor resistente a tratamiento convencional. El estudio se realizó con un diseño doble ciego, controlado con placebo. El efecto del medicamento se determinó mediante tres índices: el dolor al momento de la entrevista, el dolor habitual y el peor dolor durante la última semana, que se calificaron numéricamente. Se definió como mejoría la reducción en por lo menos el 25% del dolor manifestado al inicio del tratamiento. Unicamente en el grupo recibiendo amitriptilina se observaron mejorías del 100%, aunque las diferencias entre ambos grupos no fueron significativas. Se comentan los resultados y los problemas en la realización de este tipo de estudios


Subject(s)
Humans , Male , Female , Pain/therapy , Amitriptyline/therapeutic use , Neoplasms , Analgesics/antagonists & inhibitors , Pain/psychology
SELECTION OF CITATIONS
SEARCH DETAIL