Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-32713857

ABSTRACT

Various animal models, especially rodents, are used to study pain, due to the difficulty of studying it in humans. Many drugs that produce analgesia have been studied and there is evidence among which NSAIDs deserve to be highlighted. Dexketoprofen (DEX) provides a broad antinociceptive profile in different types of pain; therefore, this study was designed to evaluate the profile of antinociceptive potency in mice. Analgesic activity was evaluated using the acetic acid abdominal constriction test (writhing test), a chemical model of visceral pain. Dose-response curves for i.p. DEX administration (1, 3, 10, 30 and 100 mg/kg), using at least six mice in each of at least five doses, was obtained before and 30 min after pre-treatment with different pharmacological agents. Pretreatment of the mice with opioid receptor antagonists was not effective; however, the serotonin receptor antagonist and nitric oxide synthase inhibitor produce a significant increase in DEX-induced antinociception. The data from the present study shows that DEX produces antinociception in the chemical twisting test of mice, which is explained with difficulty by the simple inhibition of COX. This effect appears to be mediated by other mechanisms in which the contribution of the NO and 5-HT pathways has an important effect on DEXinduced antinociception.


Subject(s)
Ketoprofen/analogs & derivatives , Receptors, Opioid/genetics , Receptors, Serotonin/genetics , Tromethamine/pharmacology , Visceral Pain/drug therapy , Acetic Acid/pharmacology , Analgesia/methods , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dose-Response Relationship, Drug , Humans , Ketoprofen/pharmacology , Mice , Narcotic Antagonists/pharmacology , Nitric Oxide/genetics , Serotonin/genetics , Serotonin Antagonists/pharmacology , Visceral Pain/genetics , Visceral Pain/pathology
2.
Inflammopharmacology ; 27(1): 151-155, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30317449

ABSTRACT

Neuropathic pain is a complication of cancer and diabetes mellitus and the most commonly used drugs in the treatment of the diabetic neuropathic pain have only limited efficacy. The aim of this study was to evaluate the role of the biomarker interleukin-1beta (IL-1ß) in the pharmacological interaction of gabapentin with tramadol in a model of diabetic neuropathic pain. CF-1 male mice, pretreated with 200 mg/kg i.p. of streptozocin (STZ), were used and at day 3 and 7 were evaluated by the hot plate test and the spinal cord level of IL-1ß was determined. Antinociceptive interaction of the coadministration i.p. of gabapentin with tramadol, in basic of the fixed the ratio 1:1 of their ED50 values alone, was ascertained by isobolographic analysis. Tramadol was 1.13 times more potent than gabapentin in saline control mice, 1.40 times in STZ mice at 3 days and 1.28 times in STZ at 7 days. The interaction between gabapentin and tramadol was synergic, with an interaction index of 0.30 and 0.22 for mice pretreated with STZ at 3 and 7 days. The combination of gabapentin with tramadol reversed the increased concentration of IL-1ß induced by STZ in diabetic neuropathic mice. These findings could help clarify the mechanism of diabetic neuropathy.


Subject(s)
Diabetic Neuropathies/complications , Gabapentin/pharmacology , Interleukin-1beta/metabolism , Neuralgia/drug therapy , Neuralgia/genetics , Tramadol/pharmacology , Analgesics/pharmacology , Animals , Diabetic Neuropathies/metabolism , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination/methods , Male , Mice , Neuralgia/metabolism , Pain Measurement/methods , Spinal Cord/drug effects , Spinal Cord/metabolism , Streptozocin/pharmacology
4.
Inflammopharmacology ; 25(1): 91-97, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28155118

ABSTRACT

OBJECTIVE: Diabetic neuropathy (DN) is the most common complication of diabetes and pain is one of the main symptoms of diabetic neuropathy, however, currently available drugs are often ineffective and complicated by adverse events. The purpose of this research was to evaluate the antinociceptive interaction between gabapentin and minocycline in a mice experimental model of DN by streptozocin (STZ). METHODS: The interaction of gabapentin with minocycline was evaluated by the writhing and hot plate tests at 3 and 7 days after STZ injection or vehicle in male CF1 mice. RESULTS: STZ (150 mg/kg, i.p.) produced a marked increase in plasma glucose levels on day 7 (397.46 ± 29.65 mg/dL) than on day 3 (341.12 ± 35.50 mg/dL) and also developed neuropathic pain measured by algesiometric assays. Gabapentin produced similar antinociceptive activity in both writhing and hot plate tests in mice pretreated with STZ. However, minocycline was more potent in the writhing than in the hot plate test in the same type of mice. The combination of gabapentin with minocycline produced synergistic interaction in both test. CONCLUSION: The combination of gabapentin with minocycline in a 1:1 proportion fulfills all the criteria of multimodal analgesia and this finding suggests that the combination provide a therapeutic alternative that could be used for human neuropathic pain management.


Subject(s)
Amines/administration & dosage , Analgesics/administration & dosage , Cyclohexanecarboxylic Acids/administration & dosage , Diabetic Neuropathies/drug therapy , Minocycline/administration & dosage , Pain Measurement/drug effects , gamma-Aminobutyric Acid/administration & dosage , Amines/metabolism , Analgesics/metabolism , Animals , Cyclohexanecarboxylic Acids/metabolism , Diabetic Neuropathies/metabolism , Dose-Response Relationship, Drug , Drug Interactions/physiology , Drug Therapy, Combination , Gabapentin , Male , Mice , Minocycline/metabolism , Pain Measurement/methods , gamma-Aminobutyric Acid/metabolism
5.
Pharmacol Biochem Behav ; 100(1): 125-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21871913

ABSTRACT

Atorvastatin is a statin that inhibits the 3-hydroxy-methyl-glutaryl coenzyme A (HMG-CoA) reductase. Several landmark clinical trials have demonstrated the beneficial effects of statin therapy for primary and secondary prevention of cardiovascular disease. It is assumed that the beneficial effects of statin therapy are entirely due to cholesterol reduction. Statins have an additional activity (pleiotropic effect) that has been associated to their anti-inflammatory effects. The aim of the present study was to assess the antinociceptive activity of atorvastatin in five animal pain models. The daily administration of 3-100mg/kg of atorvastatin by oral gavage induced a significant dose-dependent antinociception in the writhing, tail-flick, orofacial formalin and formalin hind paw tests. However, this antinociceptive activity of atorvastatin was detectable only at high concentrations in the hot plate assay. The data obtained in the present study demonstrates the effect of atorvastatin to reduce nociception and inflammation in different animal pain models.


Subject(s)
Disease Models, Animal , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Pain Measurement/drug effects , Pain Measurement/methods , Pain/drug therapy , Pyrroles/pharmacology , Pyrroles/therapeutic use , Animals , Atorvastatin , Dose-Response Relationship, Drug , Hot Temperature/adverse effects , Male , Mice , Pain/physiopathology
6.
Pharmacol Biochem Behav ; 97(3): 423-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20887749

ABSTRACT

Animal models are used to research the mechanisms of pain and to mimic human pain. The purpose of this study was to determine the degree of interaction between dexketoprofen and dexibuprofen, by isobolographic analysis using the formalin orofacial assay in mice. This assay presents two-phase time course: an early short-lasting, phase I, starting immediately after the formalin injection producing a tonic acute pain, leaving a 15 min quiescent period, followed by a prolonged, phase II, after the formalin and representing inflammatory pain. Administration of dexketoprofen or dexibuprofen produced a dose-dependent antinociception, with different potency, either during phases I or II. The co-administration of dexketoprofen and dexibuprofen produced synergism in phase I and II. In conclusion, both dexketoprofen and dexibuprofen are able to induce antinociception in the orofacial formalin assay. Their co-administration produced a synergism, which could be related to the different degree of COX inhibition and other mechanisms of analgesics.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ibuprofen/pharmacology , Ketoprofen/pharmacology , Animals , Behavior, Animal , Drug Interactions , Face , Male , Mice , Mouth , Pain Measurement , Stereoisomerism
7.
Inflammopharmacology ; 18(2): 65-71, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127283

ABSTRACT

OBJECTIVE AND DESIGN: The antinociception induced by the intraperitoneal coadministration in mice of combinations of metamizol and paracetamol was evaluated in the tail flick test and orofacial formalin test. METHODS: The antinociception of each drugs alone and the interaction of the combinations was evaluated by isobolographic analysis in the tail-flick and in the formalin orofacial assay of mice. RESULTS: Mice pretreated with the drugs demonstrated that the antinociception of metamizol and paracetamol is dose-dependent. The potency range on the antinocifensive responses for metamizol or paracetamol was as follows: orofacial (Phase II) > orofacial (Phase I) > tail flick. In addition, the coadministration of metamizol with paracetamol induced a strong synergistic antinociception in the algesiometer assays. Both drugs showed effectiveness in inflammatory pain. CONCLUSION: These actions can be related to the differential selectivity of the drugs for inhibition of COX isoforms and also to the several additional antinociception mechanisms and pathways initiated by the analgesic drugs on pain transmission. Since the efficacy of the combination of metamizol with paracetamol has been demonstrated in the present study, this association could have a potential beneficial effect on the pharmacological treatment of clinical pain.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Pain/prevention & control , Prostaglandin-Endoperoxide Synthases/metabolism , Acetaminophen/therapeutic use , Animals , Dipyrone/therapeutic use , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Formaldehyde/pharmacology , Hot Temperature , Male , Mice , Mice, Inbred Strains , Pain/chemically induced , Pain Measurement/methods
8.
J Physiol Pharmacol ; 60(2): 71-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19617648

ABSTRACT

To enhance analgesia, combination of analgesics drugs of proven efficacy is a strategy which is accompanied by a reduction of adverse effects. The present study was undertaken to characterize the antinociceptive interaction of morphine with different non-steroidal anti-inflammatory drugs (NSAIDs) using isobolographic analysis and the writhing test of mice. One of the possible mechanisms of action of spinally administered morphine with non-steroidal antiinflammatory drugs was investigated using the DOR antagonist naltrindole. The study demonstrated a synergistic antinociception of spinal administered combinations of morphine with the following NSAIDs agents: diclofenac, ketoprofen, meloxicam, metamizol, naproxen, nimesulide, parecoxib and piroxicam. The supraadditive effect seems to be independent of the selectivity of each NSAIDs to inhibit COX-1 or COX-2. The findings of the present work suggest that the combinations of opioids and non-steroidal anti-inflammatory drugs have a direct action on spinal processing of the nociceptive information, which may achieved by additional mechanisms independent of prostaglandin synthesis inhibition and/or activation of opioid receptors. The lack of effect of naltrindole to modify the analgesic activity of the combination of opioids and NSAIDs indicates that others pain regulatory systems are involved in this central action. Therefore, these combinations could be a viable alternative to clinical pain management, especially trough multimodal analgesia.


Subject(s)
Analgesics, Opioid/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Morphine/pharmacology , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/physiology , Spinal Cord/drug effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Animals , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/therapeutic use , Drug Synergism , Injections, Spinal , Male , Mice , Mice, Inbred Strains , Morphine/administration & dosage , Morphine/therapeutic use , Naltrexone/administration & dosage , Naltrexone/pharmacology , Naltrexone/therapeutic use , Pain/drug therapy , Pain/physiopathology , Receptors, Opioid, delta/antagonists & inhibitors , Spinal Cord/physiopathology
9.
Pharmacol Res ; 60(4): 291-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19374948

ABSTRACT

The antinociceptive activity of dexketoprofen was studied in mice using the formalin assay for orofacial pain. The interaction between dexketoprofen and co-administered tramadol was studied using isobolographic analysis. The intraperitoneal administration of dexketoprofen or tramadol, showed dose-dependent antinociceptive activity in both phases of the assay. When administered together, the interaction was mildly synergistic during the first phase, and antagonistic in the second phase. Selective opioid receptor antagonists where used in order to measure the analgesic activity of tramadol in other regions of the CNS. The co-administration of dexketoprofen and tramadol, with previous administration of naltrexone, showed high synergistic activity during the first phase, and less but still synergistic during the second. When using naltrindole, the interaction was mildly more synergistic than the mixture dexketoprofen+tramadol during both phases. Using norbinaltorphimine, the interaction was synergistic in both phases, more marked in the second. These results suggest that the opioid activity of tramadol has an inhibiting effect in antinociceptive activity of the interaction between dexketoprofen and tramadol during the inflammatory (late) stages of pain.


Subject(s)
Analgesics/therapeutic use , Ketoprofen/therapeutic use , Narcotic Antagonists/pharmacology , Pain/drug therapy , Tramadol/therapeutic use , Analgesics/pharmacology , Animals , Drug Synergism , Ketoprofen/pharmacology , Male , Mice , Narcotic Antagonists/administration & dosage , Pain/chemically induced , Tramadol/pharmacology
10.
Fundam Clin Pharmacol ; 23(1): 81-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19267773

ABSTRACT

The purpose of the present study was to evaluate the nature of the antinociceptive interaction among dexketoprofen (DEX), a mixed inhibitor of the cyclo-oxygenases, and tramadol (TRAM), a weak opioid with monoaminergic activity that inhibits norepinephrine and serotonin re-uptake. We assessed antinociception in the acetic acid writhing test, the tail flick and the formalin (FT) tests, and gastrointestinal transit (GIT) after the administration of a charcoal meal. The analysis of the interaction was carried out using isobolograms and interaction indexes or the fixed-dose method GIT. The administration of DEX or TRAM individually induced dose-dependent antinociception in all the algesiometric tests. In the three tests, TRAM was between 5.2 (FT, phase I) and 35 times (FT, Phase II) more potent than DEX. When testing combinations at different potency ratios (1 : 1, 1 : 3, 3 : 1), we could demonstrate synergy in all algesiometric tests, only when drugs were combined in a 1 : 1 proportion. Interestingly, the proportion of the drugs in the combination could change the type of interaction from synergy to antagonism. On the inhibition of GIT, a dose-related inhibition was established for TRAM, but not for DEX. Using a fixed-dose protocol, we could demonstrate antagonism between DEX and TRAM on the inhibition of GIT. The results of the present study suggest that a combination of DEX and TRAM in a 1 : 1 proportion could be adequate to use in future clinical trials in humans.


Subject(s)
Gastrointestinal Transit/drug effects , Ketoprofen/analogs & derivatives , Pain/drug therapy , Tramadol/pharmacology , Tromethamine/analogs & derivatives , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Charcoal , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Ketoprofen/administration & dosage , Ketoprofen/pharmacology , Male , Mice , Pain/etiology , Pain Measurement , Tramadol/administration & dosage , Tromethamine/administration & dosage , Tromethamine/pharmacology
11.
Pharmacol Biochem Behav ; 92(2): 314-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167421

ABSTRACT

Opioids and non-steroidal anti-inflammatory drugs (NSAIDs) are used to relieve acute and chronic pain. The purpose of this study was to determine the degree of interaction between dexketoprofen and NSAID examples of COXs inhibitors using the isobolographic analysis in the formalin orofacial test in mice. The drugs, i.p., induced a dose-dependent antinociception with different potencies in both test phases. Combinations of dexketoprofen with naproxen, nimesulide, ibuprofen or paracetamol on the basis of the fixed ratio (1:1) of their ED(50)'s values alone demonstrated synergism in both phases. This is important since the orofacial pain is a test not currently used in mice; the drugs are all analgesic for humans and phase II is representative of inflammatory pain. The synergism was: COX-3>COX-2>COX-1 inhibitors, this is particularly interesting since the inhibitor of COX-3, paracetamol, displayed a robust anti-inflammatory activity in an assay of acute and inflammatory pain that mimics inflammatory pain in humans. In conclusion, the synergism of the dexketoprofen/NSAID combinations may improve this type of therapeutic profile, since with low doses of the components, side effects are not likely to occur, and they may be used in long-term treatments.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Formaldehyde/toxicity , Pain/drug therapy , Animals , Drug Synergism , Face , Female , Male , Mice , Mouth , Pain/chemically induced
12.
J Ethnopharmacol ; 112(1): 162-5, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17403589

ABSTRACT

Leaf extracts of Ugni molinae Turcz. (Myrtaceae) are used in Chilean folk medicine as analgesic and anti-inflammatory. The antinociceptive effect of dichloromethane (DCM), ethyl acetate (EA) and methanol (ME) leaf extracts was assessed by intraperitoneal, oral and topical administration in writhing, tail flick, and tail formalin tests in mice. The extracts showed a dose-dependent antinociceptive activity in all the assays under different administration routes. The ED(50) values for the different tests for the DCM, EA, ME extract and reference drug (ibuprofen) were as follows. Writhing test in acetic acid (i.p. administration): 0.21, 0.37, 1.37 and 0.85mg/kg, respectively; tail flick test (oral administration): 199, 189, 120 and 45.9mg/kg. The EC(50) values for tail flick test were (topical administration): 2.0, 0.35, 1.4 and 8.2% (w/v), respectively; and the topical analgesic effects were (formalin assay) 75.5, 77.5, 31.6 and 76.5%, respectively. Ugni molinae extracts produce antinociception in chemical and thermal pain models through a mechanism partially linked to either lipooxygenase and/or cyclooxygenase via the arachidonic acid cascade and/or opioid receptors. Flavonoid glycosides and triterpenoids have been isolated from the plant and can be associated with the observed effect. Our results corroborate the analgesic effects of Ugni molinae, and justify its traditional use for treating pain.


Subject(s)
Analgesics/therapeutic use , Myrtaceae , Pain/drug therapy , Acute Disease , Analgesics/chemistry , Animals , Flavonoids/analysis , Glycosides/analysis , Ibuprofen/therapeutic use , Male , Mice , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Plant Leaves , Triterpenes/analysis
13.
Pharmacol Res ; 50(3): 273-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15225670

ABSTRACT

The present study was undertaken to characterize the interactions between nonsteroidal anti-inflammatory drugs and the alpha(2)-adrenoceptor agonist clonidine in an acute nociceptive test. The writhing test was selected as a model of acute visceral pain. Isobolograms were constructed to assess the interactions of clonidine and each nonsteroidal anti-inflammatory drugs, when coadministered intraperitoneally and intrathecally (i.t.). The simultaneous intraperitoneal administration of fixed ratios of ED(50) fractions of all nonsteroidal anti-inflammatory drugs (naproxen, piroxicam, paracetamol, dipyrone or metamizol and nimesulide) combined with clonidine resulted in synergistic interactions. The same combinations administered intrathecally were additive. The synergistic interactions between systemic nonsteroidal anti-inflammatory drugs and clonidine may involve supraspinal mechanisms.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Clonidine/pharmacology , Pain Measurement/drug effects , Analgesics/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Clonidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions/physiology , Drug Therapy, Combination , Female , Injections, Intraperitoneal , Injections, Spinal , Male , Mice , Pain Measurement/methods
14.
Can J Physiol Pharmacol ; 82(5): 331-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15213733

ABSTRACT

The intraperitoneal administration of morphine, diclofenac, ketoprofen, meloxicam, metamizol, paracetamol and piroxicam induced dose-dependent antinociception in mice tested with the acetic acid writhing test. The isobolographic analysis of the simultaneous intraperitoneal administration of fractions of the ED50's of morphine with each nonsteroidal anti-inflammatory drug (NSAID) demonstrated the existence of a supra-additive interaction (synergy). The selective antagonist of micro -opioid receptors naltrexone partially reversed the supra-additive interactions to additive interactions; however, the combinations of morphine/metamizol and morphine/paracetamol were completely antagonized, resulting in subadditive interactions. The selective antagonist of delta-opioid receptors naltrindole failed to significantly attenuate the combinations of morphine with ketoprofen, meloxicam and piroxicam, but decreased the activity of the combinations of morphine with diclofenac, metamizol and paracetamol, transforming the interactions from supra-additive to additive. Nor-binaltorphimine was used to evaluate the involvement of kappa-opioid receptors. Nor-binaltorphimine did not modify the supra-additive interaction of morphine and NSAIDs and the additive interaction of the co-administration of morphine and metamizol. The synergy between morphine and NSAIDs could be related to different pathways of pain transmission, probably related to the different intracellular signal transduction mechanisms of action of opioid and non-opioid agents.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Morphine/pharmacology , Pain Measurement/drug effects , Animals , Dose-Response Relationship, Drug , Drug Synergism , Female , Male , Mice , Pain Measurement/methods
15.
Brain Res Bull ; 61(4): 417-25, 2003 Aug 30.
Article in English | MEDLINE | ID: mdl-12909285

ABSTRACT

The antinociceptive activity of nonsteroidal anti-inflammatory drugs (NSAIDs) has been explained mainly on the basis of their inhibition of the enzyme cyclooxygenase (COX); however, this inhibition is not enough to completely explain the analgesic efficacy of these drugs. The modulation exerted by serotonergic systems on antinociception is well known. The purpose of the present work was to further explore the role of serotonin in the antinociceptive activity of NSAIDs using the writhing test and the tail-flick test of the mice after the inhibition of serotonin biosynthesis with intraperitoneal p-chlorophenylalanine (p-CPA). Pretreatment with p-CPA produced a significant decrease in the antinociceptive activity of NSAIDs administered either by the intraperitoneal or intrathecal routes, in both algesiometric tests. These results suggest a complementary mechanism of antinociception for NSAIDs, independent of their ability to inhibit the activity of COX, involving the activation of descending serotonergic pathways. By the pharmacological nature of the study, one limitation was the absence of biochemical measurement of the synthesis of 5-HT, since the reduction of the brain 5-HT synthesis by pretreatment with p-CPA will be expressed as a diminished antinociceptive activity of NSAIDs, which would be a new argument to consider NSAIDs acting as central analgesic agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Pain Measurement/drug effects , Pain/metabolism , Serotonin/biosynthesis , Animals , Dose-Response Relationship, Drug , Drug Interactions , Female , Fenclonine/pharmacology , Male , Mice , Reaction Time/drug effects , Serotonin Antagonists/pharmacology , Time Factors
16.
Pharmacol Biochem Behav ; 74(3): 603-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12543225

ABSTRACT

The nonsteroidal anti-inflammatory drugs (NSAIDs) clonixin, diclofenac, piroxicam, ketoprofen, meloxicam, and paracetamol induced antinociception after intraperitoneal or intrathecal administration in mice submitted to an acute thermal algesiometric test without inflammation (tail-flick). Antinociception was evaluated by the increase in reaction time difference (Delta latency), between readings obtained before and after the administration of drugs. The antinociception induced by doses of NSAIDs producing between 20% and 30% of the maximum possible effect (MPE) 30 min after intraperitoneal and 15 min after intrathecal injections was compared with the antinociception obtained after pretreatment with 1 mg/kg atropine ip, 30 min before. Systemic atropine (1 mg/kg) significantly antagonized NSAID-induced antinociception in all cases, both after intraperitoneal and intrathecal administration. Cholinergic depletion by intracerebroventricular hemicholinium-3 (HC-3, 5 microg) 5 h before prevented the antinociceptive effect of all NSAIDs. These observations suggest that intrinsic muscarinic cholinergic facilitatory pathways represent an important modulating system in pain perception in this animal model of acute thermal pain. The results of the present work support the increasingly accepted notion that NSAIDs are effective analgesics even when inflammation is not present, acting by mechanisms that involve actions on spinal and supraspinal nociceptive transmission. It is suggested that, similar to morphine and clonidine, the active mechanism of NSAIDs may involve the release of acetylcholine (ACh) in the spinal cord.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Atropine/pharmacology , Pain Measurement/drug effects , Animals , Female , Male , Mice , Pain Measurement/methods , Reaction Time/drug effects , Reaction Time/physiology
17.
Inflamm Res ; 51(5): 219-22, 2002 May.
Article in English | MEDLINE | ID: mdl-12056508

ABSTRACT

OBJECTIVE: The interactions of alpha-adrenoceptors with the antinociceptive effects of non-steroidal antiinflammatory drugs (NSAIDs) were assessed in acute thermal nociception in mice. MATERIALS AND METHODS: The analgesic effect was analyzed by the tail-flick test. RESULTS: The pretreatment with yohimbine (1 mg/kg i.p.), 30 min prior to the intraperitoneal injection of ketoprofen (50 mg/kg), diclofenac (30 mg/kg) and piroxicam (50 mg/kg) antagonized the antinociception induced by these NSAIDs, significantly reducing the tail-flick latency. Yohimbine did not affect paracetamol (125 mg/kg) induced antinociception. Prazosin (1 mg/kg i.p.) antagonized only the effect of paracetamol, without affecting the latency of the other drugs. When NSAIDs were administered i.t. (ketoprofen 2 m/kg; diclofenac 0.9 mg/kg; piroxicam 1.5 mg/kg; paracetamol 3.75 mg/kg), the same results were obtained after i.p. pretreatment with yohimbine and prazosin. The pretreatment of phenoxybenzamine (1 mg/kg i.p.) antagonized all antinociceptive effects. CONCLUSIONS: NSAIDs induced antinociception in an acute thermal pain model without inflammation. The mechanism of antinociception induced by ketoprofen, diclofenac and piroxicam involves an activation of alpha2-adrenoceptors at spinal and supraspinal levels, while paracetamol-induced antinociception is probably due mainly to central activation of the descending noradrenergic inhibitory system by alpha1-adrenoceptors.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Hot Temperature/adverse effects , Nociceptors/drug effects , Sympathetic Nervous System/physiology , Adrenergic alpha-Antagonists/pharmacology , Animals , Mice , Pain Measurement/drug effects , Phenoxybenzamine/pharmacology , Prazosin/pharmacology , Reaction Time/drug effects , Receptors, Adrenergic, alpha/drug effects , Yohimbine/pharmacology
18.
Can J Physiol Pharmacol ; 80(12): 1173-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12564643

ABSTRACT

The inhibition of cyclooxygenase enzymes by nonsteroidal anti-inflammatory drugs (NSAIDs) does not completely explain the antinociceptive efficacy of these agents. It is known that cholinergic agonists are antinociceptive, and this study evaluates the interactions between carbachol and some NSAIDs. Antinociceptive activity was evaluated in mice by the acetic acid writhing test. Dose-response curves were constructed for NSAIDs and carbachol, administered either intraperitoneally (i.p.) or intrathecally (i.t.). The interactions of carbachol with NSAIDs were evaluated by isobolographic analysis after the simultaneous administration of fixed proportions of carbachol with each NSAID. All of the drugs were more potent after spinal than after systemic administration. The combinations of NSAIDs and carbachol administered i.p. were supra-additive; however, the i.t. combinations were only additive. Isobolographic analysis of the coadministration of NSAIDs and carbachol and the fact that atropine antagonized the synergistic effect suggest that carbachol may strongly modulate the antinociceptive activity of NSAIDs; thus, central cholinergic modulation would be an additional mechanism for the antinociceptive action of NSAIDs, unrelated to prostaglandin biosynthesis inhibition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbachol/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Carbachol/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions/physiology , Drug Synergism , Mice
19.
Inflammation ; 25(4): 233-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11580099

ABSTRACT

The interaction between the antinociceptive activity of ketoprofen and adrenergic agents was evaluated in the writhing test of mice. Dose-response curves were obtained for systemic and intrathecal antinociceptive effects of ketoprofen, phenylephrine, clonidine, desipramine, and prazosin; and ED50 were calculated. The interactions were evaluated by isobolographic analysis of the systemic or intrathecal co-administration of fixed-ratio combinations of ketoprofen with each adrenergic agent. The intraperitoneal combinations of ketoprofen with phenylephrine, clonidine, and prazosin showed supra-additivity, indicating that activation of alpha1 and alpha2 adrenoceptors play a role in nociceptive transmission at supraspinal levels. The same combinations given intrathecal were only additive. Desipramine intraperitoneal was also supra-additive: however, when ketoprofen was administered intrathecally with desipramine, only an additive interaction was obtained. The supra-additive interactions suggest that complementary mechanisms of antinociception have been activated, related with interference with the multiplicity of receptors and systems involved in the transmission of the nociceptive information. Racemic ketoprofen has an antinociceptive activity which is probably not only due to COX inhibition but also involves noradrenergic systems at spinal and supraspinal levels.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Analgesics/pharmacology , Ketoprofen/pharmacology , Adrenergic alpha-Agonists/administration & dosage , Analgesics/administration & dosage , Animals , Clonidine/administration & dosage , Clonidine/pharmacology , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Drug Interactions , Injections, Intraperitoneal , Ketoprofen/administration & dosage , Mice , Phenylephrine/administration & dosage , Phenylephrine/pharmacology , Prazosin/administration & dosage , Prazosin/pharmacology
20.
Anesth Analg ; 93(2): 430-5, 4th contents page, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11473875

ABSTRACT

UNLABELLED: We evaluated the noradrenergic modulation of the antinociceptive activity of diclofenac in mice using the acetic acid writhing test. Dose-response curves were obtained for the antinociceptive effect of diclofenac, phenylephrine, clonidine, desipramine, prazosin, and yohimbine administered both systemically and intrathecally, and ED(50)s were calculated. Noradrenergic modulation was evaluated by performing an isobolographic analysis of the systemic or intrathecal coadministration of fixed-ratio combinations of diclofenac with each adrenergic drug. The systemic, but not the intrathecal, combinations of diclofenac with phenylephrine or clonidine showed supraadditivity, suggesting that the activation of alpha(1) and alpha(2) adrenoceptors interfered with the nociceptive transmission at spinal and supraspinal levels. Supraadditive effects were not demonstrated for the intrathecal injection of diclofenac combined with phenylephrine, clonidine and a selective norepinephrine uptake inhibitor (desipramine) or adrenergic antagonists. We conclude that interaction between adrenoceptors and diclofenac can modulate antinociception by activating common or different mechanisms. Diclofenac has an antinociceptive activity that, in addition to cyclooxygenase inhibition, can be modulated by additive and supraadditive interactions with adrenergic drugs. IMPLICATIONS: Diclofenac analgesia in mice can be modulated by interaction with adrenergic drugs. The systemic but not the intrathecal administration of phenylephrine and clonidine produced supraadditive interactions. For desipramine, prazosin, and yohimbine, supraadditive interactions were not statistically demonstrated. The coadministration of drugs inducing supraadditive effects could be clinically relevant for the treatment of chronic pain because of reduction of doses and side effects.


Subject(s)
Adrenergic Agents/pharmacology , Analgesics/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Animals , Clonidine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Mice , Phenylephrine/pharmacology , Prazosin/pharmacology , Yohimbine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...