Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antioxidants (Basel) ; 12(1)2022 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-36670953

RESUMO

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic inflammatory disease without consistently effective treatment. Among the many mediators implicated in cystitis, the overproduction of reactive oxygen species (ROS) seems to play a key role, although the main source of ROS remains unclear. This study aimed to investigate the contribution of NADPH oxidase (NOX) isoforms in ROS generation and the voiding dysfunction of cyclophosphamide (CYP, 300 mg/Kg, ip, 24 h)-induced cystitis in adult female mice, a well-recognized animal model to study IC/BPS, by using GKT137831 (5 mg/Kg, ip, three times in a 24 h period) or GSK2795039 (5 mg/Kg, ip, three times in a 24 h period) to inhibit NOX1/4 or NOX2, respectively. Our results showed that treatment with GSK2795039 improved the dysfunctional voiding behavior induced by CYP, reduced bladder edema and inflammation, and preserved the urothelial barrier integrity and tight junction occludin expression, besides inhibiting the characteristic vesical pain and bladder superoxide anion generation. In contrast, the NOX1/4 inhibitor GKT137831 had no significant protective effects. Taken together, our in vivo and ex vivo data demonstrate that NOX2 is possibly the main source of ROS observed in cystitis-induced CYP in mice. Therefore, selective inhibition of NOX2 by GSK2795039 may be a promising target for future therapies for IC/BPS.

2.
Eur J Neurosci ; 53(2): 376-389, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32979866

RESUMO

The mechanism underlying the role of tumor necrosis factor alpha (TNF-α) in the development of inflammatory hyperalgesia has been extensively studied, mainly the role of TNF-α in the release of pro-inflammatory cytokines. The current concept relies in the fact that TNF-α stimulates the cascade release of other pro-inflammatory cytokines, such as IL-1ß, IL-6, and IL-8 (CINC-1 in rats), triggering the release of the final inflammatory mediator prostaglandin E2 (PGE2 ) and sympathetic amines that directly sensitize the nociceptors. However, this may not be the sole mechanism involved as the blockade of TNF-α synthesis by thalidomide prevents hyperalgesia without interrupting the synthesis of IL-1ß, IL-6, and CINC-1. Therefore, we hypothesized that activation of TNF-α receptor type 1 (TNFR1) by TNF-α increases nociceptors' susceptibility to the action of PGE2 and dopamine. We have found out that intrathecal administration of oligodeoxynucleotide-antisense (ODN-AS) against TNFR1 or thalidomide prevented carrageenan-induced hyperalgesia. The co-administration of TNF-α with a subthreshold dose of PGE2 or dopamine that does not induce hyperalgesia by itself in the hind paw of Wistar rats pretreated with dexamethasone (to prevent the endogenous release of cytokines) induced a robust hyperalgesia that was prevented by intrathecal treatment with ODN-AS against TNFR1. We consider that the activation of neuronal TNFR1 by TNF-α decisively increases the susceptibility of the peripheral afferent neuron to the action of final inflammatory mediators - PGE2 and dopamine - that ultimately induce hyperalgesia. This mechanism may also underlie the analgesic action of thalidomide.


Assuntos
Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa , Animais , Citocinas , Hiperalgesia/induzido quimicamente , Neurônios Aferentes , Dor , Ratos , Ratos Wistar
3.
Brain Behav Immun ; 51: 204-211, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26291402

RESUMO

This study aims to explore the contribution of endocannabinoids on the cascade of mediators involved in LPS-induced fever and to verify the participation of prostaglandins and endogenous opioids in fever induced by anandamide (AEA). Body temperature (Tc) of male Wistar rats was recorded over 6h, using a thermistor probe. Cerebrospinal fluid concentration of PGE2 and ß-endorphin were measured by ELISA after the administration of AEA. Intracerebroventricular administration of the CB1 receptor antagonist AM251 (5µg, i.c.v.), reduced the fever induced by IL-1ß (3ng, i.c.v.), TNF-α (250ng, i.c.v.), IL-6 (300ng, i.c.v.), corticotrophin release factor (CRH; 2.5µg, i.c.v.) and endothelin (ET)-1 (1pmol, i.c.v.), but not the fever induced by PGE2 (250ng, i.c.v.) or PGF2α (250ng, i.c.v.). Systemic administration of indomethacin (2mgkg(-1), i.p.) or celecoxib (5mgkg(-1), p.o.) reduced the fever induced by AEA (1µg, i.c.v.), while naloxone (1mgkg(-1), s.c.) abolished it. The increases of PGE2 and ß-endorphin concentration in the CSF induced by AEA were abolished by the pretreatment of rats with AM251. These results suggest that endocannabinoids are intrinsically involved in the pyretic activity of cytokines (IL-1ß, TNF-α, IL-6), CRH and ET-1 but not the PGE2 or PGF2α induced fevers. However, anandamide via CB1 receptor activation induces fever that is dependent on the synthesis of prostaglandin and opioids.


Assuntos
Ácidos Araquidônicos/fisiologia , Citocinas/fisiologia , Endocanabinoides/fisiologia , Febre/fisiopatologia , Prostaglandinas/fisiologia , Receptor CB1 de Canabinoide/fisiologia , beta-Endorfina/líquido cefalorraquidiano , Animais , Ácidos Araquidônicos/administração & dosagem , Temperatura Corporal/efeitos dos fármacos , Hormônio Liberador da Corticotropina/administração & dosagem , Citocinas/administração & dosagem , Endocanabinoides/administração & dosagem , Endotelina-1/administração & dosagem , Febre/induzido quimicamente , Interleucina-1beta/administração & dosagem , Interleucina-1beta/fisiologia , Interleucina-6/administração & dosagem , Interleucina-6/fisiologia , Masculino , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Piperidinas/administração & dosagem , Alcamidas Poli-Insaturadas/administração & dosagem , Prostaglandinas/administração & dosagem , Pirazóis/administração & dosagem , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/antagonistas & inibidores , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/fisiologia
4.
Pharmacol Biochem Behav ; 134: 49-56, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25902407

RESUMO

The pathophysiology of chronic inflammatory pain remains poorly understood. In this context, we developed an experimental model in which successive daily injection of prostaglandin E2 (PGE2) for 14days into rat hind paws produces a persistent state of hypernociception (i.e. decrease in mechanical nociceptive threshold). This state persists for more than 30days after discontinuing PGE2 injection. In the present study, we investigated the participation of nuclear factor kappa B (NF-κB), in the maintenance of this process. Mechanical hypernociception was evaluated using the electronic von Frey test. Activation of NF-κB signaling was measured through the determination of NF-κB p65 subunit translocation to the nucleus of dorsal root ganglion neurons (DRG) by immunofluorescence and western blotting. Herein, we detected an increase in NF-κB p65 subunit translocation to the nucleus of DRG neurons along with persistent inflammatory hypernociception compared with controls. Intrathecal treatment with either dexamethasone or PDTC (NF-κB activation inhibitor) after ending of the induction phase of the persistent inflammatory hypernociception, curtailed the hypernociception period as well as reducing NF-κB p65 subunit translocation. Treatment with antisense oligonucleotides against the NF-κB p65 subunit for 5 consecutive days also reduced persistent inflammatory hypernociception. Inhibition of PKA and PKCε reduced persistent inflammatory hypernociception, which was associated with inhibition of NF-κB p65 subunit translocation. Together these results suggest that peripheral activation of NF-κB by PKA and PKC in primary sensory neurons plays an important role in maintaining persistent inflammatory pain.


Assuntos
Inflamação/metabolismo , NF-kappa B/metabolismo , Dor Nociceptiva/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inflamação/complicações , Inflamação/enzimologia , Masculino , Dor Nociceptiva/enzimologia , Dor Nociceptiva/etiologia , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Wistar
5.
Life Sci ; 105(1-2): 7-13, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24607781

RESUMO

AIMS: Although evidence suggest that TRPA1 mediates some effects of prostaglandins, it is not known whether TRPA1 contributes to the in vivo nociceptive effects of prostaglandin E2 (PGE2), a key mediator of inflammatory pain. MAIN METHODS: To address this issue, the effect of the pharmacological blockade of TRPA1 or of its gene silencing on the hyperalgesia induced in the rat paw by PGE2 or its downstream signaling molecules, protein kinase A (PKA) or protein kinase C-epsilon (PKCε), was evaluated. TRPA1 expression on dorsal root ganglia cells was assessed by western blot. KEY FINDINGS: The pharmacological blockade of local TRPA1 by its selective antagonist, HC 030031 decreased and reversed PGE2-induced hyperalgesia. The TRPA1 gene silencing induced by intrathecal pre-treatment with antisense oligodeoxynucleotide blocked PGE2-induced hyperalgesia and strongly reduced TRPA1 expression in dorsal root ganglia cells (L5 and L6). PGE2 injection into the hind paw did not significantly increase TRPA1 expression in dorsal root ganglia cells. Treatment with either HC 030031 or antisense oligodeoxynucleotide significantly decreased the hyperalgesia induced by PKA or PKCε. Since both kinases are the major components of PGE2-induced intracellular signal transduction, the modulation of TRPA1 function by PGE2 may be downstream PKA and PKC-epsilon. SIGNIFICANCE: These findings show that TRPA1 is essential to the in vivo nociceptive effects induced by one of the most important mediators of inflammatory pain, PGE2. This is one of the crucial findings necessary to support TRPA1 as a promising target for the development of future drugs to pain treatment and control.


Assuntos
Dinoprostona/metabolismo , Gânglios Espinais/metabolismo , Nociceptividade/fisiologia , Canais de Cátion TRPC/metabolismo , Acetanilidas/farmacologia , Análise de Variância , Animais , Western Blotting , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fatores de Crescimento de Fibroblastos , Inativação Gênica , Proteína Quinase C-épsilon/metabolismo , Purinas/farmacologia , Ratos , Canal de Cátion TRPA1 , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética
6.
Anal Chem ; 84(15): 6341-5, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22741519

RESUMO

The ability of nanoassisted laser desorption-ionization mass spectrometry (NALDI-MS) imaging to provide selective chemical monitoring with proper spatial distribution of lipid profiles from tumor tissues after plate imprinting has been tested. NALDI-MS imaging identified and mapped several potential lipid biomarkers in a murine model of melanoma tumor (inoculation of B16/F10 cells). It also confirmed that the in vivo treatment of tumor bearing mice with synthetic supplement containing phosphoethanolamine (PHO-S) promoted an accentuated decrease in relative abundance of the tumor biomarkers. NALDI-MS imaging is a matrix-free LDI protocol based on the selective imprinting of lipids in the NALDI plate followed by the removal of the tissue. It therefore provides good quality and selective chemical images with preservation of spatial distribution and less interference from tissue material. The test case described herein illustrates the potential of chemically selective NALDI-MS imaging for biomarker discovery.


Assuntos
Lasers , Melanoma Experimental/patologia , Nanotecnologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Etanolaminas/uso terapêutico , Humanos , Processamento de Imagem Assistida por Computador , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Software , Transplante Homólogo
7.
Pain ; 151(2): 506-515, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20813459

RESUMO

Through activation of the A1 adenosine receptors (A1Rs) at both the central and peripheral level, adenosine produces antinociception in a wide range of tests. However, the mechanisms involved in the peripheral effect are still not fully understood. Therefore, the mechanisms by which peripheral activation of A1Rs reduces inflammatory hypernociception (a decrease in the nociceptive threshold) were addressed in the present study. Immunofluorescence of rat dorsal root ganglion revealed significant expression of A1Rs in primary sensory neurons associated with nociceptive pathways. Functionally, peripheral activation of A1Rs reduced inflammatory hypernociception because intraplantar (i.pl.) administration of an A1R antagonist (DPCPX) enhanced carrageenan-induced hypernociception. On the other hand, local (paw) administration of CPA (a selective A1R agonist) reversed mechanical hypernociception induced by carrageenan or by the directly acting hypernociceptive mediator prostaglandin E(2) (PGE(2)). Down-regulation of A1Rs expression in primary nociceptive neurons by intrathecal treatment with antisense oligodeoxinucleotides significantly reduced peripheral antinociceptive action of CPA. Direct blockade of PGE(2) inflammatory hypernociception by the activation of A1Rs depends on the nitric oxide/cGMP/Protein Kinase G/KATP signaling pathway because the peripheral antinociceptive effect of CPA was prevented by pretreatment with inhibitors of neuronal nitric oxide synthase (N-propyl-l-arginine), guanylyl cyclase (ODQ), and Protein Kinase G (KT5823) as well as with a KATP blocker (glibenclamide). However, this effect of CPA was not reduced by naloxone, excluding the participation of endogenous opioids. These results suggest that the peripheral activation of A1R plays a role in the regulation of inflammatory hypernociception by a mechanism that involves the NO/cGMP/PKG/KATP intracellular signaling pathway.


Assuntos
Inflamação/tratamento farmacológico , Óxido Nítrico/metabolismo , Nociceptores/fisiologia , Limiar da Dor/fisiologia , Receptor A1 de Adenosina/metabolismo , Transdução de Sinais/fisiologia , Antagonistas do Receptor A1 de Adenosina/uso terapêutico , Análise de Variância , Animais , Carragenina/efeitos adversos , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Vias de Administração de Medicamentos , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Inflamação/induzido quimicamente , Inflamação/patologia , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Nociceptores/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/patologia , Canais de Cátion TRPV/metabolismo , Xantinas/uso terapêutico
8.
Eur J Pharmacol ; 617(1-3): 41-7, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19567249

RESUMO

We have previously demonstrated that activation of kappa-opioid receptor located in the temporomandibular joint (TMJ) of rats induces a significantly greater TMJ antinociception in diestrus females than in proestrus females (higher estradiol serum levels than diestrus) and males. These findings indicate that gonadal hormones decrease TMJ kappa-mediated antinociception. The aim of this study was to investigate some of the mechanisms by which gonadal hormones decrease TMJ kappa-mediated antinociception. Western blot analysis demonstrated a significantly lower kappa-opioid receptor expression in the trigeminal ganglia of intact males than in intact and ovariechtomized (OVX) females and orchidectomized (ORX) males. In females, kappa-opioid receptor expression in the trigeminal ganglia was significantly lower in proestrus than in diestrus and OVX females. Taken together these findings suggest that gonadal hormones, especially male gonadal hormones, down-regulate kappa-opioid receptor expression. Co-application of the NOS inhibitor L-NMMA or the NO-sensitive guanylyl cyclase inhibitor ODQ with the kappa-opioid receptor agonist U50,488 blocked TMJ kappa-mediated antinociception in males and females. These findings suggest that antinociception induced by activation of kappa opioid receptors in the TMJ region is mediated by the L-arginine/NO/cGMP pathway in both sexes. Despite the involvement of the L-arginine/NO/cGMP pathway in TMJ kappa-mediated antinociception in both sexes, gonadal hormones do not diminish the activity of this pathway to decrease TMJ kappa-mediated antinociception. Alternatively, they significantly reduce kappa-opioid receptor expression in the trigeminal ganglia.


Assuntos
Analgésicos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Hormônios Gonadais/metabolismo , Dor/metabolismo , Receptores Opioides kappa/metabolismo , Articulação Temporomandibular/metabolismo , Gânglio Trigeminal/metabolismo , Analgésicos/uso terapêutico , Animais , Arginina/metabolismo , GMP Cíclico/metabolismo , Feminino , Humanos , Masculino , Óxido Nítrico/metabolismo , Oxidiazóis/farmacologia , Dor/tratamento farmacológico , Quinoxalinas/farmacologia , Ratos , Ratos Wistar , Receptores Opioides kappa/agonistas , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Gânglio Trigeminal/efeitos dos fármacos , ômega-N-Metilarginina/farmacologia
9.
J Leukoc Biol ; 83(4): 824-32, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18203872

RESUMO

Neutrophil migration is responsible for tissue damage observed in inflammatory diseases. Neutrophils are also implicated in inflammatory nociception, but mechanisms of their participation have not been elucidated. In the present study, we addressed these mechanisms in the carrageenan-induced mechanical hypernociception, which was determined using a modification of the Randall-Sellito test in rats. Neutrophil accumulation into the plantar tissue was determined by the contents of myeloperoxidase activity, whereas cytokines and PGE(2) levels were measured by ELISA and radioimmunoassay, respectively. The pretreatment of rats with fucoidin (a leukocyte adhesion inhibitor) inhibited carrageenan-induced hypernociception in a dose- and time-dependent manner. Inhibition of hypernociception by fucoidin was associated with prevention of neutrophil recruitment, as it did not inhibit the hypernociception induced by the direct-acting hypernociceptive mediators, PGE(2) and dopamine, which cause hypernociception, independent of neutrophils. Fucoidin had no effect on carrageenan-induced TNF-alpha, IL-1beta, and cytokine-induced neutrophil chemoattractant 1 (CINC-1)/CXCL1 production, suggesting that neutrophils were not the source of hypernociceptive cytokines. Conversely, hypernociception and neutrophil migration induced by TNF-alpha, IL-1beta, and CINC-1/CXCL1 was inhibited by fucoidin, suggesting that neutrophils are involved in the production of direct-acting hypernociceptive mediators. Indeed, neutrophils stimulated in vitro with IL-1beta produced PGE(2), and IL-1beta-induced PGE(2) production in the rat paw was inhibited by the pretreatment with fucoidin. In conclusion, during the inflammatory process, the migrating neutrophils participate in the cascade of events leading to mechanical hypernociception, at least by mediating the release of direct-acting hypernociceptive mediators, such as PGE(2). Therefore, the blockade of neutrophil migration could be a target to development of new analgesic drugs.


Assuntos
Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Neutrófilos/fisiologia , Dor/fisiopatologia , Animais , Carragenina/farmacologia , Técnicas de Cultura de Células , Quimiocina CXCL1/sangue , Citocinas/sangue , Dinoprostona/metabolismo , Membro Posterior/irrigação sanguínea , Hiperalgesia/sangue , Inflamação/sangue , Interleucina-1/sangue , Masculino , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Dor/sangue , Limiar da Dor , Polissacarídeos/farmacologia , Ratos , Ratos Wistar , Tempo de Reação , Fenômenos Fisiológicos da Pele , Fator de Necrose Tumoral alfa/sangue
10.
J Pharmacol Exp Ther ; 324(1): 313-21, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17928570

RESUMO

The 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) is an endogenous ligand of peroxisome proliferator-activated receptors gamma (PPAR-gamma) and is now recognized as a potent anti-inflammatory mediator. However, information regarding the influence of 15d-PGJ(2) on inflammatory pain is still unknown. In this study, we evaluated the effect of 15d-PGJ(2) upon inflammatory hypernociception and the mechanisms involved in this effect. We observed that intraplantar administration of 15d-PGJ(2) (30-300 ng/paw) inhibits the mechanical hypernociception induced by both carrageenan (100 mug/paw) and the directly acting hypernociceptive mediator, prostaglandin E(2) (PGE(2)). Moreover, 15d-PGJ(2) [100 ng/temporomandibular joint (TMJ)] inhibits formalin-induced TMJ hypernociception. On the other hand, the direct administration of 15d-PGJ(2) into the dorsal root ganglion was ineffective in blocking PGE(2)-induced hypernociception. In addition, the 15d-PGJ(2) antinociceptive effect was enhanced by the increase of macrophage population in paw tissue due to local injection of thioglycollate, suggesting the involvement of these cells on the 15d-PGJ(2)-antinociceptive effect. Moreover, the antinociceptive effect of 15d-PGJ(2) was also blocked by naloxone and by the PPAR-gamma antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662), suggesting the involvement of peripheral opioids and PPAR-gamma receptor in the process. Similar to opioids, the 15d-PGJ(2) antinociceptive action depends on the nitric oxide/cGMP/protein kinase G (PKG)/K(ATP)(+) channel pathway because it was prevented by the pretreatment with the inhibitors of nitric-oxide synthase (N(G)-monomethyl-l-arginine acetate), guanylate cyclase]1H-(1,2,4)-oxadiazolo(4,2-alpha)quinoxalin-1-one[, PKG [indolo[2,3-a]pyrrolo[3,4-c]carbazole aglycone (KT5823)], or with the ATP-sensitive potassium channel blocker glibenclamide. Taken together, these results demonstrate for the first time that 15d-PGJ(2) inhibits inflammatory hypernociception via PPAR-gamma activation. This effect seems to be dependent on endogenous opioids and local macrophages.


Assuntos
Analgésicos/farmacologia , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Prostaglandina D2/análogos & derivados , Receptores Opioides/imunologia , Animais , Carragenina , Citocinas/imunologia , Formaldeído , Hiperalgesia/induzido quimicamente , Hiperalgesia/imunologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/imunologia , Macrófagos/imunologia , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes , PPAR gama/imunologia , Dor/induzido quimicamente , Dor/imunologia , Prostaglandina D2/farmacologia , Ratos , Ratos Wistar , Pele/imunologia , Fator de Necrose Tumoral alfa
11.
Eur J Pharmacol ; 573(1-3): 221-9, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-17669394

RESUMO

Kinin receptors are involved in the genesis of inflammatory pain. However, there is controversy concerning the mechanism by which B(1) and B(2) kinin receptors mediate inflammatory hypernociception. In the present study, the role of these receptors on inflammatory hypernociception in mice was addressed. Mechanical hypernociception was detected with an electronic pressure meter paw test in mice and cytokines were measured by ELISA. It was observed that in naïve mice a B(2) (d-Arg-Hyp(3), d-Phe(7)-bradykinin) but not a B(1) kinin receptor antagonist (des-Arg(9)-[Leu(8)]-bradykinin, DALBK) inhibited bradykinin- and carrageenin-induced hypernociception. Bradykinin-induced hypernociception was inhibited by indomethacin (5 mg/kg) and guanethidine (30 mg/kg), while not affected by IL-1ra (10 mg/kg) or antibody against keratinocyte-derived chemokine (KC/CXCL-1, 500 ng/paw) or in TNFR1 knockout mice. By contrast, in previously lipopolysaccharide (LPS)-primed mouse paw, B(1) but not B(2) kinin receptor antagonist inhibited bradykinin hypernociception. Furthermore, B(1) kinin receptor agonist induced mechanical hypernociception in LPS-primed mice, which was inhibited by indomethacin, guanethidine, antiserum against TNF-alpha or IL-1ra. This was corroborated by the induction of TNF-alpha and IL-1beta release by B(1) kinin receptor agonist in LPS-primed mouse paws. Moreover, B(1) but not B(2) kinin receptor antagonist inhibited carrageenin-induced hypernociception, and TNF-alpha and IL-1beta release as well, in LPS-primed mice. These results suggest that in naïve mice the B(2) kinin receptor mediates inflammatory hypernociception dependent on prostanoids and sympathetic amines, through a cytokine-independent mechanism. On the other hand, in LPS-primed mice, the B(1) kinin receptor mediates hypernociception by a mechanism dependent on TNF-alpha and IL-1beta, which could stimulate prostanoid and sympathetic amine production.


Assuntos
Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Interleucina-1beta/fisiologia , Receptores da Bradicinina/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos/farmacologia , Bradicinina/administração & dosagem , Bradicinina/análogos & derivados , Bradicinina/farmacologia , Antagonistas de Receptor B1 da Bradicinina , Antagonistas de Receptor B2 da Bradicinina , Antagonistas dos Receptores da Bradicinina , Carragenina/administração & dosagem , Carragenina/toxicidade , Relação Dose-Resposta a Droga , Guanetidina/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/prevenção & controle , Indometacina/farmacologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Interleucina-1beta/imunologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandinas/fisiologia , Receptor B1 da Bradicinina/agonistas , Receptor B1 da Bradicinina/fisiologia , Receptor B2 da Bradicinina/fisiologia , Receptores da Bradicinina/agonistas , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/imunologia
12.
Eur J Pharmacol ; 562(1-2): 155-63, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17320857

RESUMO

The anti-inflammatory activities of some medicinal plants are attributed to their contents of sesquiterpene lactones. In the present study, the anti-inflammatory and anti-nociceptive activity of a sesquiterpene lactone isolated from Viguiera robusta, budlein A in mice was investigated. The treatment with budlein A dose--(1.0-10.0 mg/kg, p.o., respectively) dependently inhibited the carrageenan-induced: i. neutrophil migration to the peritoneal cavity (2-52%), ii. neutrophil migration to the paw skin tissue (32-74%), iii. paw oedema (13-74%) and iv. mechanical hypernociception (2-58%) as well as the acetic acid-induced writhings (0-66%). Additionally, budlein A (10.0 mg/kg) treatment inhibited the mechanical hypernociception-induced by tumour necrosis factor (TNF-alpha, 36%), Keratinocyte-derived chemokine (KC, 37%) and Interleukin-1beta (IL-1beta, 28%), but not of prostaglandin E(2) or dopamine. Budlein A also inhibited the carrageenan-induced release of TNF-alpha (52%), KC (70%) and IL-1beta (59%). Furthermore, an 8 days treatment with budlein A inhibited Complete Freund's adjuvant (10 microl/paw)-induced hypernociception, paw oedema and paw skin myeloperoxidase activity increase while not affecting the motor performance or myeloperoxidase activity in the stomach. Concluding, the present data suggest that budlein A presents anti-inflammatory and antinociceptive property in mice by a mechanism dependent on inhibition of cytokines production. It supports the potential beneficial effect of orally administered budlein A in inflammatory diseases involving cytokine-mediated nociception, oedema and neutrophil migration.


Assuntos
Analgésicos/farmacologia , Citocinas/metabolismo , Lactonas/farmacologia , Sesquiterpenos/farmacologia , Analgésicos/química , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Asteraceae/química , Comportamento Animal/efeitos dos fármacos , Carragenina/toxicidade , Movimento Celular/efeitos dos fármacos , Dinoprostona/toxicidade , Dopamina/toxicidade , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Edema/prevenção & controle , Adjuvante de Freund/toxicidade , Membro Posterior/efeitos dos fármacos , Membro Posterior/metabolismo , Membro Posterior/patologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Lactonas/química , Masculino , Camundongos , Estrutura Molecular , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Dor/induzido quimicamente , Dor/fisiopatologia , Dor/prevenção & controle , Cavidade Peritoneal/patologia , Sesquiterpenos/química , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia
13.
Brain Behav Immun ; 21(5): 535-43, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17194566

RESUMO

There is pre-clinical evidence that therapies targeting IL-18 might be beneficial in controlling arthropathies, which are accompanied by hypernociception (nociceptor sensitization). In the present study, we addressed the hypernociceptive role of IL-18 in a model of antigen-induced inflammation in mice and its mechanisms. In naïve mice, the intraplantar injection of IL-18 induced dose- and time-dependent mechanical hypernociception, which was inhibited in IFN-gamma deficient (-/-) mice, and by the pre-treatment with bosentan (dual endothelin [ET] receptor antagonist), BQ123 (ET(A) receptor antagonist) or indomethacin (cyclooxygenase inhibitor). IL-18 hypernociception was unaffected in TNFR1(-/-) mice or by the pre-treatment with sIL-15Ralpha (soluble form of IL-15 receptor), BQ788 (ET(B) receptor antagonist) or guanethidine (sympathetic blocker). The ovalbumin (OVA) challenge-induced mechanical hypernociception in immunized mice was inhibited by the pre-treatment with anti-IL-18 antibody or in IL-18(-/-) mice. Furthermore, IL-18 induced significant IFN-gamma production in the paw skin of naïve mice. The OVA challenge-induced IFN-gamma and ET-1 productions were inhibited in IL-18(-/-) immunized mice, as well as ET-1 production in IFN-gamma(-/-) immunized mice. In addition, significant PGE2 production was detected after IL-18 or ET-1 (via ET(A) receptors) injection in naïve mice. Taken together with previous data, these results suggest that IL-18 plays a significant role in antigen-induced inflammatory hypernociception via the production of IFN-gamma, ET-1 and PGE2. Thus, IL-18 and IL-18-downstream mediators demonstrated herein might constitute targets to inhibit antigen-induced inflammatory pain.


Assuntos
Interferon gama/imunologia , Interleucina-18/imunologia , Limiar da Dor/fisiologia , Dor/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos/imunologia , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Interferon gama/genética , Interleucina-18/administração & dosagem , Interleucina-18/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/imunologia , Prostaglandina-Endoperóxido Sintases/imunologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptor de Endotelina A/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Estresse Mecânico , Fatores de Tempo
14.
Eur J Oral Sci ; 114(5): 403-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17026506

RESUMO

Although interferon-gamma (IFN-gamma) plays a critical role in periodontitis, no information is available regarding the effect of smoking on this cytokine in the periodontium. Therefore, this study aimed to evaluate the effect of smoking on the IFN-gamma levels in gingival tissue from patients with chronic periodontitis. Sixty-two patients were assigned to three groups: healthy [non-smoking and periodontally healthy individuals (probing depth or= 5 mm and bleeding on probing; n = 25)]; and smoking [smokers (>or= 1 pack/day for at least 10 yr) diagnosed with chronic periodontitis (n = 25)]. Gingival biopsies were analyzed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Regardless of smoking status, diseased patients presented higher levels of IFN-gamma than peridontally healthy patients. In sites with comparable types of periodontitis, smoking increased both protein and mRNA levels of IFN-gamma in gingival tissue. Within the limits of this study, it can be concluded that modulation of periodontal tissue destruction by smoking may involve its effect on IFN-gamma production.


Assuntos
Gengiva/metabolismo , Interferon gama/metabolismo , Periodontite/metabolismo , Fumar/metabolismo , Adulto , Antivirais/imunologia , Doença Crônica , Métodos Epidemiológicos , Feminino , Humanos , Interferon gama/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Fumar/efeitos adversos
15.
Pharmacol Biochem Behav ; 84(2): 244-51, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16797062

RESUMO

Mice have been used as animal model to study mechanisms underlying inflammatory and immune diseases. The present study describes a model of joint inflammation-induced hypernociception to discriminate pharmacological tests in mice. A polypropylene tip probe with a large area (4.15 mm2) applied on the plantar surface of the hind paw was used to produce a dorsal flexion of tibio-tarsal joint. Experiments were performed to demonstrate that the probe application did not provoke cutaneous nociception. The decrease in the withdrawal threshold of inflamed joint was used as nociceptive parameter. Administration of zymosan in the tibio-tarsal joint induced a dose and time-dependent hypernociception elicited by articular dorsal flexion movement. Maximal joint hypernociception was detected between 7 and 24 h after zymosan injection, and matched maximal inflammation score as determined by histopathology and neutrophil migration assay. In agreement with the inflammatory hypernociceptive paradigm, flexion-elicited hypernociception induced by zymosan was dose-dependently inhibited by morphine (2-8 mg/kg) and by an effective dose of indomethacin (5 mg/kg). The present study demonstrated that the tibio-tarsal flexion reflex is a behavioral nociceptive model that allows a quantitative evaluation of inflammatory joint hypernociception in mice and its pharmacological modulation.


Assuntos
Medição da Dor/métodos , Animais , Artrite/induzido quimicamente , Comportamento Animal/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Membro Posterior , Indometacina/farmacologia , Lidocaína/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Medição da Dor/instrumentação , Pressão , Articulações Tarsianas/efeitos dos fármacos , Articulações Tarsianas/patologia , Zimosan
16.
J Neurosci ; 24(18): 4444-52, 2004 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15128858

RESUMO

The development of treatments for neuropathic pain has been hindered by our limited understanding of the basic mechanisms underlying abnormalities in nociceptor hyperexcitability. We recently showed that the polymodal receptor transient receptor potential vanilloid 4 (TRPV4), a member of the transient receptor potential (TRP) family of ion channels, may play a role in inflammatory pain (Alessandri-Haber et al., 2003). The present study tested whether TRVP4 also contributes to neuropathic pain, using a rat model of Taxol-induced painful peripheral neuropathy. Taxol is the most widely used drug for the treatment of a variety of tumor types, but the dose of Taxol that can be tolerated is limited by the development of a small-fiber painful peripheral neuropathy. We found that Taxol treatment enhanced the nociceptive behavioral responses to both mechanical and hypotonic stimulation of the hind paw. Spinal administration of antisense oligodeoxynucleotides to TRPV4, which reduced the expression of TRPV4 in sensory nerve, abolished Taxol-induced mechanical hyperalgesia and attenuated hypotonic hyperalgesia by 42%. The enhancement of osmotic nociception involves sensitization of osmotransduction in primary afferents because osmotransduction was enhanced in cultured sensory neurons isolated from Taxol-treated rats. Taxol-induced TRPV4-mediated hyperalgesia and the enhanced osmotransduction in cultured nociceptors were dependent on integrin/Src tyrosine kinase signaling. These results suggest that TRPV4 plays a crucial role in a painful peripheral neuropathy, making it a very promising target for the development of a novel class of analgesics.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Canais Iônicos/metabolismo , Neuralgia/fisiopatologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Animais , Antineoplásicos Fitogênicos , Comportamento Animal/efeitos dos fármacos , Cálcio/metabolismo , Proteínas de Transporte de Cátions/antagonistas & inibidores , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Soluções Hipotônicas , Integrinas/metabolismo , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Masculino , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Nociceptores/citologia , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Paclitaxel , Medição da Dor , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Canais de Cátion TRPV , Quinases da Família src/metabolismo
17.
Eur J Neurosci ; 19(3): 634-42, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14984413

RESUMO

Many painful conditions are associated with alterations in the extracellular matrix (ECM) of affected tissues. While several integrins, the receptors for ECM proteins, are present on sensory neurons that mediate pain, the possible role of these cell adhesion molecules in inflammatory or neuropathic pain has not been explored. We found that the intradermal injection of peptide fragments of domains of laminin and fibronectin important for adhesive signaling selectively inhibited the hyperalgesia caused by prostaglandin E2 (PGE2) and epinephrine (EPI), respectively. The block of EPI hyperalgesia was mimicked by other peptides containing the RGD integrin-binding sequence. Monoclonal antibodies (mAbs) against the alpha1 or alpha3 integrin subunits, which participate in laminin binding, selectively blocked PGE2 hyperalgesia, while a mAb against the alpha5 subunit, which participates in fibronectin binding, blocked only EPI-induced hyperalgesia. A mAb against the beta1 integrin subunit, common to receptors for both laminin and fibronectin, inhibited hyperalgesia caused by both agents, as did the knockdown of beta1 integrin expression by intrathecal injection of antisense oligodeoxynucleotides. The laminin peptide, but not the fibronectin peptides, also reversibly abolished the longer lasting inflammatory hyperalgesia induced by carrageenan. Finally, the neuropathic hyperalgesia caused by systemic administration of the cancer chemotherapy agent taxol was reversibly inhibited by antisense knockdown of beta1 integrin. These results strongly implicate specific integrins in the maintenance of inflammatory and neuropathic hyperalgesia.


Assuntos
Integrinas/metabolismo , Dor/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Geneticamente Modificados/metabolismo , Anticorpos Monoclonais/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Western Blotting/métodos , Carragenina , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Epinefrina , Proteínas da Matriz Extracelular/farmacologia , Fibronectinas/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Fibras Nervosas Amielínicas/efeitos dos fármacos , Fibras Nervosas Amielínicas/fisiologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Paclitaxel/farmacologia , Dor/induzido quimicamente , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Peptídeos , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , Subunidades Proteicas/imunologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
18.
Pain ; 105(1-2): 143-50, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14499430

RESUMO

In male rats, carrageenan (CAR)-induced inflammation or exposure to a selective protein kinase C epsilon (PKC epsilon ) agonist (psi epsilon RACK) produces prolongation of the hyperalgesia induced by a subsequent exposure to an inflammatory mediator, a phenomenon referred to as hyperalgesic priming. Since many chronic inflammatory conditions are sexually dimorphic, we tested the hypothesis that hyperalgesic priming is sexually dimorphic. Prior injection of CAR or psi epsilon RACK produced a prolongation of the hyperalgesia induced by a subsequent injection of prostaglandin E(2), from less than 3 h to greater than 24 h, but only in male rats. In ovariectomized female rats priming with CAR and psi epsilon RACK produced hyperalgesic priming effects similar to that observed in the male rat, and this effect was reversed by estrogen replacement. While gonadectomy in males had no effect on CAR and psi epsilon RACK induced hyperalgesic priming, female phenotype was observed following implantation of estrogen in males. Thus, mechanisms mediating the development of hyperalgesic priming produced by inflammation are suppressed by estrogen. This regulation of priming by estrogen appears to occur at or downstream of the activation of PKC epsilon.


Assuntos
Carragenina/farmacologia , Hiperalgesia/fisiopatologia , Oligopeptídeos/farmacologia , Caracteres Sexuais , Animais , Comportamento Animal , Carragenina/administração & dosagem , Dinoprostona/administração & dosagem , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Implantes de Medicamento , Estrogênios/administração & dosagem , Feminino , Hiperalgesia/induzido quimicamente , Hiperalgesia/psicologia , Injeções , Masculino , Oligopeptídeos/administração & dosagem , Orquiectomia , Ovariectomia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
19.
Eur J Neurosci ; 17(9): 1847-52, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12752784

RESUMO

Carrageenan-induced inflammatory pain lasting hours to days produces a protein kinase C epsilon (PKC epsilon )-dependent 'primed' state lasting several weeks, during which time injection of prostaglandin E2 induces hyperalgesia which is markedly enhanced and prolonged compared to PGE2-induced hyperalgesia in normal 'unprimed' rats. In the present study, we demonstrate that while inhibition of prostaglandin synthesis and antagonism of beta2-adrenergic receptors markedly attenuated the hyperalgesia induced by carrageenan, these interventions did not affect hyperalgesic priming. Tumor necrosis factor-alpha (rat recombinant; rrTNFalpha), another mediator of carrageenan-induced inflammation, alone produced hyperalgesia and priming, which were attenuated and prevented, respectively, by intrathecal administration of antisense to PKC epsilon. Inhibition of TNFalpha with thalidomide or a rat polyclonal anti-TNFalpha antibody attenuated carrageenan-induced hyperalgesia and prevented priming. Intrathecal administration of antisense to tumour necrosis factor receptor type-1 (TNFR1) reduced the level of TNFR1 transported toward the peripheral terminals of sensory neurons, and attenuated both carrageenan- and rrTNFalpha-induced priming. Acute hyperalgesia induced by carrageenan or rrTNFalpha remained intact in animals treated with TNFR1 antisense. Our results demonstrate that the generation of the primed state does not require production of hyperalgesia and that TNFalpha, which is generated during acute inflammation, can act on sensory neurons to induce hyperalgesic priming by activating neuronal PKC epsilon.


Assuntos
Antígenos CD/fisiologia , Hiperalgesia/metabolismo , Neurônios Aferentes/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Animais , Antígenos CD/biossíntese , Doença Crônica , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Neurônios Aferentes/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores Tipo I de Fatores de Necrose Tumoral
20.
Br J Pharmacol ; 138(7): 1351-7, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12711636

RESUMO

1. Nitric oxide has been described either as pronociceptive or antinociceptive. In this investigation, using an electronic pressure-metre, the intradermal and the subcutaneous effects of prostaglandin E(2) (PGE(2)) and agents that mimic or inhibit the arginine/NO/cGMP pathway were compared. 2. The hypernociceptive effect of the intradermal injection of PGE(2) (100 ng) was immediate, peaking within 15-30 min and returning to basal values in 45-60 min. The subcutaneous injection of PGE(2) induced a hypernociception with a delayed peak (3 h) plateauing for 4-6 h. 3. Intradermal administration of 3-morpholino-sydnonimine-hydrochloride (SIN-1) enhanced, while its subcutaneous administration inhibited, subcutaneous hypernociception induced by PGE(2). This inhibition was prevented by ODQ (8 micro g) but not by NG-monomethyl-L-arginine (L-NMMA) (50 micro g). 4. Intradermal but not subcutaneous administration of L-arginine (1-100 micro g), SIN-1 (1-100 micro g) and dibutyrylguanosine 3':5'-cyclic monophosphate (db cGMP) (0.1-100 micro g) induced an early (15-30 min) dose-dependent hypernociceptive effect. Intradermal pretreatment with NG-monomethyl-L-arginine (L-NMMA; 50 micro g) inhibited the hypernociception induced by L-Arg (10 micro g), but not that induced by SIN-1 (10 micro g) or db cGMP (10 micro g). 5. Intradermal injection of ODQ (8 micro g) antagonized the hypernociception induced by L-arginine and SIN-1, but not that induced by db cGMP. 6. Considering (a) the different time course of intradermal and subcutaneous PGE(2)-induced hypernociception, (b) the opposite nociceptive effect of intradermal and subcutaneous administration of SIN-1 (db cGMP) as well as the arginine/NO/cGMP pathway, the existence of different subsets of nociceptive primary sensory neurons in which the arginine/NO/cGMP pathway plays opposing roles is suggested. This hypothesis would explain the apparent contradictory observations described in the literature.


Assuntos
Arginina/antagonistas & inibidores , Arginina/farmacologia , GMP Cíclico/administração & dosagem , Derme/fisiopatologia , Dinoprostona/administração & dosagem , Hiperalgesia/induzido quimicamente , Molsidomina/análogos & derivados , Nociceptores/efeitos dos fármacos , Nociceptores/fisiopatologia , Medição da Dor/efeitos dos fármacos , Tela Subcutânea/fisiopatologia , Animais , Bucladesina/administração & dosagem , Bucladesina/antagonistas & inibidores , GMP Cíclico/fisiologia , Relação Dose-Resposta a Droga , Injeções Subcutâneas , Masculino , Molsidomina/administração & dosagem , Molsidomina/antagonistas & inibidores , Óxido Nítrico/fisiologia , Oxidiazóis/administração & dosagem , Quinoxalinas/administração & dosagem , Ratos , Ratos Wistar , Fatores de Tempo , ômega-N-Metilarginina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA