Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol

Detalhes bibliográficos
Autor(a) principal: Gonçalves, Gabriela Mastrangelo
Data de Publicação: 2016
Tipo de documento: Dissertação
Idioma: por
Título da fonte: Biblioteca Digital de Teses e Dissertações da UFRRJ
Texto Completo: https://rima.ufrrj.br/jspui/handle/20.500.14407/11405
Resumo: Diversos fármacos de uso corrente foram descobertos durante ensaios experimentais e mediante a observação em animais. Quando um novo composto parece promissor, geralmente este sofre alterações em sua estrutura química a fim de aperfeiçoar a sua seletividade, potência e eficácia terapêutica. O objetivo deste estudo foi avaliar as atividades antinociceptiva e anti-inflamatória de um novo composto sintético (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol (CTHP) preparado a partir de um protótipo anterior, ácido (±)-cis-(6-etil-tetrahidropirano-2-il) fórmico. O composto CTHP foi avaliado em ensaios de indução de dor aguda. A administração oral do composto foi capaz de induzir atividade antinociceptiva nos modelos de contorções abdominais induzidas por ácido acético, formalina (em ambas as fases) e retirada da cauda. Para elucidação do mecanismo de ação do composto, o modelo de retirada de cauda foi utilizado. Neste modelo foi realizada a administração prévia de naloxona (antagonista opioide não-seletivo), em que foi observada a inibição do efeito produzido pelo composto. Assim, foi então avaliada a participação seletiva de receptores opioides (μ, δ e κ), através de administração prévia de metilnaltrexona, naltrindol e nor-binaltorfimina, respectivamente, onde somente a nor-binaltorfimina foi capaz de reduzir o efeito antinociceptivo do composto. Para avaliar a possível participação da via NO/GMPc/KATP, os animais foram pré-tratados com N-nitro-arginina-L-metil éster (L-NAME), 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalina-1-ona (ODQ) (inibidor da guanilato ciclase sensível ao óxido nítrico) e glibenclamida (bloqueador de canais de potássio regulados por ATP), foi observado redução do efeito antinociceptivo com a administração prévia de todos estes. No teste de indução de tolerância, tanto a morfina quanto o composto desenvolveram tolerância, porém o composto de forma mais lenta e houve desenvolvimento de tolerância cruzada com a morfina. Para avaliar o envolvimento da via serotoninérgica na atividade do composto, foi realizada a administração diária por 3 dias de 4-cloro-DL-fenilalanina (inibidor da enzima triptofano hidroxilase). Nenhuma alteração no efeito antinociceptivo do composto foi observado, no que diz respeito ao envolvimento da via serotoninérgica. Já o modelo de campo aberto foi utilizado para avaliar a possibilidade de interferência da performance motora sobre o efeito antinociceptivo, foi demonstrada ausência desta interferência. Quanto à atividade anti-inflamatória, o resultado no teste de edema de pata indica efeito antiedematogênico do composto. Houve uma diminuição na quantidade de leucócitos totais, indicando que o composto foi capaz de reduzir a migração leucocitária na inflamação existente na bolsa de ar subcutâneo. O composto também demonstrou atividade inibitória sobre a produção de TNF-α e inibição seletiva da enzima COX-2. Esses resultados indicam atividade antinociceptiva significativa do composto, sem evidências de comprometimento motor. O composto CTHP demonstrou efeito antinociceptivo central, tendo este último contribuição dos sistemas opioide (seletivo para receptores do tipo κ) e nitrérgico em seu mecanismo de ação. E ainda, atividade anti-inflamatória, com inibição da migração leucocitária, de TNF-α e atividade inibitória seletiva sobre COX-2.
id UFRRJ-1_dd6f62c88d112b3674db9589e0ff1071
oai_identifier_str oai:rima.ufrrj.br:20.500.14407/11405
network_acronym_str UFRRJ-1
network_name_str Repositório Institucional da UFRRJ
repository_id_str
spelling Gonçalves, Gabriela MastrangeloMarinho, Bruno Guimarães077.077.277-38http://lattes.cnpq.br/2685794388394484Cortes, Wellington da SilvaMatheus, Maria Eline384.158.588-41http://lattes.cnpq.br/03000791483596742023-12-22T01:52:19Z2023-12-22T01:52:19Z2016-02-23GONÇALVES, Gabriela Mastrangelo. Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol. 2016. 63 f. Dissertação (Mestrado em Ciências Fisiológicas) - Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica - RJ, 2016.https://rima.ufrrj.br/jspui/handle/20.500.14407/11405Diversos fármacos de uso corrente foram descobertos durante ensaios experimentais e mediante a observação em animais. Quando um novo composto parece promissor, geralmente este sofre alterações em sua estrutura química a fim de aperfeiçoar a sua seletividade, potência e eficácia terapêutica. O objetivo deste estudo foi avaliar as atividades antinociceptiva e anti-inflamatória de um novo composto sintético (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol (CTHP) preparado a partir de um protótipo anterior, ácido (±)-cis-(6-etil-tetrahidropirano-2-il) fórmico. O composto CTHP foi avaliado em ensaios de indução de dor aguda. A administração oral do composto foi capaz de induzir atividade antinociceptiva nos modelos de contorções abdominais induzidas por ácido acético, formalina (em ambas as fases) e retirada da cauda. Para elucidação do mecanismo de ação do composto, o modelo de retirada de cauda foi utilizado. Neste modelo foi realizada a administração prévia de naloxona (antagonista opioide não-seletivo), em que foi observada a inibição do efeito produzido pelo composto. Assim, foi então avaliada a participação seletiva de receptores opioides (μ, δ e κ), através de administração prévia de metilnaltrexona, naltrindol e nor-binaltorfimina, respectivamente, onde somente a nor-binaltorfimina foi capaz de reduzir o efeito antinociceptivo do composto. Para avaliar a possível participação da via NO/GMPc/KATP, os animais foram pré-tratados com N-nitro-arginina-L-metil éster (L-NAME), 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalina-1-ona (ODQ) (inibidor da guanilato ciclase sensível ao óxido nítrico) e glibenclamida (bloqueador de canais de potássio regulados por ATP), foi observado redução do efeito antinociceptivo com a administração prévia de todos estes. No teste de indução de tolerância, tanto a morfina quanto o composto desenvolveram tolerância, porém o composto de forma mais lenta e houve desenvolvimento de tolerância cruzada com a morfina. Para avaliar o envolvimento da via serotoninérgica na atividade do composto, foi realizada a administração diária por 3 dias de 4-cloro-DL-fenilalanina (inibidor da enzima triptofano hidroxilase). Nenhuma alteração no efeito antinociceptivo do composto foi observado, no que diz respeito ao envolvimento da via serotoninérgica. Já o modelo de campo aberto foi utilizado para avaliar a possibilidade de interferência da performance motora sobre o efeito antinociceptivo, foi demonstrada ausência desta interferência. Quanto à atividade anti-inflamatória, o resultado no teste de edema de pata indica efeito antiedematogênico do composto. Houve uma diminuição na quantidade de leucócitos totais, indicando que o composto foi capaz de reduzir a migração leucocitária na inflamação existente na bolsa de ar subcutâneo. O composto também demonstrou atividade inibitória sobre a produção de TNF-α e inibição seletiva da enzima COX-2. Esses resultados indicam atividade antinociceptiva significativa do composto, sem evidências de comprometimento motor. O composto CTHP demonstrou efeito antinociceptivo central, tendo este último contribuição dos sistemas opioide (seletivo para receptores do tipo κ) e nitrérgico em seu mecanismo de ação. E ainda, atividade anti-inflamatória, com inibição da migração leucocitária, de TNF-α e atividade inibitória seletiva sobre COX-2.FAPERJ - Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de JaneiroSeveral drugs in current use were discovered during experimental tests and by observing animals. When a new compound looks promising, it usually undergoes changes in its chemical structure in order to perfect its selectivity, potency and therapeutic efficacy. The aim of this study was to evaluate the antinociceptive and anti-inflammatory activities of a new synthetic hybrid compound (±)-4-chloro-6-(naphthalen-1-yl)-tetrahydro-2H-pyran-2-yl-methanol (CTHP) prepared from a previous prototype acid, (±) - cis- (6-ethyl-tetrahydropyran-2-yl) Formic. The compound CTHP was evaluated in acute pain induction assays. Oral administration of the compound was able to induce antinociceptive activity in models of writhing induced by acetic acid, formalin (both stages) and tail flick. To elucidate the mechanism of action of the compound, the tail flick model was used. This model was perform by prior administration of naloxone (opioid antagonist non-selective), where we observed the inhibition of the effect produced by the compound. The selective involvement of opioid receptors (μ, δ and κ) was then evaluated by prior administration of methylnaltrexone, naltrindol, and nor-binaltorphimine, respectively, where only nor-binaltorphimine was able to reduce the analgesic effect of the compound. To evaluate the possible role of the NO/cGMP/KATP, animals were pretreated with N-nitro-L-arginine methyl ester (L-NAME), 1H- [1,2,4 ] oxadiazolo [4,3-a] quinoxalin-1-one (ODQ) (inhibitor of guanylate cyclase sensitive to nitric oxide), and glibenclamide (blocker of the ATP-regulated potassium channels), where reduction was observed with the administration of analgesic effect prior to all of these. In the tolerance induction test, both morphine and compound developed tolerance, however the compound perform at a slower rate and developed cross-tolerance with morphine. To assess the involvement of serotonin pathway in the activity of the compound, daily administration for 3 days of 4-chloro-DL-phenylalanine (inhibitor of the enzyme tryptophan hydroxylase) was performed. No changes in the analgesic effect of the compound was noted, with regard to the involvement of serotonin pathway. The open field model was used to assess the possibility of interference from motor performance on the analgesic effect, which demonstrated absence of this interference. As for anti-inflammatory activity results in paw edema test indicate anti-oedematogenic effect of compound. There was a decrease in the number of total leukocytes, indicating that the compound was able to reduce existing inflammation in leukocyte migration in the air pouch model. The compound also demonstrated an inhibitory activity on TNF-α production and selective inhibition of COX-2 enzyme. These results indicate significant antinociceptive activity of the compound without evidence of motor impairment. The compound CTHP showed central analgesic effect, which has contribution of opioid systems (selective for the κ-like receptors) and nitrergic in its mechanism of action. It has also showed an anti-inflammatory activity, with inhibition of leukocyte migration, TNF-α production and selective inhibitory activity on COX-2.application/pdfporUniversidade Federal Rural do Rio de JaneiroPrograma de Pós-Graduação em Ciências FisiológicasUFRRJBrasilInstituto de Ciências Biológicas e da Saúdeopioid system,, .leukocyte migrationnociceptionsistema opioidemigração leucocitárianocicepçãoFisiologiaFarmacologiaAvaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanolAntinociceptive and anti-inflammatory profile of (±)-4-chloro-6-(naphthalen-1-yl)-tetrahydro-2H-pyran-2-yl-methanolinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/masterThesisAIRES, M. M. Fisiologia. Rio de Janeiro: Guanabara Koogan, 2008. 1252p. AKIL, H.; OWENS, C.; GUTSTEIN, H.; TAYLOR, L.; CURRAN, E.; WATSON, S. Endogenous opioids: overview and current issues. Drug Alcohol Depend, v. 51, n. 1-2, p. 127-40, 1998. AL-HASANI, R.; BRUCHAS, M. R. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology, v. 115, n. 6, p. 1363-81, 2011. ALVARENGA, F.Q.; MOTA, B.C.F.; LEITE, M.N.; FONSECA, J.M.S.; OLIVEIRA, D.A.; ROYO, V.A.; SILVA, M.L.A.; ESPERANDIM, V.; BORGES, A.; LAURENTIZ, R.S. In vivo analgesic activity, toxicity and phytochemical screening of the hydroalcoholic extract from the leaves of Psidium cattleianum Sabine. Journal of Ethnopharmacology 150, 280–284, 2013. ALVES, D; DUARTE, I.D. Involvement of ATP-sensitive K(+) channels in the peripheral antinociceptive effect induced by dipyrone. Eur. J. Pharmacol.; 444: 47-52. 2002. AMARANTE, L.H.; DUARTE, I.D. The kappa-opioid agonist (+/-)-bremazocine elicits peripheral antinociception by activation of the L-arginine/nitric oxide/cyclic GMP pathway. Eur. J. Pharmacol., v.454, n.1, p. 19-23, 2002. AMAYA, F; IZUMI, Y; MATSUDA, M; SASAKI, M. Tissue Injury and Related Mediators of Pain Exacerbation. Curr Neuropharmacol.; 11(6): 592–597, 2013. BALAJI, B; HARIHARAN, S; SHAH, D.B; RAMANATHAN, M. Discovery of potential and selective COX-1 inhibitory leads using pharmacophore modelling, in silico screening and in vitro evaluation. Eur. J. Med. Chem. 86: 469-480. 2014. BALTIERI, D.A.; STRAIN, E.C.; DIAS, J.C.; SCIVOLETTO, S.; MALBERGIER, A.; NICASTRI, S.; JERÔNIMO, C.; ANDRADE, A.G. Brazilian guideline for the treatment of patients with opioids dependence syndrome. Rev Bras Psiquiatr 2004;26(4):259-69, 2004. BANSAL, S;BALA, M; SUTHAR, S.K;CHOUDHARY, S; BHATTACHARYA, S; BHARDWAJ, V; SINGLA, S; JOSEPH, A. Design and synthesis of novel 2-phenyl-5-(1, 3-diphenyl-1 H-pyrazol-4-yl)-1,3,4-oxadiazoles as selective COX-2inhibitors with potent anti-inflammatory activity. Eur. J. Med. Chem. 80:167- 174. 2014. BARROS, H.M; TANNHAUSER, M.A; TANNHAUSER, S.L; TANNHAUSER, M. Enhanced detection of hyperactivity after drug withdrawal with a simple modification of the open-field apparatus. J Pharmacol Methods. 26(4):269-275, 1991. 55 BOHN, L.M; GAINETDINOV, R.R; LIN, F.T; LEFKOWITZ, R.J; CARON, M.G. Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature; 408: 720–23. 2000. CALIXTO JB, MEDEIROS R, FERNANDES ES, FERREIRA J, CABRINI DA, CAMPOS MM. Kinin B1 receptors: key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J Pharmacol.; 143:803-18, 2004. CAMARATA, P.J., YAKSH, T.L. Characterization of the spinal adrenergic receptors mediating the spinal effects produced by the microinjection of morphine into the periaqueductal gray. Brain Res. Jun 10;336(1):133–142, 1985. CAPIM, S.L; CARNEIRO, P.H.P; CASTRO, P.C; BARROS, M.R.M; MARINHO, B.G; VASCONCELLOS, M.L.A.A. Design, Prins-cyclization, reaction promoting diastereoselective synthesis of 10 new tetrahydropyran derivatives and in vivo antinociceptive evaluations. European Journal of Medicinal Chemistry. 58:1-11, 2012. CARLTON, S.M.; HARGETT, G.L.; COGGESHALL, R.E. Localization and activation of glutamate receptors in unmyelinated axons of rat glabrous skin. Neurosci. Lett., 197(1), (25-28), 1995. CHUNG, E; BURKE, B; BIEBER, A.J; DOSS, J.C; OHGAMI, Y; QUOCK, R.M. Dynorphinmediated antinociceptive effects of L-arginine and SIN-1 (an NO donor) in mice. Brain Res. Bull. 70: 245–250, 2006. COMMINS, S.P; BORISH, L; STEINKE, J.W. Immunologic messenger molecules: Cytokines, interferons, and chemokines. The Journal of allergy and clinical immunology. 125(2 Suppl 2):S53-72., 2010. COSTANTINO, C. M; GOMES, I.; STOCKTON, S.D; LIM, M.P; DEVI, L.A. Opioid receptor heteromers in analgesia. Expert Rev Mol Med, v. 14, p. e9, 2012. CRUVINEL, W.M e cols. Sistema imunitário: Parte I. Fundamentos da imunidade inata com ênfase nos mecanismos moleculares e celulares da resposta inflamatória. Rev. Bras. Reumatol., São Paulo , v. 50, n. 4, p. 434-447, Aug. 2010. CUNNEEN, J.; CARTWRIGHT, M. The puzzle of sepsis: fitting the pieces of the inflammatory response with treatment. AACN Clinical Issues, v. 15, n. 1, p. 18-44, 2004. CUNHA, T.M; ROMAN-CAMPOS, D; LOTUFO, C.M; DUARTE, H.L; SOUZA, G.R; VERRI, W.A., JR; FUNEZ, M.I; DIAS, Q.M; SCHIVO, I.R; DOMINGUES, A.C; SACHS, D; CHIAVEGATTO, S; TEIXEIRA, M.M; HOTHERSALL, J.S; CRUZ, J.S; CUNHA, F.Q; FERREIRA, S.H. Morphine peripheral analgesia depends on activation of the PI3Kγ/ AKT/nNOS/NO/KATP signaling pathway. Proc. Natl. Acad. Sci. USA 107:4442–4447, 2010. 56 CURY, Y; PICOLO, G; GUTIERREZ, V.P; FERREIRA, S.H. Pain and analgesia: The dual effect of nitric oxide in the nociceptive system. Nitric Oxide 25: 243–254, 2011. D’AMOUR, F.E; SMITH, D.L. A method for determining loss of pain sensation. J. Pharmacol. Exp. Ther., 72: 74–79, 1941. DAWSON, J.; SEDGWICK, A. D.; EDWARDS, J. C.; LEES, P. A comparative study of the cellular, exudative and histological responses to carrageenan, dextran and zymosan in the mouse. International Journal of Tissue Reactions, v. 13, n. 4, p. 171– 185, 1991. DICKENSON, A.H.; SULLIVAN, A.F. Evidence for a role of the NMDA receptor in the frequency dependent potentiaton of deep rat dorsal horn nociceptive neurones following C fibre stimulation. Neuropharmacology, v.26, n. 8, p. 1235-1238, 1987. DI ROSA, M.; GIROUD, J. P.; WILLOUGHBY, D. A. Studies on the mediators of the acute inflammatory response induced in rats in different sites by carrageenan and turpentine. Journal of Pathology, Amsterdam, v. 104, n. 1, p. 15-29, 1971. DUARTE, I.D; LORENZETTI, B.B; FERREIRA, S.H. Peripheral analgesia and activation of the nitric oxide–cyclic GMP pathway. Eur. J. Pharmacol. 186: 289–293, 1990. DUARTE, D.B.; VASKO, M.R.; FEHRENBACHER, J.C. Models of inflammation: carrageenan air pouch. Current protocols in pharmacology. v. 56, p. 561-568, 2012. FEIN, A. Nociceptores: As células que sentem dor. Petrov P, Francischi JN, Ferreira SH, et al. tradutores. Ribeirão Preto – SP: Dor On Line; 2011. 106 p. Disponivel em: http://www.dol.inf.br/nociceptores Acessado em 30 de Março de 2015. FENG, Y.; XIAOZHOU, H.; YANG, Y.; CHAO, D.; LAZARUS, L.H.; XIA, Y. Current research on opioid receptor function. Curr Drug Targets, v. 13, n. 2, p. 230-46, 2012. FELIPINI, R.C. Vídeo de Inflamação Aguda – UNESP. Departamento de Patologia e Propedêutica Clínica, 2013. Disponível em: <https://www.youtube.com/watch?v=gqCIIpHIfqw>. Acesso em: 10 fev.2016. FERREIRA, S.H; VAN ARMAN, C.G. Oedema and increased vascular permeability. In: Vane JR, Van Arman CG, editors. Handbook of experimental pharmacology. New York 7 Springer-Verlag; p. 75-91, 1979. FERREIRA, S.H. A classification of peripheral analgesics based upon their mode of action. Oxford: Oxford University Press, 1990. FERREIRA, A.A; AMARAL, F.A; DUARTE, I.D.G; OLIVEIRA, P.M; ALVES, R.B; SILVEIRA, D; AZEVEDO, A.O; RASLAN, D.S; CASTRO, M.S.A. 57 Antinociceptive effect from Ipomoea cairica extract. J. Ethnopharmacol; 105: 148–53. 2006. FERREIRA, S.H; FERRARI, L.F; CUNHA, T.M; NASCIMENTO, P.G.B.D; JUNIOR, W.A.V; CUNHA, F.Q. Dor: Príncipios e Prática. Capítulo 19: Dor Inflamatória. 2010. Disponível em: < http://dol.inf.br/Html/DorInflamatoria.html>. Acesso em: 2 set. 2016. FISCHER, L.G.; SANTOS, D.; SERAFIN, C.; MALHEIROS, A.; MONACHE, F.D.; MONACHE, G.D.; FILHO, V.C.; SOUZA, M.M. Further Antinociceptive Properties of Extracts and Phenolic Compounds from Plinia glomerata (Myrtaceae) Leaves. Biol. Pharm. Bull. 31(2) 235—239, 2008. FITZGERALD, G.A; RICCIOTTI, E. Prostaglandins and Inflammation. Arterioscler Thromb Vasc Biol.; 31(5): 986–1000, 2011. FÖRSTERMANN, U; SESSA, W.C. Nitric oxide synthases: regulation and function.European Heart Journal 33, 829–837, 2012. FRANCISCHETTI, I; MORENO, J.B; SCHOLZ, M; YOSHIDA, W.B. Leukocytes and the inflammatory response in ischemia-reperfusion injury. Rev Bras Cir Cardiovasc.; 25(4): 575-584, 2010. FRENZEL, L; HERMINE, O. Mast cells and inflammation. Joint Bone Spine 80: 141-145, 2013. GREGORIAN, R.S., JR.; GASIK, A.; KWONG, W.J; VOELLER, S.; KAVANAGHZ, S. Importance of Side Effects in Opioid Treatment: A Trade-Off Analysis With Patients and Physicians. The Journal of Pain, Vol 11, No 11: pp 1095-1108, 2010. GREGORY, N.S; HARRIS, A.L; ROBINSON, C.R; DOUGHERTY, P.M, FUCHS, P.N, SLUKA, K.A. An overview of animal models of pain: disease models and outcome measures. J Pain. doi:10.1016/j.jpain.2013.06.008. November; 14(11), 2013. GRELLNER, W. Time-dependent imunohistochemicaldetection of pro-inflammatory cytokines (IL-1, IL-6, TNF) in human skin wounds. Forens Sci. Int., v. 130, n. 2-3, p. 90-96, 2002. GUGINSKI, G; LUIZ, A.P; SILVA, M.D; MASSARO, M; MARTINS, D.F; CHAVES, J; MATTOS, R.W; SILVEIRA, D; FERREIRA, V.M; CALIXTO, J.B; SANTOS, A.R. Mechanisms involved in the antinociception caused by ethanolic extract obtained from the leaves of Melissa officinalis (lemon balm) in mice. Pharmacol. Biochem. Behav. 93:10–16, 2009. GUILHON, C.C; RAYMUNDO, L.J.R.P; ALVIANO, D.S; BLANK, A.F; ARRIGONI-BLANK, M.F; CAVALCANTI, S.C; ALVIANO, C.S; FERNANDES, P.D. Characterisation of the anti-inflammatory and antinociceptive activities and mechanism of the action of Lippia gracilis essential oil. Journal Ethnopharmacology, v. 135, p. 406-413, 2011. 58 GUTIERREZ, V; ZAMBELLI, V; PICOLO, G; CHACUR, M; SAMPAIO, S; BRIGATTE, P; CURY, Y. Peripheral L-arginine-nitric oxide-cGMP pathway and ATP-sensitive K+ channels are involved in the antinociceptive effect of crotalphine on neuropathic pain in rats. Behav. Pharmacol., (1):14-24, 2012. HALLEGUA, D.S; WEISMAN, M.H. Potential therapeutic uses of interleukin1receptor antagonists in human diseases. Ann Rheum; 61: 960–967. 2002. HAN, J.; KIM, N.; KIM, E.; HO, W.K; EARM, Y.E. Modulation of ATP-sensitive potassium channels by cGMP-dependent protein kinase in rabbit ventricular myocytes. J. Biol. Chem., v.276, n. 25, p. 22140-22147, 2001. HERVERA, A; LEANEZ, S; NEGRETE, R; POL, O. The peripheral administration of a nitric oxide donor potentiates the local antinociceptive effects of a DOR agonist during chronic inflammatory pain in mice. Naunyn Schmiedebergs Arch. Pharmacol 380: 345–352, 2009. HUNSKAAR, S; HOLE, K. The formalin test in mice: dissociation between inflammatory and non-inflammatory pain. Pain 30: 103–14, 1987. JAIN, M.; PARMAR, H.S. Evaluation of antioxidative and anti-inflammatory potential of hesperidin and naringin on the rat air pouch model of inflammation. Inflammation research. v.60, p. 483-491, 2011. JAVANMARDI, K, M; PARVIZ, S.S; SADR, M; KESHAVARZ, B; MINAII; DEHPOUR, A.R. Involvement of N-methyl-D-aspartate receptors and nitric oxide in the rostral ventromedial medulla in modulating morphine pain-inhibitory signals from the periaqueductal grey matter in rats. Clin. Exp. Pharmacol. Physiol. 32: 585–589, 2005. KELLEY, N.E; TEPPER, D.E. Rescue therapy for acute migraine, part 3: opioids, NSAIDs, steroids, and post-discharge medications. Headache.;52(3):467-82, 2012. KOSTER, R; ANDERSON, M; DE BEER, EJ. Acetic acid for analgesic screening. Federation Proceedings. v. 18, p. 412, 1959. LAW, B.K.; WALTNER-LAW, M.E; ENTINGH, A.J.; CHYTIL, A; AAKRE, M.E.; NØRGAARD, P.; MOSES, H.L. Salicylate-induced growth arrest is associated with inhibition of p70s6K and down-regulation of c-myc, cyclin D1, cyclin A, and proliferating cell nuclear antigen. J. Biol. Chem., v.275, n.49, p. 38261-38267, 2000. LE BARS, D., GOZARIU, M., CADDEN, S. Animal models of nociception. Pharmacological Reviews, v. 53, p. 628-651, 2001. LESNIAK, A.; LIPKOWSKI, A. W. Opioid peptides in peripheral pain control. Acta Neurobiol Exp (Wars), v. 71, n. 1, p. 129-38, 2011. 59 LOESER, J. D.; TREEDE, R. D. The Kyoto protocol of IASP Basic Pain Terminology. Pain, v. 137, n. 3, p. 473-477, Jul 31 2008. LOISA, P.; RINNE, T.; LAINE, S.; HURME, M.; KAUKINEN, S. Anti-inflammatory cytokine response and the development of multiple organ failure in severe sepsis. Acta Anaesthesiologica Scandinavica, v. 47, n. 3, p. 319-325, 2003. LORKE, D. A new approach to practical acute toxicity testing. Arch. Toxicol; 54: 275–87. 1983. LOZANO-CUENCA, J.; CASTANEDA-HERNANDEZ, G.; GRANADOS-SOTO, V. Peripheral and spinal mechanisms of antinociceptive action of lumiracoxib. Eur. J. Pharmacol. 513: 81–91, 2005. MARINHO, B.G; MIRANDA, L.S.M.; GOMES, N.M.; MATHEUS, M.E.; LEITÃO, S.G.; VASCONCELLOS, M.L.A.A.; FERNANDES, P.D. Antinociceptive action of (±)-cis-(6-ethyl-tetrahydropyran-2-yl)-formic acid in mice. European Journal of Pharmacology. 550: 47-53, 2006. MARTINS, R.T.; ALMEIDA, D.B.; MONTEIRO, F.M.R.; KOZACS, P.A.; RAMINA, R. Opioid receptors to date. Rev Dor. São Paulo, 13(1):75-9, 2012. MATSUKAWA, A.; HOGABOAM, C.M.; LUKACS, N.W.; LINCOLIN, P.M.; EVANOFF, H.L.; KUNKEL, S.L. Pivotal role of the CC chemokine, macrophage derived chemokine, in the innate immune response. Journal of Immunology, Boston, v.164, p.5362–5368, 2000. MÁZAK, K.; HOSZTAFI, S.; RÁCZ. Á.; NOSZÁL, B. Structural and physicochemical profiling of morphine and related compounds of therapeutic interest. Mini-reviews in Med. Chem., v.9, p. 984-995, 2009. MCNAMARA CR, MANDEL-BREHM J, BAUTISTA DM, SIEMENS J, DERANIAN KL, ZHAO M, HAYWARD NJ, CHONG JA, JULIUS D, MORAN MM, FANGER CM. TRPA1 mediates formalin-induced pain. Proc. Natl. Acad. Sci. U. S. A. ; 104: 1352–3530. 2007. MENEGAZZI, M; DI PAOLA, R; MAZZON, E; GENOVESE, T; CRISAFULLI, C; DAL BOSCO, M; ZOU, Z; SUZAKI, H; CUZZOCREA S. Glycyrrhizin attenuates the development of carrageenan-induced lung injury in mice. Pharmacol Res; 58: 22–31. 2008. MERRER,J.L.; JECKER, J.A.; BEFORT, K.; KIEFFER, B.L. Reward processing by the opioid system in the brain. Physiol Rev. Oct;89(4):1379-412, 2009. MILANO, J; OLIVEIRA, S.M; ROSSATO, M.F; SAUZEM, P.D; MACHADO, P; BECK, P; ZANATTA, N; MARTINS, M.A.P; MELLO, C.F; RUBIN, M.A; FERREIRA, J; BONACORSO, H.G. Antinociceptive effect of novel trihalomethyl-substituted pyrazoline methyl esters in formalin and hot-plate tests in mice. Eur. J. Pharmacol; 581: 86–96. 2008. 60 MILLAN, M.J. The induction of pain: an integrative review. Prog. Neurobiol, v. 57, p. 161-164, 1999. MILLAN, M.J. Descending control of pain. Prog. Neurobiol. 66:355–474, 2002. MILLER, R.J. Presynaptic receptors. Annu Rev Pharmacol Toxicol; 38: 201–27. 1998. MOHAMAD, A. S.; AKHTAR, M. N.; ZAKARIA, Z. A.; PERIMAL, E.K.; KHALID, S.; MOHD, P. A.; KHALID, M. H.; ISRAF, D. A.; LAJIS, N. H.; SULAIMAN, M. R. Antinociceptive activity of a synthetic chalcone, flavokawin B on chemical and thermal models of nociception in mice. European Journal of Pharmacology. v. 647, n. 1 – 3, p. 103 – 109, 2010. NAPIMOGA, C.J.T; PELLEGRINI-DA-SILVA, A; FERREIRA, V.H; NAPIMOGA, M.H; PARADA, C.A; TAMBELI, C.H. Gonadal hormones decrease temporomandibular joint kappa-mediated antinociception through a down-regulation in the expression of kappa opioid receptors in the trigeminal ganglia. Eur. J. Pharmacol. 617: 41–47, 2009. NESS, T.J., GEBHART, G.F. Visceral pain: a review of experimental studies. Pain 41:167–234, 1990. NUGTEREN, D.H; HAZELHOF, E. Isolation and properties of intermediates in prostaglandin biosynthesis. Biochim. Biophys. Acta. 326: 448-461. 1973. OCANA, M; CENDRAN, C.M; COBOS, E.J; ENTRENA, J.M; BAEYENS, J.M. Potassium channels and pain: present realities and future opportunities. Eur J Pharmacol; 500: 203–19. 2004. OLIVEIRA, F.S.; SOUSA, D.P.; ALMEIDA, R.N. Antinociceptive effect of hydroxydihydrocarvone. Biol Pharm Bull 31(4): 588 – 591, 2008. OSSIPOV, M.H.; LAI J, VANDERAH, T.W.; PORRECA, F. Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sciences, v. 73, p.783-800, 2003. PAN, Z.Z; TERSHNER, S.A; FIELDS, H.L. Cellular mechanism for anti-analgesic action of agonists of the kappa-opioid receptor. Nature; 389: 382–5. 1997. PARADA, C.A; TAMBELI, C.H; CUNHA, F.Q; FERREIRA, S.H. The major role of peripheral release of histamine and 5-hydroxytryptamine in formalin-induced nociception. Neuroscience; 102: 937–44. 2001. PARVEEN, Z.; DENG, Y.; SAEED, M.K.; DAI, R.; AHAMAD, W.; YU, Y.H. Antiinflammatory and analgesic activities of Thesium chinese Turcz extracts and its major flavonoids, kaampferol and kaempferol-3-O-glucoside. Yakugaku Zasshi, v.127, p.1275-1279, 2007. 61 RAJA, S.; MEYER, R.A.; RINCKAMP, M.; CAMPBELL, J.N. Peripheral neural mechanism of nociception. In WALL, P.D.; MELZACK, R. (Eds). Textbook of pain. Ediburgh: Churchill Livingstone, p. 11-57, 1999. RAMANA, K.V.; TAMMALI, R.; REDDY, A.B.M.; BHATNAGAR, A.; SRIVASTAVA, S.K.; Aldose Reductase-Regulated Tumor Necrosis Factor- α Production Is Essential for High Glucose-Induced Vascular Smooth Muscle Cell Growth. Endocrinology. 148, n. 9, 4371-4384, 2007. ROBBINS & COTRAN. Patologia: Bases patológicas das doenças. 8ª Edição. Rio de Janeiro: Elsevier, 2010. 1458 p. ROCHA APC, KRAYCHETE DC, LEMONICA L, CARVALHO LR, BARROS GAM, GARCIA JBS, SAKATA RK — Pain: Current Aspects on Peripheral and Central Sensitization. Rev Bras Anestesiol Review article 57: 1: 94-105, 2007. SACHS, D.; CUNHA, F.Q.; FERREIRA, S.H. Peripheral analgesic blockade of hypernociception: activation of arginine/NO/cGMP/protein kinase G/ATP-sensitive K+ channel pathway. Proc. Natl. Acad. Sci. USA, v. 101, n. 10, p. 3680-3685, 2004. SCHRAMM, R; THORLACIUS, H. Neutrophil recruitment in mast cell-dependent inflammation inhibitory mechanisms of glucocorticoids. Inflammation research, v.53 (12), p. 644-652, 2004. SCHULTZ, J.; GROSS, G. Opioids and cardioprotection. Pharmac. Ther., v.89, p. 123-137, 2001. SILVA, J.C; SARAIVA, S.R; OLIVEIRA, R.G; ALMEIDA, J.R. Experimental models for evaluation of antinociceptive activity of natural products: a review. Rev. Bras. Farm. 94 (1): 18-23, 2013. SOARES, A.C; LEITE, R; TATSUO, M.A; DUARTE, I.D. Activation of ATP-sensitive K+ channels: mechanism of peripheral antinociceptive action of the nitric oxide donor, sodium nitroprusside. Eur. J. Pharmacol. 400: 67–71, 2000. SOARES, A.C; DUARTE, I.D. Dibutyryl-cyclic GMP induces peripheral antinociception via activation of ATP-sensitive K+ channels in the rat PGE2-induced hyperalgesic paw, Br. J. Pharmacol. 134: 127–131, 2001. SOJA, P. J.; TAEPAVARAPRUK, N.; PANG, W.; CAIRNS, B. E.; MCERLANE, S. A.; FRAGOSO, M. C. Transmission through the dorsal spinocerebellar and spinoreticular tracts: wakefulness versus thiopental anesthesia. Anesthesiology, v. 97, n. 5, p. 1178-88, 2002. SOMMER, C. Serotonin in Pain and Analgesia - Actions in the Periphery. Molecular Neurobiology, v. 30, p. 117-125, 2004. 62 STAROWICZ, K; OBARA, I; PRZEWLOCKI, R; PRZEWLOCKA, B. Inhibition of morphine tolerance by spinal melanocortin receptor blockade. Pain; 117: 401–11. 2005. STEIN, C.; CLARK, J.D.; VASKO, M.R.; WILCOX, G.L.; OVERLAND, A.C.; VANDERAH, T.W.; SPENCER, R.H. Peripheral mechanisms of pain and analgesia. Brain Res Rev. 2009 Apr;60(1):90-113, 2009. SU, X; JOSHI, S.K; KARDOS, S; GEBHART, G.F. Sodium channel blocking actions of the kappaopioid receptor agonist U50,488 contribute to its visceral antinociceptive effects. J Neurophysiol; 87: 1271–9. 2002. SU, X; CASTLE, N.A; ANTONIO, B; ROELOFFS, R; THOMAS, J.B; KRAFTE, D.S; et al. The effect of kappa-opioid receptor agonists on tetrodotoxin-resistant sodium channels in primary sensory neurons. Anesth Analg; 109: 632–40. 2009. THEOHARIDES, T.C; ALYSANDRATOS, K.D; ANGELIDOU, A; DELIVANIS, D.A; SISMANOPOULOS, N; ZHANG, B; ASADI, S; VASIADI, M; WENG, Z; MINIATI, A; KALOGEROMITROS, D. Mast cells and inflammation. Biochimica et Biophysica Acta 1822, 21–33, 2012. THOMAZZI, S. M.; SILVA, C. B.; SILVEIRA, D. C. R.; VASCONCELLOS, C. L. C.; LIRA, A. F.; CAMBUI, E. V. F.; ESTEVAM, C. S.; ANTONIOLLI, A. R. Antinociceptive and anti-inflammatory activities of Bowdichia virgilioides (sucupira). Journal of Ethnopharmacology, v. 127, p. 451 - 456, 2010. TORNOS, M.P; SÁENZ, M.T; GARCIA, M.D; FERNÁNDEZ, M.A. Antinociceptive effects of the tubercles of Anredera leptostachys. J. Ethnopharmacol; .68: 229–34. 1999. VANDERAH, T.W. Delta and kappa opioid receptors as suitable drug targets for pain. Clin J Pain; 26: 10–5. 2010. VARGA, E.V.; YAMAMURA, H.I; RUBENZIK, M.K.; STROPOVA, D.; NAVRATILOVA, E.; ROESKE, W.R. Molecular mechanisms of excitatory signaling upon chronic opioid agonist treatment. Life Sciences, v. 74, p. 299-311, 2003. VERRI, W.A; CUNHA, T.M; PARADA, C.A; POOLE, S; CUNHA, F.Q; FERREIRA, S.H. Hypernociceptive role of cytokines and chemokines: targets for analgesic drug development? Pharmacol Ther. Oct;112(1):116-38, 2006. VIGIL, S.V.G; DE LIZ, R.; MEDEIROS, Y.S.; FRÖDETS. Efficacy of tacrolimus in inhibiting inflammation caused by carrageenan in a murine model of air pouch. Transpl Immunol; 19:25-29, 2008. VOSCOPOULOS, C.; LEMA, M.; When does acute pain become chronic? Br J Anaesth, v. 105 Suppl 1, p. i69-i85. ISSN 0007-0912, 2010. 63 WINTER, C. A.; RISLEY, E. A.; NUSS, G. W. Carrageenin-induced edema in hind paw of the ratas an assay for anti-inflammatory drugs. Proc Soc Exp Biol Med, v. 111, p. 544-547, 1962. ZHOU, S.; BONASERA, L.; CARLTON, S.M. Peripheral administration of NMDA, AMPA or KA results in pain behaviors in rats. Neuroreport, 7(4), 895-900.), 1996. ZHU, Z. Z.; MA, K. J.; RAN, X.; ZHANG, H.; ZHENG, C. J.; HAN, T.; ZHANG, Q. Y.; QIN, L. P. Analgesic, anti-inflammatory and antipyretic activities of the petroleum ether fraction from the ethanol extract of Desmodium podocarpum. Journal of Ethnopharmacology. v. 133, n. 3, p. 1126 – 1131, 2011https://tede.ufrrj.br/retrieve/57244/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf.jpghttps://tede.ufrrj.br/jspui/handle/jspui/1425Submitted by Sandra Pereira (srpereira@ufrrj.br) on 2017-02-14T12:00:15Z No. of bitstreams: 1 2016 - Gabriela Mastrangelo Gonçalves.pdf: 1218648 bytes, checksum: d31754b24278f68ec75382b5ff3932c0 (MD5)Made available in DSpace on 2017-02-14T12:00:15Z (GMT). No. of bitstreams: 1 2016 - Gabriela Mastrangelo Gonçalves.pdf: 1218648 bytes, checksum: d31754b24278f68ec75382b5ff3932c0 (MD5) Previous issue date: 2016-02-23info:eu-repo/semantics/openAccessreponame:Biblioteca Digital de Teses e Dissertações da UFRRJinstname:Universidade Federal Rural do Rio de Janeiro (UFRRJ)instacron:UFRRJTHUMBNAIL2016 - Gabriela Mastrangelo Gonçalves.pdf.jpgGenerated Thumbnailimage/jpeg1943https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/1/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf.jpgcc73c4c239a4c332d642ba1e7c7a9fb2MD51TEXT2016 - Gabriela Mastrangelo Gonçalves.pdf.txtExtracted Texttext/plain155911https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/2/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf.txtb56ec69846bfd4203c717ef0dbb08471MD52ORIGINAL2016 - Gabriela Mastrangelo Gonçalves.pdfDocumento principalapplication/pdf1218648https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/3/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdfd31754b24278f68ec75382b5ff3932c0MD53LICENSElicense.txttext/plain2089https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/4/license.txt7b5ba3d2445355f386edab96125d42b7MD5420.500.14407/114052023-12-21 22:52:19.495oai:rima.ufrrj.br:20.500.14407/11405Tk9UQTogQ09MT1FVRSBBUVVJIEEgU1VBIFBSP1BSSUEgTElDRU4/QQpFc3RhIGxpY2VuP2EgZGUgZXhlbXBsbyA/IGZvcm5lY2lkYSBhcGVuYXMgcGFyYSBmaW5zIGluZm9ybWF0aXZvcy4KCkxJQ0VOP0EgREUgRElTVFJJQlVJPz9PIE4/Ty1FWENMVVNJVkEKCkNvbSBhIGFwcmVzZW50YT8/byBkZXN0YSBsaWNlbj9hLCB2b2M/IChvIGF1dG9yIChlcykgb3UgbyB0aXR1bGFyIGRvcyBkaXJlaXRvcyBkZSBhdXRvcikgY29uY2VkZSA/IFVuaXZlcnNpZGFkZSAKWFhYIChTaWdsYSBkYSBVbml2ZXJzaWRhZGUpIG8gZGlyZWl0byBuP28tZXhjbHVzaXZvIGRlIHJlcHJvZHV6aXIsICB0cmFkdXppciAoY29uZm9ybWUgZGVmaW5pZG8gYWJhaXhvKSwgZS9vdSAKZGlzdHJpYnVpciBhIHN1YSB0ZXNlIG91IGRpc3NlcnRhPz9vIChpbmNsdWluZG8gbyByZXN1bW8pIHBvciB0b2RvIG8gbXVuZG8gbm8gZm9ybWF0byBpbXByZXNzbyBlIGVsZXRyP25pY28gZSAKZW0gcXVhbHF1ZXIgbWVpbywgaW5jbHVpbmRvIG9zIGZvcm1hdG9zID91ZGlvIG91IHY/ZGVvLgoKVm9jPyBjb25jb3JkYSBxdWUgYSBTaWdsYSBkZSBVbml2ZXJzaWRhZGUgcG9kZSwgc2VtIGFsdGVyYXIgbyBjb250ZT9kbywgdHJhbnNwb3IgYSBzdWEgdGVzZSBvdSBkaXNzZXJ0YT8/byAKcGFyYSBxdWFscXVlciBtZWlvIG91IGZvcm1hdG8gcGFyYSBmaW5zIGRlIHByZXNlcnZhPz9vLgoKVm9jPyB0YW1iP20gY29uY29yZGEgcXVlIGEgU2lnbGEgZGUgVW5pdmVyc2lkYWRlIHBvZGUgbWFudGVyIG1haXMgZGUgdW1hIGM/cGlhIGEgc3VhIHRlc2Ugb3UgCmRpc3NlcnRhPz9vIHBhcmEgZmlucyBkZSBzZWd1cmFuP2EsIGJhY2stdXAgZSBwcmVzZXJ2YT8/by4KClZvYz8gZGVjbGFyYSBxdWUgYSBzdWEgdGVzZSBvdSBkaXNzZXJ0YT8/byA/IG9yaWdpbmFsIGUgcXVlIHZvYz8gdGVtIG8gcG9kZXIgZGUgY29uY2VkZXIgb3MgZGlyZWl0b3MgY29udGlkb3MgCm5lc3RhIGxpY2VuP2EuIFZvYz8gdGFtYj9tIGRlY2xhcmEgcXVlIG8gZGVwP3NpdG8gZGEgc3VhIHRlc2Ugb3UgZGlzc2VydGE/P28gbj9vLCBxdWUgc2VqYSBkZSBzZXUgCmNvbmhlY2ltZW50bywgaW5mcmluZ2UgZGlyZWl0b3MgYXV0b3JhaXMgZGUgbmluZ3U/bS4KCkNhc28gYSBzdWEgdGVzZSBvdSBkaXNzZXJ0YT8/byBjb250ZW5oYSBtYXRlcmlhbCBxdWUgdm9jPyBuP28gcG9zc3VpIGEgdGl0dWxhcmlkYWRlIGRvcyBkaXJlaXRvcyBhdXRvcmFpcywgdm9jPyAKZGVjbGFyYSBxdWUgb2J0ZXZlIGEgcGVybWlzcz9vIGlycmVzdHJpdGEgZG8gZGV0ZW50b3IgZG9zIGRpcmVpdG9zIGF1dG9yYWlzIHBhcmEgY29uY2VkZXIgPyBTaWdsYSBkZSBVbml2ZXJzaWRhZGUgCm9zIGRpcmVpdG9zIGFwcmVzZW50YWRvcyBuZXN0YSBsaWNlbj9hLCBlIHF1ZSBlc3NlIG1hdGVyaWFsIGRlIHByb3ByaWVkYWRlIGRlIHRlcmNlaXJvcyBlc3Q/IGNsYXJhbWVudGUgCmlkZW50aWZpY2FkbyBlIHJlY29uaGVjaWRvIG5vIHRleHRvIG91IG5vIGNvbnRlP2RvIGRhIHRlc2Ugb3UgZGlzc2VydGE/P28gb3JhIGRlcG9zaXRhZGEuCgpDQVNPIEEgVEVTRSBPVSBESVNTRVJUQT8/TyBPUkEgREVQT1NJVEFEQSBURU5IQSBTSURPIFJFU1VMVEFETyBERSBVTSBQQVRST0M/TklPIE9VIApBUE9JTyBERSBVTUEgQUc/TkNJQSBERSBGT01FTlRPIE9VIE9VVFJPIE9SR0FOSVNNTyBRVUUgTj9PIFNFSkEgQSBTSUdMQSBERSAKVU5JVkVSU0lEQURFLCBWT0M/IERFQ0xBUkEgUVVFIFJFU1BFSVRPVSBUT0RPUyBFIFFVQUlTUVVFUiBESVJFSVRPUyBERSBSRVZJUz9PIENPTU8gClRBTUI/TSBBUyBERU1BSVMgT0JSSUdBPz9FUyBFWElHSURBUyBQT1IgQ09OVFJBVE8gT1UgQUNPUkRPLgoKQSBTaWdsYSBkZSBVbml2ZXJzaWRhZGUgc2UgY29tcHJvbWV0ZSBhIGlkZW50aWZpY2FyIGNsYXJhbWVudGUgbyBzZXUgbm9tZSAocykgb3UgbyhzKSBub21lKHMpIGRvKHMpIApkZXRlbnRvcihlcykgZG9zIGRpcmVpdG9zIGF1dG9yYWlzIGRhIHRlc2Ugb3UgZGlzc2VydGE/P28sIGUgbj9vIGZhcj8gcXVhbHF1ZXIgYWx0ZXJhPz9vLCBhbD9tIGRhcXVlbGFzIApjb25jZWRpZGFzIHBvciBlc3RhIGxpY2VuP2EuCg==Biblioteca Digital de Teses e Dissertaçõeshttps://tede.ufrrj.br/PUBhttps://tede.ufrrj.br/oai/requestbibliot@ufrrj.br||bibliot@ufrrj.bropendoar:2023-12-22T01:52:19Biblioteca Digital de Teses e Dissertações da UFRRJ - Universidade Federal Rural do Rio de Janeiro (UFRRJ)false
dc.title.por.fl_str_mv Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
dc.title.alternative.eng.fl_str_mv Antinociceptive and anti-inflammatory profile of (±)-4-chloro-6-(naphthalen-1-yl)-tetrahydro-2H-pyran-2-yl-methanol
title Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
spellingShingle Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
Gonçalves, Gabriela Mastrangelo
opioid system,, .
leukocyte migration
nociception
sistema opioide
migração leucocitária
nocicepção
Fisiologia
Farmacologia
title_short Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
title_full Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
title_fullStr Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
title_full_unstemmed Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
title_sort Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol
author Gonçalves, Gabriela Mastrangelo
author_facet Gonçalves, Gabriela Mastrangelo
author_role author
dc.contributor.author.fl_str_mv Gonçalves, Gabriela Mastrangelo
dc.contributor.advisor1.fl_str_mv Marinho, Bruno Guimarães
dc.contributor.advisor1ID.fl_str_mv 077.077.277-38
dc.contributor.advisor1Lattes.fl_str_mv http://lattes.cnpq.br/2685794388394484
dc.contributor.referee1.fl_str_mv Cortes, Wellington da Silva
dc.contributor.referee2.fl_str_mv Matheus, Maria Eline
dc.contributor.authorID.fl_str_mv 384.158.588-41
dc.contributor.authorLattes.fl_str_mv http://lattes.cnpq.br/0300079148359674
contributor_str_mv Marinho, Bruno Guimarães
Cortes, Wellington da Silva
Matheus, Maria Eline
dc.subject.eng.fl_str_mv opioid system,, .
leukocyte migration
nociception
topic opioid system,, .
leukocyte migration
nociception
sistema opioide
migração leucocitária
nocicepção
Fisiologia
Farmacologia
dc.subject.por.fl_str_mv sistema opioide
migração leucocitária
nocicepção
dc.subject.cnpq.fl_str_mv Fisiologia
Farmacologia
description Diversos fármacos de uso corrente foram descobertos durante ensaios experimentais e mediante a observação em animais. Quando um novo composto parece promissor, geralmente este sofre alterações em sua estrutura química a fim de aperfeiçoar a sua seletividade, potência e eficácia terapêutica. O objetivo deste estudo foi avaliar as atividades antinociceptiva e anti-inflamatória de um novo composto sintético (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol (CTHP) preparado a partir de um protótipo anterior, ácido (±)-cis-(6-etil-tetrahidropirano-2-il) fórmico. O composto CTHP foi avaliado em ensaios de indução de dor aguda. A administração oral do composto foi capaz de induzir atividade antinociceptiva nos modelos de contorções abdominais induzidas por ácido acético, formalina (em ambas as fases) e retirada da cauda. Para elucidação do mecanismo de ação do composto, o modelo de retirada de cauda foi utilizado. Neste modelo foi realizada a administração prévia de naloxona (antagonista opioide não-seletivo), em que foi observada a inibição do efeito produzido pelo composto. Assim, foi então avaliada a participação seletiva de receptores opioides (μ, δ e κ), através de administração prévia de metilnaltrexona, naltrindol e nor-binaltorfimina, respectivamente, onde somente a nor-binaltorfimina foi capaz de reduzir o efeito antinociceptivo do composto. Para avaliar a possível participação da via NO/GMPc/KATP, os animais foram pré-tratados com N-nitro-arginina-L-metil éster (L-NAME), 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalina-1-ona (ODQ) (inibidor da guanilato ciclase sensível ao óxido nítrico) e glibenclamida (bloqueador de canais de potássio regulados por ATP), foi observado redução do efeito antinociceptivo com a administração prévia de todos estes. No teste de indução de tolerância, tanto a morfina quanto o composto desenvolveram tolerância, porém o composto de forma mais lenta e houve desenvolvimento de tolerância cruzada com a morfina. Para avaliar o envolvimento da via serotoninérgica na atividade do composto, foi realizada a administração diária por 3 dias de 4-cloro-DL-fenilalanina (inibidor da enzima triptofano hidroxilase). Nenhuma alteração no efeito antinociceptivo do composto foi observado, no que diz respeito ao envolvimento da via serotoninérgica. Já o modelo de campo aberto foi utilizado para avaliar a possibilidade de interferência da performance motora sobre o efeito antinociceptivo, foi demonstrada ausência desta interferência. Quanto à atividade anti-inflamatória, o resultado no teste de edema de pata indica efeito antiedematogênico do composto. Houve uma diminuição na quantidade de leucócitos totais, indicando que o composto foi capaz de reduzir a migração leucocitária na inflamação existente na bolsa de ar subcutâneo. O composto também demonstrou atividade inibitória sobre a produção de TNF-α e inibição seletiva da enzima COX-2. Esses resultados indicam atividade antinociceptiva significativa do composto, sem evidências de comprometimento motor. O composto CTHP demonstrou efeito antinociceptivo central, tendo este último contribuição dos sistemas opioide (seletivo para receptores do tipo κ) e nitrérgico em seu mecanismo de ação. E ainda, atividade anti-inflamatória, com inibição da migração leucocitária, de TNF-α e atividade inibitória seletiva sobre COX-2.
publishDate 2016
dc.date.issued.fl_str_mv 2016-02-23
dc.date.accessioned.fl_str_mv 2023-12-22T01:52:19Z
dc.date.available.fl_str_mv 2023-12-22T01:52:19Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/masterThesis
format masterThesis
status_str publishedVersion
dc.identifier.citation.fl_str_mv GONÇALVES, Gabriela Mastrangelo. Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol. 2016. 63 f. Dissertação (Mestrado em Ciências Fisiológicas) - Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica - RJ, 2016.
dc.identifier.uri.fl_str_mv https://rima.ufrrj.br/jspui/handle/20.500.14407/11405
identifier_str_mv GONÇALVES, Gabriela Mastrangelo. Avaliação farmacológica das atividades antinociceptiva e anti-inflamatória do composto (±)-4-cloro-6-(naftaleno-1-il)-tetrahidro-2h-pirano-2-il-metanol. 2016. 63 f. Dissertação (Mestrado em Ciências Fisiológicas) - Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica - RJ, 2016.
url https://rima.ufrrj.br/jspui/handle/20.500.14407/11405
dc.language.iso.fl_str_mv por
language por
dc.relation.references.por.fl_str_mv AIRES, M. M. Fisiologia. Rio de Janeiro: Guanabara Koogan, 2008. 1252p. AKIL, H.; OWENS, C.; GUTSTEIN, H.; TAYLOR, L.; CURRAN, E.; WATSON, S. Endogenous opioids: overview and current issues. Drug Alcohol Depend, v. 51, n. 1-2, p. 127-40, 1998. AL-HASANI, R.; BRUCHAS, M. R. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology, v. 115, n. 6, p. 1363-81, 2011. ALVARENGA, F.Q.; MOTA, B.C.F.; LEITE, M.N.; FONSECA, J.M.S.; OLIVEIRA, D.A.; ROYO, V.A.; SILVA, M.L.A.; ESPERANDIM, V.; BORGES, A.; LAURENTIZ, R.S. In vivo analgesic activity, toxicity and phytochemical screening of the hydroalcoholic extract from the leaves of Psidium cattleianum Sabine. Journal of Ethnopharmacology 150, 280–284, 2013. ALVES, D; DUARTE, I.D. Involvement of ATP-sensitive K(+) channels in the peripheral antinociceptive effect induced by dipyrone. Eur. J. Pharmacol.; 444: 47-52. 2002. AMARANTE, L.H.; DUARTE, I.D. The kappa-opioid agonist (+/-)-bremazocine elicits peripheral antinociception by activation of the L-arginine/nitric oxide/cyclic GMP pathway. Eur. J. Pharmacol., v.454, n.1, p. 19-23, 2002. AMAYA, F; IZUMI, Y; MATSUDA, M; SASAKI, M. Tissue Injury and Related Mediators of Pain Exacerbation. Curr Neuropharmacol.; 11(6): 592–597, 2013. BALAJI, B; HARIHARAN, S; SHAH, D.B; RAMANATHAN, M. Discovery of potential and selective COX-1 inhibitory leads using pharmacophore modelling, in silico screening and in vitro evaluation. Eur. J. Med. Chem. 86: 469-480. 2014. BALTIERI, D.A.; STRAIN, E.C.; DIAS, J.C.; SCIVOLETTO, S.; MALBERGIER, A.; NICASTRI, S.; JERÔNIMO, C.; ANDRADE, A.G. Brazilian guideline for the treatment of patients with opioids dependence syndrome. Rev Bras Psiquiatr 2004;26(4):259-69, 2004. BANSAL, S;BALA, M; SUTHAR, S.K;CHOUDHARY, S; BHATTACHARYA, S; BHARDWAJ, V; SINGLA, S; JOSEPH, A. Design and synthesis of novel 2-phenyl-5-(1, 3-diphenyl-1 H-pyrazol-4-yl)-1,3,4-oxadiazoles as selective COX-2inhibitors with potent anti-inflammatory activity. Eur. J. Med. Chem. 80:167- 174. 2014. BARROS, H.M; TANNHAUSER, M.A; TANNHAUSER, S.L; TANNHAUSER, M. Enhanced detection of hyperactivity after drug withdrawal with a simple modification of the open-field apparatus. J Pharmacol Methods. 26(4):269-275, 1991. 55 BOHN, L.M; GAINETDINOV, R.R; LIN, F.T; LEFKOWITZ, R.J; CARON, M.G. Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature; 408: 720–23. 2000. CALIXTO JB, MEDEIROS R, FERNANDES ES, FERREIRA J, CABRINI DA, CAMPOS MM. Kinin B1 receptors: key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J Pharmacol.; 143:803-18, 2004. CAMARATA, P.J., YAKSH, T.L. Characterization of the spinal adrenergic receptors mediating the spinal effects produced by the microinjection of morphine into the periaqueductal gray. Brain Res. Jun 10;336(1):133–142, 1985. CAPIM, S.L; CARNEIRO, P.H.P; CASTRO, P.C; BARROS, M.R.M; MARINHO, B.G; VASCONCELLOS, M.L.A.A. Design, Prins-cyclization, reaction promoting diastereoselective synthesis of 10 new tetrahydropyran derivatives and in vivo antinociceptive evaluations. European Journal of Medicinal Chemistry. 58:1-11, 2012. CARLTON, S.M.; HARGETT, G.L.; COGGESHALL, R.E. Localization and activation of glutamate receptors in unmyelinated axons of rat glabrous skin. Neurosci. Lett., 197(1), (25-28), 1995. CHUNG, E; BURKE, B; BIEBER, A.J; DOSS, J.C; OHGAMI, Y; QUOCK, R.M. Dynorphinmediated antinociceptive effects of L-arginine and SIN-1 (an NO donor) in mice. Brain Res. Bull. 70: 245–250, 2006. COMMINS, S.P; BORISH, L; STEINKE, J.W. Immunologic messenger molecules: Cytokines, interferons, and chemokines. The Journal of allergy and clinical immunology. 125(2 Suppl 2):S53-72., 2010. COSTANTINO, C. M; GOMES, I.; STOCKTON, S.D; LIM, M.P; DEVI, L.A. Opioid receptor heteromers in analgesia. Expert Rev Mol Med, v. 14, p. e9, 2012. CRUVINEL, W.M e cols. Sistema imunitário: Parte I. Fundamentos da imunidade inata com ênfase nos mecanismos moleculares e celulares da resposta inflamatória. Rev. Bras. Reumatol., São Paulo , v. 50, n. 4, p. 434-447, Aug. 2010. CUNNEEN, J.; CARTWRIGHT, M. The puzzle of sepsis: fitting the pieces of the inflammatory response with treatment. AACN Clinical Issues, v. 15, n. 1, p. 18-44, 2004. CUNHA, T.M; ROMAN-CAMPOS, D; LOTUFO, C.M; DUARTE, H.L; SOUZA, G.R; VERRI, W.A., JR; FUNEZ, M.I; DIAS, Q.M; SCHIVO, I.R; DOMINGUES, A.C; SACHS, D; CHIAVEGATTO, S; TEIXEIRA, M.M; HOTHERSALL, J.S; CRUZ, J.S; CUNHA, F.Q; FERREIRA, S.H. Morphine peripheral analgesia depends on activation of the PI3Kγ/ AKT/nNOS/NO/KATP signaling pathway. Proc. Natl. Acad. Sci. USA 107:4442–4447, 2010. 56 CURY, Y; PICOLO, G; GUTIERREZ, V.P; FERREIRA, S.H. Pain and analgesia: The dual effect of nitric oxide in the nociceptive system. Nitric Oxide 25: 243–254, 2011. D’AMOUR, F.E; SMITH, D.L. A method for determining loss of pain sensation. J. Pharmacol. Exp. Ther., 72: 74–79, 1941. DAWSON, J.; SEDGWICK, A. D.; EDWARDS, J. C.; LEES, P. A comparative study of the cellular, exudative and histological responses to carrageenan, dextran and zymosan in the mouse. International Journal of Tissue Reactions, v. 13, n. 4, p. 171– 185, 1991. DICKENSON, A.H.; SULLIVAN, A.F. Evidence for a role of the NMDA receptor in the frequency dependent potentiaton of deep rat dorsal horn nociceptive neurones following C fibre stimulation. Neuropharmacology, v.26, n. 8, p. 1235-1238, 1987. DI ROSA, M.; GIROUD, J. P.; WILLOUGHBY, D. A. Studies on the mediators of the acute inflammatory response induced in rats in different sites by carrageenan and turpentine. Journal of Pathology, Amsterdam, v. 104, n. 1, p. 15-29, 1971. DUARTE, I.D; LORENZETTI, B.B; FERREIRA, S.H. Peripheral analgesia and activation of the nitric oxide–cyclic GMP pathway. Eur. J. Pharmacol. 186: 289–293, 1990. DUARTE, D.B.; VASKO, M.R.; FEHRENBACHER, J.C. Models of inflammation: carrageenan air pouch. Current protocols in pharmacology. v. 56, p. 561-568, 2012. FEIN, A. Nociceptores: As células que sentem dor. Petrov P, Francischi JN, Ferreira SH, et al. tradutores. Ribeirão Preto – SP: Dor On Line; 2011. 106 p. Disponivel em: http://www.dol.inf.br/nociceptores Acessado em 30 de Março de 2015. FENG, Y.; XIAOZHOU, H.; YANG, Y.; CHAO, D.; LAZARUS, L.H.; XIA, Y. Current research on opioid receptor function. Curr Drug Targets, v. 13, n. 2, p. 230-46, 2012. FELIPINI, R.C. Vídeo de Inflamação Aguda – UNESP. Departamento de Patologia e Propedêutica Clínica, 2013. Disponível em: <https://www.youtube.com/watch?v=gqCIIpHIfqw>. Acesso em: 10 fev.2016. FERREIRA, S.H; VAN ARMAN, C.G. Oedema and increased vascular permeability. In: Vane JR, Van Arman CG, editors. Handbook of experimental pharmacology. New York 7 Springer-Verlag; p. 75-91, 1979. FERREIRA, S.H. A classification of peripheral analgesics based upon their mode of action. Oxford: Oxford University Press, 1990. FERREIRA, A.A; AMARAL, F.A; DUARTE, I.D.G; OLIVEIRA, P.M; ALVES, R.B; SILVEIRA, D; AZEVEDO, A.O; RASLAN, D.S; CASTRO, M.S.A. 57 Antinociceptive effect from Ipomoea cairica extract. J. Ethnopharmacol; 105: 148–53. 2006. FERREIRA, S.H; FERRARI, L.F; CUNHA, T.M; NASCIMENTO, P.G.B.D; JUNIOR, W.A.V; CUNHA, F.Q. Dor: Príncipios e Prática. Capítulo 19: Dor Inflamatória. 2010. Disponível em: < http://dol.inf.br/Html/DorInflamatoria.html>. Acesso em: 2 set. 2016. FISCHER, L.G.; SANTOS, D.; SERAFIN, C.; MALHEIROS, A.; MONACHE, F.D.; MONACHE, G.D.; FILHO, V.C.; SOUZA, M.M. Further Antinociceptive Properties of Extracts and Phenolic Compounds from Plinia glomerata (Myrtaceae) Leaves. Biol. Pharm. Bull. 31(2) 235—239, 2008. FITZGERALD, G.A; RICCIOTTI, E. Prostaglandins and Inflammation. Arterioscler Thromb Vasc Biol.; 31(5): 986–1000, 2011. FÖRSTERMANN, U; SESSA, W.C. Nitric oxide synthases: regulation and function.European Heart Journal 33, 829–837, 2012. FRANCISCHETTI, I; MORENO, J.B; SCHOLZ, M; YOSHIDA, W.B. Leukocytes and the inflammatory response in ischemia-reperfusion injury. Rev Bras Cir Cardiovasc.; 25(4): 575-584, 2010. FRENZEL, L; HERMINE, O. Mast cells and inflammation. Joint Bone Spine 80: 141-145, 2013. GREGORIAN, R.S., JR.; GASIK, A.; KWONG, W.J; VOELLER, S.; KAVANAGHZ, S. Importance of Side Effects in Opioid Treatment: A Trade-Off Analysis With Patients and Physicians. The Journal of Pain, Vol 11, No 11: pp 1095-1108, 2010. GREGORY, N.S; HARRIS, A.L; ROBINSON, C.R; DOUGHERTY, P.M, FUCHS, P.N, SLUKA, K.A. An overview of animal models of pain: disease models and outcome measures. J Pain. doi:10.1016/j.jpain.2013.06.008. November; 14(11), 2013. GRELLNER, W. Time-dependent imunohistochemicaldetection of pro-inflammatory cytokines (IL-1, IL-6, TNF) in human skin wounds. Forens Sci. Int., v. 130, n. 2-3, p. 90-96, 2002. GUGINSKI, G; LUIZ, A.P; SILVA, M.D; MASSARO, M; MARTINS, D.F; CHAVES, J; MATTOS, R.W; SILVEIRA, D; FERREIRA, V.M; CALIXTO, J.B; SANTOS, A.R. Mechanisms involved in the antinociception caused by ethanolic extract obtained from the leaves of Melissa officinalis (lemon balm) in mice. Pharmacol. Biochem. Behav. 93:10–16, 2009. GUILHON, C.C; RAYMUNDO, L.J.R.P; ALVIANO, D.S; BLANK, A.F; ARRIGONI-BLANK, M.F; CAVALCANTI, S.C; ALVIANO, C.S; FERNANDES, P.D. Characterisation of the anti-inflammatory and antinociceptive activities and mechanism of the action of Lippia gracilis essential oil. Journal Ethnopharmacology, v. 135, p. 406-413, 2011. 58 GUTIERREZ, V; ZAMBELLI, V; PICOLO, G; CHACUR, M; SAMPAIO, S; BRIGATTE, P; CURY, Y. Peripheral L-arginine-nitric oxide-cGMP pathway and ATP-sensitive K+ channels are involved in the antinociceptive effect of crotalphine on neuropathic pain in rats. Behav. Pharmacol., (1):14-24, 2012. HALLEGUA, D.S; WEISMAN, M.H. Potential therapeutic uses of interleukin1receptor antagonists in human diseases. Ann Rheum; 61: 960–967. 2002. HAN, J.; KIM, N.; KIM, E.; HO, W.K; EARM, Y.E. Modulation of ATP-sensitive potassium channels by cGMP-dependent protein kinase in rabbit ventricular myocytes. J. Biol. Chem., v.276, n. 25, p. 22140-22147, 2001. HERVERA, A; LEANEZ, S; NEGRETE, R; POL, O. The peripheral administration of a nitric oxide donor potentiates the local antinociceptive effects of a DOR agonist during chronic inflammatory pain in mice. Naunyn Schmiedebergs Arch. Pharmacol 380: 345–352, 2009. HUNSKAAR, S; HOLE, K. The formalin test in mice: dissociation between inflammatory and non-inflammatory pain. Pain 30: 103–14, 1987. JAIN, M.; PARMAR, H.S. Evaluation of antioxidative and anti-inflammatory potential of hesperidin and naringin on the rat air pouch model of inflammation. Inflammation research. v.60, p. 483-491, 2011. JAVANMARDI, K, M; PARVIZ, S.S; SADR, M; KESHAVARZ, B; MINAII; DEHPOUR, A.R. Involvement of N-methyl-D-aspartate receptors and nitric oxide in the rostral ventromedial medulla in modulating morphine pain-inhibitory signals from the periaqueductal grey matter in rats. Clin. Exp. Pharmacol. Physiol. 32: 585–589, 2005. KELLEY, N.E; TEPPER, D.E. Rescue therapy for acute migraine, part 3: opioids, NSAIDs, steroids, and post-discharge medications. Headache.;52(3):467-82, 2012. KOSTER, R; ANDERSON, M; DE BEER, EJ. Acetic acid for analgesic screening. Federation Proceedings. v. 18, p. 412, 1959. LAW, B.K.; WALTNER-LAW, M.E; ENTINGH, A.J.; CHYTIL, A; AAKRE, M.E.; NØRGAARD, P.; MOSES, H.L. Salicylate-induced growth arrest is associated with inhibition of p70s6K and down-regulation of c-myc, cyclin D1, cyclin A, and proliferating cell nuclear antigen. J. Biol. Chem., v.275, n.49, p. 38261-38267, 2000. LE BARS, D., GOZARIU, M., CADDEN, S. Animal models of nociception. Pharmacological Reviews, v. 53, p. 628-651, 2001. LESNIAK, A.; LIPKOWSKI, A. W. Opioid peptides in peripheral pain control. Acta Neurobiol Exp (Wars), v. 71, n. 1, p. 129-38, 2011. 59 LOESER, J. D.; TREEDE, R. D. The Kyoto protocol of IASP Basic Pain Terminology. Pain, v. 137, n. 3, p. 473-477, Jul 31 2008. LOISA, P.; RINNE, T.; LAINE, S.; HURME, M.; KAUKINEN, S. Anti-inflammatory cytokine response and the development of multiple organ failure in severe sepsis. Acta Anaesthesiologica Scandinavica, v. 47, n. 3, p. 319-325, 2003. LORKE, D. A new approach to practical acute toxicity testing. Arch. Toxicol; 54: 275–87. 1983. LOZANO-CUENCA, J.; CASTANEDA-HERNANDEZ, G.; GRANADOS-SOTO, V. Peripheral and spinal mechanisms of antinociceptive action of lumiracoxib. Eur. J. Pharmacol. 513: 81–91, 2005. MARINHO, B.G; MIRANDA, L.S.M.; GOMES, N.M.; MATHEUS, M.E.; LEITÃO, S.G.; VASCONCELLOS, M.L.A.A.; FERNANDES, P.D. Antinociceptive action of (±)-cis-(6-ethyl-tetrahydropyran-2-yl)-formic acid in mice. European Journal of Pharmacology. 550: 47-53, 2006. MARTINS, R.T.; ALMEIDA, D.B.; MONTEIRO, F.M.R.; KOZACS, P.A.; RAMINA, R. Opioid receptors to date. Rev Dor. São Paulo, 13(1):75-9, 2012. MATSUKAWA, A.; HOGABOAM, C.M.; LUKACS, N.W.; LINCOLIN, P.M.; EVANOFF, H.L.; KUNKEL, S.L. Pivotal role of the CC chemokine, macrophage derived chemokine, in the innate immune response. Journal of Immunology, Boston, v.164, p.5362–5368, 2000. MÁZAK, K.; HOSZTAFI, S.; RÁCZ. Á.; NOSZÁL, B. Structural and physicochemical profiling of morphine and related compounds of therapeutic interest. Mini-reviews in Med. Chem., v.9, p. 984-995, 2009. MCNAMARA CR, MANDEL-BREHM J, BAUTISTA DM, SIEMENS J, DERANIAN KL, ZHAO M, HAYWARD NJ, CHONG JA, JULIUS D, MORAN MM, FANGER CM. TRPA1 mediates formalin-induced pain. Proc. Natl. Acad. Sci. U. S. A. ; 104: 1352–3530. 2007. MENEGAZZI, M; DI PAOLA, R; MAZZON, E; GENOVESE, T; CRISAFULLI, C; DAL BOSCO, M; ZOU, Z; SUZAKI, H; CUZZOCREA S. Glycyrrhizin attenuates the development of carrageenan-induced lung injury in mice. Pharmacol Res; 58: 22–31. 2008. MERRER,J.L.; JECKER, J.A.; BEFORT, K.; KIEFFER, B.L. Reward processing by the opioid system in the brain. Physiol Rev. Oct;89(4):1379-412, 2009. MILANO, J; OLIVEIRA, S.M; ROSSATO, M.F; SAUZEM, P.D; MACHADO, P; BECK, P; ZANATTA, N; MARTINS, M.A.P; MELLO, C.F; RUBIN, M.A; FERREIRA, J; BONACORSO, H.G. Antinociceptive effect of novel trihalomethyl-substituted pyrazoline methyl esters in formalin and hot-plate tests in mice. Eur. J. Pharmacol; 581: 86–96. 2008. 60 MILLAN, M.J. The induction of pain: an integrative review. Prog. Neurobiol, v. 57, p. 161-164, 1999. MILLAN, M.J. Descending control of pain. Prog. Neurobiol. 66:355–474, 2002. MILLER, R.J. Presynaptic receptors. Annu Rev Pharmacol Toxicol; 38: 201–27. 1998. MOHAMAD, A. S.; AKHTAR, M. N.; ZAKARIA, Z. A.; PERIMAL, E.K.; KHALID, S.; MOHD, P. A.; KHALID, M. H.; ISRAF, D. A.; LAJIS, N. H.; SULAIMAN, M. R. Antinociceptive activity of a synthetic chalcone, flavokawin B on chemical and thermal models of nociception in mice. European Journal of Pharmacology. v. 647, n. 1 – 3, p. 103 – 109, 2010. NAPIMOGA, C.J.T; PELLEGRINI-DA-SILVA, A; FERREIRA, V.H; NAPIMOGA, M.H; PARADA, C.A; TAMBELI, C.H. Gonadal hormones decrease temporomandibular joint kappa-mediated antinociception through a down-regulation in the expression of kappa opioid receptors in the trigeminal ganglia. Eur. J. Pharmacol. 617: 41–47, 2009. NESS, T.J., GEBHART, G.F. Visceral pain: a review of experimental studies. Pain 41:167–234, 1990. NUGTEREN, D.H; HAZELHOF, E. Isolation and properties of intermediates in prostaglandin biosynthesis. Biochim. Biophys. Acta. 326: 448-461. 1973. OCANA, M; CENDRAN, C.M; COBOS, E.J; ENTRENA, J.M; BAEYENS, J.M. Potassium channels and pain: present realities and future opportunities. Eur J Pharmacol; 500: 203–19. 2004. OLIVEIRA, F.S.; SOUSA, D.P.; ALMEIDA, R.N. Antinociceptive effect of hydroxydihydrocarvone. Biol Pharm Bull 31(4): 588 – 591, 2008. OSSIPOV, M.H.; LAI J, VANDERAH, T.W.; PORRECA, F. Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sciences, v. 73, p.783-800, 2003. PAN, Z.Z; TERSHNER, S.A; FIELDS, H.L. Cellular mechanism for anti-analgesic action of agonists of the kappa-opioid receptor. Nature; 389: 382–5. 1997. PARADA, C.A; TAMBELI, C.H; CUNHA, F.Q; FERREIRA, S.H. The major role of peripheral release of histamine and 5-hydroxytryptamine in formalin-induced nociception. Neuroscience; 102: 937–44. 2001. PARVEEN, Z.; DENG, Y.; SAEED, M.K.; DAI, R.; AHAMAD, W.; YU, Y.H. Antiinflammatory and analgesic activities of Thesium chinese Turcz extracts and its major flavonoids, kaampferol and kaempferol-3-O-glucoside. Yakugaku Zasshi, v.127, p.1275-1279, 2007. 61 RAJA, S.; MEYER, R.A.; RINCKAMP, M.; CAMPBELL, J.N. Peripheral neural mechanism of nociception. In WALL, P.D.; MELZACK, R. (Eds). Textbook of pain. Ediburgh: Churchill Livingstone, p. 11-57, 1999. RAMANA, K.V.; TAMMALI, R.; REDDY, A.B.M.; BHATNAGAR, A.; SRIVASTAVA, S.K.; Aldose Reductase-Regulated Tumor Necrosis Factor- α Production Is Essential for High Glucose-Induced Vascular Smooth Muscle Cell Growth. Endocrinology. 148, n. 9, 4371-4384, 2007. ROBBINS & COTRAN. Patologia: Bases patológicas das doenças. 8ª Edição. Rio de Janeiro: Elsevier, 2010. 1458 p. ROCHA APC, KRAYCHETE DC, LEMONICA L, CARVALHO LR, BARROS GAM, GARCIA JBS, SAKATA RK — Pain: Current Aspects on Peripheral and Central Sensitization. Rev Bras Anestesiol Review article 57: 1: 94-105, 2007. SACHS, D.; CUNHA, F.Q.; FERREIRA, S.H. Peripheral analgesic blockade of hypernociception: activation of arginine/NO/cGMP/protein kinase G/ATP-sensitive K+ channel pathway. Proc. Natl. Acad. Sci. USA, v. 101, n. 10, p. 3680-3685, 2004. SCHRAMM, R; THORLACIUS, H. Neutrophil recruitment in mast cell-dependent inflammation inhibitory mechanisms of glucocorticoids. Inflammation research, v.53 (12), p. 644-652, 2004. SCHULTZ, J.; GROSS, G. Opioids and cardioprotection. Pharmac. Ther., v.89, p. 123-137, 2001. SILVA, J.C; SARAIVA, S.R; OLIVEIRA, R.G; ALMEIDA, J.R. Experimental models for evaluation of antinociceptive activity of natural products: a review. Rev. Bras. Farm. 94 (1): 18-23, 2013. SOARES, A.C; LEITE, R; TATSUO, M.A; DUARTE, I.D. Activation of ATP-sensitive K+ channels: mechanism of peripheral antinociceptive action of the nitric oxide donor, sodium nitroprusside. Eur. J. Pharmacol. 400: 67–71, 2000. SOARES, A.C; DUARTE, I.D. Dibutyryl-cyclic GMP induces peripheral antinociception via activation of ATP-sensitive K+ channels in the rat PGE2-induced hyperalgesic paw, Br. J. Pharmacol. 134: 127–131, 2001. SOJA, P. J.; TAEPAVARAPRUK, N.; PANG, W.; CAIRNS, B. E.; MCERLANE, S. A.; FRAGOSO, M. C. Transmission through the dorsal spinocerebellar and spinoreticular tracts: wakefulness versus thiopental anesthesia. Anesthesiology, v. 97, n. 5, p. 1178-88, 2002. SOMMER, C. Serotonin in Pain and Analgesia - Actions in the Periphery. Molecular Neurobiology, v. 30, p. 117-125, 2004. 62 STAROWICZ, K; OBARA, I; PRZEWLOCKI, R; PRZEWLOCKA, B. Inhibition of morphine tolerance by spinal melanocortin receptor blockade. Pain; 117: 401–11. 2005. STEIN, C.; CLARK, J.D.; VASKO, M.R.; WILCOX, G.L.; OVERLAND, A.C.; VANDERAH, T.W.; SPENCER, R.H. Peripheral mechanisms of pain and analgesia. Brain Res Rev. 2009 Apr;60(1):90-113, 2009. SU, X; JOSHI, S.K; KARDOS, S; GEBHART, G.F. Sodium channel blocking actions of the kappaopioid receptor agonist U50,488 contribute to its visceral antinociceptive effects. J Neurophysiol; 87: 1271–9. 2002. SU, X; CASTLE, N.A; ANTONIO, B; ROELOFFS, R; THOMAS, J.B; KRAFTE, D.S; et al. The effect of kappa-opioid receptor agonists on tetrodotoxin-resistant sodium channels in primary sensory neurons. Anesth Analg; 109: 632–40. 2009. THEOHARIDES, T.C; ALYSANDRATOS, K.D; ANGELIDOU, A; DELIVANIS, D.A; SISMANOPOULOS, N; ZHANG, B; ASADI, S; VASIADI, M; WENG, Z; MINIATI, A; KALOGEROMITROS, D. Mast cells and inflammation. Biochimica et Biophysica Acta 1822, 21–33, 2012. THOMAZZI, S. M.; SILVA, C. B.; SILVEIRA, D. C. R.; VASCONCELLOS, C. L. C.; LIRA, A. F.; CAMBUI, E. V. F.; ESTEVAM, C. S.; ANTONIOLLI, A. R. Antinociceptive and anti-inflammatory activities of Bowdichia virgilioides (sucupira). Journal of Ethnopharmacology, v. 127, p. 451 - 456, 2010. TORNOS, M.P; SÁENZ, M.T; GARCIA, M.D; FERNÁNDEZ, M.A. Antinociceptive effects of the tubercles of Anredera leptostachys. J. Ethnopharmacol; .68: 229–34. 1999. VANDERAH, T.W. Delta and kappa opioid receptors as suitable drug targets for pain. Clin J Pain; 26: 10–5. 2010. VARGA, E.V.; YAMAMURA, H.I; RUBENZIK, M.K.; STROPOVA, D.; NAVRATILOVA, E.; ROESKE, W.R. Molecular mechanisms of excitatory signaling upon chronic opioid agonist treatment. Life Sciences, v. 74, p. 299-311, 2003. VERRI, W.A; CUNHA, T.M; PARADA, C.A; POOLE, S; CUNHA, F.Q; FERREIRA, S.H. Hypernociceptive role of cytokines and chemokines: targets for analgesic drug development? Pharmacol Ther. Oct;112(1):116-38, 2006. VIGIL, S.V.G; DE LIZ, R.; MEDEIROS, Y.S.; FRÖDETS. Efficacy of tacrolimus in inhibiting inflammation caused by carrageenan in a murine model of air pouch. Transpl Immunol; 19:25-29, 2008. VOSCOPOULOS, C.; LEMA, M.; When does acute pain become chronic? Br J Anaesth, v. 105 Suppl 1, p. i69-i85. ISSN 0007-0912, 2010. 63 WINTER, C. A.; RISLEY, E. A.; NUSS, G. W. Carrageenin-induced edema in hind paw of the ratas an assay for anti-inflammatory drugs. Proc Soc Exp Biol Med, v. 111, p. 544-547, 1962. ZHOU, S.; BONASERA, L.; CARLTON, S.M. Peripheral administration of NMDA, AMPA or KA results in pain behaviors in rats. Neuroreport, 7(4), 895-900.), 1996. ZHU, Z. Z.; MA, K. J.; RAN, X.; ZHANG, H.; ZHENG, C. J.; HAN, T.; ZHANG, Q. Y.; QIN, L. P. Analgesic, anti-inflammatory and antipyretic activities of the petroleum ether fraction from the ethanol extract of Desmodium podocarpum. Journal of Ethnopharmacology. v. 133, n. 3, p. 1126 – 1131, 2011
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Universidade Federal Rural do Rio de Janeiro
dc.publisher.program.fl_str_mv Programa de Pós-Graduação em Ciências Fisiológicas
dc.publisher.initials.fl_str_mv UFRRJ
dc.publisher.country.fl_str_mv Brasil
dc.publisher.department.fl_str_mv Instituto de Ciências Biológicas e da Saúde
publisher.none.fl_str_mv Universidade Federal Rural do Rio de Janeiro
dc.source.none.fl_str_mv reponame:Biblioteca Digital de Teses e Dissertações da UFRRJ
instname:Universidade Federal Rural do Rio de Janeiro (UFRRJ)
instacron:UFRRJ
instname_str Universidade Federal Rural do Rio de Janeiro (UFRRJ)
instacron_str UFRRJ
institution UFRRJ
reponame_str Biblioteca Digital de Teses e Dissertações da UFRRJ
collection Biblioteca Digital de Teses e Dissertações da UFRRJ
bitstream.url.fl_str_mv https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/1/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf.jpg
https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/2/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf.txt
https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/3/2016%20-%20Gabriela%20Mastrangelo%20Gon%c3%a7alves.pdf
https://rima.ufrrj.br/jspui/bitstream/20.500.14407/11405/4/license.txt
bitstream.checksum.fl_str_mv cc73c4c239a4c332d642ba1e7c7a9fb2
b56ec69846bfd4203c717ef0dbb08471
d31754b24278f68ec75382b5ff3932c0
7b5ba3d2445355f386edab96125d42b7
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
MD5
repository.name.fl_str_mv Biblioteca Digital de Teses e Dissertações da UFRRJ - Universidade Federal Rural do Rio de Janeiro (UFRRJ)
repository.mail.fl_str_mv bibliot@ufrrj.br||bibliot@ufrrj.br
_version_ 1810108201982492672