Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Brain Res Bull ; 135: 157-162, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29054696

RESUMO

Since a significant proportion of diabetic patients have clinical or subclinical neuropathy, there may be concerns about the use of local anaesthetics. The present study was designed to determine and compare the effects of articaine, a widely used anaesthetic in dental practice, and lidocaine on the resting and axonal stimulation-evoked release of [3H]noradrenaline ([3H]NA) in prefrontal cortex slices and the release of [3H]NA in spinal cord slices prepared from non-diabetic and streptozocin (STZ)-induced diabetic (glucose level=22.03±2.31mmol/l) rats. The peak of allodynia was achieved 9 weeks after STZ-treatment. Articaine and lidocaine inhibited the stimulation-evoked release in a concentration-dependent manner and increased the resting release by two to six times. These effects indicate an inhibitory action of these anaesthetics on Na+- and K+-channels. There was no difference in clinically important nerve conduction between non-diabetic and diabetic rats, as measured by the release of transmitter in response to axonal stimulation. The uptake and resting release of NA was significantly higher in the brain slices prepared from diabetic rats, but there were no differences in the spinal cord. For the adverse effects, the effects of articaine on K+ channels (resting release) are more pronounced compared to lidocaine. In this respect, articaine has a thiophene ring with high lipid solubility, which may present potential risks for some patients.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Carticaína/farmacologia , Norepinefrina/fisiologia , Anestesia Local , Animais , Axônios/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Carticaína/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Lidocaína/metabolismo , Lidocaína/farmacologia , Masculino , Condução Nervosa/efeitos dos fármacos , Norepinefrina/metabolismo , Córtex Olfatório/citologia , Ratos , Ratos Wistar , Medula Espinal/citologia , Estreptozocina/farmacologia
2.
Brain Res Bull ; 90: 66-71, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22995282

RESUMO

Since the discovery of opioid receptor dimers their possible roles in opioid actions were intensively investigated. Here we suggest a mechanism that may involve the µ-δ opioid heterodimers. The exact role of δ opioid receptors in antinociception and in the development of opioid tolerance is still unclear. While receptor up-regulation can be observed during the development of opioid tolerance no µ receptor down-regulation could be detected within five days. In our present work we investigated how the selective δ opioid receptor agonists and antagonists influence the antinociceptive effect of the selective µ receptor agonist DAMGO in naïve and morphine-tolerant mice. We treated male NMRI mice with 200 µmol/kg subcutaneous (s.c.) morphine twice daily for three days. On the fourth day we measured the antinociceptive effect of DAMGO alone and combined with delta ligands: DPDPE, deltorphin II (agonists), TIPP and TICPψ (antagonists), respectively, administered intrathecally (i.t.) in mouse tail-flick test. In naive control mice none of the δ ligands caused significant changes in the antinociceptive action of DAMGO. The treatment with s.c. morphine resulted in approximately four-fold tolerance to i.t. DAMGO, i.e. the ED50 value of DAMGO was four times as high as in naive mice. 500 and 1000 pmol/mouse of the δ1 selective agonist DPDPE enhanced the tolerance to DAMGO while 1000 pmol/mouse of the δ2 selective agonist deltorphin II did not influence the degree of tolerance. However, both δ antagonists TIPP and TICPψ potentiated the antinociceptive effect of i.t. DAMGO, thus they restored the potency of DAMGO to the control level. The inhibitory action of DPDPE against the antinociceptive effect of DAMGO could be antagonized by TIPP and TICPψ. We hypothesize that during the development of morphine tolerance the formation of µÎ´ heterodimers may contribute to the spinal opioid tolerance. δ ligands may affect the dimer formation differently. Those, like DPDPE may facilitate the dimer formation hence inhibit the antinociceptive effect of DAMGO by causing virtual µ receptor down-regulation. Ligands that do not affect the dimer formation do not influence antinociception either but ligands with the presumed capability of disconnecting the dimers may decrease the spinal tolerance to DAMGO.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Morfina/farmacologia , Receptores Opioides delta/metabolismo , Medula Espinal/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Esquema de Medicação , Interações Medicamentosas , Ligantes , Masculino , Camundongos , Medição da Dor/efeitos dos fármacos , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Medula Espinal/efeitos dos fármacos , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/patologia
3.
Curr Med Chem ; 19(27): 4699-707, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22934777

RESUMO

The aims of this study were to synthesize 14-O-Methylmorphine-6-O-sulfate (14-O-MeM6SU) and examine its opioid properties (potency, affinity, efficacy) in receptor ligand binding and isolated tissues (mouse vas deferens, MVD and rat vas deferens, RVD bioassays). The results were then compared to the parent compounds morphine-6-O-sulfate (M6SU) and morphine, as well as the �- opioid receptor (MOR) selective agonist peptide [D-Ala2,N-Me-Phe4,Gly-ol5]enkephalin (DAMGO). An additional objective was to compare the effect of subcutaneously (s.c.) or intracerebroventricularly (i.c.v.) administered 14-O-MeM6SU, M6SU and morphine in thermal nociception, rat tail-flick (RTF) test. In MVD, the EC50 (nM) value was 4.38 for 14-O-MeM6SU, 102.81 for M6SU, 346.63 for morphine and 238.47 for DAMGO. The effect of 14-O-MeM6SU and DAMGO was antagonized by naloxone (NAL) with Ke value 1-2.00 nM. The Emax values (%) were 99.10, 36.87, 42.51 and 96.99 for 14-O-MeM6SU, M6SU, morphine and DAMGO, respectively. In RVD 14-O-MeM6SU and DAMGO but not M6SU or morphine showed agonist activity. In binding experiments the affinity of 14-OMeM6SU, M6SU, morphine and DAMGO for MOR was 1.12, 11.48, 4.37 and 3.24 nM, respectively. The selectivity of 14-O-MeM6SU was κ/µ= 269 and δ/µ= 9. In G-protein activation experiments, 14-O-MeM6SU and DAMGO showed higher Emax values than M6SU or morphine. S.c. or i.c.v-injected 14-O-MeM6SU, M6SU and morphine produced a dose and time-dependent increase in RTF response latency. 14-O-MeM6SU was the most potent. Our results showed that introduction of 14-O-Me in M6SU increased the binding affinity, agonist potency, and most importantly, the intrinsic efficacy (Emax).


Assuntos
Codeína/análogos & derivados , Ligantes , Receptores Opioides mu/agonistas , Analgésicos/química , Analgésicos/farmacologia , Animais , Codeína/síntese química , Codeína/química , Codeína/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Cobaias , Técnicas In Vitro , Cinética , Masculino , Camundongos , Morfina/farmacologia , Derivados da Morfina/farmacologia , Contração Muscular/efeitos dos fármacos , Ligação Proteica , Ratos , Ratos Wistar , Receptores Opioides mu/metabolismo , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/fisiologia
4.
Eur J Pain ; 16(5): 690-705, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22337491

RESUMO

The contribution of supraspinal, spinal or peripheral mu-opioid receptors (MORs) to the overall antinociception of systemic centrally penetrating versus peripherally restricted opioids has not been thoroughly investigated. Therefore, we examined paw pressure thresholds in Wistar rats with complete Freund's adjuvant hindpaw inflammation following different doses of intraplantar (i.pl.) as well as intravenous (i.v.) fentanyl (6.25-50 µg/kg), morphine (1-7.5 mg/kg) or loperamide (1-7.5 mg/kg). Antagonism of the i.v. mu-opioid agonists by intracerebroventricular (i.c.v.), intrathecal (i.t.) or i.pl. naloxone-methiodide (NLXM) revealed the relative contributions of supraspinal, spinal and peripheral MOR to the overall antinociceptive effects. In parallel, the MOR density at these three levels of pain transmission was assessed by radioligand binding. Antinociceptive effects of i.v. fentanyl and morphine, but not of the peripherally restricted loperamide were two- to threefold greater and longer lasting compared with their i.pl. administration. I.c.v. but not i.pl. NLXM significantly antagonized fentanyl's and morphine's antinociception by 70-80%, whereas i.t. NLXM reduced it by 20-30%. In contrast, antinociception of i.v. loperamide was abolished by i.pl. but not by i.c.v. or i.t. NLXM. In parallel, a respective 32- and sixfold higher MOR density in supraspinal and spinal versus peripheral sensory neurons was detected. In conclusion, in comparison with supraspinal and spinal opioid receptors, peripheral opioid receptors do not significantly contribute to the antinociception of systemic fentanyl and morphine during inflammatory pain. Antinociception of their i.v. administration was superior over both i.v and i.pl. loperamide, acting exclusively via peripheral MOR. These findings may guide the future development of novel peripherally restricted opioids.


Assuntos
Analgésicos Opioides/uso terapêutico , Fentanila/uso terapêutico , Loperamida/uso terapêutico , Morfina/uso terapêutico , Dor/tratamento farmacológico , Receptores Opioides/metabolismo , Medula Espinal/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Fentanila/administração & dosagem , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Loperamida/administração & dosagem , Masculino , Morfina/administração & dosagem , Dor/metabolismo , Ratos , Ratos Wistar , Células Receptoras Sensoriais , Medula Espinal/efeitos dos fármacos
5.
Neurochem Int ; 58(6): 708-13, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21333706

RESUMO

The role of α(2)-adrenoceptors in regulation of gastric motility has been well documented. However, only few data are available on the adrenoceptor subtype that mediates this effect. The purpose of the present work was to identify the α(2)-adrenoceptor subtype(s) responsible for the inhibition of gastric motor activity in isolated fundus strip of the mouse. It was shown that (i) the electrically evoked contraction of the gastric fundus strip of the mouse was inhibited by the non-selective α(2)-adrenoceptor stimulant clonidine (EC(50): 0.019±0.001µM), the α(2A)-adrenoceptor subtype selective agonist oxymetazoline (EC(50): 0.004±0.001µM) and the α(2B)-adrenoceptor subtype preferring ST-91 (EC(50): 0.029±0.004µM), (ii) the inhibitory effect of clonidine (1µM), oxymetazoline (0.1µM) and ST-91 (1µM) on the contractions of gastric fundus strip was reversed by the non-selective α(2)-adrenoceptor antagonist idazoxan and α(2A)-adrenoceptor antagonist BRL 44408, but not by the α(2B/2C)-adrenoceptor antagonist ARC-239. (iii) Clonidine and ST-91 inhibited the electrically induced gastric contractions in C57BL/6 wild type mice as well as in α(2B)- and α(2C)-adrenoceptor deficient mice in a concentration-dependent manner; however, neither of them was effective in α(2A)-deficient mice. As a conclusion, it was first demonstrated that the inhibitory effect of α(2)-adrenoceptor agonists on the gastric motor activity of isolated stomach strip of the mouse is mediated purely by α(2A)-adrenoceptors.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Atividade Motora/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Estômago/efeitos dos fármacos , Animais , Clonidina/análogos & derivados , Clonidina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Estômago/fisiologia
6.
Inflammopharmacology ; 17(3): 171-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19526313

RESUMO

Our previous findings suggest that alpha(2)-adrenoceptor stimulants induce gastroprotective action, the effect is likely to be mediated by alpha(2B)-adrenoceptor subtype. Clonidine (0.094 micromol/kg p.o.) and rilmenidine (0.014 micromol/kg p.o.) in gastroprotective dose range, as well as ST-91 (2.2 micromol/kg p.o.), a clonidine analogue showing higher affinity to alpha(2B)-adrenoceptor subtype than to alpha(2A)-one, inhibited the carrageenan-induced hyperalgesia in Randall-Selitto test, the antinociceptive action was reversed by yohimbine (5 micromol/kg s.c.) and the alpha(2B)-adrenoceptor antagonist prazosin (0.24 micromol/kg i.p.). Similarly, clonidine and rilmenidine in the same dose range reduced the oedema formation induced by carrageenan, yohimbine and the alpha(2A)-adrenoceptor antagonist BRL-44408 (3 micromol/kg i.p.) inhibited the anti-inflammatory effect; however, prazosin failed to affect it. These results suggest that alpha(2B/C)-like adrenoceptor subtype may be involved in the antihyperalgesic action, but not in the antiphlogistic effect of alpha(2)-adrenoceptor stimulants. The later effect may be mediated by alpha(2A)-like adrenoceptor subtype.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2 , Analgésicos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Antiulcerosos/uso terapêutico , Edema/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Úlcera Gástrica/tratamento farmacológico , Antagonistas de Receptores Adrenérgicos alfa 2 , Agonistas alfa-Adrenérgicos/uso terapêutico , Animais , Clonidina/análogos & derivados , Clonidina/uso terapêutico , Modelos Animais de Doenças , Edema/induzido quimicamente , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Hiperalgesia/induzido quimicamente , Masculino , Oxazóis/uso terapêutico , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 2/fisiologia , Rilmenidina , Úlcera Gástrica/induzido quimicamente
7.
Biochem Biophys Res Commun ; 290(1): 153-61, 2002 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11779147

RESUMO

Side chain modifications were introduced to endomorphin 2 (E2) to improve its binding properties and biological activity. A number of C-terminal modifications decreased the binding affinity to the mu-opioid receptor and the intrinsic activity in rat brain membranes. The exception was E2-ol, which showed increased binding affinity to MOR and higher potency in stimulating [(35)S]GTPgammaS binding. N-methylation of Phe(3) (MePhe(3)) attenuated the binding affinity and produced a rightward shift of [(35)S]GTPgammaS binding curves. All derivatives had lower intrinsic activity than E2. Some of the modified peptides partially inhibited, while YPF-benzyl-allyl-amide fully inhibited, the E2 or [d-Ala(2),MePhe(4),Gly(5)ol]enkephalin stimulated [(35)S]GTPgammaS binding. Marked differences were found between the results obtained using tritiated E2, tritiated naloxone, and [(35)S]GTPgammaS binding, indicating the possible involvement of multiple binding sites. The data presented demonstrate that the C-terminal amide group has an essential role in the regulation of the binding and the agonist/antagonist properties of E2.


Assuntos
Oligopeptídeos/química , Oligopeptídeos/metabolismo , Animais , Sítios de Ligação , Encéfalo/metabolismo , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Cinética , Ligantes , Modelos Químicos , Mutagênese Sítio-Dirigida , Naloxona/farmacologia , Peptídeos/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Receptores Opioides mu/química
8.
Eur J Pharmacol ; 421(1): 61-7, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11408050

RESUMO

The opioid properties of endomorphin derivatives containing a C-terminal alcoholic(-ol) function were compared to the parent amidated compounds in isolated organs (longitudinal muscle strip of guinea-pig ileum and mouse vas deferens). Similar data were also generated for the mu-opioid receptor selective agonist synthetic peptide (D-Ala2, MePhe4, Gly5-ol)-enkephalin (DAMGO) and its Gly5-NH2 congener (DAMGA). Endomorphin-1-ol (Tyr-Pro-Trp-Phe-ol) had an IC50 of 80.6 nM in mouse vas deferens and 61.2 nM in guinea-pig ileum; the corresponding values for endomorphin-2-ol (Tyr-Pro-Phe-Phe-ol) were 49.6 and 48.2 nM, for DAMGO 59.8 and 29.2 nM, respectively. As it was indicated by the antagonism by naltrexone, the agonist actions were exerted exclusively at mu-opioid receptors in both organs. The -ol derivatives were slightly (2.3-4.3 times) less potent than the parent amides in the bioassays: all peptides had, apparently, full agonist properties in intact preparations. With the aim of revealing potential partial agonist properties among the investigated peptides, we partially inactivated the mu-opioid receptor pool in mouse vas deferens by 5x10(-7) M beta-funaltrexamine. The calculated receptor constants indicated a "high-affinity, low intrinsic efficacy" profile (i.e. a potential partial agonist property) for endomorphin-1, an intermediate character for endomorpin-1-ol and full agonism for DAMGA and DAMGO. Apparently, a higher receptor fraction remained accessible for endomorphin-1 (42.8%) than for the -ol congener (14.0%), DAMGO (20.2%) and DAMGA (14.1%) after partial inactivation.


Assuntos
Oligopeptídeos/metabolismo , Receptores Opioides mu/metabolismo , Ducto Deferente/metabolismo , Analgésicos Opioides/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Cobaias , Técnicas In Vitro , Masculino , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Receptores Opioides mu/agonistas , Ducto Deferente/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA