Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Reprod Biol Endocrinol ; 14(1): 33, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27301276

RESUMO

BACKGROUND: The adrenergic system and progesterone play major roles in the control of the uterine function. Our aims were to clarify the changes in function and expression of the α2-adrenergic receptor (AR) subtypes after progesterone pretreatment in late pregnancy. METHODS: Sprague Dawley rats from pregnancy day 15 were treated with progesterone for 7 days. The myometrial expressions of the α2-AR subtypes were determined by RT-PCR and Western blot analysis. In vitro contractions were stimulated with (-)-noradrenaline, and its effect was modified with the selective antagonists BRL 44408 (α2A), ARC 239 (α2B/C) and spiroxatrine (α2A). The accumulation of myometrial cAMP was also measured. The activated G-protein level was investigated via GTPγS binding assays. RESULTS: Progesterone pretreatment decreased the contractile effect of (-)-noradrenaline through the α2-ARs. The most significant reduction was found through the α2B-ARs. The mRNA of all of the α2-AR subtypes was increased. Progesterone pretreatment increased the myometrial cAMP level in the presence of BRL 44408 (p < 0.001), spiroxatrine (p < 0.001) or the spiroxatrine + BRL 44408 combination (p < 0.05). Progesterone pretreatment increased the G-protein-activating effect of (-)-noradrenaline in the presence of the spiroxatrine + BRL 44408 combination. CONCLUSIONS: The expression of the α2-AR subtypes is progesterone-sensitive. It decreases the contractile response of (-)-noradrenaline through the α2B-AR subtype, blocks the function of α2A-AR subtype and alters the G protein coupling of these receptors, promoting a Gs-dependent pathway. A combination of α2C-AR agonists and α2B-AR antagonists with progesterone could be considered for the treatment or prevention of preterm birth.


Assuntos
Miométrio/efeitos dos fármacos , Progesterona/farmacologia , Receptores Adrenérgicos alfa 2/metabolismo , Contração Uterina/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Animais , AMP Cíclico/metabolismo , Feminino , Imidazóis/farmacologia , Isoindóis/farmacologia , Miométrio/metabolismo , Norepinefrina/farmacologia , Gravidez , Ratos , Ratos Sprague-Dawley
2.
Croat Med J ; 57(2): 100-9, 2016 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-27106352

RESUMO

AIM: To assess the effect of 17ß-estradiol pretreatment on the function and expression of α2- adrenergic receptors (ARs) subtypes in late pregnancy in rats. METHODS: Sprague-Dawley rats (n=37) were treated with 17ß-estradiol for 4 days starting from the 18th day of pregnancy. The myometrial expression of the α2-AR subtypes was determined by real time polymerase chain reaction and Western blot analysis. In vitro contractions were stimulated with (-)-noradrenaline, and its effect was modified with the selective antagonists BRL 44408 (α2A), ARC 239 (α2B/C), and spiroxatrine (α2A). The cyclic adenosine monophosphate (cAMP) accumulation was also measured. The activated G-protein level was investigated by guanosine 5'-O-[gamma-thio]triphosphate (GTPγS) binding assay. RESULTS: 17ß-estradiol pretreatment decreased the contractile effect of (-)-noradrenaline via the α2-ARs, and abolished the contractile effect via the α2B-ARs. All the α2-AR subtypes' mRNA was significantly decreased. 17ß-estradiol pretreatment significantly increased the myometrial cAMP level in the presence of BRL 44408 (P=0.001), ARC 239 (P=0.007), and spiroxatrine (P=0.045), but did not modify it in the presence of spiroxatrine + BRL 44408 combination (P=0.073). It also inhibited the G-protein-activating effect of (-)-noradrenaline by 25% in the presence of BRL 44408 + spiroxatrine combination. CONCLUSIONS: The expression of the α2-AR subtypes is sensitive to 17ß-estradiol, which decreases the contractile response of (-)-noradrenaline via the α2B-AR subtype, and might cause changes in G-protein signaling pathway. Estrogen dysregulation may be responsible for preterm labor or uterine inertia via the α2-ARs.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Estradiol/farmacologia , Trabalho de Parto Prematuro/fisiopatologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Animais , Feminino , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Contração Uterina/efeitos dos fármacos
3.
Anesth Analg ; 121(6): 1488-94, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26465932

RESUMO

BACKGROUND: Hemopressin, so-called because of its hypotensive effect, belongs to the derivatives of the hemoglobin α-chain. It was isolated from rat brain membrane homogenate by the use of catalytically inactive forms of endopeptidase 24.15 and neurolysin. Hemopressin has antihyperalgesic features that cannot be prevented by the opioid receptor antagonist, naloxone. METHODS: In the present study, we investigated whether hemopressin (PVNFKFLSH) and its C-terminally truncated fragment hemopressin 1-7 (PVNFKFL) have any influence on opioid-dependent signaling. Peptides have been analyzed using G-protein-stimulating functional and receptor bindings in this experimental setup. RESULTS: These 2 compounds efficiently activated the G-proteins, and naloxone slightly blocked this stimulation. At the same time, they were able to displace radiolabeled [3H]DAMGO, a selective ligand for µ-opioid system, at micromolar concentrations. Displacement caused by the heptapeptide was more modest compared with hemopressin. Experiments performed on cell lines overexpressing µ-opioid receptors verified the opioid activity of both hemopressins. Moreover, the CB1 cannabinoid receptor antagonist, AM251, significantly decreased their G-protein stimulatory effect. CONCLUSIONS: Here, we further confirm that hemopressins can modulate CB1 receptors and can have a slight modulatory effect on the opioid system.


Assuntos
Canabinoides/metabolismo , Hemoglobinas/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores Opioides mu/metabolismo , Animais , Células CHO , Canabinoides/farmacologia , Cricetinae , Cricetulus , Cobaias , Hemoglobinas/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/fisiologia , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas
4.
Mol Divers ; 17(1): 19-31, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23124951

RESUMO

This study reports on new pharmacologically active endomorphin-2 analogues, incorporating ß(2)-hPhe, ß(3)-hPhe and ß(3)-hTic unnatural amino acids in the place of the Phe(3)-Phe(4)residues. Such α, ß-hybrid analogues were designed to exploit the great potential of ß-amino acids in generating conformational variation at the key positions 3 and 4, with the aim of evaluating the effect on the opioid binding affinity. Ligand-stimulated binding assays indicated that some analogues retained a significant affinity, especially for the δ receptor. (1)H NMR and molecular modelling suggested the predominance of bent structures for all compounds. The molecular docking with the µ-opioid receptor model was also performed, highlighting a common binding mode for active compounds and helping to rationalize the observed structure-activity data.


Assuntos
Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Aminoácidos/química , Modelos Moleculares , Conformação Molecular , Simulação de Acoplamento Molecular , Mimetismo Molecular , Ressonância Magnética Nuclear Biomolecular , Oligopeptídeos/química , Receptores Opioides/metabolismo , Relação Estrutura-Atividade
5.
Neurobiol Dis ; 45(1): 499-507, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21964255

RESUMO

Experiments were designed to evaluate different variables of the dopaminergic system in the temporal cortex of surgically treated patients with temporal lobe epilepsy (TLE) associated with mesial sclerosis (MTLE, n=12) or with cerebral tumor or lesion (n=8). In addition, we sought to identify dopaminergic abnormalities in those patients with epilepsy that had comorbid anxiety and depression. Specifically, we investigated changes in dopamine and its metabolites, D1 and D2 receptors, tyrosine hydroxylase (TH) and dopamine transporter. Results obtained from patients with epilepsy were compared with those found in experiments using autopsy material. The neocortex of patients with MTLE demonstrated high D1 expression (1680%, p<0.05) and binding (layers I-II, 31%, p<0.05; layers V-VI, 28%, p<0.05), and decreased D2 expression (77%, p<0.05). The neocortex of patients with TLE secondary to cerebral tumor or lesion showed high expression of D1 receptors (1100%, p<0.05), and D2-like induced activation of G proteins (layers I-II, 503%; layers III-IV, 557%; layers V-VI, 964%, p<0.05). Both epileptic groups presented elevated binding to the dopamine transporter and low tissue content of dopamine and its metabolites. Analysis revealed the following correlations: a) D1 receptor binding correlated negatively with seizure onset age and seizure frequency, and positively with duration of epilepsy; b) D2 receptor binding correlated positively with age of seizure onset and negatively with duration of epilepsy; c) dopamine transporter binding correlated positively with duration of epilepsy and frequency of seizures; d) D2-like induced activation of G proteins correlated positively with the age of patients. When compared with autopsies and patients with anxiety and depression, patients without neuropsychiatric disorders showed high D2-like induced activation of G proteins, an effect that correlated positively with age of patient and seizure onset age, and negatively with duration of epilepsy. The present study suggests that alterations of the dopaminergic system result from epileptic activity and could be involved in the physiopathology of TLE and the comorbid anxiety and depression.


Assuntos
Dopamina/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Neocórtex/metabolismo , Neurônios/metabolismo , Lobo Temporal/metabolismo , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Adulto , Feminino , Ácido Homovanílico/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Hippocampus ; 22(2): 122-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21049484

RESUMO

Mu opioid receptors (MOR) are known to be involved in seizure activity. The main goal of the present study was to characterize the MOR mRNA expression, binding, as well as G protein activation mediated by these receptors in epileptic hippocampus of patients with pharmacoresistant mesial temporal lobe epilepsy (TLE). In contrast with autopsy samples, hippocampus obtained from patients with mesial TLE demonstrated enhanced MOR mRNA expression (116%). Saturation binding experiments revealed significantly higher (60%) B(max) values for the mesial TLE group, whereas the K(d) values were not statistically different. Although mesial TLE group demonstrated high levels of basal binding for the G proteins (136%), DAMGO-stimulated [(35)S]GTPγS binding did not demonstrate significant alterations. In conclusion, our present data provide strong evidence that the epileptic hippocampus of patients with pharmacoresistant mesial TLE presents significant alterations in MOR. Such changes may represent adaptive mechanisms to compensate for other as yet unknown alterations.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Hipocampo/metabolismo , RNA Mensageiro/análise , Receptores Opioides mu/metabolismo , Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
7.
J Neural Transm (Vienna) ; 117(4): 421-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20213428

RESUMO

Besides generally accepted lower analgesic potencies of opioids in neuropathic pain, our recent pharmacological reports have demonstrated that the effectiveness of the micro-opioid receptor (MOR) agonists in neuropathy might depends upon the chemical/structural property of these compounds (alkaloid vs. peptides). Such findings prompted us to investigate the changes in MOR mRNA expression (estimated by PCR) as well as MOR functional activity (examined by [(35)S]GTPgammaS binding) in the dorsal horn of the spinal cord and the dorsal root ganglia (DRG) of neuropathic rats at different time points after sciatic nerve ligation. We found that the spinal MOR mRNA level and agonist-stimulated [(35)S]GTPgammaS binding were not affected by nerve injury. In contrast, down-regulation of MOR mRNA in the ipsilateral side of DRG developed 3 (approximately 63% reduction) and 14 (approximately 89% reduction) days after the ligation. The decrease was paralleled with pronounced reduction in the stimulation of [(35)S]GTPgammaS binding by morphine (approximately 37-39%). Thus, neuropathy-induced specific dysfunction of MOR to activate G-protein together with changes in the MOR synthesis might be related, at least in part, to diminish analgesic efficacy of morphine in neuropathic pain. Interesting observations from current studies are linked to endomorphins (EMs), which do not affect the G protein stimulation of MOR after nerve ligation. This intriguing property of EMs, together with previously reported high analgesic efficacy of these compounds indicate that chemically/structurally different MOR agonists, particularly morphine versus EMs, may differentially interact with receptors causing distinct pharmacological effects in chronic pain.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Gânglios Espinais/metabolismo , Neuralgia/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacologia , Animais , Lateralidade Funcional , Gânglios Espinais/efeitos dos fármacos , Masculino , Morfina/farmacologia , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Nervo Isquiático/lesões , Neuropatia Ciática/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Fatores de Tempo
8.
Neurochem Int ; 138: 104752, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32445659

RESUMO

Kynurenic acid (KYNA) is one of the most significant metabolite of the kynurenine pathway both in terms of functional and potential therapeutic value. It is an N-methyl-D-aspartate (NMDA) receptor antagonist, but it can also activate the G-protein coupled receptor 35 (GPR35), which shares several structural and functional properties with cannabinoid receptors. Previously our group demonstrated that systemic chronic KYNA treatment altered opioid receptor G-protein activity. Opioid receptors also overlap in many features with cannabinoid receptors. Thus, our aim was to examine the direct in vitro and systemic, chronic in vivo effect of KYNA on type 1 cannabinoid receptor (CB1R) binding and G-protein activity. Based on competition and [35S]GTPγS G-protein binding assays in rat brain, KYNA alone did not show significant binding towards the CB1R, nor did it alter CB1R ligand binding and agonist activity in vitro. When rats were chronically treated with KYNA (single daily, i.p., 128 mg/kg for 9 days), the KYNA plasma and cerebrospinal fluid levels significantly increased compared to vehicle treated group. Furthermore, in G-protein binding assays, in the whole brain the amount of G-proteins in basal and in maximum activity coupled to the CB1R also increased due to the treatment. At the same time, the overall stimulatory properties of the receptor remained unaltered in vehicle and KYNA treated samples. Similar observations were made in rat hippocampus, but not in the cortex and brainstem. In saturation binding assays the density of CB1Rs in rat whole brain and hippocampus were also significantly enhanced after the same treatment, without significantly affecting ligand binding affinity. Thus, KYNA indirectly and brain region specifically increases the abundance of functional CB1Rs, without modifying the overall binding and activity of the receptor. Supposedly, this can be a compensatory mechanism on the part of the endocannabinoid system induced by the long-term KYNA exposure.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ácido Cinurênico/administração & dosagem , Ácido Cinurênico/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Animais , Benzoxazinas/metabolismo , Benzoxazinas/farmacologia , Bloqueadores dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/metabolismo , Masculino , Morfolinas/metabolismo , Morfolinas/farmacologia , Naftalenos/metabolismo , Naftalenos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley
9.
Neurobiol Dis ; 35(3): 466-73, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19573600

RESUMO

There is no information concerning signal transduction mechanisms downstream of the opioid/nociceptin receptors in the human epileptic brain. The aim of this work was to evaluate the level of G-proteins activation mediated by DAMGO (a mu receptor selective peptide) and nociceptin, and the binding to mu and nociceptin (NOP) receptors and adenylyl cyclase (AC) in neocortex of patients with pharmacoresistant temporal lobe epilepsy. Patients with temporal lobe epilepsy associated with mesial sclerosis (MTLE) or secondary to tumor or vascular lesion showed enhanced [3H]DAMGO and [3H]forskolin binding, lower DAMGO-stimulated [35S]GTPgammaS binding and no significant changes in nociceptin-stimulated G-protein. [3H]Nociceptin binding was lower in patients with MTLE. Age of seizure onset correlated positively with [3H]DAMGO binding and DAMGO-stimulated [35S]GTPgammaS binding, whereas epilepsy duration correlated negatively with [3H]DAMGO and [3H]nociceptin binding, and positively with [3H]forskolin binding. In conclusion, our present data obtained from neocortex of epileptic patients provide strong evidence that a) temporal lobe epilepsy is associated with alterations in mu opioid and NOP receptor binding and signal transduction mechanisms downstream of these receptors, and b) clinical aspects may play an important role on these receptor changes.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Neocórtex/metabolismo , Receptores Opioides mu/metabolismo , Receptores Opioides/metabolismo , Lobo Temporal/metabolismo , Adenilil Ciclases/metabolismo , Adulto , Fármacos do Sistema Nervoso Central/farmacologia , Colforsina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos Opioides/metabolismo , Radioisótopos de Enxofre , Trítio , Adulto Jovem , Receptor de Nociceptina , Nociceptina
10.
Neurochem Int ; 52(1-2): 321-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17698254

RESUMO

We examined the occurrence of possible changes in mRNA expression and the functional activity of opioid receptors after acute in vivo and in vitro treatment with the putative endogenous cannabinoid noladin ether. While noladin ether (NE) demonstrates agonist activity at CB1 cannabinoid receptors, recent data indicate that NE acts as a full agonist at CB2 cannabinoid receptors too. Considering the functional interactions between opioids and cannabinoids, it is of interest to examine whether NE affects the opioid system. To that end, we studied the influence of NE on mu-opioid receptor (MOR) mRNA expression and MOR mediated G-protein signaling. We used real-time PCR and [35S]GTPgammaS binding assays to examine the changes of MOR mRNA levels and the capability of the mu-opioid agonist peptide ([D-Ala2,(NMe)Phe4,Gly5-ol]enkephalin (DAMGO) in activating regulatory G-proteins via MORs in forebrain membrane fractions of wild-type (w.t., CB1+/+) and CB1 receptor deficient transgenic mice (knockout, CB1-/-). We found, that the expression of MOR mRNAs significantly decreased both in CB1+/+ and CB1-/- forebrain after a single injection of NE at 1 mg/kg when compared to control. Consequently, MOR-mediated signaling is attenuated after acute in vivo treatment with NE in both CB1+/+ and CB1-/- mice. Inhibition on MOR mediated activation is observed after in vitro NE administration as well. Radioligand binding competition studies showed that the noticed effect of NE on MOR signaling is not mediated through MORs. Both in vivo and in vitro attenuations of NE can be antagonized by the CB2 selective antagonist SR144528. Taken together, our data suggest that the NE caused pronounced decrease in the activity of MOR is mediated via CB2 cannabinoid receptors.


Assuntos
Glicerídeos/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Receptores Opioides mu/antagonistas & inibidores , Animais , Sequência de Bases , Primers do DNA , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Ensaio Radioligante , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
11.
Neurochem Int ; 53(6-8): 309-16, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18804501

RESUMO

Formerly considered as an exclusively peripheral receptor, it is now accepted that CB(2) cannabinoid receptor is also present in limited amounts and distinct locations in the brain of several animal species, including mice. However, the possible roles of CB(2) receptors in the brain need to be clarified. The aim of our work was to study the mu-opioid receptor (MOR) mRNA expression level and functional activity after acute in vivo and in vitro treatments with the endocannabinoid noladin ether (NE) and with the CB(2) receptor antagonist SR144528 in brainstem of mice deficient in either CB(1) or CB(2) receptors. This study is based on our previous observations that noladin ether (NE) produces decrease in the activity of MOR in forebrain and this attenuation can be antagonized by the CB(2) cannabinoid antagonist SR144528, suggesting a CB(2) receptor mediated effect. We used quantitative real-time PCR to examine the changes of MOR mRNA levels, [(35)S]GTPgammaS binding assay to analyze the capability of mu-opioid agonist DAMGO to activate G-proteins and competition binding assays to directly measure the ligand binding to MOR in mice brainstem. After acute NE administration no significant changes were observed on MOR signaling. Nevertheless pretreatment of mice with SR144528 prior to the administration of NE significantly decreased MOR signaling suggesting the involvement of SR144528 in mediating the effect of MOR. mRNA expression of MORs significantly decreased both in CB(1) wild-type and CB(1) knockout mice after a single injection of SR144528 at 0.1mg/kg when compared to the vehicle treated controls. Consequently, MOR-mediated signaling was attenuated after acute in vivo treatment with SR144528 in both CB(1) wild-type and CB(1) knockout mice. In vitro addition of 1microM SR144528 caused a decrease in the maximal stimulation of DAMGO in [(35)S]GTPgammaS binding assays in CB(2) wild-type brainstem membranes whereas no significant changes were observed in CB(2) receptor knockouts. Radioligand binding competition studies showed that the noticed effect of SR144528 on MOR signaling is not mediated through MORs. Our data demonstrate that the SR144528 caused pronounced decrease in the activity of MOR is mediated via CB(2) cannabinoid receptors.


Assuntos
Tronco Encefálico/metabolismo , Canfanos/farmacologia , Moduladores de Receptores de Canabinoides/metabolismo , Dor/metabolismo , Pirazóis/farmacologia , Receptor CB2 de Canabinoide/metabolismo , Receptores Opioides mu/genética , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Moduladores de Receptores de Canabinoides/agonistas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Glicerídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Dor/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ensaio Radioligante , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/antagonistas & inibidores
12.
J Med Chem ; 50(2): 328-33, 2007 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-17228874

RESUMO

The opioid peptide TIPP (H-Tyr-Tic-Phe-Phe-OH, Tic:1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid) was substituted with Dmt (2',6'-dimethyltyrosine) and a new unnatural amino acid, beta-MeCha (beta-methyl-cyclohexylalanine). This double substitution led to a new series of opioid peptides displaying subnanomolar delta antagonist activity and mu agonist or antagonist properties depending on the configuration of the beta-MeCha residue. The most promising analog, H-Dmt-Tic-(2S,3S)-beta-MeCha-Phe-OH was a very selective delta antagonist both in the mouse vas deferens (MVD) assay (Ke = 0.241 +/- 0.05 nM) and in radioligand binding assay (K i delta = 0.48 +/- 0.05 nM, K i mu/K i delta = 2800). The epimeric peptide H-Dmt-Tic-(2S,3R)-beta-MeCha-Phe-OH and the corresponding peptide amide turned out to be mixed partial mu agonist/delta antagonists in the guinea pig ileum and MVD assays. Our results constitute further examples of the influence of Dmt and beta-methyl substitution as well as C-terminal amidation on the potency, selectivity, and signal transduction properties of TIPP related peptides. Some of these compounds represent valuable pharmacological tools for opioid research.


Assuntos
Oligopeptídeos/síntese química , Fenilalanina/análogos & derivados , Receptores Opioides delta/antagonistas & inibidores , Animais , Ligação Competitiva , Encéfalo/metabolismo , Cobaias , Íleo/efeitos dos fármacos , Íleo/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Fenilalanina/química , Ensaio Radioligante , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Estereoisomerismo , Relação Estrutura-Atividade , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/fisiologia
13.
Brain Res Bull ; 71(6): 628-32, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17292806

RESUMO

Prodynorphins (PDYNs) from the African clawed frog (Xenopus laevis), originally described as 'proxendorphins', are novel members of the family of opioid-like precursor polypeptides and were recently discovered based on polymerase chain reaction (PCR) isolates from a Xenopus brain cDNA library. This amphibian prodynorphin was found in two isoforms, (Xen)PDYN-A and (Xen)PDYN-B, consisting of 247 and 279 amino acids, respectively. Each prepropeptide contains five potential opioid-like peptides, collectively named xendorphins. One of these, xendorphin B1 ((Xen)PDYN-B sequence 96-111: YGGFIRKPDKYKFLNA), is a hexadecapeptide that displaced [3H]naloxone and the radiolabelled kappa opioid, [3H]dynorphin A (1-17), with nanomolar affinity from rat brain membranes. Using the acetic acid pain test, the present study examined the antinociceptive effects of spinally administered xendorphin B1 in amphibians. Xendorphin B1 produced a long-lasting and dose-dependent antinociceptive effect in the Northern grass frog (Rana pipiens) with an ED50 value of 44.5 nmol/frog. The antinociceptive effects of xendorphin B1 were significantly blocked by pretreatment with the non-selective opioid antagonist, naltrexone. This is the first report of the in vivo characterization of a non-mammalian prodynorphin-derived peptide and suggests that xendorphin peptides may play a role in the modulation of noxious information in vertebrates.


Assuntos
Anfíbios/metabolismo , Neuropeptídeos/farmacologia , Nociceptores/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Dor/tratamento farmacológico , Hormônios Peptídicos/farmacologia , Precursores de Proteínas/farmacologia , Medula Espinal/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Química Encefálica/genética , Relação Dose-Resposta a Droga , Encefalinas/genética , Encefalinas/isolamento & purificação , Encefalinas/farmacologia , Biblioteca Gênica , Neuropeptídeos/genética , Nociceptores/metabolismo , Peptídeos Opioides/genética , Peptídeos Opioides/isolamento & purificação , Dor/metabolismo , Dor/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Hormônios Peptídicos/genética , Hormônios Peptídicos/isolamento & purificação , Precursores de Proteínas/genética , Precursores de Proteínas/isolamento & purificação , Rana pipiens , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Proteínas de Xenopus/genética , Proteínas de Xenopus/isolamento & purificação , Proteínas de Xenopus/farmacologia , Xenopus laevis
14.
Brain Res Bull ; 74(1-3): 119-29, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17683797

RESUMO

A novel opioid peptide antagonist analogue, [3H]Dmt-Tic-(2S,3R)betaMePhe-Phe, derived from the potent, delta-receptor selective TIPP tetrapeptide (Tyr-Tic-Phe-Phe) series was synthesized and radiolabeled by catalytic tritiation of its iodinated precursor peptide. The purified radioprobe exhibited a specific activity of 2.15 TBq/mmol (58 Ci/mmol). The novelty of this compound is that it contains structurally modified tyrosine residue (2',6'-dimethyltyrosine, Dmt1) replacing tyrosine (Tyr1) at the N-terminus, and beta-methyl substituted phenylalanine (betaMePhe3) at the third position. As the configuration of betaMePhe3 side-chain might be different due to diastereomerism, and accordingly can alter the biological activity, both unlabeled threo (2S,3R and 2R,3S) diastereomeric analogues were also prepared and included in this study. The affinity and selectivity (delta-opioid versus mu-opioid receptor) were evaluated by radioreceptor binding assays. Agonist or antagonist potencies were determined in [35S]GTPgammaS binding experiments using Chinese Hamster Ovary (CHO) cells selectively expressing delta- or mu-opioid receptors. The equilibrium binding of the radiolabeled peptide derivative [3H]Dmt-Tic-(2S,3R)betaMePhe-Phe to rat brain membranes was saturable and the Scatchard analysis indicated a single binding site with a Kd of 0.3 nM and a Bmax of 127 fmol/mg protein. A study of [3H]Dmt-Tic-(2S,3R)betaMePhe-Phe binding displacement by various receptor-type specific opioid ligands showed the rank order of competitor's potency delta > mu > kappa, suggesting selective labeling of opioid delta-sites. In the functional tests, the (2S,3R) and (2R,3S) peptides exhibited partial agonist behaviour by weakly stimulating regulatory G-proteins in CHO cell membranes transfected with different receptors. Both isomers were quite weak partial agonists at the delta-receptor and reasonable partial agonists at the mu-receptor, with a prevalence of (2S,3R) over (2R,3S) for the mu-receptor. Consistent with these observations both stereomers competitively inhibited the stimulation of [35S]GTPgammaS binding induced by the prototype delta-agonist peptide (pClPhe4)-DPDPE in delta(m) CHO cell membranes, and still the (2S,3R) compound exerted more potent delta-antagonist effect. [3H]Dmt-Tic-(2S,3R)betaMePhe-Phe represents a high affinity new radioligand and also constitute further example of the influence of beta-methyl substitution on the potency and selectivity of TIPP analogues, thus becoming a valuable biochemical and pharmacological tool in opioid research.


Assuntos
Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Receptores Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Células CHO , Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/química , Ensaio Radioligante , Ratos , Ratos Wistar , Receptores Opioides/efeitos dos fármacos , Estereoisomerismo , Relação Estrutura-Atividade , Tetra-Hidroisoquinolinas/farmacologia , Trítio , Tirosina/metabolismo
15.
Epilepsy Res ; 77(2-3): 75-84, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913464

RESUMO

The effects with pretreatment with nociceptin (0.03-30nmol, i.c.v.) were evaluated on the threshold for eliciting afterdischarge (ADT), generation and spread of seizure activity and postictal depression in rats with kindling stimulation. Nociceptin produced a decrease in ADT (32-45%) in rats with partial seizures (PS, stage II-III), and an increase (61-92%) in rats with generalized seizures (GS, kindled state). Nociceptin did not modify the behavioral changes, spike frequency and duration of afterdischarge elicited at ADT in both experimental groups. In rats with GS, nociceptin enhanced postictal depression (34-44%) evaluated with a recycling paradigm. Autoradiography experiments revealed enhanced nociceptin opioid receptor (NOP) binding in medial amygdala (22-26%), frontal (21-23%) and entorhinal (27-32%) cortices, and reduced binding in the substantia nigra pars compacta (28%) and medial central gray (29%) of rats with PS. The GS group displayed significant decreased NOP binding (40-70%) in most of the brain areas evaluated. These results suggest that nociceptin facilitates ictal activity in rats with PS, whereas in animals with GS, it induces inhibitory effects on ADT and enhances the postictal period. These effects correlate with significant changes in NOP binding.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiopatologia , Excitação Neurológica/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Peptídeos Opioides/farmacologia , Convulsões/fisiopatologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Estimulação Elétrica , Eletrodos Implantados , Epilepsias Parciais/fisiopatologia , Epilepsia Generalizada/fisiopatologia , Injeções Intraventriculares , Masculino , Ratos , Ratos Wistar , Nociceptina
16.
Seizure ; 16(7): 645-52, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17560811

RESUMO

Opioid receptor binding was evaluated in parahippocampal cortex (PHC) obtained from patients with intractable mesial temporal lobe epilepsy (MTLE) with and without subacute high frequency electrical stimulation (HFS) in this brain area. Mu, delta and nociceptin receptor binding was determined by autoradiography in PHC of five patients (ESAE group) with MTLE history of 14.8 +/- 2.5 years and seizure frequency of 11 +/- 2.9 per month, two of them (40%) with mesial sclerosis. This group demonstrated antiepileptic effects following subacute HFS (130 Hz, 450 micros, 200-400 microA), applied continuously during 16-20 days in PHC. Values were compared with those obtained from patients with severe MTLE (history of 21.7 +/- 2.8 years and seizure frequency of 28.2 +/- 14 per month) in whom electrical stimulation did not induce antiepileptic effects (ESWAE group, n = 4), patients with MTLE in whom no electrical stimulation was applied (MTLE group, n = 4) and autopsy material acquired from subjects without epilepsy (n = 4 obtained from three subjects). Enhanced 3H-DAMGO (MTLE, 755%; ESAE, 375%; ESWAE, 693%), 3H-DPDPE (MTLE, 242%; ESAE, 80%; ESWAE, 346%) and 3H-nociceptin (MTLE, 424%; ESAE, 217%; ESWAE, 451%) binding was detected in the PHC of all epileptic groups. However, tissue obtained from ESAE group demonstrated lower opioid receptor binding (3H-DAMGO, 44.5%, p < 0.05; 3H-DPDPE, 47%, p < 0.05; 3H-nociceptin, 39.3%, p < 0.5) when compared with MTLE group. The present results indicate that a high effectiveness to the antiepileptic effects induced by HFS is associated with reduced opioid peptide binding.


Assuntos
Terapia por Estimulação Elétrica , Epilepsia do Lobo Temporal/metabolismo , Giro Para-Hipocampal/metabolismo , Receptores Opioides/metabolismo , Convulsões/prevenção & controle , Adulto , Anticonvulsivantes/uso terapêutico , Autorradiografia , Eletrofisiologia , Epilepsia do Lobo Temporal/patologia , Feminino , Humanos , Ligantes , Imageamento por Ressonância Magnética , Masculino , Giro Para-Hipocampal/patologia , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Receptor de Nociceptina
17.
Acta Biol Hung ; 58 Suppl: 113-29, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18297798

RESUMO

Numerous studies have shown functional links between the cannabinoid and opioid systems. The goal of this study was to evaluate whether acute treatments by endogenous cannabinoid agonist, selective CB1 or CB2 receptor antagonists modulate the expression of mu- (MOR) and delta- (DOR) opioid receptor mRNA levels and functional activity in the cerebellum of transgenic mice deficient in the CB1 type of cannabis receptors. We examined the effect of noladin ether (endogenous cannabinoid agonist) pretreatment on MOR and DOR mRNA expression by using reverse transcription and real-time polimerase chain reaction (PCR) and the ability of subsequent application of the opioid agonists to activate G-proteins, as measured by [35S]GTPgammaS binding, in wild-type (CB1+/+) and CB1 cannabinoid receptor deficient (CB1-/-, 'knockout', K.O.) mice. The acute administration of noladin ether markedly reduced MOR-mediated G-protein activation and caused a significant increase in the level of MOR mRNAs in the cerebella of wildtype, but not in the CB1-/- mice. No significant differences were observed in DOR functional activity and mRNA expression in wild-type animals. In CB1-/- mice the expression of DOR mRNA increased after noladin ether treatment, but no changes were found in DOR functional activity. In addition, Rimonabant (selective central cannabinoid CB1 receptor antagonist) and SR144528 (selective peripheral cannabinoid CB2 receptor antagonist) caused significant potentiation in MOR functional activity in the wild-type animals, whereas DOR mediated G-protein activation was increased in the CB1-/- mice. In contrast, Rimonabant and SR144528 decreased the MOR and DOR mRNA expressions in both CB1+/+ and CB1-/- mice. Taken together, these results indicate that acute treatment with cannabinoids causes alterations in MOR and DOR mRNA expression and functional activity in the cerebella of wild-type and CB1 knockout mice indicating indirect interactions between these two signaling systems.


Assuntos
Canabinoides/farmacologia , Cerebelo/efeitos dos fármacos , Receptor CB1 de Canabinoide/fisiologia , Receptores Opioides/genética , Receptores Opioides/fisiologia , Animais , Sequência de Bases , Cerebelo/metabolismo , Primers do DNA , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Receptor CB1 de Canabinoide/genética
18.
Neurosci Lett ; 643: 16-21, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28192197

RESUMO

Oxymorphone, one of oxycodone's metabolic products, is a potent opioid receptor agonist which is thought to contribute to the analgesic effect of its parent compound and may have high potential abuse liability. Nonetheless, the in vivo pharmacological binding profile of this drug is still unclear. This study uses mice lacking mu (MOP), kappa (KOP) or delta (DOP) opioid receptors as well as mice lacking all three opioid receptors to provide full characterisation of oxymorphone binding sites in the brain. Saturation binding studies using [3H]oxymorphone revealed high affinity binding sites in mouse brain displaying Kd of 1.7nM and Bmax of 147fmol/mg. Furthermore, we performed quantitative autoradiography binding studies using [3H]oxymorphone in mouse brain. The distribution of [3H]oxymorphone binding sites was found to be similar to the selective MOP agonist [3H]DAMGO in the mouse brain. [3H]Oxymorphone binding was completely abolished across the majority of the brain regions in mice lacking MOP as well as in mice lacking all three opioid receptors. DOP and KOP knockout mice retained [3H]oxymorphone binding sites suggesting oxymorphone may not target DOP or KOP. These results confirm that the MOP, and not the DOP or the KOP is the main high affinity binding target for oxymorphone.


Assuntos
Encéfalo/metabolismo , Oximorfona/farmacologia , Receptores Opioides/metabolismo , Animais , Autorradiografia/métodos , Sítios de Ligação , Camundongos Knockout , Receptores Opioides/deficiência , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
19.
J Neurol Sci ; 376: 63-70, 2017 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-28431630

RESUMO

Previously, we have shown that the N-methyl d-aspartate (NMDA)-receptor antagonist kynurenic acid (KYNA) and its analogue KYNA1 do not bind directly to mu, kappa and delta opioid receptors in vitro. On the other hand, chronic administration of KYNA and KYNA1 resulted in region (cortex vs striatum) and opioid receptor-type specific alterations in G-protein activation of mouse brain homogenates. Here we describe for the first time the acute effect of KYNA and KYNA1 on opioid receptor function with the possible involvement of the NMDA receptor. The acute 30minute in vivo KYNA1 and KYNA treatments altered opioid receptor G-protein signaling or ligand potency depending on the opioid receptor type and brain region (rat cortex vs striatum) using [35S]GTPγS binding assays. Pretreatment with the NMDA receptor antagonist MK-801 impaired or reversed the effects of KYNA1 and KYNA. These results suggest an NMDA receptor mediated effect. After acute 30minute treatment HPLC measurements revealed a similar KYNA1 and a higher KYNA plasma concentration compared to cerebrospinal fluid concentrations. Finally, KYNA, KYNA1 and MK-801 showed comparable results in opioid receptor G-protein activity and ligand potency with acute in vivo treatments when they were administered in vitro for 30min on isolated cortex and striatum slices. We previously demonstrated that KYNA1 and KYNA acutely altered opioid receptor function in vivo and in vitro through the NMDA receptor depending on the opioid receptor type and brain region. This study may lead to a new, indirect approach to influence opioid receptor signaling.


Assuntos
Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Ácido Cinurênico/análogos & derivados , Ácido Cinurênico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Córtex Cerebral/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Corpo Estriado/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ácido Cinurênico/farmacologia , Masculino , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Técnicas de Cultura de Tecidos
20.
Eur J Pharmacol ; 539(1-2): 39-48, 2006 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-16682024

RESUMO

It was recently reported that the hexapeptide Ac-RYYRIK-ol binds with high affinity nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptors and competitively antagonizes N/OFQ actions in the mouse vas deferens assay. Here we further describe the in vitro and in vivo pharmacological features of this NOP receptor ligand. In mouse brain homogenate the degradation half life of Ac-RYYRIK-ol (2.48 min) was significantly higher than that of the parent compound Ac-RYYRIK-NH2 (1.20 min). In the electrically stimulated mouse vas deferens, Ac-RYYRIK-ol (10-1000 nM) competitively antagonized the inhibitory effect of N/OFQ (pA2=8.46), while in the isolated mouse colon the hexapeptide mimicked N/OFQ contractile effects thus behaving as a NOP receptor agonist (pEC50=9.09). This latter effect was no longer evident in colon tissues taken from mice knock out for the NOP receptor gene (NOP-/-). In vivo in mice, similarly to N/OFQ, Ac-RYYRIK-ol (dose range 0.001-1 nmol) produced: i) pronociceptive effects after intracerebroventricular (i.c.v.) administration and antinociceptive actions when given intrathecally (i.t.) in the tail withdrawal assay; ii) inhibition of locomotor activity and iii) stimulation of food intake after supraspinal administration. Finally, in the forced swimming test, Ac-RYYRIK-ol was inactive per se, but reversed the antidepressant-like effects elicited by the NOP receptor selective antagonist UFP-101 ([Nphe(1),Arg(14),Lys(15)]N/OFQ-NH2). Thus, in all these in vivo assays Ac-RYYRIK-ol mimicked the actions of N/OFQ showing however higher potency. In conclusion, Ac-RYYRIK-ol displayed a complex pharmacological profile which is likely due to the low efficacy agonist nature of this novel ligand of the NOP receptor. The high potency, selectivity of action, and in vivo effectiveness make Ac-RYYRIK-ol a useful pharmacological tool for future studies in the field of N/OFQ and its NOP receptor.


Assuntos
Oligopeptídeos/farmacologia , Receptores Opioides/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Colo/efeitos dos fármacos , Colo/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Estimulação Elétrica , Técnicas In Vitro , Ligantes , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Oligopeptídeos/farmacocinética , Receptores Opioides/fisiologia , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/fisiologia , Receptor de Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA